Skip to main content

REVIEW article

Front. Cardiovasc. Med., 14 January 2025
Sec. Cardiovascular Metabolism
This article is part of the Research Topic Inflammatory Pathways in Cardiometabolic Diseases: Mechanisms, Biomarkers, and Therapeutic Insights View all 3 articles

Mechanisms of inflammatory microenvironment formation in cardiometabolic diseases: molecular and cellular perspectives

\r\nMenghua Liu,&#x;Menghua Liu1,†Rumeng Chen,&#x;Rumeng Chen1,†Zhiwei Zheng,&#x;Zhiwei Zheng1,†Shuling Xu,&#x;Shuling Xu1,†Chunyan Hou,&#x;Chunyan Hou1,†Yining DingYining Ding1Mengling ZhangMengling Zhang2Meihua Bao
Meihua Bao3*Binsheng He
Binsheng He3*Sen Li
\r\nSen Li1*
  • 1School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
  • 2School of Stomatology, Changsha Medical University, Changsha, China
  • 3Hunan key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China

Cardiometabolic diseases (CMD) are leading causes of death and disability worldwide, with complex pathophysiological mechanisms in which inflammation plays a crucial role. This review aims to elucidate the molecular and cellular mechanisms within the inflammatory microenvironment of atherosclerosis, hypertension and diabetic cardiomyopathy. In atherosclerosis, oxidized low-density lipoprotein (ox-LDL) and pro-inflammatory cytokines such as Interleukin-6 (IL-6) and Tumor Necrosis Factor-alpha (TNF-α) activate immune cells contributing to foam cell formation and arterial wall thickening. Hypertension involves the activation of the renin-angiotensin system (RAS) alongside oxidative stress-induced endothelial dysfunction and local inflammation mediated by T cells. In diabetic cardiomyopathy, a high-glucose environment leads to the accumulation of advanced glycation end products (AGEs), activating the Receptor for Advanced Glycation Endproducts (RAGE) and triggering inflammatory responses that further damage cardiac and microvascular function. In summary, the inflammatory mechanisms in different types of metabolic cardiovascular diseases are complex and diverse; understanding these mechanisms deeply will aid in developing more effective individualized treatment strategies.

Introduction

Cardiometabolic diseases (CMD) represent a complex cluster of disorders, primarily encompassing atherosclerosis, coronary artery disease, and hypertension, all of which are intricately linked to metabolic dysfunction (13). These diseases are frequently associated with metabolic syndrome (MetS), a condition characterized by a constellation of symptoms including central obesity, insulin resistance, hyperglycemia, dyslipidemia, and hypertension (47). As societies continue to urbanize and modernize, the adverse lifestyle practices have become increasingly prevalent, leading to a rising incidence of cardiometabolic diseases globally (811). According to a large number of relevant literature findings, CMD have become one of the leading causes of mortality worldwide, exerting a considerable burden on individuals, families, and public health systems (12, 13). Given that these diseases often progress insidiously, with many patients remaining asymptomatic until severe cardiovascular events such as myocardial infarction, heart failure, or stroke occur, there is an urgent need to unravel the underlying mechanisms of CMD to develop effective preventive and therapeutic strategies that can improve patient outcomes and quality of life (14, 15).

The inflammatory microenvironment plays a critical role in the onset and progression of CMD (1618). A growing body of research indicates that chronic low-grade inflammation in the cardiovascular system is a pivotal driver of the onset and progression of these diseases (19, 20). Inflammation impacts cardiovascular health through its regulation of processes such as lipid uptake, glucose metabolism, endothelial function, and vascular remodeling (2124). During the development of atherosclerosis, inflammatory cells such as macrophages, T cells, and mast cells, along with various inflammatory mediators they secrete—including cytokines and chemokines—contribute to the formation and progression of arterial plaques (25, 26). These cells and molecules form a complex inflammatory network that affects multiple aspects of the cardiovascular system, not only causing direct damage to blood vessel walls but also modulating metabolic functions and activating immune responses (17, 26). Despite substantial evidence supporting the critical role of inflammation in CMD, the precise mechanisms by which it is initiated and sustained, and how it interacts with metabolic processes to accelerate disease progression, remain inadequately understood and warrant further investigation.

To address these challenges, this review aims to systematically explore the molecular mechanisms and cellular foundations underpinning the development and maintenance of the inflammatory microenvironment in cardiometabolic diseases. By synthesizing current research advancements, the article seeks to elucidate the specific roles of various inflammatory mediators in CMD, and how their interplay with metabolic abnormalities influences disease progression and outcomes. As our understanding of these pathological mechanisms deepens, we hope to lay a robust scientific foundation for future therapeutic innovations designed to improve the prognosis of patients with cardiometabolic diseases and alleviate the global health burden they pose. By enhancing our understanding of the pathological mechanisms at play, we aim to pave the way for the development of innovative interventions that improve patient outcomes and reduce the global burden of cardiometabolic diseases.

Metabolic factors are involved in the occurrence and development of cardiovascular diseases

Cardiovascular disease (CVD) is the leading cause of death worldwide and is characterized by atherosclerosis, endothelial dysfunction, inflammation, and oxidative stress (2730). Metabolic factors play a crucial role in the onset and progression of CVD, primarily including hyperlipidemia, hyperglycemia, and insulin resistance (31). Mendelian randomization (MR) is an analytical method that employs genetic variants as instrumental variables and can be widely used to study the causal associations between exposures and outcomes (3235). In a MR study, MetS exhibits significant causal relationships with various CVD (36). In clinical trials, patients with MetS have a significantly higher risk of developing CVD compared to those without MetS (37). In patients with hypopituitarism, the high prevalence of MetS is primarily linked to abdominal fat deposition, dyslipidemia, and insulin resistance, with growth hormone replacement therapy showing benefits for lipid profiles and body fat but potentially transiently worsening glucose tolerance (38).

Recent molecular and cellular mechanisms linking inflammation and metabolism

The interplay between inflammation and metabolism is crucial for physiological balance and disease pathogenesis, including type 2 diabetes (T2D), cardiovascular diseases, obesity, and autoimmune disorders (39). Silent Information Regulator 2 Ortholog 1 (SIRT1) regulates inflammation through various pathways, including affecting metabolic pathways, inflammatory cells and mediators, as well as key signaling pathways, thus playing a significant role in metabolic and immune diseases and serving as a potential therapeutic target (40). Research indicates that small dense low-density lipoprotein-cholesterol (sdLDL-C) and remnant-like particle cholesterol (RLP-C) induce inflammation by activating immune cells, while hyperlipidemia exacerbates inflammatory responses, leading to dysregulated lipid metabolism and alterations in lipoprotein profiles (41). These insights provide new directions for developing targeted therapies for chronic inflammatory and metabolic disorders, potentially leading to more precise and effective treatments.

By targeting these key biological processes, it is possible to develop more precise and effective interventions to improve patient health in the future. Therefore, an in-depth understanding of the mechanisms underlying the formation of the inflammatory microenvironment is essential for disease prevention and treatment. This review will focus on the formation mechanism of the inflammatory microenvironment of atherosclerosis, hypertension and diabetic cardiomyopathy from the perspective of molecular and cellular levels, focusing on the specific factors affecting the inflammatory response.

Inflammatory mechanisms in atherosclerosis

Initial events and the role of oxidized Low-density lipoprotein

Atherosclerosis is a chronic inflammatory disease characterized by the formation of lipid deposits, immune cell infiltration, and fibrosis in the arterial wall, eventually leading to atherosclerotic plaque formation (42, 43). One of the initial events in atherosclerosis is the accumulation of LDL in the subendothelial space of arteries (44, 45). In these regions, LDL becomes susceptible to oxidation by ROS, forming ox-LDL (46). ox-LDL not only exhibits pro-inflammatory properties but also activates various immune responses (46). ox-LDL binds to scavenger receptors on endothelial cell (EC), macrophages, and smooth muscle cells, triggering intracellular signaling pathways that initiate inflammatory responses (47). For instance, ox-LDL activates the Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway, promoting the expression of various pro-inflammatory cytokines and chemokines like IL-6 and TNF-α (47, 48). Ox-LDL has different effects on vascular smooth muscle cells (VSMCs) and ECs (49, 50). In ECs, ox-LDL activates the NF-κB pathway through Toll-like receptors (TLRs) and other receptors, increasing the expression of pro-inflammatory cytokines such as IL-6, Interleukin-8 (IL-8), and Monocyte Chemoattractant Protein-1 (MCP-1), which attract monocytes to the vessel wall and promote plaque formation (51). In VSMCs, ox-LDL also activates the NF-κB pathway but primarily promotes cell proliferation and migration, influencing plaque stability or instability (52). Additionally, the phenotypic switch of VSMCs from a contractile to a synthetic phenotype is associated with NF-κB activation (52).

Inflammation amplification and immune cell recruitment

The presence of ox-LDL and lipid accumulation in the arterial wall further induces the overproduction of pro-inflammatory cytokines (such as IL-6 and TNF-α) and chemokines, such as Chemokine (C-C motif) ligand 2 (CCL2) and Chemokine (C-X-C motif) ligand 10 (CXCL10) (53, 54). These molecules bind to their respective receptors on EC and immune cells, further amplifying the inflammatory response. IL-6 and TNF-α activate the Janus Kinase—Signal Transducer and Activator of Transcription (JAK-STAT) and NF-κB signaling pathways, promoting inflammatory responses and stimulating the proliferation and migration of vascular wall cells (55, 56). Chemokines (like CCL2) recruit monocytes to the arterial wall, enhancing the local immune response (57). CCL2 binds to the Chemokine (C-C motif) receptor 2 (CCR2) on monocytes, facilitating their adhesion to the endothelium and subsequent infiltration into the vascular wall (57). As atherosclerosis progresses, the accumulation of cholesterol and its metabolites triggers additional inflammatory responses by activating the NLR family, pyrin domain containing 3 (NLRP3) inflammasome, a key intracellular inflammatory signaling complex (58). Activation of the NLRP3 inflammasome leads to Cysteine-aspartic protease 1 (caspase-1) activation, which promotes the conversion of Pro-Interleukin-1β (pro-IL-1β) and Pro-Interleukin-18 (pro-IL-18) into their mature forms, Interleukin-1β (IL-1β) and Interleukin-18 (IL-18) (59, 60). These cytokines further amplify local inflammation and induce apoptosis and necrosis of vascular wall cells (60). Activation of the inflammasome is also associated with plaque instability, potentially leading to plaque rupture and thrombosis, which can trigger acute cardiovascular events such as myocardial infarction and stroke (61).

Endothelial cell activation and foam cell formation

In the early stages of atherosclerosis, EC become activated due to various risk factors such as ox-LDL, hyperglycemia, and hypertension (62). Activated EC express various adhesion molecules, including intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and Endothelial Selectin (E-selectin) (63). The upregulation of ICAM-1 and VCAM-1 is a hallmark of endothelial cell activation (64). These molecules bind to integrins on monocytes, promoting firm adhesion of monocytes to the endothelium and their subsequent infiltration into the subendothelial space (65, 66). Endothelial cell activation also involves the release of pro-inflammatory cytokines like IL-1β and TNF-α, which further enhance endothelial adhesion and permeability, promoting monocyte migration (67). Monocytes that adhere to activated EC, guided by chemokines, cross the endothelial layer and infiltrate the intima (68). Once in the intima, monocytes differentiate into macrophages, which then phagocytose large amounts of ox-LDL through scavenger receptors (47). These macrophages, laden with excessive lipid, transform into foam cells (69). The formation of foam cells is a hallmark of atherosclerotic plaques and involves the release of various inflammatory mediators, further promoting the development of local inflammation (69, 70). The apoptosis and death of foam cells contribute to plaque instability by releasing large amounts of lipids and cellular debris, increasing the risk of plaque rupture and thrombosis (71).

Smooth muscle cell phenotypic switching and changes in plaque stability

In the progression of atherosclerosis, smooth muscle cells undergo proliferation and a shift towards a synthetic phenotype under the influence of inflammatory mediators (72). This process involves a phenotypic change that enables smooth muscle cells to produce extracellular matrix components such as collagen, forming fibrous caps and thickening the arterial intima (73). Smooth muscle cell proliferation and synthetic phenotype transformation play a critical role in stabilizing plaques (74). However, as the disease progresses, the apoptosis and necrosis of smooth muscle cells can lead to the rupture of fibrous caps, increasing plaque instability and the risk of cardiovascular events (75). In summary, the inflammatory mechanisms of atherosclerosis involve complex interactions among multiple cells and molecules. In Figure 1, the molecules and cells involved in the inflammatory microenvironment of atherosclerosis can be seen. The oxidation of LDL, the release of pro-inflammatory cytokines, the role of cholesterol metabolites, and the dynamic changes in EC, monocytes, and smooth muscle cells all contribute significantly to the progression of this disease.

Figure 1
www.frontiersin.org

Figure 1. Diagram of inflammatory mechanisms in cardiometabolic diseases. LDL, low-density lipoprotein; ox-LDL, oxidized low-density lipoprotein; TNF-α, Tumor Necrosis Factor-alpha; IL-6, Interleukin-6; ICAM-1, intercellular adhesion molecule-1; VCAM-1, vascular cell adhesion molecule-1; NF-κB, Nuclear Factor kappa-light-chain-enhancer of activated B cells; NLRP3, NLR family, pyrin domain containing 3; pro-IL-1β, Pro-Interleukin-1β; pro-IL-18, Pro-Interleukin-18; IL-1β, Interleukin-1β; IL-18, Interleukin-18; ROS, reactive oxygen species; ACE, Angiotensin-Converting Enzyme; AGEs, advanced glycation end products; RAGE, Receptor for Advanced Glycation Endproducts. This figure was created with BioRender.com.

Inflammatory mechanisms in hypertension

Role of RAS in hypertension-related inflammation

Hypertension is a complex chronic disease with pathophysiological mechanisms involving multiple aspects, with inflammatory mechanisms receiving increasing attention in recent years (76, 77). Inflammation not only serves as a consequence of hypertension but also plays a crucial role in its progression and maintenance. The Renin-Angiotensin System (RAS) plays a pivotal role in the pathogenesis and progression of hypertension, with Angiotensin II (Ang II) being the key effector molecule (78). Ang II, by binding to the Angiotensin II type 1 receptor (AT1R), activates multiple signaling pathways, leading to vasoconstriction, sodium and water retention, and increased blood pressure (79). These inflammatory mediators further attract and activate immune cells, leading to vascular wall inflammation. ROS, including superoxide, hydrogen peroxide, and hydroxyl radicals, play significant roles in the inflammatory mechanisms of hypertension (80). Ang II increases ROS production via the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase system, which can directly damage vascular EC and activate inflammatory signaling pathways (81). In hypertensive patients, the activity of antioxidant enzymes such as superoxide dismutase (SOD) and glutathione peroxidase (GPx) often decreases, leading to weakened ROS clearance and further exacerbating oxidative stress and inflammation (82).

Oxidative stress and endothelial damage

Endothelial dysfunction is a critical aspect of inflammatory mechanisms in hypertension (83). Upon exposure to Ang II, ROS, and other stimuli, EC release molecules like endothelin-1 (ET-1) and VCAM-1, which promote vasoconstriction and increase leukocyte adhesion and infiltration, leading to vascular wall inflammation (84, 85). Under normal conditions, EC produce nitric oxide (NO) through endothelial nitric oxide synthase (eNOS), which has vasodilatory and anti-inflammatory effects (86, 87). However, in hypertension, eNOS activity declines, reducing NO production, resulting in vasoconstriction and heightened inflammation (88). The immune system plays a crucial role in the inflammatory mechanisms of hypertension, with lymphocytes, particularly T cells, being significantly involved (89). In hypertensive patients and animal models, there is significant infiltration of T cells around blood vessels and in renal tissues (90). These T cells release cytokines such as interferon-γ (IFN-γ) and interleukin-17 (IL-17), further promoting local inflammatory responses (91). T Helper 1 (Th1) cells primarily secrete IFN-γ, which activates macrophages and promotes inflammation; T Helper 17 (Th17) cells mainly secrete IL-17, which is involved in neutrophil recruitment and activation (91, 92). These cytokines work together to cause chronic inflammation in blood vessels and kidneys, further maintaining and exacerbating hypertension.

The inflammatory mechanisms in hypertension are a complex process involving multiple interactions at both molecular and cellular levels. Figure 1 shows the molecules and cells involved in the inflammatory microenvironment of hypertension. At the molecular level, activation of the RAS, particularly the pro-inflammatory effects of Ang II, and exacerbation of oxidative stress, are crucial drivers of inflammation. At the cellular level, endothelial dysfunction and the involvement of lymphocytes, particularly T cells, further promote local and systemic inflammatory responses. A deeper understanding of these mechanisms not only elucidates the pathophysiological basis of hypertension but also provides a theoretical foundation for developing new anti-inflammatory therapeutic strategies.

Inflammatory mechanisms in diabetic heart disease

Advanced glycation end products and inflammation in diabetic cardiomyopathy

Diabetes is classified into three primary types: type 1, type 2, and gestational diabetes (9396). Diabetes is a long-term condition associated with various complications (97105). Among these, diabetic cardiomyopathy (DCM) is a common and severe complication, characterized by myocardial dysfunction, cardiac remodeling, and heart failure (106). Inflammation plays a crucial role in the initiation and progression of DCM (107). In a hyperglycemic state, excess glucose can undergo non-enzymatic reactions with proteins, lipids, or nucleic acids to form advanced glycation end products (AGEs) (108). The formation of AGEs is a significant trigger of chronic inflammation in diabetic patients (109). In high glucose conditions, glucose reacts with proteins and lipids through the Maillard reaction to form Schiff bases, which then undergo Amadori rearrangement to produce stable AGEs (110). AGEs bind to their specific receptor, the receptor for advanced glycation end products (RAGE), and activate various pro-inflammatory signaling pathways (111). RAGE, a member of the immunoglobulin superfamily, is widely expressed in cardiomyocytes, EC, and macrophages (112). The binding of AGEs to RAGE can initiate multiple pro-inflammatory signaling cascades (113). Upon AGE-RAGE binding, the IκB kinase (IKK) complex is activated, leading to the phosphorylation and degradation of the inhibitory protein Inhibitor of κB alpha (IκBα) (114). This releases NF-κB, enabling its translocation to the nucleus and initiating the transcription of pro-inflammatory genes such as TNF-α and IL-6 (114). The AGE-RAGE complex can also activate the mitogen-activated protein kinase (MAPK) pathway (115). These kinases promote the expression of inflammatory factors and contribute to apoptosis (115).

Insulin resistance and oxidative stress in diabetic cardiomyopathy

Insulin resistance, a hallmark of T2D, is closely associated with inflammatory responses (116). Insulin resistance induces oxidative stress, leading to increased production of ROS, which in turn activates the expression of pro-inflammatory factors (117). In insulin-resistant states, the activity of NADPH oxidase and the mitochondrial electron transport chain increases, leading to elevated ROS production (118). ROS can activate multiple signaling pathways, including NF-κB and MAPK, promoting the expression of pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6, which further exacerbate myocardial inflammation and damage (119). Myocardial cells are severely damaged under the dual impact of high glucose toxicity and inflammatory factors (120). A high glucose environment directly causes metabolic disturbances in cardiomyocytes, including imbalances in glycolysis and fatty acid oxidation, leading to energy metabolism disorders and cell damage (121). Pro-inflammatory cytokines such as TNF-α and IL-6 activate apoptotic signaling pathways, including the Caspase family and B-cell Lymphoma 2 (Bcl-2) family, leading to myocardial cell apoptosis (122). High glucose and AGEs can activate EC through the NF-κB pathway (123). The cardiac microvascular system in diabetic patients is severely affected by inflammation, leading to coronary microcirculatory disorders, which further exacerbate myocardial ischemia and injury (124). As depicted in Figure 1, in diabetic cardiomyopathy, a high-glucose environment induces the formation of AGEs, which activate pro-inflammatory pathways through the RAGE. Additionally, insulin resistance increases pro-inflammatory factors related to oxidative stress. Myocardial cells are damaged, and microvascular inflammation leads to coronary microcirculatory dysfunction, exacerbating heart injury.

The role of gut microbiota in obesity-related cardiovascular metabolic disorders

Obesity-related CMD are closely associated with chronic low-grade inflammation (125). Key mechanisms include inflammation in adipose tissue, insulin resistance, and cardiac inflammation (126). Recent research has highlighted the role of gut microbiota in the inflammatory microenvironment of CMD (127). Impaired gut barrier function, reduced production of short-chain fatty acids (SCFAs), and increased generation of microbial metabolites like trimethylamine N-oxide (TMAO) are all linked to the development and progression of CMD (128, 129). Future research needs to further explore individual variations in gut microbiota, interactions with the host immune system, and potential interventions based on gut microbiota.

Systems biology integration of multi-omics data to elucidate mechanisms of CMD inflammatory microenvironment

From a systems biology perspective, integrating multi-omics data can provide a comprehensive theoretical framework and data support for elucidating the mechanisms underlying the formation of the inflammatory microenvironment in CMD. Firstly, genomics analysis can reveal genetic variations associated with CMD inflammation and identify potential risk genes (130). Secondly, proteomics data can elucidate the functions and interaction networks of key proteins in the inflammatory microenvironment, revealing the roles of proteins in signal transduction and functional regulation (131). Simultaneously, metabolomics research can capture the dynamic changes in metabolites during the inflammatory process, revealing the critical roles of metabolic pathways in inflammation regulation (132). This integrated analysis not only helps to uncover the complex regulatory network of the CMD inflammatory microenvironment but also provides a theoretical foundation and experimental basis for developing precision intervention strategies based on specific targets, thereby promoting the advancement of CMD-related research to a deeper level.

Conclusion

This review has explored the complex inflammatory mechanisms in CMD diseases, including atherosclerosis, hypertension, and DCM. These conditions involve intricate interactions among various inflammatory factors, cell types, and signaling pathways. In atherosclerosis, ox-LDL and cytokines like IL-6 and TNF-α drive foam cell formation and arterial thickening. Hypertension involves RAS activation, oxidative stress, and T cell-mediated inflammation. DCM results from AGEs accumulation, RAGE activation, and cardiac inflammation induced by high glucose levels. By understanding these mechanisms, we can develop targeted treatments that manage and potentially reverse disease progression. Personalized strategies based on specific inflammatory pathways are crucial for improving patient outcomes, and precision medicine offers promising avenues to enhance the quality of life and prognosis for individuals with these conditions.

Author contributions

ML: Writing – original draft. RC: Writing – original draft. ZZ: Writing – original draft. SX: Writing – original draft. CH: Writing – original draft. YD: Writing – original draft. MZ: Writing – original draft. MB: Writing – review & editing. BH: Writing – review & editing. SL: Writing – review & editing.

Funding

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This study was supported by the National Natural Science Foundation of China (Grant No. 81973698 and 81703942), the BUCM Precision Cultivation Program (Grant No. JZPY-202205), the BUCM Research Development Fund (Grant No. 2024-JYB-900203-009), and the Key Research Projects of Hunan Provincial Department of Education (Grant No. 23A0662).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Silveira Rossi JL, Barbalho SM, Reverete de Araujo R, Bechara MD, Sloan KP, Sloan LA. Metabolic syndrome and cardiovascular diseases: going beyond traditional risk factors. Diabetes Metab Res Rev. (2022) 38(3):e3502. doi: 10.1002/dmrr.3502

PubMed Abstract | Crossref Full Text | Google Scholar

2. Qiao Q, Gao W, Zhang L, Nyamdorj R, Tuomilehto J. Metabolic syndrome and cardiovascular disease. Ann Clin Biochem. (2007) 44(3):232–63. doi: 10.1258/000456307780480963

PubMed Abstract | Crossref Full Text | Google Scholar

3. Bonora E. The metabolic syndrome and cardiovascular disease. Ann Med. (2006) 38(1):64–80. doi: 10.1080/07853890500401234

PubMed Abstract | Crossref Full Text | Google Scholar

4. Han TS, Lean ME. Metabolic syndrome. Medicine. (2015) 43(2):80–7. doi: 10.1016/j.mpmed.2014.11.006

Crossref Full Text | Google Scholar

5. Cornier M-A, Dabelea D, Hernandez TL, Lindstrom RC, Steig AJ, Stob NR, et al. The metabolic syndrome. Endocr Rev. (2008) 29(7):777–822. doi: 10.1210/er.2008-0024

PubMed Abstract | Crossref Full Text | Google Scholar

6. Huang PL. A comprehensive definition for metabolic syndrome. Dis Model Mech. (2009) 2(5-6):231–7. doi: 10.1242/dmm.001180

PubMed Abstract | Crossref Full Text | Google Scholar

7. Li JM, Li X, Chan LWC, Hu R, Zheng T, Li H, et al. Lipotoxicity-polarised macrophage-derived exosomes regulate mitochondrial fitness through Miro1-mediated mitophagy inhibition and contribute to type 2 diabetes development in mice. Diabetologia. (2023) 66(12):2368–86. doi: 10.1007/s00125-023-05992-7

PubMed Abstract | Crossref Full Text | Google Scholar

8. O'Neill S, O'Driscoll L. Metabolic syndrome: a closer look at the growing epidemic and its associated pathologies. Obes Rev. (2015) 16(1):1–12. doi: 10.1111/obr.12229

PubMed Abstract | Crossref Full Text | Google Scholar

9. Reddy KS, Yusuf S. Emerging epidemic of cardiovascular disease in developing countries. Circulation. (1998) 97(6):596–601. doi: 10.1161/01.CIR.97.6.596

PubMed Abstract | Crossref Full Text | Google Scholar

10. Wang H, Liu J, Feng Y, Ma A, Wang T. The burden of cardiovascular diseases attributable to metabolic risk factors and its change from 1990 to 2019: a systematic analysis and prediction. Front Epidemiol. (2023) 3:1048515. doi: 10.3389/fepid.2023.1048515

PubMed Abstract | Crossref Full Text | Google Scholar

11. Wang K, Ma J, Li Y, Han Q, Yin Z, Zhou M, et al. Effects of essential oil extracted from Artemisia argyi leaf on lipid metabolism and gut microbiota in high-fat diet-fed mice. Front Nutr. (2022) 9:1024722. doi: 10.3389/fnut.2022.1024722

PubMed Abstract | Crossref Full Text | Google Scholar

12. Ferreira S, Chiavegatto Filho A, Lebrão M, Duarte Y, Laurenti R. Cardiometabolic diseases. Rev Bras Epidemiol. (2019) 21(Suppl 02):e180008.

Google Scholar

13. Rao G. Cardiometabolic diseases: a global perspective. J Cardiol Cardiovasc Ther. (2018) 12(2):555834.

Google Scholar

14. Flora GD, Nayak MK. A brief review of cardiovascular diseases, associated risk factors and current treatment regimes. Curr Pharm Des. (2019) 25(38):4063–84. doi: 10.2174/1381612825666190925163827

PubMed Abstract | Crossref Full Text | Google Scholar

15. Steven S, Frenis K, Oelze M, Kalinovic S, Kuntic M, Bayo Jimenez MT, et al. Vascular inflammation and oxidative stress: major triggers for cardiovascular disease. Oxid Med Cell Longevity. (2019) 2019(1):7092151.

Google Scholar

16. Manabe I. Chronic inflammation links cardiovascular, metabolic and renal diseases. Circ J. (2011) 75(12):2739–48. doi: 10.1253/circj.CJ-11-1184

PubMed Abstract | Crossref Full Text | Google Scholar

17. Tsoupras A, Lordan R, Zabetakis I. Inflammation and cardiovascular diseases. In: Zabetakis I, Lordan R, Tsoupras A, editors. The Impact of Nutrition and Statins on Cardiovascular Diseases. Amsterdam: Elsevier (2019). p. 53–117.

Google Scholar

18. Hamjane N, Benyahya F, Nourouti NG, Mechita MB, Barakat A. Cardiovascular diseases and metabolic abnormalities associated with obesity: what is the role of inflammatory responses? A systematic review. Microvasc Res. (2020) 131:104023. doi: 10.1016/j.mvr.2020.104023

PubMed Abstract | Crossref Full Text | Google Scholar

19. Nishida K, Otsu K. Inflammation and metabolic cardiomyopathy. Cardiovasc Res. (2017) 113(4):389–98. doi: 10.1093/cvr/cvx012

PubMed Abstract | Crossref Full Text | Google Scholar

20. Lasselin J, Capuron L. Chronic low-grade inflammation in metabolic disorders: relevance for behavioral symptoms. Neuroimmunomodulation. (2014) 21(2-3):95–101. doi: 10.1159/000356535

PubMed Abstract | Crossref Full Text | Google Scholar

21. Poznyak A, Grechko AV, Poggio P, Myasoedova VA, Alfieri V, Orekhov AN. The diabetes mellitus–atherosclerosis connection: the role of lipid and glucose metabolism and chronic inflammation. Int J Mol Sci. (2020) 21(5):1835. doi: 10.3390/ijms21051835

PubMed Abstract | Crossref Full Text | Google Scholar

22. Lamb R, Goldstein B. Modulating an oxidative-inflammatory cascade: potential new treatment strategy for improving glucose metabolism, insulin resistance, and vascular function. Int J Clin Pract. (2008) 62(7):1087–95. doi: 10.1111/j.1742-1241.2008.01789.x

PubMed Abstract | Crossref Full Text | Google Scholar

23. Bao MH, Li JM, Zhou QL, Li GY, Zeng J, Zhao J, et al. Effects of miR-590 on oxLDL-induced endothelial cell apoptosis: roles of p53 and NF-κB. Mol Med Rep. (2016) 13(1):867–73. doi: 10.3892/mmr.2015.4606

PubMed Abstract | Crossref Full Text | Google Scholar

24. Yi J, Li L, Yin ZJ, Quan YY, Tan RR, Chen SL, et al. Polypeptide from Moschus suppresses lipopolysaccharide-induced inflammation by inhibiting NF-κ B-ROS/NLRP3 pathway. Chin J Integr Med. (2023) 29(10):895–904. doi: 10.1007/s11655-023-3598-z

PubMed Abstract | Crossref Full Text | Google Scholar

25. Kritikou E, Kuiper J, Kovanen PT, Bot I. The impact of mast cells on cardiovascular diseases. Eur J Pharmacol. (2016) 778:103–15. doi: 10.1016/j.ejphar.2015.04.050

PubMed Abstract | Crossref Full Text | Google Scholar

26. Amin MN, Siddiqui SA, Ibrahim M, Hakim ML, Ahammed MS, Kabir A, et al. Inflammatory cytokines in the pathogenesis of cardiovascular disease and cancer. SAGE Open Med. (2020) 8:2050312120965752. doi: 10.1177/2050312120965752

PubMed Abstract | Crossref Full Text | Google Scholar

27. Incalza MA, D'Oria R, Natalicchio A, Perrini S, Laviola L, Giorgino F. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vasc Pharmacol. (2018) 100:1–19. doi: 10.1016/j.vph.2017.05.005

PubMed Abstract | Crossref Full Text | Google Scholar

28. Donia T, Khamis A. Management of oxidative stress and inflammation in cardiovascular diseases: mechanisms and challenges. Environ Sci Pollut Res. (2021) 28(26):34121–53. doi: 10.1007/s11356-021-14109-9

PubMed Abstract | Crossref Full Text | Google Scholar

29. Victor VM, Rocha M, Sola E, Banuls C, Garcia-Malpartida K, Hernandez-Mijares A. Oxidative stress, endothelial dysfunction and atherosclerosis. Curr Pharm Des. (2009) 15(26):2988–3002. doi: 10.2174/138161209789058093

PubMed Abstract | Crossref Full Text | Google Scholar

30. Fu Q, Chen R, Ding Y, Xu S, Huang C, He B, et al. Sodium intake and the risk of various types of cardiovascular diseases: a Mendelian randomization study. Front Nutr. (2023) 10:1250509. doi: 10.3389/fnut.2023.1250509

PubMed Abstract | Crossref Full Text | Google Scholar

31. Meshkani R, Adeli K. Hepatic insulin resistance, metabolic syndrome and cardiovascular disease. Clin Biochem. (2009) 42(13–14):1331–46. doi: 10.1016/j.clinbiochem.2009.05.018

PubMed Abstract | Crossref Full Text | Google Scholar

32. Sanderson E, Glymour MM, Holmes MV, Kang H, Morrison J, Munafò MR, et al. Mendelian Randomization. Nat Rev Methods Primers. (2022) 2:1–21.

Google Scholar

33. Jiang Y, Chen R, Xu S, Ding Y, Zhang M, Bao M, et al. Assessing causal associations of hyperparathyroidism with blood counts and biochemical indicators: a Mendelian randomization study. Front Endocrinol. (2023) 14:1295040. doi: 10.3389/fendo.2023.1295040

PubMed Abstract | Crossref Full Text | Google Scholar

34. Huang C, Xu S, Chen R, Ding Y, Fu Q, He B, et al. Assessing causal associations of bile acids with obesity indicators: a Mendelian randomization study. Medicine. (2024) 103(25):e38610. doi: 10.1097/MD.0000000000038610

PubMed Abstract | Crossref Full Text | Google Scholar

35. Han L, Xu S, Zhou D, Chen R, Ding Y, Zhang M, et al. Unveiling the causal link between metabolic factors and ovarian cancer risk using Mendelian randomization analysis. Front Endocrinol. (2024) 15:1401648. doi: 10.3389/fendo.2024.1401648

PubMed Abstract | Crossref Full Text | Google Scholar

36. Wu Z, Luo S, Cai D, Lin W, Hu X, Zhou T, et al. The causal relationship between metabolic syndrome and its components and cardiovascular disease: a Mendelian randomization study. Diabetes Res Clin Pract. (2024) 211:111679. doi: 10.1016/j.diabres.2024.111679

PubMed Abstract | Crossref Full Text | Google Scholar

37. Ueno K, Kaneko H, Suzuki Y, Okada A, Matsuoka S, Fujiu K, et al. Metabolic syndrome and cardiovascular disease in cancer survivors. J Cachexia Sarcopenia Muscle. (2024) 15(3):1062–71. doi: 10.1002/jcsm.13443

PubMed Abstract | Crossref Full Text | Google Scholar

38. Laway BA, Baba MS. Sheehan syndrome: cardiovascular and metabolic comorbidities. Front Endocrinol. (2023) 14:1086731. doi: 10.3389/fendo.2023.1086731

PubMed Abstract | Crossref Full Text | Google Scholar

39. Varra F-N, Varras M, Varra V-K, Theodosis-Nobelos P. Molecular and pathophysiological relationship between obesity and chronic inflammation in the manifestation of metabolic dysfunctions and their inflammation-mediating treatment options. Mol Med Rep. (2024) 29(6):95. doi: 10.3892/mmr.2024.13219

PubMed Abstract | Crossref Full Text | Google Scholar

40. Li X, Li Y, Hao Q, Jin J, Wang Y. Metabolic mechanisms orchestrated by Sirtuin family to modulate inflammatory responses. Front Immunol. (2024) 15:1448535. doi: 10.3389/fimmu.2024.1448535

PubMed Abstract | Crossref Full Text | Google Scholar

41. Xiong P, Zhang F, Liu F, Zhao J, Huang X, Luo D, et al. Metaflammation in glucolipid metabolic disorders: pathogenesis and treatment. Biomed Pharmacother. (2023) 161:114545. doi: 10.1016/j.biopha.2023.114545

PubMed Abstract | Crossref Full Text | Google Scholar

42. Tuttolomondo A, Di Raimondo D, Pecoraro R, Arnao V, Pinto A, Licata G. Atherosclerosis as an inflammatory disease. Curr Pharm Des. (2012) 18(28):4266–88. doi: 10.2174/138161212802481237

PubMed Abstract | Crossref Full Text | Google Scholar

43. Bao MH, Luo HQ, Chen LH, Tang L, Ma KF, Xiang J, et al. Impact of high fat diet on long non-coding RNAs and messenger RNAs expression in the aortas of ApoE(−/−) mice. Sci Rep. (2016) 6:34161. doi: 10.1038/srep34161

PubMed Abstract | Crossref Full Text | Google Scholar

44. Tabas I, Williams KJ, Borén J. Subendothelial lipoprotein retention as the initiating process in atherosclerosis: update and therapeutic implications. Circulation. (2007) 116(16):1832–44. doi: 10.1161/CIRCULATIONAHA.106.676890

PubMed Abstract | Crossref Full Text | Google Scholar

45. Jiang Y, Chen R, Xu S, Ding Y, Zhang M, Bao M, et al. Endocrine and metabolic factors and the risk of idiopathic pulmonary fibrosis: a Mendelian randomization study. Front Endocrinol. (2023) 14:1321576. doi: 10.3389/fendo.2023.1321576

PubMed Abstract | Crossref Full Text | Google Scholar

46. Vekic J, Zeljkovic A, Cicero AF, Janez A, Stoian AP, Sonmez A, et al. Atherosclerosis development and progression: the role of atherogenic small, dense LDL. Medicina (B Aires). (2022) 58(2):299. doi: 10.3390/medicina58020299

Crossref Full Text | Google Scholar

47. Malekmohammad K, Bezsonov EE, Rafieian-Kopaei M. Role of lipid accumulation and inflammation in atherosclerosis: focus on molecular and cellular mechanisms. Front Cardiovasc Med. (2021) 8:707529. doi: 10.3389/fcvm.2021.707529

PubMed Abstract | Crossref Full Text | Google Scholar

48. Li W, Zhi W, Zhao J, Yao Q, Liu F, Niu X. Cinnamaldehyde protects VSMCs against ox-LDL-induced proliferation and migration through S arrest and inhibition of p38, JNK/MAPKs and NF-κB. Vasc Pharmacol. (2018) 108:57–66. doi: 10.1016/j.vph.2018.05.005

Crossref Full Text | Google Scholar

49. Guo J, Du L. An update on ox-LDL-inducing vascular smooth muscle cell-derived foam cells in atherosclerosis. Front Cell Dev Biol. (2024) 12:1481505. doi: 10.3389/fcell.2024.1481505

PubMed Abstract | Crossref Full Text | Google Scholar

50. Li T, Wang B, Ding H, Chen S, Cheng W, Li Y, et al. Effect of extracellular vesicles from multiple cells on vascular smooth muscle cells in atherosclerosis. Front Pharmacol. (2022) 13:857331. doi: 10.3389/fphar.2022.857331

PubMed Abstract | Crossref Full Text | Google Scholar

51. Yan B, Yu X, Cai X, Huang X, Xie B, Lian D, et al. A review: the significance of toll-like receptors 2 and 4, and NF-κB signaling in endothelial cells during atherosclerosis. Front Biosci. (2024) 29(4):161. doi: 10.31083/j.fbl2904161

Crossref Full Text | Google Scholar

52. Yu Y, Cai Y, Yang F, Yang Y, Cui Z, Shi D, et al. Vascular smooth muscle cell phenotypic switching in atherosclerosis. Heliyon. (2024) 10(18):e37727.39309965

PubMed Abstract | Google Scholar

53. Gibson MS, Domingues N, Vieira OV. Lipid and non-lipid factors affecting macrophage dysfunction and inflammation in atherosclerosis. Front Physiol. (2018) 9:654. doi: 10.3389/fphys.2018.00654

PubMed Abstract | Crossref Full Text | Google Scholar

54. Han Z, Liu Q, Li H, Zhang M, You L, Lin Y, et al. The role of monocytes in thrombotic diseases: a review. Front Cardiovasc Med. (2023) 10:1113827. doi: 10.3389/fcvm.2023.1113827

PubMed Abstract | Crossref Full Text | Google Scholar

55. Feng Y, Ye D, Wang Z, Pan H, Lu X, Wang M, et al. The role of interleukin-6 family members in cardiovascular diseases. Front Cardiovasc Med. (2022) 9:818890. doi: 10.3389/fcvm.2022.818890

PubMed Abstract | Crossref Full Text | Google Scholar

56. Su J-H, Luo M-Y, Liang N-, Gong S-X, Chen W, Huang W-Q, et al. Interleukin-6: a novel target for cardio-cerebrovascular diseases. Front Pharmacol. (2021) 12:745061. doi: 10.3389/fphar.2021.745061

PubMed Abstract | Crossref Full Text | Google Scholar

57. Gencer S, Evans BR, van der Vorst EP, Döring Y, Weber C. Inflammatory chemokines in atherosclerosis. Cells. (2021) 10(2):226. doi: 10.3390/cells10020226

PubMed Abstract | Crossref Full Text | Google Scholar

58. Duez H, Pourcet B. Nuclear receptors in the control of the NLRP3 inflammasome pathway. Front Endocrinol. (2021) 12:630536. doi: 10.3389/fendo.2021.630536

PubMed Abstract | Crossref Full Text | Google Scholar

59. Van de Veerdonk FL, Netea MG, Dinarello CA, Joosten LA. Inflammasome activation and IL-1β and IL-18 processing during infection. Trends Immunol. (2011) 32(3):110–6. doi: 10.1016/j.it.2011.01.003

PubMed Abstract | Crossref Full Text | Google Scholar

60. Barker BR, Taxman DJ, Ting JP. Cross-regulation between the IL-1β/IL-18 processing inflammasome and other inflammatory cytokines. Curr Opin Immunol. (2011) 23(5):591–7. doi: 10.1016/j.coi.2011.07.005

PubMed Abstract | Crossref Full Text | Google Scholar

61. An N, Gao Y, Si Z, Zhang H, Wang L, Tian C, et al. Regulatory mechanisms of the NLRP3 inflammasome, a novel immune-inflammatory marker in cardiovascular diseases. Front Immunol. (2019) 10:1592. doi: 10.3389/fimmu.2019.01592

PubMed Abstract | Crossref Full Text | Google Scholar

62. Jiang H, Zhou Y, Nabavi SM, Sahebkar A, Little PJ, Xu S, et al. Mechanisms of oxidized LDL-mediated endothelial dysfunction and its consequences for the development of atherosclerosis. Front Cardiovasc Med. (2022) 9:925923. doi: 10.3389/fcvm.2022.925923

PubMed Abstract | Crossref Full Text | Google Scholar

63. Haraldsen G, Kvale D, Lien B, Farstad IN, Brandtzaeg P. Cytokine-regulated expression of E-selectin, intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1) in human microvascular endothelial cells. J Immun. (1996) 156(7):2558–65. doi: 10.4049/jimmunol.156.7.2558

PubMed Abstract | Crossref Full Text | Google Scholar

64. Singh V, Kaur R, Kumari P, Pasricha C, Singh R. ICAM-1 and VCAM-1: gatekeepers in various inflammatory and cardiovascular disorders. Clin Chim Acta. (2023) 548:117487. doi: 10.1016/j.cca.2023.117487

PubMed Abstract | Crossref Full Text | Google Scholar

65. Carlos TM, Harlan JM. Leukocyte-endothelial adhesion molecules. Blood. (1994) 84(7):2068–101. doi: 10.1182/blood.V84.7.2068.2068

PubMed Abstract | Crossref Full Text | Google Scholar

66. Van Gils JM, Zwaginga JJ, Hordijk PL. Molecular and functional interactions among monocytes, platelets, and endothelial cells and their relevance for cardiovascular diseases. J Leukoc Biol. (2009) 85(2):195–204. doi: 10.1189/jlb.0708400

PubMed Abstract | Crossref Full Text | Google Scholar

67. Medrano-Bosch M, Simón-Codina B, Jiménez W, Edelman ER, Melgar-Lesmes P. Monocyte-endothelial cell interactions in vascular and tissue remodeling. Front Immunol. (2023) 14:1196033. doi: 10.3389/fimmu.2023.1196033

PubMed Abstract | Crossref Full Text | Google Scholar

68. Čejková S, Králová-Lesná I, Poledne R. Monocyte adhesion to the endothelium is an initial stage of atherosclerosis development. Cor Vasa. (2016) 58(4):e419–25. doi: 10.1016/j.crvasa.2015.08.002

Crossref Full Text | Google Scholar

69. Lee-Rueckert M, Lappalainen J, Kovanen PT, Escola-Gil JC. Lipid-laden macrophages and inflammation in atherosclerosi s and cancer: an integrative view. Front Cardiovasc Med. (2022) 9:777822. doi: 10.3389/fcvm.2022.777822

PubMed Abstract | Crossref Full Text | Google Scholar

70. Farahi L, Sinha SK, Lusis AJ. Roles of macrophages in atherogenesis. Front Pharmacol. (2021) 12:785220. doi: 10.3389/fphar.2021.785220

PubMed Abstract | Crossref Full Text | Google Scholar

71. da Luz PL, Chagas ACP, Dourado PMM, Laurindo FR. Endothelium in atherosclerosis: plaque formation and its complications. In: Da Luz PL, Libby P, Chagas ACP, Laurindo FRM, editors. Endothelium and Cardiovascular Diseases. Amsterdam: Elsevier (2018). p. 493–512.

Google Scholar

72. Orr AW, Hastings NE, Blackman BR, Wamhoff BR. Complex regulation and function of the inflammatory smooth muscle cell phenotype in atherosclerosis. J Vasc Res. (2010) 47(2):168–80. doi: 10.1159/000250095

PubMed Abstract | Crossref Full Text | Google Scholar

73. Jaminon A, Reesink K, Kroon A, Schurgers L. The role of vascular smooth muscle cells in arterial remodeling: focus on calcification-related processes. Int J Mol Sci. (2019) 20(22):5694. doi: 10.3390/ijms20225694

PubMed Abstract | Crossref Full Text | Google Scholar

74. Harman JL, Jørgensen HF. The role of smooth muscle cells in plaque stability: therapeutic targeting potential. Br J Pharmacol. (2019) 176(19):3741–53. doi: 10.1111/bph.14779

PubMed Abstract | Crossref Full Text | Google Scholar

75. Badimon L, Vilahur G. Thrombosis formation on atherosclerotic lesions and plaque rupture. J Intern Med. (2014) 276(6):618–32. doi: 10.1111/joim.12296

PubMed Abstract | Crossref Full Text | Google Scholar

76. Solak Y, Afsar B, Vaziri ND, Aslan G, Yalcin CE, Covic A, et al. Hypertension as an autoimmune and inflammatory disease. Hypertens Res. (2016) 39(8):567–73. doi: 10.1038/hr.2016.35

PubMed Abstract | Crossref Full Text | Google Scholar

77. Totoń-Żurańska J, Mikolajczyk TP, Saju B, Guzik TJ. Vascular remodelling in cardiovascular diseases: hypertension, oxidation, and inflammation. Clin Sci. (2024) 138(13):817–50. doi: 10.1042/CS20220797

PubMed Abstract | Crossref Full Text | Google Scholar

78. Su C, Xue J, Ye C, Chen A. Role of the central renin-angiotensin system in hypertension. Int J Mol Med. (2021) 47(6):1–16.

Google Scholar

79. Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, et al. Angiotensin II signal transduction: an update on mechanisms of physiology and pathophysiology. Physiol Rev. (2018) 98(3):1627–738. doi: 10.1152/physrev.00038.2017

PubMed Abstract | Crossref Full Text | Google Scholar

80. Harrison DG, Gongora MC. Oxidative stress and hypertension. Med Clin. (2009) 93(3):621–35.

Google Scholar

81. Dikalov SI, Nazarewicz RR. Angiotensin II-induced production of mitochondrial reactive oxygen species: potential mechanisms and relevance for cardiovascular disease. Antioxid Redox Signaling. (2013) 19(10):1085–94. doi: 10.1089/ars.2012.4604

PubMed Abstract | Crossref Full Text | Google Scholar

82. Wang W, Kang PM. Oxidative stress and antioxidant treatments in cardiovascular diseases. Antioxidants. (2020) 9(12):1292. doi: 10.3390/antiox9121292

PubMed Abstract | Crossref Full Text | Google Scholar

83. Brandes RP. Endothelial dysfunction and hypertension. Hypertension. (2014) 64(5):924–8. doi: 10.1161/HYPERTENSIONAHA.114.03575

PubMed Abstract | Crossref Full Text | Google Scholar

84. Goncharov N, Popova P, Avdonin P, Kudryavtsev I, Serebryakova M, Korf E, et al. Markers of endothelial cells in normal and pathological conditions. Biochem (Mosc) Suppl A Membr Cell Biol. (2020) 14:167–83. doi: 10.1134/S1990747819030140

PubMed Abstract | Crossref Full Text | Google Scholar

85. Xiao L, Liu Y, Wang N. New paradigms in inflammatory signaling in vascular endothelial cells. Am J Physiol Heart Circ Physiol. (2014) 306(3):H317–25. doi: 10.1152/ajpheart.00182.2013

PubMed Abstract | Crossref Full Text | Google Scholar

86. Tran N, Garcia T, Aniqa M, Ali S, Ally A, Nauli SM. Endothelial nitric oxide synthase (eNOS) and the cardiovascular system: in physiology and in disease states. Am J Biomed Sci Res. (2022) 15(2):153.35072089

PubMed Abstract | Google Scholar

87. Yang YY, Shi LX, Li JH, Yao LY, Xiang DX. Piperazine ferulate ameliorates the development of diabetic nephropathy by regulating endothelial nitric oxide synthase. Mol Med Rep. (2019) 19(3):2245–53.30664213

PubMed Abstract | Google Scholar

88. Li Q, Youn J-Y, Cai H. Mechanisms and consequences of endothelial nitric oxide synthase dysfunction in hypertension. J Hypertens. (2015) 33(6):1128–36. doi: 10.1097/HJH.0000000000000587

PubMed Abstract | Crossref Full Text | Google Scholar

89. Zhang Z, Zhao L, Zhou X, Meng X, Zhou X. Role of inflammation, immunity, and oxidative stress in hypertension: new insights and potential therapeutic targets. Front Immunol. (2023) 13:1098725. doi: 10.3389/fimmu.2022.1098725

PubMed Abstract | Crossref Full Text | Google Scholar

90. Itani HA, McMaster WG Jr, Saleh MA, Nazarewicz RR, Mikolajczyk TP, Kaszuba AM, et al. Activation of human T cells in hypertension: studies of humanized mice and hypertensive humans. Hypertension. (2016) 68(1):123–32. doi: 10.1161/HYPERTENSIONAHA.116.07237

PubMed Abstract | Crossref Full Text | Google Scholar

91. Eid RE, Rao DA, Zhou J, Lo S-fL, Ranjbaran H, Gallo A, et al. Interleukin-17 and interferon-γ are produced concomitantly by human coronary artery–infiltrating T cells and act synergistically on vascular smooth muscle cells. Circulation. (2009) 119(10):1424–32. doi: 10.1161/CIRCULATIONAHA.108.827618

PubMed Abstract | Crossref Full Text | Google Scholar

92. Mills KH. IL-17 and IL-17-producing cells in protection versus pathology. Nat Rev Immunol. (2023) 23(1):38–54. doi: 10.1038/s41577-022-00746-9

PubMed Abstract | Crossref Full Text | Google Scholar

93. Liang D, Cai X, Guan Q, Ou Y, Zheng X, Lin X. Burden of type 1 and type 2 diabetes and high fasting plasma glucose in Europe, 1990–2019: a comprehensive analysis from the global burden of disease study 2019. Front Endocrinol. (2023) 14:1307432. doi: 10.3389/fendo.2023.1307432

Crossref Full Text | Google Scholar

94. Zuberi Z, Sauli E, Cun L, Deng J, Li WJ, He XL, et al. Insulin-delivery methods for children and adolescents with type 1 diabetes. Ther Adv Endocrinol Metab. (2020) 11:2042018820906016. doi: 10.1177/2042018820906016

PubMed Abstract | Crossref Full Text | Google Scholar

95. Zhou Y, Chai X, Yang G, Sun X, Xing Z. Changes in body mass index and waist circumference and heart failure in type 2 diabetes mellitus. Front Endocrinol. (2023) 14:1305839. doi: 10.3389/fendo.2023.1305839

PubMed Abstract | Crossref Full Text | Google Scholar

96. Fu Q, Chen R, Xu S, Ding Y, Huang C, He B, et al. Assessment of potential risk factors associated with gestational diabetes mellitus: evidence from a Mendelian randomization study. Front Endocrinol. (2023) 14:1276836. doi: 10.3389/fendo.2023.1276836

PubMed Abstract | Crossref Full Text | Google Scholar

97. He K, Chen R, Xu S, Ding Y, Wu Z, Bao M, et al. Environmental endocrine disruptor-induced mitochondrial dysfunction: a potential mechanism underlying diabetes and its complications. Front Endocrinol. (2024) 15:1422752. doi: 10.3389/fendo.2024.1422752

PubMed Abstract | Crossref Full Text | Google Scholar

98. Chen R, Xu S, Ding Y, Li L, Huang C, Bao M, et al. Dissecting causal associations of type 2 diabetes with 111 types of ocular conditions: a Mendelian randomization study. Front Endocrinol. (2023) 14:1307468. doi: 10.3389/fendo.2023.1307468

PubMed Abstract | Crossref Full Text | Google Scholar

99. Chen J, Li X, Liu H, Zhong D, Yin K, Li Y, et al. Bone marrow stromal cell-derived exosomal circular RNA improves diabetic foot ulcer wound healing by activating the nuclear factor erythroid 2-related factor 2 pathway and inhibiting ferroptosis. Diabet Med. (2023) 40(7):e15031. doi: 10.1111/dme.15031

PubMed Abstract | Crossref Full Text | Google Scholar

100. Su M, Hu R, Tang T, Tang W, Huang C. Review of the correlation between Chinese medicine and intestinal microbiota on the efficacy of diabetes mellitus. Front Endocrinol. (2022) 13:1085092. doi: 10.3389/fendo.2022.1085092

PubMed Abstract | Crossref Full Text | Google Scholar

101. Xu Z, Zhang P, Chen Y, Jiang J, Zhou Z, Zhu H. Comparing SARC-CalF with SARC-F for screening sarcopenia in adults with type 2 diabetes Mellitus. Front Nutr. (2022) 9:803924. doi: 10.3389/fnut.2022.803924

PubMed Abstract | Crossref Full Text | Google Scholar

102. Yu T, Xu B, Bao M, Gao Y, Zhang Q, Zhang X, et al. Identification of potential biomarkers and pathways associated with carotid atherosclerotic plaques in type 2 diabetes mellitus: a transcriptomics study. Front Endocrinol. (2022) 13:981100. doi: 10.3389/fendo.2022.981100

PubMed Abstract | Crossref Full Text | Google Scholar

103. Luo M, Cao Q, Wang D, Tan R, Shi Y, Chen J, et al. The impact of diabetes on postoperative outcomes following spine surgery: a meta-analysis of 40 cohort studies with 2.9 million participants. Int J Surg. (2022) 104:106789. doi: 10.1016/j.ijsu.2022.106789

PubMed Abstract | Crossref Full Text | Google Scholar

104. Mi W, Xia Y, Bian Y. Meta-analysis of the association between aldose reductase gene (CA)n microsatellite variants and risk of diabetic retinopathy. Exp Ther Med. (2019) 18(6):4499–509.31777552

PubMed Abstract | Google Scholar

105. Wei Y, Xu S, Wu Z, Zhang M, Bao M, He B. Exploring the causal relationships between type 2 diabetes and neurological disorders using a Mendelian randomization strategy. Medicine. (2024) 103(46):e40412. doi: 10.1097/MD.0000000000040412

PubMed Abstract | Crossref Full Text | Google Scholar

106. Salvatore T, Pafundi PC, Galiero R, Albanese G, Di Martino A, Caturano A, et al. The diabetic cardiomyopathy: the contributing pathophysiological mechanisms. Front Med. (2021) 8:695792. doi: 10.3389/fmed.2021.695792

PubMed Abstract | Crossref Full Text | Google Scholar

107. Luo B, Huang F, Liu Y, Liang Y, Wei Z, Ke H, et al. NLRP3 Inflammasome as a molecular marker in diabetic cardiomyopathy. Front Physiol. (2017) 8:519. doi: 10.3389/fphys.2017.00519

PubMed Abstract | Crossref Full Text | Google Scholar

108. Mengstie MA, Chekol Abebe E, Behaile Teklemariam A, Tilahun Mulu A, Agidew MM, Teshome Azezew M, et al. Endogenous advanced glycation end products in the pathogenesis of chronic diabetic complications. Front Mol Biosci. (2022) 9:1002710. doi: 10.3389/fmolb.2022.1002710

PubMed Abstract | Crossref Full Text | Google Scholar

109. Hu H, Jiang H, Ren H, Hu X, Wang X, Han C. AGEs and chronic subclinical inflammation in diabetes: disorders of immune system. Diabetes Metab Res Rev. (2015) 31(2):127–37. doi: 10.1002/dmrr.2560

PubMed Abstract | Crossref Full Text | Google Scholar

110. Peng H, Gao Y, Zeng C, Hua R, Guo Y, Wang Y, et al. Effects of Maillard reaction and its product AGEs on aging and age-related diseases. Food Sci Hum Wellness. (2024) 13(3):1118–34. doi: 10.26599/FSHW.2022.9250094

Crossref Full Text | Google Scholar

111. Teissier T, Boulanger É. The receptor for advanced glycation end-products (RAGE) is an important pattern recognition receptor (PRR) for inflammaging. Biogerontology. (2019) 20(3):279–301. doi: 10.1007/s10522-019-09808-3

PubMed Abstract | Crossref Full Text | Google Scholar

112. Dong H, Zhang Y, Huang Y, Deng H. Pathophysiology of RAGE in inflammatory diseases. Front Immunol. (2022) 13:931473. doi: 10.3389/fimmu.2022.931473

PubMed Abstract | Crossref Full Text | Google Scholar

113. Bhattacharya R, Alam MR, Kamal MA, Seo KJ, Singh LR. AGE-RAGE axis culminates into multiple pathogenic processes: a central road to neurodegeneration. Front Mol Neurosci. (2023) 16:1155175. doi: 10.3389/fnmol.2023.1155175

PubMed Abstract | Crossref Full Text | Google Scholar

114. Gąsiorowski K, Brokos B, Echeverria V, Barreto GE, Leszek J. RAGE-TLR crosstalk sustains chronic inflammation in neurodegeneration. Mol Neurobiol. (2018) 55:1463–76. doi: 10.1007/s12035-017-0419-4

PubMed Abstract | Crossref Full Text | Google Scholar

115. Waghela BN, Vaidya FU, Ranjan K, Chhipa AS, Tiwari BS, Pathak C. AGE-RAGE synergy influences programmed cell death signaling to promote cancer. Mol Cell Biochem. (2021) 476:585–98. doi: 10.1007/s11010-020-03928-y

PubMed Abstract | Crossref Full Text | Google Scholar

116. Rehman K, Akash MSH. Mechanisms of inflammatory responses and development of insulin resistance: how are they interlinked? J Biomed Sci. (2016) 23:1–18. doi: 10.1186/s12929-016-0303-y

PubMed Abstract | Crossref Full Text | Google Scholar

117. Wei Y, Chen K, Whaley-Connell AT, Stump CS, Ibdah JA, Sowers JR. Skeletal muscle insulin resistance: role of inflammatory cytokines and reactive oxygen species. Am J Physiol Regul Integr Comp Physiol. (2008) 294(3):R673–80. doi: 10.1152/ajpregu.00561.2007

PubMed Abstract | Crossref Full Text | Google Scholar

118. Di Meo S, Iossa S, Venditti P. Skeletal muscle insulin resistance: role of mitochondria and other ROS sources. J Endocrinol. (2017) 233(1):R15–42. doi: 10.1530/JOE-16-0598

PubMed Abstract | Crossref Full Text | Google Scholar

119. Zhang H, Dhalla NS. The role of pro-inflammatory cytokines in the pathogenesis of cardiovascular disease. Int J Mol Sci. (2024) 25(2):1082. doi: 10.3390/ijms25021082

PubMed Abstract | Crossref Full Text | Google Scholar

120. Palomer X, Salvadó L, Barroso E, Vázquez-Carrera M. An overview of the crosstalk between inflammatory processes and metabolic dysregulation during diabetic cardiomyopathy. Int J Cardiol. (2013) 168(4):3160–72. doi: 10.1016/j.ijcard.2013.07.150

PubMed Abstract | Crossref Full Text | Google Scholar

121. Chen Z, Jin Z-X, Cai J, Li R, Deng K-Q, Ji Y-X, et al. Energy substrate metabolism and oxidative stress in metabolic cardiomyopathy. J Mol Med. (2022) 100(12):1721–39. doi: 10.1007/s00109-022-02269-1

PubMed Abstract | Crossref Full Text | Google Scholar

122. Li J, Yang Y, Wang H, Ma D, Wang H, Chu L, et al. Baicalein ameliorates myocardial ischemia through reduction of oxidative stress, inflammation and apoptosis via TLR4/MyD88/MAPKS/NF-κB pathway and regulation of Ca2 + homeostasis by L-type Ca2 + channels. Front Pharmacol. (2022) 13:842723. doi: 10.3389/fphar.2022.842723

PubMed Abstract | Crossref Full Text | Google Scholar

123. Shu M, Cheng W, Jia X, Bai X, Zhao Y, Lu Y, et al. AGEs promote atherosclerosis by increasing LDL transcytosis across endothelial cells via RAGE/NF-κB/caveolin-1 pathway. Mol Med. (2023) 29(1):113. doi: 10.1186/s10020-023-00715-5

PubMed Abstract | Crossref Full Text | Google Scholar

124. Salvatore T, Galiero R, Caturano A, Vetrano E, Loffredo G, Rinaldi L, et al. Coronary microvascular dysfunction in diabetes mellitus: pathogenetic mechanisms and potential therapeutic options. Biomedicines. (2022) 10(9):2274. doi: 10.3390/biomedicines10092274

PubMed Abstract | Crossref Full Text | Google Scholar

125. Liu S, Gu Y. INFLA Score: a novel inflammatory marker for assessing cardiometabolic disease risk in obese individuals. Diabetol Metab Syndr. (2024) 16(1):151. doi: 10.1186/s13098-024-01396-8

PubMed Abstract | Crossref Full Text | Google Scholar

126. Butcko AJ, Putman AK, Mottillo EP. The intersection of genetic factors, aberrant nutrient metabolism and oxidative stress in the progression of cardiometabolic disease. Antioxidants. (2024) 13(1):87. doi: 10.3390/antiox13010087

PubMed Abstract | Crossref Full Text | Google Scholar

127. Guiducci L, Nicolini G, Forini F. Dietary patterns, gut microbiota remodeling, and cardiometabolic disease. Metabolites. (2023) 13(6):760. doi: 10.3390/metabo13060760

PubMed Abstract | Crossref Full Text | Google Scholar

128. Hijová E. Benefits of biotics for cardiovascular diseases. Int J Mol Sci. (2023) 24(7):6292. doi: 10.3390/ijms24076292

Crossref Full Text | Google Scholar

129. Wang A, Guan B, Zhang H, Xu H. Danger-associated metabolites trigger metaflammation: a crowbar in cardiometabolic diseases. Pharmacol Res. (2023) 198:106983.37931790

PubMed Abstract | Google Scholar

130. Zheng S, Tsao PS, Pan C. Abdominal aortic aneurysm and cardiometabolic traits share strong genetic susceptibility to lipid metabolism and inflammation. Nat Commun. (2024) 15(1):5652. doi: 10.1038/s41467-024-49921-7

PubMed Abstract | Crossref Full Text | Google Scholar

131. Börschel CS, Ortega-Alonso A, Havulinna AS, Jousilahti P, Salmi M, Jalkanen S, et al. Inflammatory proteomics profiling for prediction of incident atrial fibrillation. Heart. (2023) 109(13):1000–6. doi: 10.1136/heartjnl-2022-321959

PubMed Abstract | Crossref Full Text | Google Scholar

132. Rath S, Hawsawi YM, Alzahrani F, Khan MI. Epigenetic regulation of inflammation: the metabolomics connection. Semin Cell Dev Biol. (2024) 154(Pt C):355–63. doi: 10.1016/j.semcdb.2022.09.008

PubMed Abstract | Crossref Full Text | Google Scholar

Glossary

CMD            Cardiometabolic diseases

ox-LD            Loxidized low-density lipoprotein

IL-6            Interleukin-6

TNF-α            Tumor Necrosis Factor-alpha

RAS            renin-angiotensin system

AGE            sadvanced glycation end products

RAGE            Receptor for Advanced Glycation Endproducts

MetS            metabolic syndrome

CVD            Cardiovascular disease

MR            Mendelian randomization

T2D            type 2 diabetes

SIRT1            Silent Information Regulator 2 Ortholog 1

sdLDL-C            small dense LDL cholesterol

RLP-C            remnant-like particle cholesterol

VSMCs            vascular smooth muscle cells

EC            endothelial cells

TLRs            Toll-like receptors

IL-8            Interleukin-8

MCP-1            Monocyte Chemoattractant Protein-1

LDL-C            low-density lipoprotein cholesterol

ROS            reactive oxygen species

DNA            Deoxyribonucleic Acid

LDL            low-density lipoprotein

FFA            free fatty acids

NF-κB            Nuclear Factor kappa-light-chain-enhancer of activated B cells

CCL2            Chemokine (C-C motif) ligand 2

CXCL10            Chemokine (C-X-C motif) ligand 10

JAK-STAT            Janus Kinase—Signal Transducer and Activator of Transcription

CCR2            Chemokine (C-C motif) receptor 2

NLRP3            NLR family, pyrin domain containing 3

caspase-1            Cysteine-aspartic protease 1

pro-IL-1β            Pro-Interleukin-1β

pro-IL-18            Pro-Interleukin-18

IL-1β            Interleukin-1β

IL-18            Interleukin-18

ICAM-1            intercellular adhesion molecule-1

VCAM-1            vascular cell adhesion molecule-1

E-selectin            Endothelial Selectin

Ang II            Angiotensin II

AT1R            Angiotensin II type 1 receptor

NADPH            nicotinamide adenine dinucleotide phosphate

SOD            superoxide dismutase

GPx            glutathione peroxidase

ET-1            endothelin-1

NO            nitric oxide

eNOS            nitric oxide synthase

IFN-γ            interferon-γ

IL-17            interleukin-17

Th1            T Helper 1

Th17            T Helper 17

DCM            Diabetic cardiomyopathy

IKKI            κB kinase

IκBα            inhibitory protein Inhibitor of κB alpha

MAPK            mitogen-activated protein kinase

Bcl-2            B-cell Lymphoma 2

SCFAs            short-chain fatty acids

TMAO            trimethylamine N-oxide

Keywords: cardiometabolic diseases, inflammatory microenvironment, atherosclerosis, hypertension, diabetic cardiomyopathy

Citation: Liu M, Chen R, Zheng Z, Xu S, Hou C, Ding Y, Zhang M, Bao M, He B and Li S (2025) Mechanisms of inflammatory microenvironment formation in cardiometabolic diseases: molecular and cellular perspectives. Front. Cardiovasc. Med. 11:1529903. doi: 10.3389/fcvm.2024.1529903

Received: 18 November 2024; Accepted: 26 December 2024;
Published: 14 January 2025.

Edited by:

Lei Wang, Guangdong Provincial Hospital of Chinese Medicine, China

Reviewed by:

Deng Bo, Guangzhou Medical University, China
Bin Liu, Guangzhou Medical University, China

Copyright: © 2025 Liu, Chen, Zheng, Xu, Hou, Ding, Zhang, Bao, He and Li. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Meihua Bao, bWhiYW83OEAxNjMuY29t; Binsheng He, aGJzY3NtdUAxNjMuY29t; Sen Li, c2VubGlAYnVjbS5lZHUuY24=

These authors have contributed equally to this work and share first authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.