Skip to main content

EDITORIAL article

Front. Cardiovasc. Med., 27 April 2022
Sec. Cardiovascular Therapeutics
This article is part of the Research Topic Highlights in Cardiovascular Therapeutics: 2021 View all 8 articles

Editorial: Highlights for Cardiovascular Therapeutics in 2021 – Trained Immunity, Immunometabolism, Gender Differences of Cardiovascular Diseases, and Novel Targets of Cardiovascular Therapeutics

\nWilliam Y. YangWilliam Y. Yang1Bonnie NguyenBonnie Nguyen1Sheng WuSheng Wu1Jun YuJun Yu1Hong WangHong Wang1Xiaofeng Yang,
Xiaofeng Yang1,2*
  • 1Centers for Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
  • 2Cardiovascular Research, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States

Editorial on the Research Topic
Highlights in Cardiovascular Therapeutics: 2021

Introduction

Due to the highly collaborative efforts of the authors, editorial office, and editorial team, the Frontiers in Cardiovascular Medicine - Cardiovascular Therapeutics section has made significant achievements in 2021, with a five-fold increase in manuscript submission. We all greatly appreciate it. In 2022, we will continue our efforts to build an outstanding platform for cardiologists and translational cardiovascular scientists to exchange novel findings and data in clinical cardiology and cardiovascular therapeutic fields. Here, we would like to highlight some excellent articles published in our section as well as new progress in the field with significant potential in cardiovascular therapeutics. In addition, these highlights may also serve as the foundation for some new special topics in our Cardiovascular Therapeutics section in 2022.

Trained Immunity, an Innate Immune Memory, is a New Inflammation Amplifying Mechanism

Cardiovascular disease (CVD) is a leading cause of death in the USA and worldwide. Numerous risk factors for triggering the onset and promoting CVD progression have been identified, including hyperlipidemia (1), hyperglycemia, hyperhomocysteinemia (2), smoking, metabolic syndrome, hypertension, obesity (35), and infections (1, 69). On the other hand, many danger-associated molecular patterns (DAMPs) and conditional DAMPs (10) have been characterized in triggering the recruitment of monocytes, macrophages, T cells, B cells, and other immune cells into arteries to amplify the atherosclerotic process, suggesting that innate and adaptive immunity contribute to the atherosclerotic plaque formation. However, several important issues remain poorly characterized: first, what compounds in the environment and endogenous metabolic process in the host are qualified to become risk factors to stimulate atherosclerotic CVD (11); second, many disease risk factors are synergistic in promoting atherosclerotic CVD, including a) hyperhomocysteinemia and hyperlipidemia (12, 13); b) hyperhomocysteinemia and hyperglycemia (14, 15); c) multiple DAMPs stimulation in chronic kidney disease (CKD) (1619); d) hyperlipidemia and CKD (17); e) COVID-19 infection (coronavirus disease-2019, a viral respiratory illness caused by the severe acute respiratory syndrome-coronavirus-2, SARS-CoV-2) (20) and CVD; and f) sex hormone dysfunction and CVD (21); third, what cellular processes contribute to the quality and synergy of disease risk factors? Innate immune cells can develop exacerbated long-term immune responses and inflammatory phenotype following brief exposure to endogenous or exogenous DAMPs, which results in a primed and significantly enhanced inflammatory response toward a second challenge after returning to a non-activated state. This phenomenon is known as innate immune memory or trained immunity (TI). TI is important for host defense, vaccine response, and promoting the pathogenesis of chronic inflammations including metabolic CVDs such as atherosclerosis. In contrast to the memory function in the adaptive immune system with special cell subsets to carry out memory function such as memory T cells and memory B cells (22), TI can occur in innate immune cells such as monocytes/macrophages (23), natural killer cells, endothelial cells (ECs) (6, 11, 24), vascular smooth muscle cells (25, 26), and nonimmune cells, such as fibroblasts (7) and hepatocytes (27, 28). Of note, we recently reported that CD4+Foxp3+ regulatory T cells (Tregs) have many active innate immunity pathways (29, 30), and can sustain their immunosuppressive functions (31) in a proinflammatory atherogenic environment, although Tregs plasticity in atherosclerosis has been reported (32, 33). It has been reported that increased energy metabolism pathways and electron transport chain (34), including glycolysis, acetyl-CoA generation, mevalonate synthesis, glutaminolysis, and epigenetic modification (11, 35), contribute significantly to the establishment of TI. Extensive characterization of TI relative to CVD would provide novel insights into CVD pathogenesis and new therapeutic targets. To demonstrate the therapeutic potential of inhibiting new TI-related metabolic pathways such as glycolysis, Gager et al. reported that sodium–glucose co-transporter 2 (SGLT2) inhibitors, an emerging class of glucose-lowering drugs, have become increasingly relevant for the treatment and prevention of heart failure (HF). SGLT2 inhibitors are associated with improved cardiovascular outcomes in patients with HF (Gager et al.). In addition, Chen et al. reported that 2-Deoxy-D-glucose (2-DG), a glycolysis inhibitor, can alleviate cardiac fibrosis after myocardial infarction (MI) (36).

Trained Immunity May Be Underlying the Synergy of Multiple Risk Factors in Promoting the Pathogenesis of Cardiovascular Disease

A significant attribute of trained immunity in promoting cardiovascular diseases is that TI pathways' synergy among multiple risk factors contributes to CVD progression, which provides novel guidance for cardiovascular therapeutics. A recent report showed that COVID-19 may predispose patients to thrombotic disease, both in the venous and arterial circulations, attributed to excessive inflammation, platelet activation, endothelial dysfunction, and stasis (37). These findings emphasize that inflammation induced by COVID-19 infection may serve as the first stimuli in the TI setting and predispose patients to thrombotic disease (second stimuli). Zhao et al. reported that antithrombotic management for atrial fibrillation (AF) in patients undergoing percutaneous coronary intervention or with acute coronary syndrome [an evidence-based update combined antithrombotic regimens for AF in coronary artery disease patients, particularly in acute coronary syndrome (ACS) patients or patients undergoing percutaneous coronary intervention (PCI)], presents a great challenge in the real-world clinical scenario. The results of these studies have impacted the recommendations of current international guidelines, which favor a dual antithrombotic therapy (DAT) with a non-vitamin K antagonist (VKA) oral anticoagulant (NOAC) and classic antiplatelet drug and potential inflammasome inhibitor (38) P2Y12 inhibitor (39) (especially clopidogrel) in the clinical setting. Aspirin, a nonsteroidal anti-inflammatory drug, can be administered during the periprocedural period, while triple antithrombotic therapy (TAT) treatment duration should be as short as possible (Zhao et al.). Similarly, Bitar et al. conducted a systematic review of 326 articles and a meta-analysis on eight randomized clinical trials to evaluate the efficacy and safety of direct oral anticoagulants (DOACs) vs. warfarin (brand names Coumadin and Jantoven) in the treatment of AF and valvular heart disease (VHD). They found that DOACs remained with similar efficacy and safety compared to warfarin in thromboprophylaxis for AF and VHD (Bitar et al.). Since the interplay has been reported between inflammation and thrombosis in cardiovascular pathology (40), once again, these findings support our argument that blocking trained immunity as an inflammation amplifying pathway could be a benefit for cardiovascular therapies.

Insulin resistance (IR) has been identified as a risk factor and metabolic stress to accelerate vascular dysfunction and cardiovascular disease relative to type 2 diabetes mellitus (T2DM) (41). Controversies concerning the association between insulin therapy and atherosclerotic lesions in T2DM remain. Ke et al. investigated whether insulin therapy in T2DM patients is linked with the increased risk of carotid atherosclerosis, by retrospectively enrolling 2,356 hospitalized patients with T2DM, including 1,716 subjects receiving insulin therapy and 640 subjects without insulin therapy. This report showed that insulin therapy is associated with a markedly increased risk of carotid atherosclerotic lesions in T2DM, partly contributing to the more severe insulin resistance in T2DM patients receiving insulin therapy (Ke et al.). These findings correlate well with the previous findings from other teams that hyperinsulinemia is strongly associated with T2DM and is an early indicator of metabolic dysfunction (42). A potential mechanism underlying these findings may be insulin signaling and IR functions in promoting trained immunity in macrophages through metabolic and epigenetic changes (43).

NAD+ As an Immunomodulator May Inhibit Trained Immunity

Nicotinamide adenine dinucleotide (NAD+) has a direct inhibitory effect on poly(ADP-ribose) polymerase 1 (PARP-1) (44) and can prevent pro-inflammatory cytokines' over-activation. Increasing the NAD+ level will also stabilize telomeres, which positively impacts immune cells' function (45). We recently reported that liver ischemia reperfusion injury (IRI) is enhanced by trained immunity but is attenuated in the deficiency of pro-inflammatory DAMP/conditional DAMP (10) sensor (38) caspase-1/caspase-11 (human caspase-4) pathways in gene knockout mice (27, 46). Xiao et al. reported the cardioprotective properties of known agents in rat IRI model under clinically relevant conditions: only the NAD precursor nicotinamide riboside (NR) reduces myocardial infarct size in the presence of fentanyl (a synthetic opioid that is 80–100 times stronger than morphine), midazolam (produces sleepiness or drowsiness and relieves anxiety), and cangrelor (purinergic receptor P2Y12 inhibitor), but not propofol (P2Y12 antagonist). This observational study suggests that NR is a promising cardioprotective agent to target cardiac IRI in clinical conditions employing opioid agonists, benzodiazepines, and platelet P2Y12 inhibitors, but not propofol (Xiao et al.).

Rna Therapeutics Becomes a New Front in Cardiovascular Therapeutics and Regulators for Trained Immunity

The unprecedented expansion of data and information on RNA biology has led to new RNA classes with unique functions and unexpected modifications. The biggest challenge is to transfer the large number of findings in basic RNA biology into corresponding clinical RNA-based therapeutics. Lately, this research has begun to yield positive outcomes. Schellinger et al. reviewed significant progress on this front. RNA drugs advance to the final phases of clinical trials or even receive U.S. Food and Drug Administration (FDA) approval. Furthermore, the introduction of the RNA-guided gene-editing technology, the clustered regularly interspaced short palindromic repeats (CRISPR), and the advances in the delivery of messenger RNAs have triggered a significant progression in the field of RNA-therapeutics. Short interfering RNAs and antisense oligonucleotides especially are promising examples for novel categories of therapeutics. However, several issues need to be resolved, including intracellular delivery, toxicity, and immune responses, before utilizing RNAs in a clinical setting. Schellinger et al. provided an overview of opportunities and challenges for clinical translation of RNA-based therapeutics, emphasizing advances in novel delivery technologies and abdominal aortic aneurysm (AAA) disease where non-coding RNAs have been shown to play a crucial regulatory role (Schellinger et al.). To facilitate the studies on TI, a comprehensive database (47) was established, including 118 trained immunity regulators. Further characterization of master regulators, such as NADPH oxidase 2 (NOX2) (48), nuclear factor erythroid 2 (EFE2) like basic leucine zipper (bZIP) transcription factor 2 (NRF2) (20), hypoxia inducible factor 1 subunit alpha (HIF1a), mechanistic target of rapamycin kinase (mTOR), and SET domain containing 7 histone lysine methyltransferase (SET7) (49), will allow us to use RNA therapeutics to inhibit TI facilitated cardiovascular diseases.

Cell Death May Orchestrate a Balance Between Trained Immunity- Associated Chronic Cardiovascular Inflammation and Resolution

Cell death may orchestrate a balance between trained immunity-promoted chronic cardiovascular inflammation and resolution (50). Several new forms of cell death have been identified recently in infections, inflammation (51), cancers, and cardiovascular diseases (52), including panoptosis (53), pyroptosis (38), necroptosis, and ferroptosis (54). Ferroptosis is a type of regulated necrosis triggered by iron toxicity, lipid peroxidation, and plasma membrane damage. It is distinct from apoptosis, necroptosis, autophagy, and other types of cell death in morphology and function. The upstream inducers of ferroptosis can be divided into two categories (biological and chemical) and activate two major pathways (the extrinsic/transporter and the intrinsic/enzymatic pathways). Excessive or deficient ferroptotic cell death is implicated in a growing list of physiological and pathophysiological processes, coupled to a dysregulated immune response (55). Ferroptosis is regulated by various factors and controlled by several mechanisms, including mitochondrial activity and metabolism of iron, lipid, and amino acids. Accumulating evidence shows that ferroptosis is closely related to a majority of CVDs, including cardiomyopathy, myocardial infarction, ischemia/reperfusion injury, heart failure, and atherosclerosis. Hu et al. summarized the current status of ferroptosis and discusses ferroptosis as a potential therapeutic target for CVDs (Hu et al.). Further characterization of novel cell death pathways would lead to the future development of therapeutics for CVD.

Sex Hormones May Modulate Trained Immunity and Cardiovascular Diseases

Most studies on CVD have been conducted on male subjects, and assumed that women and men have similar physiological responses. However, the effects of sex hormones and their respective receptors in modulating cardiovascular functions are not uniform between men and women. The women (56) or female rodents (57) were associated with elevated androgen experience insulin resistance and increased risk of CVD (58). In contrast, men are at higher risk for CVD than women before the ages of 60 years old (59). It has been reported that low estrogen levels in younger females are associated with an increased risk of CVD. In addition, decreased estrogen levels after menopause are associated with dyslipidemia, increased blood pressure, and CVD. Moreover, pregnancy complications, including gestational diabetes and preeclampsia, and endocrine disorders such as polycystic ovarian syndrome promote CVD (60, 61). A recent report showed distinct sex differences in neutrophil biology related to responses to type I interferons (IFNs), immunometabolism, and maturation status that may have prominent functional and pathogenic implications (62). More directly, 17β-Estradiol was reported to promote TI in females against sepsis via regulating nucleus translocation of pro-inflammatory transcription factor RelB (48). Testosterone was identified to play a very important role in modulating the innate and adaptive immune systems (63). Further characterization of the functions of sex hormones in modulating TI relative to CVD pathogenesis would significantly improve our understanding of sex differences in CVDs and lead to the future development of therapeutics for CVD.

Author Contributions

WY, BN, SW, JY, and HW provided material input and participated in writing. XY supervised and edited the manuscript. All authors contributed to the article and approved the submitted version.

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.

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.

Acknowledgments

This work was supported by the National Institutes of Health Grants to XY (HL132399-01A1; HL138749-01 and HL147565-01), HW (DK104116; DK113775 and HL131460), SW (HD095512), and JY (HL153599).

References

1. Yang XF, Yin Y, Wang H. Vascular inflammation and atherogenesis are activated via receptors for pamps and suppressed by regulatory T cells. Drug Discov Today Ther Strateg. (2008) 5:125–42. doi: 10.1016/j.ddstr.2008.11.003

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Wang H, Jiang X, Yang F, Gaubatz JW, Ma L, Magera MJ, et al. Hyperhomocysteinemia accelerates atherosclerosis in cystathionine beta-synthase and apolipoprotein E double knock-out mice with and without dietary perturbation. Blood. (2003) 101:3901–7. doi: 10.1182/blood-2002-08-2606

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Virtue A, Johnson C, Lopez-Pastrana J, Shao Y, Fu H, Li X, et al. MicroRNA-155 deficiency leads to decreased atherosclerosis, increased white adipose tissue obesity, and non-alcoholic fatty liver disease: a novel mouse model of obesity paradox. J Biol Chem. (2017) 292:1267–87. doi: 10.1074/jbc.M116.739839

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Johnson C, Drummer Iv C, Shan H, Shao Y, Sun Y, Lu Y, et al. Novel subset of CD95(+) pro-inflammatory macrophages overcome miR155 deficiency and may serve as a switch from metabolically healthy obesity to metabolically unhealthy obesity. Front Immunol. (2020) 11:619951. doi: 10.3389/fimmu.2020.619951

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Johnson C, Drummer Ct, Virtue A, Gao T, Wu S, Hernandez M, et al. Increased Expression of Resistin in MicroRNA-155-Deficient White Adipose Tissues May Be a Possible Driver of Metabolically Healthy Obesity Transition to Classical Obesity. Front Physiol. (2018) 9:1297. doi: 10.3389/fphys.2018.01297

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Shao Y, Saredy J, Yang WY, Sun Y, Lu Y, Saaoud F, et al. Vascular endothelial cells and innate immunity. Arterioscler Thromb Vasc Biol. (2020) 40:e138–52. doi: 10.1161/ATVBAHA.120.314330

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Drummer Ct, Saaoud F, Shao Y, Sun Y, Xu K, Lu Y, et al. Trained immunity and reactivity of macrophages and endothelial cells. Arterioscler Thromb and Vasc Biol. (2021) 41:1032–46. doi: 10.1161/ATVBAHA.120.315452

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Zhong C, Yang X, Feng Y, Yu J. Trained immunity: an underlying driver of inflammatory atherosclerosis. Front Immunol. (2020) 11:284. doi: 10.3389/fimmu.2020.00284

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Yang Y, Xiong Z, Zhang S, Yan Y, Nguyen J, Ng B, et al. Bcl-xL inhibits T-cell apoptosis induced by expression of SARS coronavirus E protein in the absence of growth factors. Biochem J. (2005) 392:135–43. doi: 10.1042/BJ20050698

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Wang X, Li YF, Nanayakkara G, Shao Y, Liang B, Cole L, et al. Lysophospholipid receptors, as novel conditional danger receptors and homeostatic receptors modulate inflammation-novel paradigm and therapeutic potential. J Cardiovasc Transl Res. (2016) 9:343–59. doi: 10.1007/s12265-016-9700-6

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Lu Y, Sun Y., Drummer Ct, Nanayakkara GK, Shao Y, Saaoud F, et al. Increased acetylation of H3K14 in the genomic regions that encode trained immunity enzymes in lysophosphatidylcholine-activated human aortic endothelial cells - Novel qualification markers for chronic disease risk factors and conditional. DAMPs Redox Biol. (2019) 24:101221. doi: 10.1016/j.redox.2019.101221

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Zhang D, Jiang X, Fang P, Yan Y, Song J, Gupta S, et al. Hyperhomocysteinemia promotes inflammatory monocyte generation and accelerates atherosclerosis in transgenic cystathionine beta-synthase-deficient mice. Circulation. (2009) 120:1893–902. doi: 10.1161/CIRCULATIONAHA.109.866889

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Zhang D, Fang P, Jiang X, Nelson J, Moore JK, Kruger WD, et al. Severe hyperhomocysteinemia promotes bone marrow-derived and resident inflammatory monocyte differentiation and atherosclerosis in LDLr/CBS-deficient mice. Circ Res. (2012) 111:37–49. doi: 10.1161/CIRCRESAHA.112.269472

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Fang P, Li X, Shan H, Saredy JJ, Cueto R, Xia J, et al. Ly6C(+) inflammatory monocyte differentiation partially mediates hyperhomocysteinemia-induced vascular dysfunction in type 2 diabetic db/db mice. Arterioscler Thromb Vasc Biol. (2019) 39:2097–119. doi: 10.1161/ATVBAHA.119.313138

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Fang P, Zhang D, Cheng Z, Yan C, Jiang X, Kruger WD, et al. Hyperhomocysteinemia potentiates hyperglycemia-induced inflammatory monocyte differentiation and atherosclerosis. Diabetes. (2014) 63:4275–90. doi: 10.2337/db14-0809

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Sun Y, Johnson C, Zhou J, Wang L, Li YF, Lu Y, et al. Uremic toxins are conditional danger- or homeostasis-associated molecular patterns. Front Biosci (Landmark Ed). (2018) 23:348–87. doi: 10.2741/4595

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Yang J, Fang P, Yu D, Zhang L, Zhang D, Jiang X, et al. Chronic kidney disease induces inflammatory CD40+ monocyte differentiation via homocysteine elevation and DNA hypomethylation. Circ Res. (2016) 119:1226–41. doi: 10.1161/CIRCRESAHA.116.308750

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Zhang R, Saredy J, Shao Y, Yao T, Liu L, Saaoud F, et al. End-stage renal disease is different from chronic kidney disease in upregulating ROS-modulated proinflammatory secretome in PBMCs - A novel multiple-hit model for disease progression. Redox Biol. (2020) 34:101460. doi: 10.1016/j.redox.2020.101460

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Ferrer LM, Monroy AM, Lopez-Pastrana J, Nanayakkara G, Cueto R, Li YF Li X, et al. Caspase-1 plays a critical role in accelerating chronic kidney disease-promoted neointimal hyperplasia in the carotid artery. J Cardiovasc Transl Res. (2016). doi: 10.1007/s12265-016-9683-3

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Shao Y, Saredy J, Xu K, Sun Y, Saaoud F, Drummer Ct, et al. Endothelial immunity trained by coronavirus infections, DAMP stimulations and regulated by anti-oxidant nrf2 may contribute to inflammations, myelopoiesis, COVID-19 cytokine storms and thromboembolism. Front Immunol. (2021) 12:653110. doi: 10.3389/fimmu.2021.653110

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Wang C, Jackson G, Jones TH, Matsumoto AM, Nehra A, Perelman MA, et al. Low testosterone associated with obesity and the metabolic syndrome contributes to sexual dysfunction and cardiovascular disease risk in men with type 2 diabetes. Diabetes Care. (2011) 34:1669–75. doi: 10.2337/dc10-2339

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Shen H, Wu N, Nanayakkara G, Fu H, Yang Q, Yang WY Li A, et al. Co-signaling receptors regulate T-cell plasticity and immune tolerance. Front Biosci (Landmark Ed). (2019) 24:96–132. doi: 10.2741/4710

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Lai B, Wang J, Fagenson A, Sun Y, Saredy J, Lu Y, et al. Twenty novel disease group-specific and 12 new shared macrophage pathways in eight groups of 34 diseases including 24 inflammatory organ diseases and 10 types of tumors. Front Immunol. (2019) 10:2612. doi: 10.3389/fimmu.2019.02612

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Mai J, Virtue A, Shen J, Wang H, Yang XF. An evolving new paradigm: endothelial cells–conditional innate immune cells. J Hematol Oncol. (2013) 6:61. doi: 10.1186/1756-8722-6-61

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Flores-Gomez D, Bekkering S, Netea MG, Riksen NP. Trained Immunity in Atherosclerotic Cardiovascular Disease. Arterioscler Thromb Vasc Biol. (2021) 41:62–9. doi: 10.1161/ATVBAHA.120.314216

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Schnack L, Sohrabi Y, Lagache SMM, Kahles F, Bruemmer D, Waltenberger J, et al. Mechanisms of trained innate immunity in oxLDL primed human coronary smooth muscle cells. Front Immunol. (2019) 10:13. doi: 10.3389/fimmu.2019.00013

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Fagenson AM, Xu K, Saaoud F, Nanayakkara G, Jhala NC, Liu L, et al. Liver ischemia reperfusion injury, enhanced by trained immunity, is attenuated in caspase 1/caspase 11 double gene knockout mice. Pathogens. (2020) 9. doi: 10.3390/pathogens9110879

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Drummer CIV, Saaoud F, Sun Y, Atar D, Xu K, Lu Y, et al. Hyperlipidemia may synergize with hypomethylation in establishing trained immunity and promoting inflammation in NASH and NAFLD. J Immunol Res. (2021) 2021:3928323. doi: 10.1155/2021/3928323

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Zhang R, Xu K, Shao Y, Sun Y, Saredy J, Cutler E, et al. Tissue Treg secretomes and transcription factors shared with stem cells contribute to a Treg niche to maintain Treg-ness with 80% innate immune pathways, and functions of immunosuppression and tissue repair. Front Immunol. (2020) 11: 632239. doi: 10.3389/fimmu.2020.632239

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Ni D, Tang T, Lu Y, Xu K, Shao Y, Saaoud F, et al. Canonical Secretomes, Innate Immune Caspase-1-, 4/11-Gasdermin D Non-Canonical Secretomes and Exosomes May Contribute to Maintain Treg-Ness for Treg Immunosuppression, Tissue Repair and Modulate Anti-Tumor Immunity via ROS Pathways. Front Immunol. (2021) 12:678201. doi: 10.3389/fimmu.2021.678201

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Shao Y, Yang WY, Saaoud F, Drummer Ct, Sun Y, Xu K, et al. IL-35 promotes CD4+Foxp3+ Tregs and inhibits atherosclerosis via maintaining CCR5-amplified Treg-suppressive mechanisms. JCI Insight. (2021) 6. doi: 10.1172/jci.insight.152511

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Xu K, Yang WY, Nanayakkara GK, Shao Y, Yang F, Hu W, et al. GATA3, HDAC6, and BCL6 regulate FOXP3+ treg plasticity and determine treg conversion into either novel antigen-presenting cell-like Treg or Th1-Treg. Front Immunol. (2018) 9:45. doi: 10.3389/fimmu.2018.00045

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Ali AJ, Makings J, Ley K. Regulatory T cell stability and plasticity in atherosclerosis. Cells. (2020) 9. doi: 10.3390/cells9122665

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Yin M, O'Neill LAJ. The role of the electron transport chain in immunity. FASEB J. (2021) 35:e21974. doi: 10.1096/fj.202101161R

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Ferreira AV, Domiguez-Andres J, Netea MG. The role of cell metabolism in innate immune memory. J Innate Immun. (2022) 14:42–50. doi: 10.1159/000512280

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Chen ZT, Gao QY, Wu MX, Wang M, Sun RL, Jiang Y, et al. Glycolysis inhibition alleviates cardiac fibrosis after myocardial infarction by suppressing cardiac fibroblast activation. Front Cardiovasc Med. (2021) 8:701745. doi: 10.3389/fcvm.2021.701745

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Bikdeli B, Madhavan MV, Jimenez D, Chuich T, Dreyfus I, Driggin E, et al. COVID-19 and thrombotic or thromboembolic disease: implications for prevention, antithrombotic therapy, and follow-up: JACC State-of-the-Art Review. J Am Coll Cardiol. (2020) 75:2950–73. doi: 10.1016/j.jacc.2020.04.031

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Yin Y, Pastrana JL Li X, Huang X, Mallilankaraman K, Choi ET, Madesh M, et al. Inflammasomes: sensors of metabolic stresses for vascular inflammation. Front Biosci. (2013) 18:638–49. doi: 10.2741/4127

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Huang B, Qian Y, Xie S, Ye X, Chen H, Chen Z, et al. Ticagrelor inhibits the NLRP3 inflammasome to protect against inflammatory disease independent of the P2Y12 signaling pathway. Cell Mol Immunol. (2021) 18:1278–89. doi: 10.1038/s41423-020-0444-5

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Stark K, Massberg S. Interplay between inflammation and thrombosis in cardiovascular pathology. Nat Rev Cardiol. (2021) 18:666–82. doi: 10.1038/s41569-021-00552-1

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Pansuria M, Xi H, Li L, Yang XF, Wang H. Insulin resistance, metabolic stress, and atherosclerosis. Front Biosci (Schol Ed). (2012) 4:916–31. doi: 10.2741/s308

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Thomas DD, Corkey BE, Istfan NW, Apovian CM. Hyperinsulinemia: an early indicator of metabolic dysfunction. J Endocr Soc. (2019) 3:1727–47. doi: 10.1210/js.2019-00065

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Ieronymaki E, Daskalaki MG, Lyroni K, Tsatsanis C. Insulin signaling and insulin resistance facilitate trained immunity in macrophages through metabolic and epigenetic changes. Front Immunol. (2019) 10:1330. doi: 10.3389/fimmu.2019.01330

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Zhu H, Tang YD, Zhan G, Su C, Zheng C. The critical role of PARPS in regulating innate immune responses. Front Immunol. (2021) 12:712556. doi: 10.3389/fimmu.2021.712556

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Omran HM, Almaliki MS. Influence of NAD+ as an ageing-related immunomodulator on COVID 19 infection: a hypothesis. J Infect Public Health. (2020) 13:1196–201. doi: 10.1016/j.jiph.2020.06.004

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Yin Y, Li X, Sha X, Xi H, Li Y-F, Shao Y, et al. Early hyperlipidemia promotes endothelial activation via a caspase-1-sirtuin 1 pathway. Arterioscler Thromb Vasc Biol. (2015) 35:804–16. doi: 10.1161/ATVBAHA.115.305282

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Cao Y, Dong Q, Wang D, Liu Y, Zhang P, Yu X, Niu C. TIDB: a comprehensive database of trained immunity. Database (Oxford). (2021) 2021. doi: 10.1093/database/baab041

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Sun Z, Pan Y, Qu J, Xu Y, Dou H, Hou Y. 17beta-estradiol promotes trained immunity in females against sepsis via regulating nucleus translocation of RelB. Front Immunol. (2020) 11:1591. doi: 10.3389/fimmu.2020.01591

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Lu Y, Sun Y, Xu K, Saaoud F, Shao Y, Drummer Ct, et al. Aorta in pathologies may function as an immune organ by upregulating secretomes for immune and vascular cell activation, differentiation and trans-differentiation-early secretomes may serve as drivers for trained immunity. Front Immunol. (2022) 13:858256. doi: 10.3389/fimmu.2022.858256

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Soloff AC, Lotze MT. A peaceful death orchestrates immune balance in a chaotic environment. Proc Natl Acad Sci U S A. (2019) 116:22901–3. doi: 10.1073/pnas.1916211116

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Wang J, Lai B, Nanayakkara G, Yang Q, Sun Y, Lu Y, et al. Experimental data-mining analyses reveal new roles of low-intensity ultrasound in differentiating cell death regulatome in cancer and non-cancer cells via potential modulation of chromatin long-range interactions. Front Oncol. (2019) 600. doi: 10.3389/fonc.2019.00600

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Chakraborty A, Li Y, Zhang C, Li Y, LeMaire SA, Shen YH. Programmed cell death in aortic aneurysm and dissection: A potential therapeutic target. J Mol Cell Cardiol. (2022) 163:67–80. doi: 10.1016/j.yjmcc.2021.09.010

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Nguyen LN, Kanneganti TD. PANoptosis in viral infection: the missing puzzle piece in the cell death field. J Mol Biol. (2022) 434:167249. doi: 10.1016/j.jmb.2021.167249

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. (2018) 25:486–541. doi: 10.1038/s41418-018-0102-y

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Chen X, Kang R, Kroemer G, Tang D. Ferroptosis in infection, inflammation, and immunity. J Exp Med. (2021) 218. doi: 10.1084/jem.20210518

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Azziz R. Polycystic ovary syndrome. Obstet Gynecol. (2018) 132:321–36. doi: 10.1097/AOG.0000000000002698

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Andrisse S, Feng M, Wang Z, Awe O, Yu L, Zhang H, et al. Androgen-induced insulin resistance is ameliorated by deletion of hepatic androgen receptor in females. FASEB J. (2021) 35:e21921. doi: 10.1096/fj.202100961R

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Stone T, Stachenfeld NS. Pathophysiological effects of androgens on the female vascular system. Biol Sex Differ. (2020) 11:45. doi: 10.1186/s13293-020-00323-6

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Rodgers JL, Jones J, Bolleddu SI, Vanthenapalli S, Rodgers LE, Shah K, et al. Cardiovascular risks associated with gender and aging. J Cardiovasc Dev Dis. (2019) 6. doi: 10.3390/jcdd6020019

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Connelly PJ, Azizi Z, Alipour P, Delles C, Pilote L, Raparelli V. The importance of gender to understand sex differences in cardiovascular disease. Can J Cardiol. (2021) 37:699–710. doi: 10.1016/j.cjca.2021.02.005

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Veltman-Verhulst SM, van Rijn BB, Westerveld HE, Franx A, Bruinse HW, Fauser BC, et al. Polycystic ovary syndrome and early-onset preeclampsia: reproductive manifestations of increased cardiovascular risk. Menopause. (2010) 17:990–6. doi: 10.1097/gme.0b013e3181ddf705

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Gupta S, Nakabo S, Blanco LP, O'Neil LJ, Wigerblad G, Goel RR, et al. Sex differences in neutrophil biology modulate response to type I interferons and immunometabolism. Proc Natl Acad Sci U S A. (2020) 117:16481–91. doi: 10.1073/pnas.2003603117

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Trigunaite A, Dimo J, Jorgensen TN. Suppressive effects of androgens on the immune system. Cell Immunol. (2015) 294:87–94. doi: 10.1016/j.cellimm.2015.02.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: trained immunity, cardiovascular therapeutics, cardiovascular diseases, gender differences, novel targets

Citation: Yang WY, Nguyen B, Wu S, Yu J, Wang H and Yang X (2022) Editorial: Highlights for Cardiovascular Therapeutics in 2021 – Trained Immunity, Immunometabolism, Gender Differences of Cardiovascular Diseases, and Novel Targets of Cardiovascular Therapeutics. Front. Cardiovasc. Med. 9:892288. doi: 10.3389/fcvm.2022.892288

Received: 08 March 2022; Accepted: 25 March 2022;
Published: 27 April 2022.

Edited and reviewed by: Mark W. Feinberg, Brigham and Women's Hospital and Harvard Medical School, United States

Copyright © 2022 Yang, Nguyen, Wu, Yu, Wang and Yang. 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: Xiaofeng Yang, xfyang@temple.edu

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.