Next Article in Journal
Zebrafish as a Model for Cardiovascular and Metabolic Disease: The Future of Precision Medicine
Previous Article in Journal
Functions of Differentially Regulated miRNAs in Breast Cancer Progression: Potential Markers for Early Detection and Candidates for Therapy
Previous Article in Special Issue
Different Coactivator Recruitment to Human PPARα/δ/γ Ligand-Binding Domains by Eight PPAR Agonists to Treat Nonalcoholic Fatty Liver Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bridging Metabolic-Associated Steatotic Liver Disease and Cardiovascular Risk: A Potential Role for Ketogenesis

by
Rafael Suárez del Villar-Carrero
1,2,3,
Agustín Blanco
1,2,4,
Lidia Daimiel Ruiz
5,6,
Maria J. García-Blanco
1,2,7,8,
Ramón Costa Segovia
1,2,4,
Rocío García de la Garza
1,2,9 and
Diego Martínez-Urbistondo
1,2,9,*
1
Grupo de Riesgo Vascular, Sociedad Española de Medicina Interna (SEMI), 28016 Madrid, Spain
2
Grupo de Trabajo Prevención Secundaria y Alto Riesgo Vascular, Sociedad Española Arteriosclersosis (SEA), 08029 Barcelona, Spain
3
Servicio de Urgencias, Hospital Universitario HM Monteprincipe, 28660 Madrid, Spain
4
Unidad de Riesgo Cardiovascular, Departamento de Medicina Interna, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
5
Grupo de Estudio de Nutrigenómica y Nutrición Personalizada, IMDEA Alimentación, 28049 Madrid, Spain
6
CIBEROBN, Instituto Carlos III, 28029 Madrid, Spain
7
Servicio de Medicina Interna, Hospital Central de la Defensa Gómez Ulla, 28047 Madrid, Spain
8
Facultad de Medicina, Universidad de Alcalá, 28801 Alcalá de Henares (Madrid), Spain
9
Área de Medicina Vascular, Departamento de Medicina Interna, Clínica Universidad de Navarra, 28027 Madrid, Spain
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(3), 692; https://doi.org/10.3390/biomedicines12030692
Submission received: 6 February 2024 / Revised: 5 March 2024 / Accepted: 13 March 2024 / Published: 20 March 2024
(This article belongs to the Special Issue Metabolic- and Genetic-Associated Fatty Liver Diseases Volume II)

Abstract

:
The prevalence of cardiovascular diseases (CVDs) is a growing global health concern. Recent advances have demonstrated significant reductions in acute cardiovascular events through the management of modifiable cardiovascular risk factors. However, these factors are responsible for about 50% of the global cardiovascular disease burden. Considering that CVDs are one of the top mortality causes worldwide, the concept of residual cardiovascular risk is an important emerging area of study. Different factors have been proposed as sources of residual risk markers, including non-HDL particles characterization, as well as inflammation measured by serum and imaging technics. Among these, metabolic-associated steatotic liver disease (MASLD) remains controversial. Two opposing viewpoints contend: one positing that fatty liver disease merely reflects classical risk factors and thus adds no additional risk and another asserting that fatty liver disease independently impacts cardiovascular disease incidence. To address this dilemma, one hypothetical approach is to identify specific hepatic energy-yielding mechanisms and assess their impact on the cardiovascular system. Ketogenesis, a metabolic intermediate process particularly linked to energy homeostasis during fasting, might help to link these concepts. Ketogenic metabolism has been shown to vary through MASLD progression. Additionally, newer evidence supports the significance of circulating ketone bodies in cardiovascular risk prediction. Furthermore, ketogenic metabolism modification seems to have a therapeutic impact on cardiovascular and endothelial damage. Describing the relationship, if any, between steatotic liver disease and cardiovascular disease development through ketogenesis impairment might help to clarify MASLD’s role in cardiovascular risk. Furthermore, this evidence might help to solve the controversy surrounding liver steatosis impact in CVD and might lead to a more accurate risk assessment and therapeutic targets in the pursuit of precision medicine.

Graphical Abstract

1. Introduction

Cardiovascular diseases (CVD) represent a major concern in preventive health. An estimated 17.9 million people died from CVDs in 2019, representing 32% of global deaths worldwide. Of these deaths, 85% were due to heart attack and stroke. In the United States, CVD remains the leading cause of death for both men and women, accounting for approximately one in every four deaths [1]. According to the American Heart Association, in 2020, nearly half of U.S. adults (48%) were estimated to have some form of CVD, with the prevalence slightly higher in men (50.5%) than in women (47.3%) [2]. In Europe, the burden of CVD is similarly worrying, with cardiovascular diseases being the leading cause of death, accounting for 45% of all deaths in Europe and 37% in the European Union. The prevalence of CVD in European adults is estimated to be around 49% in men and 38% in women [3].
The prevention of CVD is a critical aspect of public health strategies, providing a Global action plan for the prevention and control of non-communicable diseases (NCD). This plan aims to reduce the number of premature deaths from NCDs by 25% by 2025 through different global targets [4]. Lifestyle modifications, such as adopting a balanced diet, engaging in regular physical activity, avoiding nicotine exposure, and maintaining a healthy weight, play a pivotal role in reducing the risk of CVD. These measures address key modifiable risk factors, including hypertension, dyslipidemia, diabetes, and obesity, which are known to significantly increase the likelihood of developing CVD. Together, the control of hypertension, dyslipidemia, dysglucemia, sleep care, physical activity performance, taking care about nutrition, avoiding tobacco, and maintaining a healthy weight are known as life’s essential eight [5]. Investigations of the pathogenesis of CVD have identified several potential pathways involving inflammation, endothelial function, atherosclerosis, cardiac stress and remodeling, hemostatic factors, microbiota, and epigenetics, among others [6,7,8,9,10].
At present, classic cardiovascular risk factors can only explain approximately 57% of cardiovascular events in women and 52% in men, accounting for a 10-year all-cause mortality of 22.2% and 19.1%, respectively [11]. This limitation highlights the growing importance of clinical and translational research in studying residual risk due to other characteristics. In this context, non-HDL molecules and the triglyceride content of cholesterol-carrying particles have demonstrated a significant and likely causal role in cardiovascular risk. Non-HDL cholesterol includes all atherogenic lipoproteins and is considered a better marker of risk than LDL cholesterol alone [12,13,14]. Elevated levels of non-HDL cholesterol are associated with an increased risk of atherosclerotic cardiovascular disease. Lipoprotein (a), or Lp (a), is another lipid-related risk factor receiving increased attention. Lp (a) is a unique lipoprotein particle with a structure similar to LDL cholesterol, but with an additional protein called apolipoprotein (a). Elevated levels of Lp (a) have been independently associated with increased risk of cardiovascular diseases, including coronary heart disease and stroke [15]. Lp (a) is considered a genetically determined risk factor, with concentrations largely unaffected by lifestyle changes or most lipid-lowering medications [16,17].
Local and systemic inflammation plays a significant role in the pathogenesis of CVD [18,19,20,21,22,23,24,25,26]. Hs-CRP has been widely recognized as a marker of systemic inflammation and an independent predictor of cardiovascular events. Furthermore, IL-1 and IL-6 are key cytokines involved in inflammatory processes and have been linked to atherosclerosis progression [19,20,21]. These findings underscore the need for a broader approach to cardiovascular risk assessment and management, encompassing both traditional and emerging risk factors. Identifying and targeting these residual risks could lead to more effective strategies for preventing cardiovascular diseases.

2. Liver Steatotic Disease as a Potential Residual Cardiovascular Risk Factor

The progressive increase in the global prevalence of hepatic steatosis disease raises the question of whether this condition might play a role in predicting residual cardiovascular risk [27]. Steatotic liver disease (SLD), previously known as non-alcoholic fatty liver disease (NAFLD), has become the most common cause of liver disease in developed countries today, as up to 30–40% of the global population may be affected by NAFLD [28,29]. This incidence and persistence of simple steatosis in patients is associated with the development of more advanced forms of the disease with a higher morbimortality, such as non-alcoholic steatohepatitis (NASH), cirrhosis, or the development of hepatocellular carcinoma [29]. Furthermore, the increasing prevalence of this condition among young individuals confers additional risk due to the prolonged duration of disease exposure [29]. However, the number of deaths directly attributable to liver disease itself is relatively limited [29].
Interestingly, steatotic liver disease is deeply linked to metabolic and cardiovascular disease [30]. Insulin resistance, a hallmark of metabolic syndrome, plays a central role in the development of NAFLD by promoting the accumulation of fat in the liver and exacerbating liver inflammation and damage [31,32,33]. Liver steatosis also contributes to alterations in lipid metabolism and glucose regulation (Figure 1) [33]. As research has advanced, the association between NAFLD and classic cardiovascular risk factors has become more evident, including hypertension, dyslipidemia, obesity, and type 2 diabetes mellitus [34]. Thus, the conceptualization of steatotic liver disease (SLD) has significantly progressed, with a particular focus on distinguishing between metabolic and alcohol-related factors while avoiding stigmatizing terms for patients. The term non-alcoholic fatty liver disease (NAFLD) has been re-evaluated due to concerns that it might overlook the nuances of alcohol’s role in liver steatosis [35]. Acknowledging that both excessive and moderate alcohol consumption can influence liver health, a more refined diagnostic approach has been advocated. This shift to the broader category of SLD allows for a more inclusive understanding, taking into account varying levels of alcohol intake. The new nomenclature distinguishes between excessive (Metabolic and Alcohol Steatotic Liver Disease—MetALD) and moderate or no alcohol intake (Metabolic-Associated Steatotic Liver Disease—MASLD) [35]. Although the repercussions on the disease epidemiology are still under evaluation, with some data pointing to a low reclassification capacity of the new definition [36], the inclusion of a metabolic disturbance basis on SLD categorization reaffirms a new paradigm in the comprehension of this systemic disease. However, the question remains open: Does MASLD contribute independently to cardiovascular residual risk beyond traditional factors?
Cardiovascular events are the most frequent cause of morbimortality in patients with NAFLD [29]. Consequently, interest in studying the potential causal relationship between SLD and cardiovascular disease has surged [27,37,38]. Initially, some epidemiological assessments pointed out that hepatic lipid accumulation mirrors an individual’s metabolic milieu but does not represent an independent risk factor for CV events [39,40]. This concept is also included in the current European Society of Cardiology guidelines for cardiovascular risk prevention [41]. In these cohorts, although some effects of steatosis and liver fibrosis could be addressed, this prediction capacity was lost when further adjustments were applied. However, other investigations suggest that NAFLD might proffer an additional, independent prognostic value for cardiovascular events, including a biopsy-based NAFLD stage correlation with the probability of cardiovascular arrest [42] and a mendelian randomization study, which found an independent link between NAFLD and CVD when gene function adjustments were included [43]. Nevertheless, biopsy or genetic studies are too risky, expensive, and complicated to be efficient in the risk factor scenario [44]. Thus, hepatic steatosis may be considered a surrogate metric for metabolic risk when evaluated through non-invasive techniques such as ultrasound, MRI, and transient elastography [45,46] and even validated serologic indices [47]. Several epidemiological studies have explored the association between non-invasively assessed SLD and metabolic and cardiovascular risk regarding quality of life [48], as well as the impact of lifestyle modification [49], incidence of T2DM [50], and incidence of cardiovascular events [51]. Various prospective cohort analyses and meta-analyses, grounded in liver biopsy results [42] and non-invasive biomarkers [52,53], have buttressed this hypothesis and precipitated a specific AHA statement on this subject [27] (Table 1).
Although further meticulous epidemiological research should be performed in the field, the pursuit of specific hepatic metabolic pathways linking MASLD to CVD is settled. In this context, the production of ketone bodies through the Randle cycle presents several interesting features: (i) liver exclusiveness, (ii) ketone synthesis variation along the MASLD spectrum [75], and (iii) potential prediction capacity in cardiovascular disease [76]. Ketogenesis predominantly occurs in the liver, where fatty acids are converted into ketone bodies, namely acetoacetate, beta-hydroxybutyrate, and acetone. Ketone bodies exhibit biphasic alterations in MASLD patients, escalating during initial disease phases and waning during advanced stages [75]. Ketone bodies have been related to CVD in two ways. On the one hand, their reducing effect on oxidative stress and inflammation was suggested to protect from CVD [77,78]; on the other hand, they were found to be associated with cardiovascular risk in CVD-naïve patients [76]. Thus, further elucidation of the role of ketogenesis may contribute to the understanding of the link between SLD and cardiovascular risk. Thus, assessing ketogenesis may contribute to the understanding of the link between SLD and cardiovascular risk.

3. Metabolic Associated Steatotic Liver Disease Role in Ketogenesis Impairment

Ketogenesis is initiated in the mitochondrial matrix of hepatocytes and entails a cascade of enzymatic reactions, catalyzed by lipases, enzymes of betaoxidation, and HMG-CoA synthase [79]. The generated ketone bodies acetoacetate, betahydroxybutyrate (BHB) and acetone can serve as energy source, particularly furnishing essential energy to the brain and the cardiovascular system during periods of fasting or carbohydrate restriction [80]. The influence of MASLD on ketogenesis is bi-phasic. Initially, ketone body production is augmented due to an increase in beta-oxidation while other hepatic features remain clinically normal [81]. Subsequently, as steatohepatitis and fibrosis unfold, production is reduced due to a decrease in HMG-CoA activity [80,82] (Figure 2).

3.1. Ketogenesis Impairment in MASLD

In the early stage of MASLD, hepatic lipogenesis is increased, resulting in liver steatosis. This is associated with increased fatty acid uptake and synthesis [83] and insufficient fatty acid conversion to ketone bodies [84]. This impaired ketogenesis could deleteriously impact mitochondrial functionality, given the intimate relationship between the ketogenesis pathway and mitochondrial integrity [85]. The accumulation of fatty acids within hepatocytes can provoke mitochondrial dysfunction and oxidative stress, igniting a vicious cycle that exacerbates hepatic lipid accumulation, induces hepatic injury, and propels inflammation and fibrosis, favoring the progression from simple steatosis to metabolic-associated steatohepatitis (MASH) [86]. Ketogenesis can be influenced by multiple factors, including insulin resistance, nutrient availability, and signaling cascades across various cellular receptors and pathways. Insulin resistance, a pivotal actor in SLD onset and progression, notably exerts a suppressive impact upon ketogenesis [87]. Additionally, the peroxisome proliferator-activated receptor-alpha (PPARα), instrumental in regulating fatty acid metabolism, becomes substantively involved in the regulatory mechanisms of this process comprising hepatic fatty acid and plasma lipoprotein metabolism during nutritional transition and the regulation of hepatic inflammatory response [88].
When NAFLD advances to NASH, the interplay between impaired ketogenesis and hepatic lipid metabolism diversifies, adding inflammation and cellular injury to lipid accumulation. In this context, impaired ketogenesis may intensify hepatic lipid accumulation, oxidative stress, and mitochondrial dysfunction, thereby contributing to hepatocellular injury and inflammation [75]. Then, as the pathophysiological process evolves through the progression toward fibrosis and cirrhosis, ketone production is diminished. This metabolic disturbance might affect systemic energy homeostasis especially in the brain and heart [79], which are the main targets of CVD. Thus, circulating ketone bodies might serve as indirect hepatic function markers with specific cardiovascular implications, but these should be completely understood, compelling the interaction between SLD progression, ketone body production, and endothelial dysfunction (Figure 3).

3.2. Ketogenesis Impairment in Cardiovascular Disease

Beta-hydroxybutyrate is the most abundant in plasma, constituting more than 90% of the total ketone bodies, though acetoacetate and acetone also occur in blood samples [80]. Particularly during glucose-limiting conditions such as prolonged fasting or rigorous physical exertion, these ketone bodies are indispensable, ensuring energy provision predominantly for the cerebrum and myocardium [80]. The affinity of these fuel transfer metabolites for the major organs involved in cardiovascular events is still intriguing. Emerging evidence underscores the advantageous impact of ketone bodies on cardiac metabolic adaptability under pathological scenarios like heart failure (HF) [89,90,91,92]. Hence, during energy deficits, the malfunctioning heart may escalate ketone utilization, acting as a compensatory response to its amplified energy demands [91,92]. Yet the question remains: does reduced ketogenesis improve or deteriorate CVD prognosis?
Preliminary findings suggest that ketone synthesis in apparently healthy individuals is associated with higher cardiovascular risk. Inadequate ketone body synthesis might aggravate cardiac malfunction by depriving the heart muscle of an essential energy source during metabolic distress periods [91,92]. A similar theory has been advanced concerning arteriosclerosis [93].
Conversely, dysregulated ketone metabolism and heightened BHB concentrations, frequently observed in diabetic cohorts, might elevate cardiovascular risk by fostering oxidative stress and endothelial malfunction [94]. It appears that maintaining equilibrium in ketone metabolism, evading both deficiency and surplus, is imperative for cardiovascular integrity [95]. Some investigations, specifically from the MESA cohort, have appraised the nexus between circulating ketone bodies and CVD occurrence. Preliminary results show that augmented endogenous ketone body levels correlate with a higher incidence and mortality rate of CVD among populations without prior events due to CVD. This potentially positions ketone bodies as prospective biomarkers for cardiovascular risk evaluation [76].
Thus, the interpretation of ketone bodies in the clinical setting remains unclear. On the one hand, further ketone synthesis in apparently healthy individuals leads to higher cardiovascular risk, while on the other hand, ketone body insufficiency in diseased patients seems to be associated with further morbidity and mortality. Hence, different hypotheses might arise from this controversy: Are ketone bodies an early biomarker of cardiovascular disease? This approach might mean that people generating a relative excess of ketones while being apparently healthy are adapting to a thinner metabolic health equilibrium through flexible homeostatic resources such as ketone bodies. However, an increased ketogenesis could be a consequence of other metabolic disturbances such as SLD progression, providing further cardiovascular and metabolic risk through other pathways such as dyslipidemia or serum glucose control, but then, how is mild ketone body increase associated with a better prognosis in secondary cardiovascular prevention? Although this fact could be elucidated by a higher capacity of these patients to provide energetic balance through accessory pathways, this would not correlate with the apparent harmful effect of liver steatosis progression, which is linked to a higher production of ketones during the intermediate phase of MASH. In addition to its role in energy supply, BHB operates as an epigenetic regulator, curbing histone deacetylases and subsequently modifying gene transcription [96]. This modulatory capability suggests that ketogenesis perturbations altering BHB concentrations could potentially impact cardiac gene expression, introducing another layer of complexity to cardiovascular pathophysiology. Therefore, an in-depth exploration of the relationships between hepatic ketogenesis and cardiovascular health is key to uncovering clinically significant ties between SLD and CVD.

3.3. Impaired Ketogenesis: Bridging Non-Alcoholic Fatty Liver Disease (NAFLD) and Cardiovascular Disease (CVD)

Mediation analyses to prove the connection between SLD and CVD through circulating ketone bodies have been appraised. Post A et al. investigated the association between NAFLD and circulating ketone bodies in a cohort of 6297 participants and determined the extent to which NAFLD and circulating ketone bodies were associated with all-cause mortality [97]. An elevated FLI as a marker of liver steatosis was independently associated with an increased risk of mortality. Higher total ketone bodies were also associated with an increased mortality risk. Mediation analysis suggested that the association of elevated FLI with all-cause mortality was in part mediated by ketone bodies (proportion mediated: 10%, p < 0.001).
This association might be mechanistically rooted in impaired lipid metabolism, which fosters dyslipidemia, typified by an augmentation of triglycerides and low-density lipoprotein cholesterol (LDL-C), and a decrease in high-density lipoprotein cholesterol (HDL-C) levels [98]. Furthermore, the intricate web intertwining NAFLD and CVD might be influenced by inflammatory pathways, especially in the context of the more aggressive non-alcoholic steatohepatitis (NASH). This latter condition is associated with systemic inflammation and a surge in pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), which are pivotal in atherosclerosis and CVD pathogenesis [99,100]. Moreover, the potential role of impaired ketogenesis in modulating nitric oxide (NO) bioavailability and, consequently, impacting endothelial function, which is pivotal for vascular well-being, cannot be ignored. A decrease in endothelial NO production is intrinsically linked with endothelial dysfunction, a harbinger of atherosclerosis and subsequent CVD [101].
Intriguingly, the nuanced effect of impaired ketogenesis is somewhat illuminated by the impact of externally provided ketone production on a myriad of markers like dyslipemia [102,103], dysglycemia [104,105,106], appetite and obesity [107], blood pressure [108], meta inflammation [78,109], and endothelial dysfunction [77,110], providing regulation of insulin and glucagon role in beta-oxidation, glycolysis, gluconeogenesis, and NO synthase in the hemodynamic control of vascular system. These findings, as well as the predictive value of circulating ketone bodies for CVD, point to an independent effect of impaired ketogenesis on CVD and a potential benefit of circulating ketone bodies for assessing cardiovascular risk (Figure 3).
Some novel therapeutic tools to treat metabolic disturbances, such as sodium-glucose co-transporter 2 inhibitors (SGLT2i), might also help to elucidate the consequential impact of ketogenesis in CVD. By obstructing glucose reabsorption in proximal renal tubules and by amplifying urinary glucose excretion, SGLT2i not only mitigates hyperglycemia but also reduces cardiovascular events and mortality in diabetic patients with close monitoring to avoid euglycemic diabetic ketoacidosis [111,112,113]. Furthermore, SGLT2i might augment glucagon secretion [114]. On a contrasting note, SGLT2i, while inducing a mild ketosis, paves the way for several metabolic adaptations, potentially conferring assorted benefits. For example, mild ketosis, propelled by an elevation in circulating ketone bodies like BHB, could offer an alternative energy substrate for various tissues when glucose utilization is jeopardized [115]. BHB also plays roles in signaling activities, potentially exerting anti-inflammatory and anti-aging effects by inhibiting the NLRP3 inflammasome and reducing oxidative stress [86,116,117,118]. For patients with type 2 diabetes, the utilization of ketone bodies as an alternate fuel might aid in safeguarding cardiac and renal functions. These benefits have translated into a potential role of ketogenesis in SGLT2i protection against heart failure, decompensation, and kidney failure [116,117,118].
The clinical approach to cardiovascular disease represents na urgent for the research of new pathways that may elucidate the connection between different pathogenic mechanisms, deriving causal biomarkers and potential therapeutic targets. The finding of new pathways may help to finally control the cardiovascular pandemic. Thus, the available pathophysiological and epidemiological data hint at the impact of impaired ketogenesis as a potential mediator between SLD and cardiovascular disease. This statement could be set based on liver exclusiveness in ketone production, the fluctuations in ketogenesis according to MASLD status, and evidence for the role of ketone bodies in cardiovascular disease. These concepts might pinpoint impaired ketogenesis as a distinct biological mechanism linking MASLD with CVD. For these reasons, the authors consider that future research should support the effort to develop and perform specific studies to furnish mechanistic evidence of this connection. Subsequent evidence may aid in refining our understanding of residual cardiovascular risk, providing further knowledge of both processes, and thereby enhancing diagnostic and therapeutic precision and fostering the progress of precision medicine.

Author Contributions

Conceptualization, D.M.-U., R.G.d.l.G. and R.S.d.V.-C.; Writing-original draft preparation, R.S.d.V.-C., A.B., L.D.R., M.J.G.-B., R.C.S., R.G.d.l.G. and D.M.-U.; Writing- review and editing, R.S.d.V.-C., A.B., L.D.R., M.J.G.-B., R.C.S., R.G.d.l.G. and D.M.-U.; Supervision, D.M.-U.; All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external founding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. Cardiovascular Diseases (CVDs). 2021. Available online: https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds) (accessed on 30 January 2024).
  2. Tsao, C.W.; Aday, A.W.; Almarzooq, Z.I.; Alonso, A.; Beaton, A.Z.; Bittencourt, M.S.; Boehme, A.K.; Buxton, A.E.; Carson, A.P.; Commodore-Mensah, Y.; et al. Heart Disease and Stroke Statistics-2022 Update: A Report from the American Heart Association. Circulation 2022, 145, E153–E639. [Google Scholar] [CrossRef]
  3. Timmis, A.; Vardas, P.; Townsend, N.; Torbica, A.; Katus, H.; De Smedt, D.; Gale, C.P.; Maggioni, A.P.; Petersen, S.E.; Huculeci, R.; et al. European Society of Cardiology: Cardiovascular disease statistics 2021. Eur. Heart J. 2022, 43, 716–799. [Google Scholar] [CrossRef]
  4. 2013–2020 Global Action Plan for the Prevention and Control of Noncommunicable Diseases. 2013. Available online: www.who.int (accessed on 30 January 2024).
  5. Lloyd-Jones, D.M.; Allen, N.B.; Anderson, C.A.M.; Black, T.; Brewer, L.C.; Foraker, R.E.; Grandner, M.A.; Lavretsky, H.; Marma Perak, A.; Sharma, G.; et al. Life’s Essential 8: Updating and Enhancing the American Heart Association’s Construct of Cardiovascular Health: A Presidential Advisory from the American Heart Association. Circulation 2022, 146, E18–E43. [Google Scholar] [CrossRef]
  6. Gaye, B.; Tafflet, M.; Arveiler, D.; Montaye, M.; Wagner, A.; Ruidavets, J.B.; Ke, F.; Evans, A.; Amouyel, P.; Ferrieres, J.; et al. Ideal Cardiovascular Health and Incident Cardiovascular Disease: Heterogeneity across Event Subtypes and Mediating Effect of Blood Biomarkers: The PRIME Study. J. Am. Heart Assoc. 2017, 6, e006389. [Google Scholar] [CrossRef]
  7. Xanthakis, V.; Enserro, D.M.; Murabito, J.M.; Polak, J.F.; Wollert, K.C.; Januzzi, J.L.; Wang, T.J.; Tofler, G.; Vasan, R. Ideal cardiovascular health: Associations with biomarkers and subclinical disease and impact on incidence of cardiovascular disease in the Framingham Offspring Study. Circulation 2014, 130, 1676–1683. [Google Scholar] [CrossRef]
  8. Polonsky, T.S.; Ning, H.; Daviglus, M.L.; Liu, K.; Burke, G.L.; Cushman, M.; Eng, J.; Folsom, A.R.; Lutsey, P.L.; Nettleton, J.A.; et al. Association of Cardiovascular Health with Subclinical Disease and Incident Events: The Multi-Ethnic Study of Atherosclerosis. J. Am. Heart Assoc. 2017, 6, e004894. [Google Scholar] [CrossRef] [PubMed]
  9. Pottinger, T.D.; Khan, S.S.; Zheng, Y.; Zhang, W.; Tindle, H.A.; Allison, M.; Wells, G.; Shadyab, A.H.; Nassir, R.; Warsinger Martin, L.; et al. Association of cardiovascular health and epigenetic age acceleration. Clin. Epigenet. 2021, 13, 42. [Google Scholar] [CrossRef] [PubMed]
  10. Joyce, B.T.; Gao, T.; Zheng, Y.; Ma, J.; Hwang, S.J.; Liu, L.; Nannini, D.; Horvath, S.; Lu, A.T.; Bai Allen, N.; et al. Epigenetic Age Acceleration Reflects Long-Term Cardiovascular Health. Circ. Res. 2021, 129, 770–781. [Google Scholar] [CrossRef] [PubMed]
  11. Magnussen, C.; Ojeda, F.M.; Leong, D.P.; Alegre-Diaz, J.; Amouyel, P.; Aviles-Santa, L.; De Bacquer, D.; Ballantyne, C.M.; Bernabe-Ortiz, A.; Bobak, M.; et al. Global Impact of Modifiable Risk Factors on Cardiovascular Disease and Mortality. N. Engl. J. Med. 2023, 389, 1273. [Google Scholar] [PubMed]
  12. Hegele, R.A.; Ginsberg, H.N.; Chapman, M.J.; Nordestgaard, B.G.; Kuivenhoven, J.A.; Averna, M.; Borén, J.; Bruckert, E.; Catapano, A.L.; Descamps, O.S.; et al. The polygenic nature of hypertriglyceridaemia: Implications for definition, diagnosis, and management. Lancet Diabetes Endocrinol. 2014, 2, 655–666. [Google Scholar] [CrossRef]
  13. Laufs, U.; Parhofer, K.G.; Ginsberg, H.N.; Hegele, R.A. Clinical review on triglycerides. Eur. Heart J. 2020, 41, 99–109. [Google Scholar] [CrossRef]
  14. Raja, V.; Aguiar, C.; Alsayed, N.; Chibber, Y.S.; Elbadawi, H.; Ezhov, M.; Hermans, M.P.; Chandra Pandey, R.; Ray, K.K.; Tokgözoglu, L.; et al. Non-HDL-cholesterol in dyslipidemia: Review of the state-of-the-art literature and outlook. Atherosclerosis 2023, 383, 117312. [Google Scholar] [CrossRef] [PubMed]
  15. Willeit, P.; Yeang, C.; Moriarty, P.M.; Tschiderer, L.; Varvel, S.A.; McConnell, J.P.; Tsimikas, S. Low-Density Lipoprotein Cholesterol Corrected for Lipoprotein(a) Cholesterol, Risk Thresholds, and Cardiovascular Events. J. Am. Heart Assoc. 2020, 9, 16318. [Google Scholar] [CrossRef]
  16. Trinder, M.; DeCastro, M.L.; Azizi, H.; Cermakova, L.; Jackson, L.M.; Frohlich, J.; John Mancini, G.B.; Francis, G.A.; Brunham, L.R. Ascertainment Bias in the Association between Elevated Lipoprotein(a) and Familial Hypercholesterolemia. J. Am. Coll. Cardiol. 2020, 75, 2682–2693. [Google Scholar] [CrossRef]
  17. Langsted, A.; Kamstrup, P.R.; Benn, M.; Tybjærg-Hansen, A.; Nordestgaard, B.G. High lipoprotein(a) as a possible cause of clinical familial hypercholesterolaemia: A prospective cohort study. Lancet Diabetes Endocrinol. 2016, 4, 577–587. [Google Scholar] [CrossRef] [PubMed]
  18. Ross, R. Atherosclerosis—An inflammatory disease. N. Engl. J. Med. 1999, 340, 302–309. [Google Scholar] [CrossRef] [PubMed]
  19. Swerdlow, D.I.; Holmes, M.V.; Kuchenbaecker, K.B.; Engmann, J.E.L.; Shah, T.; Sofat, R.; Guo, Y.; Chung, C.; Peasey, A.; Pfister, T.; et al. The interleukin-6 receptor as a target for prevention of coronary heart disease: A mendelian randomisation analysis. Lancet 2012, 379, 1214–1224. [Google Scholar]
  20. Ridker, P.M.; Hennekens, C.H.; Buring, J.E.; Rifai, N. C-reactive protein and other markers of inflammation in the prediction of cardiovascular disease in women. N. Engl. J. Med. 2000, 342, 836–843. [Google Scholar] [CrossRef]
  21. Antonopoulos, A.S.; Angelopoulos, A.; Papanikolaou, P.; Simantiris, S.; Oikonomou, E.K.; Vamvakaris, K.; Koumpoura, A.; Farmaki, M.; Trivella, M.; Vlachopoulos, C.; et al. Biomarkers of Vascular Inflammation for Cardiovascular Risk Prognostication: A Meta-Analysis. JACC Cardiovasc Imaging 2022, 15, 460–471. [Google Scholar] [CrossRef]
  22. Antoniades, C.; Antonopoulos, A.S.; Deanfield, J. Imaging Residual Inflammatory Cardiovascular Risk. Available online: https://academic.oup.com/eurheartj/article/41/6/748/5533079 (accessed on 30 January 2024).
  23. Figueroa, A.L.; Abdelbaky, A.; Truong, Q.A.; Corsini, E.; MacNabb, M.H.; Lavender, Z.R.; Lawler, M.A.; Grinspoon, S.K.; Brady, T.J.; Nasir, K.; et al. Measurement of Arterial Activity on Routine FDG PET/CT Images Improves Prediction of Risk of Future CV Events. JACC Cardiovasc. Imaging 2013, 6, 1250–1259. [Google Scholar] [CrossRef]
  24. Ibanez, B.; Fernández-Ortiz, A.; Fernández-Friera, L.; García-Lunar, I.; Andrés, V.; Fuster, V. Progression of Early Subclinical Atherosclerosis (PESA) Study: JACC Focus Seminar 7/8. J. Am. Coll. Cardiol. 2021, 78, 156–179. [Google Scholar] [CrossRef]
  25. Fernández-Friera, L.; Fuster, V.; López-Melgar, B.; Oliva, B.; Sánchez-González, J.; Macías, A.; Pérez-Asenjo, B.; Zamudio, D.; Alonso-Farto, J.C.; España, S.; et al. Vascular Inflammation in Subclinical Atherosclerosis Detected by Hybrid PET/MRI. J. Am. Coll. Cardiol. 2019, 73, 1371–1382. [Google Scholar] [CrossRef]
  26. Martínez-Urbistondo, D.; Beltrán, A.; Beloqui, O.; Huerta, A. The neutrophil-to-lymphocyte ratio as a marker of systemic endothelial dysfunction in asymptomatic subjects. Nefrologia 2016, 36, 397–403. [Google Scholar] [CrossRef]
  27. Duell, P.B.; Welty, F.K.; Miller, M.; Chait, A.; Hammond, G.; Ahmad, Z.; Cohen, D.E.; Horton, J.D.; Pressman, G.S.; Toth, P.P.; et al. Nonalcoholic Fatty Liver Disease and Cardiovascular Risk: A Scientific Statement from the American Heart Association. Arterioscler. Thromb. Vasc. Biol. 2022, 42, e168–e185. [Google Scholar] [CrossRef]
  28. Younossi, Z.; Tacke, F.; Arrese, M.; Chander Sharma, B.; Mostafa, I.; Bugianesi, E.; Wai-Sun Wong, V.; Yilmaz, Y.; George, J.; Fan, J.; et al. Global Perspectives on Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis. Hepatology 2019, 69, 2672–2682. [Google Scholar] [CrossRef]
  29. Younossi, Z.; Anstee, Q.M.; Marietti, M.; Hardy, T.; Henry, L.; Eslam, M.; George, J.; Bugianesi, E. Global burden of NAFLD and NASH: Trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 11–20. [Google Scholar] [CrossRef]
  30. Samuel, V.T.; Shulman, G.I. Nonalcoholic Fatty Liver Disease as a Nexus of Metabolic and Hepatic Diseases. Cell Metab. 2018, 27, 22–41. [Google Scholar] [CrossRef]
  31. Samuel, V.T.; Shulman, G.I. The pathogenesis of insulin resistance: Integrating signaling pathways and substrate flux. J. Clin. Investig. 2016, 126, 12–22. [Google Scholar] [CrossRef]
  32. Samuel, V.T.; Liu, Z.X.; Qu, X.; Elder, B.D.; Bilz, S.; Befroy, D.; Romanelli, A.J.; Shulman, G.I. Mechanism of hepatic insulin resistance in non-alcoholic fatty liver disease. J. Biol. Chem. 2004, 279, 32345–32353. [Google Scholar] [CrossRef]
  33. Samuel, V.T.; Liu, Z.X.; Wang, A.; Beddow, S.A.; Geisler, J.G.; Kahn, M.; Zhang, X.-m.; Monia, B.P.; Bhanot, S.; Shulman, G.I. Inhibition of protein kinase Cepsilon prevents hepatic insulin resistance in nonalcoholic fatty liver disease. J. Clin. Investig. 2007, 117, 739–745. [Google Scholar] [CrossRef]
  34. Long, M.T.; Zhang, X.; Xu, H.; Liu, C.T.; Corey, K.E.; Chung, R.T.; Loomba, R.; Benjamin, E.J. Hepatic Fibrosis Associates with Multiple Cardiometabolic Disease Risk Factors: The Framingham Heart Study. Hepatology 2021, 73, 548–559. [Google Scholar] [CrossRef]
  35. Rinella, M.E.; Lazarus, J.V.; Ratziu, V.; Francque, S.M.; Sanyal, A.J.; Kanwal, F.; Romero, D.; Abdelmalek, M.F.; Anstee, Q.M.; Arab, J.P.; et al. A multisociety Delphi consensus statement on new fatty liver disease nomenclature. J. Hepatol. 2023, 79, 1542–1556. [Google Scholar] [CrossRef]
  36. Ciardullo, S.; Carbone, M.; Invernizzi, P.; Perseghin, G. Exploring the landscape of steatotic liver disease in the general US population. Liver Int. 2023, 43, 2425–2433. [Google Scholar] [CrossRef]
  37. Zhou, X.D.; Targher, G.; Byrne, C.D.; Somers, V.; Kim, S.U.; Chahal, C.A.A.; Wong, V.W.; Cai, J.; Shapiro, M.D.; Eslam, M.; et al. An international multidisciplinary consensus statement on MAFLD and the risk of CVD. Hepatol. Int. 2023, 17, 773–791. [Google Scholar] [CrossRef]
  38. Hutchison, A.L.; Tavaglione, F.; Romeo, S.; Charlton, M. Endocrine aspects of metabolic dysfunction-associated steatotic liver disease (MASLD): Beyond insulin resistance. J. Hepatol. 2023, 79, 1524–1541. [Google Scholar] [CrossRef]
  39. Ciardullo, S.; Cannistraci, R.; Mazzetti, S.; Mortara, A.; Perseghin, G. Nonalcoholic Fatty Liver Disease, Liver Fibrosis and Cardiovascular Disease in the Adult US Population. Front. Endocrinol. 2021, 12, 711484. [Google Scholar] [CrossRef]
  40. Alexander, M.; Loomis, A.K.; Van Der Lei, J.; Duarte-Salles, T.; Prieto-Alhambra, D.; Ansell, D.; Pasqua, A.; Lapi, F.; Rijnbeek, P.; Mosseveld, M.; et al. Non-alcoholic fatty liver disease and risk of incident acute myocardial infarction and stroke: Findings from matched cohort study of 18 million European adults. BMJ 2019, 367, l5367. [Google Scholar] [CrossRef]
  41. Visseren, F.; Mach, F.; Smulders, Y.M.; Carballo, D.; Koskinas, K.C.; Bäck, M.; Benetos, A.; Biffi, A.; Boavida, J.M.; Capodanno, D.; et al. 2021 ESC Guidelines on cardiovascular disease prevention in clinical practice. Eur. Heart J. 2021, 42, 3227–3337. [Google Scholar] [CrossRef]
  42. Simon, T.G.; Roelstraete, B.; Hagström, H.; Sundström, J.; Ludvigsson, J.F. Non-alcoholic fatty liver disease and incident major adverse cardiovascular events: Results from a nationwide histology cohort. Gut 2022, 71, 1867–1875. [Google Scholar] [CrossRef]
  43. Ren, Z.; Simons, P.I.H.G.; Wesselius, A.; Stehouwer, C.D.A.; Brouwers, M.C.G.J. Relationship between NAFLD and coronary artery disease: A Mendelian randomization study. Hepatology 2023, 77, 230–238. [Google Scholar] [CrossRef]
  44. Marchesini, G.; Day, C.P.; Dufour, J.F.; Canbay, A.; Nobili, V.; Ratziu, V.; Tilg, H.; Roden, M.; Gastaldelli, A.; Yki-Järvinen, H.; et al. EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. J. Hepatol. 2016, 64, 1388–1402. [Google Scholar] [CrossRef]
  45. Pennisi, G.; Enea, M.; Romero-Gomez, M.; Viganò, M.; Bugianesi, E.; Wong, V.W.S.; Fracanzani, A.L.; Sebastiani, G.; Boursier, J.; Berzigotti, A.; et al. Liver-related and extrahepatic events in patients with non-alcoholic fatty liver disease: A retrospective competing risks analysis. Aliment. Pharmacol. Ther. 2022, 55, 604–615. [Google Scholar] [CrossRef]
  46. Castera, L.; Friedrich-Rust, M.; Loomba, R. Noninvasive Assessment of Liver Disease in Patients with Nonalcoholic Fatty Liver Disease. Gastroenterology 2019, 156, 1264–1281.e4. [Google Scholar] [CrossRef]
  47. Perez-Diaz-Del-Campo, N.; Martínez-Urbistondo, D.; Bugianesi, E.; Martínez, J.A. Diagnostic scores and scales for appraising Nonalcoholic fatty liver disease and omics perspectives for precision medicine. Curr. Opin. Clin. Nutr. Metab. Care 2022, 25, 285–291. [Google Scholar] [CrossRef]
  48. Martinez-Urbistondo, D.; del Villar, R.S.; Argemí, J.; Daimiel, L.; Ramos-López, O.; San-Cristobal, R.; Villares, P.; Martinez, J.A. Antioxidant Lifestyle, Co-Morbidities and Quality of Life Empowerment Concerning Liver Fibrosis. Antioxidants 2020, 9, 1125. [Google Scholar] [CrossRef]
  49. Martínez-Urbistondo, D.; San Cristóbal, R.; Villares, P.; Martínez-González, M.A.; Babio, N.; Corella, D.; del Val, J.L.; Ordovás, J.M.; Alonso-Gómez, A.M.; Wärnberg, J.; et al. Role of NAFLD on the Health Related QoL Response to Lifestyle in Patients with Metabolic Syndrome: The PREDIMED Plus Cohort. Front. Endocrinol. 2022, 13, 868795. [Google Scholar] [CrossRef]
  50. Martinez-Urbistondo, D.; Huerta, A.; Navarro-González, D.; Sánchez-Iñigo, L.; Fernandez-Montero, A.; Landecho, M.F.; Martinez, J.A.; Pastrana-Delgado, J.C. Estimation of fatty liver disease clinical role on glucose metabolic remodelling phenotypes and T2DM onset. Eur. J. Clin. Investig. 2023, 53, e14036. [Google Scholar] [CrossRef]
  51. Martinez-Urbistondo, D.; D’Avola, D.; Navarro-González, D.; Sanchez-Iñigo, L.; Fernandez-Montero, A.; Perez-Diaz-del-Campo, N.; Bugianesi, E.; Martinez, J.A.; Pastrana, J.C. Interactive Role of Surrogate Liver Fibrosis Assessment and Insulin Resistance on the Incidence of Major Cardiovascular Events. J. Clin. Med. 2022, 11, 5190. [Google Scholar] [CrossRef]
  52. Mantovani, A.; Csermely, A.; Petracca, G.; Beatrice, G.; Corey, K.E.; Simon, T.G.; Byrne, C.D.; Targher, G. Non-alcoholic fatty liver disease and risk of fatal and non-fatal cardiovascular events: An updated systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 2021, 6, 903–913. [Google Scholar] [CrossRef]
  53. Mantovani, A.; Byrne, C.D.; Bonora, E.; Targher, G. Nonalcoholic Fatty Liver Disease and Risk of Incident Type 2 Diabetes: A Meta-analysis. Diabetes Care 2018, 41, 372–382. [Google Scholar] [CrossRef]
  54. Stepanova, M.; Younossi, Z.M. Independent association between nonalcoholic fatty liver disease and cardiovascular disease in the US population. Clin. Gastroenterol. Hepatol. 2012, 10, 646–650. [Google Scholar] [CrossRef]
  55. Haring, R.; Wallaschofski, H.; Nauck, M.; Dörr, M.; Baumeister, S.E.; Völzke, H. Ultrasonographic hepatic steatosis increases prediction of mortality risk from elevated serum gamma-glutamyl transpeptidase levels. Hepatology 2009, 50, 1403–1411. [Google Scholar] [CrossRef]
  56. Kim, D.; Choi, S.Y.; Park, E.H.; Lee, W.; Kang, J.H.; Kim, W.; Kim, Y.J.; Yoon, J.-H.; Jeong, S.-H.; Lee, D.H.; et al. Nonalcoholic fatty liver disease is associated with coronary artery calcification. Hepatology 2012, 56, 605–613. [Google Scholar] [CrossRef]
  57. Targher, G.; Bertolini, L.; Padovani, R.; Rodella, S.; Tessari, R.; Zenari, L.; Day, C.; Arcaro, G. Prevalence of nonalcoholic fatty liver disease and its association with cardiovascular disease among type 2 diabetic patients. Diabetes Care 2007, 30, 1212–1218. [Google Scholar] [CrossRef]
  58. Tsutsumi, T.; Eslam, M.; Kawaguchi, T.; Yamamura, S.; Kawaguchi, A.; Nakano, D.; Koseki, M.; Yoshinaga, S.; Takahashi, H.; Anzai, K.; et al. MAFLD better predicts the progression of atherosclerotic cardiovascular risk than NAFLD: Generalized estimating equation approach. Hepatol. Res. 2021, 51, 1115–1128. [Google Scholar] [CrossRef]
  59. Hamaguchi, M.; Kojima, T.; Takeda, N.; Nagata, C.; Takeda, J.; Sarui, H.; Kawahito, Y.; Yoshida, N.; Suetsugu, A.; Kato, T.; et al. Nonalcoholic fatty liver disease is a novel predictor of cardiovascular disease. World J. Gastroenterol. 2007, 13, 1579–1584. [Google Scholar] [CrossRef]
  60. Yoshitaka, H.; Hamaguchi, M.; Kojima, T.; Fukuda, T.; Ohbora, A.; Fukui, M. Nonoverweight nonalcoholic fatty liver disease and incident cardiovascular disease: A post hoc analysis of a cohort study. Medicine 2017, 96, e6712. [Google Scholar] [CrossRef]
  61. Wong, V.W.S.; Wong, G.L.H.; Yip, G.W.K.; Lo, A.O.S.; Limquiaco, J.; Chu, W.C.W.; Chim, A.M.-L.; Yu, C.M.; Yu, J.; Chan, F.K.-L.; et al. Coronary artery disease and cardiovascular outcomes in patients with non-alcoholic fatty liver disease. Gut 2011, 60, 1721–1727. [Google Scholar] [CrossRef]
  62. Santos, R.D.; Nasir, K.; Conceição, R.D.; Sarwar, A.; Carvalho, J.A.M.; Blumenthal, R.S. Hepatic steatosis is associated with a greater prevalence of coronary artery calcification in asymptomatic men. Atherosclerosis 2007, 194, 517–519. [Google Scholar] [CrossRef]
  63. Mantovani, A.; Mingolla, L.; Rigolon, R.; Pichiri, I.; Cavalieri, V.; Zoppini, G.; Lippi, G.; Bonora, E.; Targher, G. Nonalcoholic fatty liver disease is independently associated with an increased incidence of cardiovascular disease in adult patients with type 1 diabetes. Int. J. Cardiol. 2016, 225, 387–391. [Google Scholar] [CrossRef]
  64. Mahfood Haddad, T.; Hamdeh, S.; Kanmanthareddy, A.; Alla, V.M. Nonalcoholic fatty liver disease and the risk of clinical cardiovascular events: A systematic review and meta-analysis. Diabetes Metab. Syndr. 2017, 11 (Suppl. S1), S209–S216. [Google Scholar] [CrossRef]
  65. Zhou, Y.Y.; Zhou, X.D.; Wu, S.J.; Hu, X.Q.; Tang, B.; Van Poucke, S.; Pan, X.Y.; Wu, W.J.; Gu, X.M.; Fu, S.W.; et al. Synergistic increase in cardiovascular risk in diabetes mellitus with nonalcoholic fatty liver disease: A meta-analysis. Eur. J. Gastroenterol. Hepatol. 2018, 30, 631–636. [Google Scholar] [CrossRef]
  66. Mellinger, J.L.; Pencina, K.M.; Massaro, J.M.; Hoffmann, U.; Seshadri, S.; Fox, C.S.; O´Donnell, C.J.; Speliotes, E.K. Hepatic steatosis and cardiovascular disease outcomes: An analysis of the Framingham Heart Study. J. Hepatol. 2015, 63, 470–476. [Google Scholar] [CrossRef]
  67. Assy, N.; Djibre, A.; Farah, R.; Grosovski, M.; Marmor, A. Presence of coronary plaques in patients with nonalcoholic fatty liver disease. Radiology 2010, 254, 393–400. [Google Scholar] [CrossRef]
  68. Chen, C.H.; Nien, C.K.; Yang, C.C.; Yeh, Y.H. Association between nonalcoholic fatty liver disease and coronary artery calcification. Dig. Dis. Sci. 2010, 55, 1752–1760. [Google Scholar] [CrossRef]
  69. Park, J.H.; Koo, B.K.; Kim, W.; Kim, W.H. Histological severity of nonalcoholic fatty liver disease is associated with 10-year risk for atherosclerotic cardiovascular disease. Hepatol. Int. 2021, 15, 1148–1159. [Google Scholar] [CrossRef]
  70. Ekstedt, M.; Hagström, H.; Nasr, P.; Fredrikson, M.; Stål, P.; Kechagias, S.; Hultcrantz, R. Fibrosis stage is the strongest predictor for disease-specific mortality in NAFLD after up to 33 years of follow-up. Hepatology 2015, 61, 1547–1554. [Google Scholar] [CrossRef]
  71. Chun, H.S.; Lee, M.; Lee, J.S.; Lee, H.W.; Kim, B.K.; Park, J.Y.; Kim, D.Y.; Ahn, S.H.; Lee, Y.H.; Kim, J.H.; et al. Metabolic dysfunction associated fatty liver disease identifies subjects with cardiovascular risk better than non-alcoholic fatty liver disease. Liver Int. 2023, 43, 608–625. [Google Scholar] [CrossRef]
  72. Pais, R.; Giral, P.; Khan, J.F.; Rosenbaum, D.; Housset, C.; Poynard, T.; Ratziu, V.; LIDO Study Group. Fatty liver is an independent predictor of early carotid atherosclerosis. J. Hepatol. 2016, 65, 95–102. [Google Scholar] [CrossRef]
  73. Lee, J.; Kim, H.S.; Cho, Y.K.; Kim, E.H.; Lee, M.J.; Bae, I.Y.; Jung, C.H.; Park, J.Y.; Kim, H.K.; Lee, W.J. Association between noninvasive assessment of liver fibrosis and coronary artery calcification progression in patients with nonalcoholic fatty liver disease. Sci. Rep. 2020, 10, 18323. [Google Scholar] [CrossRef]
  74. Pennisi, G.; Di Marco, V.; Buscemi, C.; Mazzola, G.; Randazzo, C.; Spatola, F.; Craxi, A.; Buscemi, S.; Petta, S. Interplay between non-alcoholic fatty liver disease and cardiovascular risk in an asymptomatic general population. J. Gastroenterol. Hepatol. 2021, 36, 2389–2396. [Google Scholar] [CrossRef]
  75. Mooli, R.G.R.; Ramakrishnan, S.K. Emerging Role of Hepatic Ketogenesis in Fatty Liver Disease. Front. Physiol. 2022, 13, 946474. [Google Scholar] [CrossRef]
  76. Shemesh, E.; Chevli, P.A.; Islam, T.; German, C.A.; Otvos, J.; Yeboah, J.; Rodriguez, F.; deFilippi, C.; Lima, J.A.C.; Blaha, M.; et al. Circulating ketone bodies and cardiovascular outcomes: The MESA study. Eur. Heart J. 2023, 44, 1636–1646. [Google Scholar] [CrossRef]
  77. Nielsen, R.; Møller, N.; Gormsen, L.C.; Tolbod, L.P.; Hansson, N.H.; Sorensen, J.; Harms, H.J.; Frokiaer, J.; Eiskjaer, H.; Jespersen, N.R.; et al. Cardiovascular Effects of Treatment with the Ketone Body 3-Hydroxybutyrate in Chronic Heart Failure Patients. Circulation 2019, 139, 2129–2141. [Google Scholar] [CrossRef] [PubMed]
  78. Bae, H.R.; Kim, D.H.; Park, M.H.; Lee, B.; Kim, M.J.; Lee, E.K.; Chung, K.W.; Kim, S.M.; Im, D.S.; Chung, H.Y. β-Hydroxybutyrate suppresses inflammasome formation by ameliorating endoplasmic reticulum stress via AMPK activation. Oncotarget 2016, 7, 66444–66454. [Google Scholar] [CrossRef] [PubMed]
  79. Cotter, D.G.; Schugar, R.C.; Crawford, P.A. Ketone body metabolism and cardiovascular disease. Am. J. Physiol. Heart Circ. Physiol. 2013, 304, H1060–H1076. [Google Scholar] [CrossRef] [PubMed]
  80. Puchalska, P.; Crawford, P.A. Multi-dimensional Roles of Ketone Bodies in Fuel Metabolism, Signaling, and Therapeutics. Cell Metab. 2017, 25, 262–284. [Google Scholar] [CrossRef] [PubMed]
  81. Koliaki, C.; Roden, M. Hepatic energy metabolism in human diabetes mellitus, obesity and non-alcoholic fatty liver disease. Mol. Cell Endocrinol. 2013, 379, 35–42. [Google Scholar] [CrossRef]
  82. Cotter, D.G.; Ercal, B.; Huang, X.; Leid, J.M.; D’Avignon, D.A.; Graham, M.J.; Dietzen, D.J.; Brunt, E.M.; Patti, G.J.; Crawford, P.A. Ketogenesis prevents diet-induced fatty liver injury and hyperglycemia. J. Clin. Investig. 2014, 124, 5175–5190. [Google Scholar] [CrossRef] [PubMed]
  83. Sunny, N.E.; Parks, E.J.; Browning, J.D.; Burgess, S.C. Excessive hepatic mitochondrial TCA cycle and gluconeogenesis in humans with nonalcoholic fatty liver disease. Cell Metab. 2011, 14, 804–810. [Google Scholar] [CrossRef]
  84. Fu, X.; Fletcher, J.A.; Deja, S.; Inigo-Vollmer, M.; Burgess, S.C.; Browning, J.D. Persistent fasting lipogenesis links impaired ketogenesis with citrate synthesis in humans with nonalcoholic fatty liver. J. Clin. Investig. 2023, 133, e167442. [Google Scholar] [CrossRef]
  85. Schoiswohl, G.; Stefanovic-Racic, M.; Menke, M.N.; Wills, R.C.; Surlow, B.A.; Basantani, M.K.; Sitnick, M.T.; Cai, L.; Yazbeck, C.F.; Stolz, D.B.; et al. Impact of reduced ATGL-mediated adipocyte lipolysis on obesity-associated insulin resistance and inflammation in male mice. Endocrinology 2015, 156, 3610–3624. [Google Scholar] [CrossRef] [PubMed]
  86. Begriche, K.; Igoudjil, A.; Pessayre, D.; Fromenty, B. Mitochondrial dysfunction in NASH: Causes, consequences and possible means to prevent it. Mitochondrion 2006, 6, 1–28. [Google Scholar] [CrossRef]
  87. Marra, F.; Gastaldelli, A.; Svegliati Baroni, G.; Tell, G.; Tiribelli, C. Molecular basis and mechanisms of progression of non-alcoholic steatohepatitis. Trends Mol. Med. 2008, 14, 72–81. [Google Scholar] [CrossRef]
  88. Pawlak, M.; Lefebvre, P.; Staels, B. Molecular mechanism of PPARα action and its impact on lipid metabolism, inflammation and fibrosis in non-alcoholic fatty liver disease. J. Hepatol. 2015, 62, 720–733. [Google Scholar] [CrossRef]
  89. Wang, H.; Shen, M.; Shu, X.; Guo, B.; Jia, T.; Feng, J.; Lu, Z.; Chen, Y.; Lin, J.; Liu, Y.; et al. Cardiac Metabolism, Reprogramming, and Diseases. J. Cardiovasc. Transl. Res. 2023, 17, 71–84. [Google Scholar] [CrossRef] [PubMed]
  90. Matsuura, T.R.; Puchalska, P.; Crawford, P.A.; Kelly, D.P. Ketones and the Heart: Metabolic Principles and Therapeutic Implications. Circ. Res. 2023, 132, 882–898. [Google Scholar] [CrossRef]
  91. Packer, M. Critical Reanalysis of the Mechanisms Underlying the Cardiorenal Benefits of SGLT2 Inhibitors and Reaffirmation of the Nutrient Deprivation Signaling/Autophagy Hypothesis. Circulation 2022, 146, 1383–1405. [Google Scholar] [CrossRef] [PubMed]
  92. Vargas-Delgado, A.P.; Arteaga Herrera, E.; Tumbaco Mite, C.; Delgado Cedeno, P.; Van Loon, M.C.; Badimon, J.J. Renal and Cardiovascular Metabolic Impact Caused by Ketogenesis of the SGLT2 Inhibitors. Int. J. Mol. Sci. 2023, 24, 4144. [Google Scholar] [CrossRef]
  93. Kolwicz, S.C. Ketone Body Metabolism in the Ischemic Heart. Front. Cardiovasc. Med. 2021, 8, 789458. [Google Scholar] [CrossRef]
  94. Kanikarla-Marie, P.; Jain, S.K. Hyperketonemia and ketosis increase the risk of complications in type 1 diabetes. Free. Radic. Biol. Med. 2016, 95, 268–277. [Google Scholar] [CrossRef]
  95. Ritterhoff, J.; Tian, R. Metabolic mechanisms in physiological and pathological cardiac hypertrophy: New paradigms and challenges. Nat. Rev. Cardiol. 2023, 20, 812–829. [Google Scholar] [CrossRef]
  96. Shimazu, T.; Hirschey, M.D.; Newman, J.; He, W.; Shirakawa, K.; Le Moan, N.; Grueter, C.A.; Lim, H.; Saunders, L.R.; Stevens, R.D.; et al. Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 2013, 339, 211–214. [Google Scholar] [CrossRef] [PubMed]
  97. Post, A.; Garcia, E.; van den Berg, E.H.; Flores-Guerrero, J.L.; Gruppen, E.G.; Groothof, D.; Westenbrink, B.D.; Connelly, M.A.; Bakker, S.J.L.; Dullart, R.P.F. Nonalcoholic fatty liver disease, circulating ketone bodies and all-cause mortality in a general population-based cohort. Eur. J. Clin. Investig. 2021, 51, e13627. [Google Scholar] [CrossRef]
  98. Gaggini, M.; Morelli, M.; Buzzigoli, E.; DeFronzo, R.A.; Bugianesi, E.; Gastaldelli, A. Non-alcoholic fatty liver disease (NAFLD) and its connection with insulin resistance, dyslipidemia, atherosclerosis and coronary heart disease. Nutrients 2013, 5, 1544–1560. [Google Scholar] [CrossRef]
  99. Navab, M.; Gharavi, N.; Watson, A.D. Inflammation and metabolic disorders. Curr. Opin. Clin. Nutr. Metab. Care 2008, 11, 459–464. [Google Scholar] [CrossRef]
  100. Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 2006, 444, 860–867. [Google Scholar] [CrossRef]
  101. Antonopoulos, A.S.; Margaritis, M.; Lee, R.; Channon, K.; Antoniades, C. Statins as Anti-Inflammatory Agents in Atherogenesis: Molecular Mechanisms and Lessons from the Recent Clinical Trials. Curr. Pharm. Des. 2012, 18, 1519–1530. [Google Scholar] [CrossRef] [PubMed]
  102. Stubbs, B.J.; Cox, P.J.; Evans, R.D.; Santer, P.; Miller, J.J.; Faull, O.K.; Magor-Elliot, S.; Hiyama, S.; Stirling, M.; Clarke, K. On the metabolism of exogenous ketones in humans. Front. Physiol. 2017, 8, 848. [Google Scholar] [CrossRef] [PubMed]
  103. Caminhotto, R.D.O.; Komino, A.C.M.; De Fatima Silva, F.; Andreotti, S.; Sertié, R.A.L.; Boltes Reis, G.; Lima, F.B. Oral β-hydroxybutyrate increases ketonemia, decreases visceral adipocyte volume and improves serum lipid profile in Wistar rats. Nutr. Metab. 2017, 14, 31. [Google Scholar] [CrossRef]
  104. Myette-Côté, É.; Caldwell, H.G.; Ainslie, P.N.; Clarke, K.; Little, J.P. A ketone monoester drink reduces the glycemic response to an oral glucose challenge in individuals with obesity: A randomized trial. Am. J. Clin. Nutr. 2019, 110, 1491–1501. [Google Scholar] [CrossRef] [PubMed]
  105. Myette-Côté, É.; Neudorf, H.; Rafiei, H.; Clarke, K.; Little, J.P. Prior ingestion of exogenous ketone monoester attenuates the glycaemic response to an oral glucose tolerance test in healthy young individuals. J. Physiol. 2018, 596, 1385–1395. [Google Scholar] [CrossRef] [PubMed]
  106. Ari, C.; Murdun, C.; Koutnik, A.P.; Goldhagen, C.R.; Rogers, C.; Park, C.; Bharwani, S.; Diamond, D.M.; Kindy, M.S.; D´Agostino, D.P.; et al. Exogenous ketones lower blood glucose level in rested and exercised rodent models. Nutrients 2019, 11, 2330. [Google Scholar] [CrossRef]
  107. Samaha, F.F.; Iqbal, N.; Seshadri, P.; Chicano, K.L.; Daily, D.A.; McGrory, J.; Williams, T.; Williams, M.; Gracely, E.J.; Stern, L. A Low-Carbohydrate as Compared with a Low-Fat Diet in Severe Obesity. N. Engl. J. Med. 2003, 348, 2074–2081. [Google Scholar] [CrossRef]
  108. Cicero, A.F.G.; Benelli, M.; Brancaleoni, M.; Dainelli, G.; Merlini, D.; Negri, R. Middle and Long-Term Impact of a Very Low-Carbohydrate Ketogenic Diet on Cardiometabolic Factors: A Multi-Center, Cross-Sectional, Clinical Study. High Blood Press. Cardiovasc. Prev. 2015, 22, 389–394. [Google Scholar] [CrossRef]
  109. Mezzaroma, E.; Toldo, S.; Farkas, D.; Seropian, I.M.; Van Tassell, B.W.; Salloum, F.N.; Kannan, H.R.; Menna, A.C.; Voelkel, N.F.; Abbate, A. The inflammasome promotes adverse cardiac remodeling following acute myocardial infarction in the mouse. Proc. Natl. Acad. Sci. USA 2011, 108, 19725–19730. [Google Scholar] [CrossRef]
  110. Gormsen, L.C.; Svart, M.; Thomsen, H.H.; Søndergaard, E.; Vendelbo, M.H.; Christensen, N.; Tolbod, L.P.; Harms, H.J.; Nielsen, R.; Wiggers, H.; et al. Ketone body infusion with 3-hydroxybutyrate reduces myocardial glucose uptake and increases blood flow in humans: A positron emission tomography study. J. Am. Heart Assoc. 2017, 6, e005066. [Google Scholar] [CrossRef] [PubMed]
  111. Steiner, S. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. Z. Fur Gefassmedizin 2016, 13, 2117–2128. [Google Scholar]
  112. Ou, H.Y.; Karnchanasorn, R.; Chuang, L.M.; Chiu, K. Diabetic Ketoacidosis and Related Events in the Canagliflozin Type 2 Diabetes Clinical Program. Diabetes Care 2015, 38, 1680–1686. [Google Scholar]
  113. Peters, A.L.; Buschur, E.O.; Buse, J.B.; Cohan, P.; Diner, J.C.; Hirsch, I.B. Euglycemic diabetic ketoacidosis: A potential complication of treatment with sodium-glucose cotransporter 2 inhibition. Diabetes Care 2015, 38, 1687–1693. [Google Scholar] [CrossRef] [PubMed]
  114. Ferrannini, E.; Muscelli, E.; Frascerra, S.; Baldi, S.; Mari, A.; Heise, T.; Broedl, U.C.; Woerle, H.J. Metabolic response to sodium-glucose cotransporter 2 inhibition in type 2 diabetic patients. J. Clin. Investig. 2014, 124, 499–508. [Google Scholar] [CrossRef] [PubMed]
  115. Newman, J.C.; Verdin, E. β-hydroxybutyrate: Much more than a metabolite. Diabetes Res. Clin. Pract. 2014, 106, 173–181. [Google Scholar] [CrossRef] [PubMed]
  116. EMPA-Kidney Collaborative Group. Empagliflozin in Patients with Chronic Kidney Disease. N. Engl. J. Med. 2023, 388, 117–127. [Google Scholar] [CrossRef] [PubMed]
  117. Verma, S.; Dhingra, N.K.; Butler, J.; Anker, S.D.; Ferreira, J.P.; Filippatos, G.S.; Januzzi, J.; Lam, C.; Sattar, N.; Pell, B.; et al. Empagliflozin in the treatment of heart failure with reduced ejection fraction in addition to background therapies and therapeutic combinations (EMPEROR-Reduced): A post-hoc analysis of a randomised, double-blind trial. Lancet Diabetes Endocrinol. 2022, 10, 35–45. [Google Scholar] [CrossRef]
  118. Butler, J.; Filippatos, G.; Jamal Siddiqi, T.; Brueckmann, M.; Böhm, M.; Chopra, V.K.; Ferreira, J.P.; Januzzi, J.L.; Kaul, S.; Piña, I.L.; et al. Empagliflozin, Health Status, and Quality of Life in Patients with Heart Failure and Preserved Ejection Fraction: The EMPEROR-Preserved Trial. Circulation 2022, 145, 184–193. [Google Scholar] [CrossRef]
Figure 1. Role of simplified MASLD pathogenesis in classic cardiovascular risk factor development. TCA: Tricarboxylic acid; TAG: Triglycerides; ROS: Reactive oxygen species; AG: Acylglycerides; CV: Cardiovascular. Upwards arrows: increases; downwards arrows: decreases; red cross: blocks; red lighting symbol: interferes or impacts. Legend: Both pernicious genes and lifestyle have a role in the incremental concentration of glucose in the liver, which is metabolized through the TCA cycle and the glucogen synthesis pathways, among others. An excess in Acetyl CoA provides a higher concentration of intrahepatic acyl glycerides, which is directly linked to insulin resistance, providing reduced glycogen synthesis, a need for higher levels of insulin to provide glucose for energetic purposes (effective liver glucose), which activates intrahepatic gluconeogenesis and an excess in triglycerides, leading to a less efficient metabolization, with Radical Oxygen Species production and a higher plasma concentration of triglycerides and cholesterol particles contributing to a higher cardiovacular risk through classical risk factors [30,31,32,33].
Figure 1. Role of simplified MASLD pathogenesis in classic cardiovascular risk factor development. TCA: Tricarboxylic acid; TAG: Triglycerides; ROS: Reactive oxygen species; AG: Acylglycerides; CV: Cardiovascular. Upwards arrows: increases; downwards arrows: decreases; red cross: blocks; red lighting symbol: interferes or impacts. Legend: Both pernicious genes and lifestyle have a role in the incremental concentration of glucose in the liver, which is metabolized through the TCA cycle and the glucogen synthesis pathways, among others. An excess in Acetyl CoA provides a higher concentration of intrahepatic acyl glycerides, which is directly linked to insulin resistance, providing reduced glycogen synthesis, a need for higher levels of insulin to provide glucose for energetic purposes (effective liver glucose), which activates intrahepatic gluconeogenesis and an excess in triglycerides, leading to a less efficient metabolization, with Radical Oxygen Species production and a higher plasma concentration of triglycerides and cholesterol particles contributing to a higher cardiovacular risk through classical risk factors [30,31,32,33].
Biomedicines 12 00692 g001
Figure 2. TCA: Tricarboxylic acid; PEP: Phosphonyl pyruvate. Legend: A dual result of triglyceride exposure might be expected depending on the MASLD status, as a higher exposure to beta-oxidation products leads to a higher production of ketone bodies in the first stages of SLD, while, as fibrosis progresses, ketone metabolism is reduced as a potential early marker of liver metabolic dysfunction [75]. Upward arrows indicate an increase in the synthesis of ketone bodies, activation of the TCA cycle, and gluconeogenesis. Downward arrows would represent a decrease in the processes of lipogenesis and the accumulation of fatty acids. The red cross would indicate the inhibition of the hydroxy-methyl-glutaril-CoA (HMG-CoA) complex pathway, thus decreasing the production of ketone bodies. The red lightning symbol suggests stress or damage leading to steatosis and possibly hepatic fibrosis.
Figure 2. TCA: Tricarboxylic acid; PEP: Phosphonyl pyruvate. Legend: A dual result of triglyceride exposure might be expected depending on the MASLD status, as a higher exposure to beta-oxidation products leads to a higher production of ketone bodies in the first stages of SLD, while, as fibrosis progresses, ketone metabolism is reduced as a potential early marker of liver metabolic dysfunction [75]. Upward arrows indicate an increase in the synthesis of ketone bodies, activation of the TCA cycle, and gluconeogenesis. Downward arrows would represent a decrease in the processes of lipogenesis and the accumulation of fatty acids. The red cross would indicate the inhibition of the hydroxy-methyl-glutaril-CoA (HMG-CoA) complex pathway, thus decreasing the production of ketone bodies. The red lightning symbol suggests stress or damage leading to steatosis and possibly hepatic fibrosis.
Biomedicines 12 00692 g002
Figure 3. Potential interrelationship between traditional cardiovascular risk factors and SLD in the progression of endothelial dysfunction and ketone body production. Legend: The red line describes the evolution of ketone body production through NAFL evolution, while the yellow line describes the evolution of endothelial dysfunction [42,77].
Figure 3. Potential interrelationship between traditional cardiovascular risk factors and SLD in the progression of endothelial dysfunction and ketone body production. Legend: The red line describes the evolution of ketone body production through NAFL evolution, while the yellow line describes the evolution of endothelial dysfunction [42,77].
Biomedicines 12 00692 g003
Table 1. Studies providing evidence on NAFLD invasive and non-invasive assessment in the prediction of cardiovascular disease [27].
Table 1. Studies providing evidence on NAFLD invasive and non-invasive assessment in the prediction of cardiovascular disease [27].
Diagnostic NAFLDReferencePatients, nType of StudyImpact of the NAFLD
Ultrasound
Stepanova and Younossi, 2012 [54]20,050ProspectiveOR, 1.23 for CVD events
Haring et al., 2009 [55]4160ProspectiveHR, 6.22 for all-cause mortality and CVD
Kim et al., 2012 [56]4023Cross-sectionalOR, 1.32 for CAC > 10
Targher et al., 2007 [57]2839Cross-sectionalOR, 1.49 for DKA DBP, and cerebrovascular disease in type 2 DM
Tsutsumi T et al., 2021 [58]2306ProspectiveHR, 1.08 independently with worsening CVD
Hamaguchi et al., 2007 [59]1637ProspectiveHR, 4.1 for nonfatal CVD events
Yoshitaka and al, 2017 [60]1647ProspectiveHR, 10.4 not overweight, 3.1 overweight for incident CV events
Wong et al., 2011 [61]612ProspectiveOR, 2.31 for significant coronary artery disease (>50% obstruction)
Santos et al., 2007 [62]505Cross-sectionalOR, 1.73 for coronary calcification
Mantovani et al., 2016 [63]286RetrospectiveOR, 6.73 for incident cardiovascular events in type 1 diabetes
CT
Mahfood Hadad et al., 2016 [64]25,837 (11 studies)Meta-analysisRR, 1.77 for incident CVD, 1.43 for cardiovascular mortality
Zhou et al., 2018 [65]8346 (6 studies)Meta-analysisOR, 2.20 for incident CVD in patients with diabetes
Mellinger et al., 2015 [66]3014Cross-sectionalOR, 1.20 for CAC score >90th percentile for age
Assy et al., 2010 [67]61Cross-sectionalOR, 2.03 for coronary calcification
Ultrasound/CT
Chen et al., 2010 [68]295Cross-sectionalOR, 2.46 for CAC > 100
Liver biopsy
Simon et al., 2022 [42]422ProspectiveHR, 2.15 for MACE
Ji Hye Park et al., 2021 [69]398Cross-sectionalOR, 4.07 increased risk of ASCVD for NASH OR, 8.11 increased risk of ASCVD for advanced fibrosis
Ekstedt et al., 2015 [70]229RetrospectiveHR, 1.55 for CVD mortality
Fatty Liver Index
Chun HS et al., 2023 [71]78,762Cross-sectionalOR, 1.10 for CVD history in MAFLD
OR, 1.40 for high probability of ASCVD in MAFLD
OR, 1.22 for high probability of ASCVD in NAFLD
Pais et al., 2016 [72]5671RetrospectiveThe severity of NAFLD correlates with CIMT and the severity of carotid plaque
Lee J et al., 2020 [73]1173ProspectiveOR, 1.70 for CAC progression in patients with NAFLD
Pennisi et al., 2021 [74]542Cross-sectionalOR, 1.62 risk factors for ASCVD in patients with steatosis
OR, 1.67 risk factors for ASCVD in patients with severe fibrosis
NAFLD: non-alcoholic fatty liver disease; OR: Odds ratio; CVD: cardiovascular disease; HR: hazard ratio; CAC: coronary artery calcium; DKA: diabetic ketoacidosis; DBP: diastolic blood pressure; RR: relative risk; MACE: major adverse cardiovascular event; ASCVD: atherosclerotic cardiovascular disease; NASH: non-alcoholic steatohepatitis; MAFLD: metabolic dysfunction-associated fatty liver disease.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

del Villar-Carrero, R.S.; Blanco, A.; Ruiz, L.D.; García-Blanco, M.J.; Segovia, R.C.; de la Garza, R.G.; Martínez-Urbistondo, D. Bridging Metabolic-Associated Steatotic Liver Disease and Cardiovascular Risk: A Potential Role for Ketogenesis. Biomedicines 2024, 12, 692. https://doi.org/10.3390/biomedicines12030692

AMA Style

del Villar-Carrero RS, Blanco A, Ruiz LD, García-Blanco MJ, Segovia RC, de la Garza RG, Martínez-Urbistondo D. Bridging Metabolic-Associated Steatotic Liver Disease and Cardiovascular Risk: A Potential Role for Ketogenesis. Biomedicines. 2024; 12(3):692. https://doi.org/10.3390/biomedicines12030692

Chicago/Turabian Style

del Villar-Carrero, Rafael Suárez, Agustín Blanco, Lidia Daimiel Ruiz, Maria J. García-Blanco, Ramón Costa Segovia, Rocío García de la Garza, and Diego Martínez-Urbistondo. 2024. "Bridging Metabolic-Associated Steatotic Liver Disease and Cardiovascular Risk: A Potential Role for Ketogenesis" Biomedicines 12, no. 3: 692. https://doi.org/10.3390/biomedicines12030692

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

Article Metrics

Back to TopTop