Coronary Restenosis
Introduction
Section titled “Introduction”Coronary restenosis refers to the re-narrowing of a coronary artery following an interventional procedure, such as percutaneous coronary intervention (PCI), which is commonly performed to treat coronary artery disease (CAD).[1], [2]Despite initial success in restoring blood flow, the treated vessel can gradually constrict again, leading to recurrent symptoms and potentially serious cardiovascular events like myocardial infarction (MI).[3]This phenomenon represents a significant challenge in the long-term management of cardiovascular health.
Biological Basis
Section titled “Biological Basis”The biological basis of coronary restenosis is complex, involving a cascade of cellular and molecular events triggered by vascular injury during the revascularization procedure. This injury initiates an inflammatory response and promotes the proliferation and migration of vascular smooth muscle cells into the intimal layer of the artery, a process known as neointimal hyperplasia. The deposition of extracellular matrix further contributes to the progressive narrowing of the vessel lumen. Genetic factors are understood to influence an individual’s susceptibility to restenosis, with various single nucleotide polymorphisms (SNPs) implicated in pathways affecting inflammation, cell growth, and vascular remodeling. For example, genetic variants have been associated with coronary artery disease and myocardial infarction, suggesting underlying genetic predispositions to cardiovascular pathologies that may also impact restenosis risk.[2], [3] Studies have explored biological pathways such as protein kinase A signaling and the Wnt/Ca2+ pathway, which are involved in the regulation of cardiac traits and could play a role in the cellular processes underlying restenosis.[3]
Clinical Relevance
Section titled “Clinical Relevance”Clinically, coronary restenosis can lead to a return of angina, reduced exercise tolerance, and in severe cases, acute coronary syndromes. Patients who experience restenosis often require repeat revascularization procedures, such as another PCI or coronary artery bypass grafting (CABG), to maintain adequate blood flow to the heart muscle.[1]The presence of other cardiovascular risk factors, including hypertension, hypercholesterolemia, diabetes mellitus, and smoking, can further increase the likelihood of restenosis and impact its severity.[1], [4] Understanding the genetic predispositions and biological mechanisms is crucial for developing more effective preventive strategies and targeted therapies to improve long-term patient outcomes.
Social Importance
Section titled “Social Importance”The social importance of coronary restenosis stems from its significant impact on public health and healthcare systems. As a common complication of frequently performed cardiac interventions, restenosis contributes substantially to healthcare costs through the need for repeat procedures, prolonged hospital stays, and ongoing medical management. Beyond economic implications, it diminishes the quality of life for affected individuals, causing chronic symptoms, anxiety, and a reduced capacity for daily activities. Efforts to identify genetic markers and understand the underlying biology of restenosis are vital for personalizing treatment approaches and ultimately reducing the burden of cardiovascular disease on individuals and society.
Methodological and Statistical Rigor
Section titled “Methodological and Statistical Rigor”The accurate identification of genetic variants associated with coronary restenosis faces inherent methodological and statistical challenges. Initial findings from discovery cohorts, particularly those with smaller sample sizes, may exhibit inflated effect sizes, which can lead to an overestimation of the genetic contribution to the trait.[5] Therefore, the ultimate validation of any observed genetic associations necessitates robust replication in independent and sufficiently powered cohorts to confirm true positive signals and precisely estimate their magnitude.[5] Without such widespread replication, distinguishing genuine genetic determinants from chance findings remains a fundamental hurdle in genome-wide association studies, impacting the confidence in identified risk loci and their potential clinical utility.
Generalizability and Population Specificity
Section titled “Generalizability and Population Specificity”The generalizability of genetic findings for coronary restenosis across diverse human populations is a significant limitation. While studies may employ sophisticated methods, such as adjusting for ancestry principal components, to mitigate bias from population stratification, findings derived primarily from genetically homogeneous cohorts may not be directly transferable to populations with different ancestral backgrounds.[4]This restricted representation can lead to an incomplete understanding of the global genetic architecture of coronary restenosis, potentially obscuring variants that are more prevalent or exert differential effects in underrepresented ethnic groups. Consequently, the applicability of risk prediction models or therapeutic strategies based on such findings may be limited to specific populations.
Environmental Confounders and Functional Elucidation
Section titled “Environmental Confounders and Functional Elucidation”A comprehensive understanding of the genetic landscape of coronary restenosis is further complicated by the intricate interplay between genetic predispositions and environmental or clinical factors. Established cardiovascular risk factors, including but not limited to hypertension, hypercholesterolemia, diabetes mellitus, atrial fibrillation, myocardial infarction, and smoking status, can act as powerful confounders or gene-environment modifiers.[4]These complex interactions make it challenging to isolate the independent genetic effects and fully delineate the pathways through which genetic variants contribute to disease risk. Moreover, even when statistically significant genetic associations are identified, the precise biological mechanisms and functional consequences by which these variants influence restenosis often remain largely unknown, underscoring the critical need for extensive follow-up and functional studies to bridge the gap between statistical association and mechanistic insight.[5]
Variants
Section titled “Variants”The genomic region encompassing _C12orf42_ and _LINC02401_is of interest due to its potential role in cardiovascular health, particularly in conditions like coronary restenosis._C12orf42_, or Chromosome 12 open reading frame 42, is a protein-coding gene whose specific biological functions are still being actively researched. Many genes in this category are found to subtly influence cellular processes and signaling pathways, contributing to the complexity of human diseases. Closely associated with this region is `_LINC02401_`, a long intergenic non-coding RNA. LncRNAs are crucial regulators of gene expression, acting through mechanisms such as chromatin remodeling, transcriptional control, and post-transcriptional processing.In the context of vascular biology, lncRNAs are increasingly recognized for their involvement in fundamental processes like inflammation, endothelial cell function, and vascular smooth muscle cell proliferation and migration, all of which are pivotal in the development and progression of coronary restenosis. Therefore, variations affecting the expression or function of_C12orf42_ or _LINC02401_ could modulate the cellular environment within arterial walls, influencing the body’s response to vascular injury.
The single nucleotide polymorphism (SNP)rs10861032 is situated within this genomic locus, potentially impacting the regulation or function of _C12orf42_ and _LINC02401_. Depending on its precise location relative to these genes, rs10861032 could affect gene expression by altering enhancer activity, transcription factor binding sites, or mRNA stability. Such changes in gene activity could, in turn, influence the biological processes underlying coronary restenosis, which is the re-narrowing of an artery after an interventional procedure like angioplasty. Restenosis involves a complex interplay of inflammation, the excessive proliferation and migration of vascular smooth muscle cells, and the remodeling of the extracellular matrix within the vessel wall. A genetic variant like rs10861032 could modify an individual’s susceptibility to these events, potentially leading to a higher or lower risk of restenosis by fine-tuning the cellular responses involved in arterial healing and remodeling.
Key Variants
Section titled “Key Variants”| RS ID | Gene | Related Traits |
|---|---|---|
| rs10861032 | C12orf42 - LINC02401 | coronary restenosis C-type lectin domain family 11 member A level |
Causes
Section titled “Causes”Coronary restenosis, the re-narrowing of a coronary artery after an interventional procedure such as angioplasty, is a complex process influenced by a confluence of genetic predispositions, metabolic imbalances, systemic conditions, and age-related physiological changes. These factors contribute to the underlying vascular disease and the subsequent biological responses to arterial injury that lead to vessel re-occlusion.
Genetic Predisposition and Molecular Pathways
Section titled “Genetic Predisposition and Molecular Pathways”Inherited genetic variants significantly influence an individual’s susceptibility to coronary artery disease (CAD), which serves as the foundation for interventions that can lead to restenosis. Genome-wide association studies (GWAS) have identified numerous polygenic risk loci across European populations. For instance, a new susceptibility locus for CAD has been identified on chromosome 3q22.3.[2]Similarly, common variants at 6p21.1 are associated with large artery atherosclerotic stroke, a related cardiovascular condition.[4]Specific single nucleotide polymorphisms (SNPs) such asrs17696696 , rs17608766 , and rs10774625 are significantly associated with CAD, with rs10774625 also strongly linked to myocardial infarction.[3]These genetic predispositions collectively contribute to an individual’s overall cardiovascular risk profile, influencing the likelihood of developing disease requiring intervention.
Beyond individual variants, gene-gene interactions and the resulting molecular pathways are critical in modulating disease progression. Genetic determinants influence circulating lipid levels, including sphingolipid concentrations, which are implicated in coronary heart disease risk.[6] For example, FADSgenotypes impact desaturase activity, thereby affecting the ratio of arachidonic acid to linoleic acid, and are associated with inflammation and CAD.[7] Pathway analyses of associated genetic variants reveal significant enrichment in canonical pathways like protein kinase A signaling, death receptor signaling, the Wnt/Ca2+ pathway, and P2Y purigenic receptor signaling. These findings highlight the complex genetic regulatory networks that govern cardiac structure and function, thereby indirectly influencing the vascular environment that predisposes to restenosis.[3]
Metabolic and Systemic Influences
Section titled “Metabolic and Systemic Influences”Environmental factors and comorbidities substantially modulate the risk of coronary restenosis by exacerbating underlying cardiovascular disease processes. Lifestyle choices, dietary patterns, and exposure to certain environmental triggers contribute to the development of conditions such as hypertension, hypercholesterolemia, and diabetes mellitus, all recognized as significant risk factors for coronary artery disease.[4]These conditions foster a pro-inflammatory and pro-thrombotic milieu within the vasculature, which promotes atherosclerosis and increases the likelihood of requiring a coronary intervention that could subsequently lead to restenosis. Smoking status is another critical environmental factor that profoundly impacts cardiovascular health and contributes to this risk.[4]The interplay between genetic predisposition and environmental factors is a key determinant in disease manifestation. Genetic variants that influence lipid metabolism, such as those affecting sphingolipid concentrations, can interact with dietary fat intake to influence overall lipid profiles and insulin resistance.[6]This interaction can render individuals more susceptible to the adverse effects of certain diets, leading to metabolic disease and heightened cardiovascular risk.[8]Furthermore, systemic conditions like atrial fibrillation and a history of myocardial infarction act as significant comorbidities that can influence the progression of vascular disease and the response to coronary interventions, thereby contributing to the incidence of restenosis.[4] The involvement of ceramide in triggering cardiomyocyte apoptosis during ischemia and reperfusion further underscores the cellular damage processes that contribute to adverse vascular remodeling.[9]
Epigenetic Regulation and Age-Related Changes
Section titled “Epigenetic Regulation and Age-Related Changes”Developmental and epigenetic factors play an influential role in shaping an individual’s susceptibility to cardiovascular disease and its complications, including restenosis. Epigenetic mechanisms, which involve modifications to gene expression without altering the underlying DNA sequence, are suggested by the identification of genetic variants located within enhancer histone marks and DNase-hypersensitive sites.[3] These regulatory elements can impact the long-term programming of vascular cells and their reparative responses to injury, thereby influencing the propensity for restenosis following an intervention. Such epigenetic alterations can modulate the expression of genes involved in inflammation, cell proliferation, and extracellular matrix remodeling, all of which are central to the restenotic process.
Age-related changes constitute another significant contributing factor to the development and progression of coronary artery disease and, consequently, restenosis. The cumulative effects of genetic predispositions, environmental exposures, and physiological wear and tear over a lifetime contribute to the aging of the cardiovascular system. Longitudinal studies have identified novel genetic variants associated with various age-related diseases, including those affecting cardiac morphology and function.[10]The aging process itself can impair cellular repair mechanisms, heighten chronic inflammation, and increase arterial stiffness, making older individuals more prone to both primary coronary disease and the post-interventional complications such as restenosis. Factors like sex-related differences and systolic blood pressure also contribute to the complex age-dependent risk profile for cardiovascular events.[10]
Vascular Remodeling and Cellular Signaling Pathways
Section titled “Vascular Remodeling and Cellular Signaling Pathways”Coronary restenosis involves complex cellular signaling cascades that drive vascular smooth muscle cell (VSMC) phenotypic modulation, proliferation, and migration in response to arterial injury. Receptor activation and subsequent intracellular signaling are critical in mediating these processes, leading to the thickening of the arterial wall. For instance, a coronary artery disease-associated variant can affect the cleavage ofADAMTS7, influencing VSMC migration, a key event in neointimal formation.[11] Furthermore, Notchsignaling plays a role in cardiovascular disease and vascular calcification, which can contribute to the altered mechanical properties of the vessel wall.[12] The dysregulation of these pathways, including the JAK2/STAT3 pathway, can promote VEGFupregulation under conditions like high glucose, potentially through mitochondrialROS pathways, linking metabolic stress to proliferative signals.[13] Endothelial responses to hemodynamic forces are also regulated by specific genetic variants, highlighting how mechanical stimuli interact with genetic predispositions to influence vascular remodeling.[14]
Epigenetic and Transcriptional Regulatory Mechanisms
Section titled “Epigenetic and Transcriptional Regulatory Mechanisms”Gene regulation, primarily through epigenetic modifications such as histone acetylation, plays a central role in controlling VSMC phenotype and the progression of restenosis. Histone deacetylases (HDACs) are crucial enzymes that regulate chromatin structure and gene transcription, with specific classes like class IIa HDACs (HDAC5 and HDAC9) being implicated in the heart’s response to stress signals and in developmental processes.[15] Notably, HDAC9is involved in atherosclerotic aortic calcification and impacts the VSMC phenotype, suggesting its critical role in disease pathogenesis.[16] These regulatory mechanisms involve transcription factor activation and feedback loops that fine-tune gene expression, dictating whether VSMCs adopt a proliferative, synthetic, or contractile state. Therapeutic strategies targeting selective class IIa HDAC inhibition are being explored to modulate these processes.[17] The interplay of these regulatory elements ultimately shapes the cellular responses that lead to restenosis.
Metabolic Reprogramming and Lipid Dysregulation
Section titled “Metabolic Reprogramming and Lipid Dysregulation”Metabolic pathways are significantly altered in the context of coronary restenosis, contributing to the disease’s progression through changes in energy metabolism, biosynthesis, and catabolism. Metabolite profiling has identified specific pathways associated with metabolic risk factors in humans, underscoring the importance of metabolic regulation.[18]Sphingolipids, for example, are crucial lipid mediators implicated in insulin resistance and metabolic diseases, withceramide specifically involved in triggering cardiomyocyte apoptosis during ischemia and reperfusion.[8] Furthermore, genetic variations in enzymes like FADS(fatty acid desaturase) influence desaturase activity, reflected by the ratio of arachidonic acid to linoleic acid, which is associated with inflammation and coronary artery disease.[7]Tryptophan metabolism and its downstream products, such askynurenine, can also impact vascular function; kynurenine acts as an endothelium-derived relaxing factor produced during inflammation.[19]Uremic toxins derived from tryptophan metabolism can activate the aryl hydrocarbon receptor, contributing to cardiovascular complications.[20]
Protein Homeostasis and Stress Responses
Section titled “Protein Homeostasis and Stress Responses”The ubiquitin-proteasome system (UPS), a critical regulatory mechanism for protein modification and degradation, is dysregulated in cardiovascular diseases like carotid atherosclerosis.[21] Increased UPS activity has been linked to enhanced inflammation and can destabilize atherosclerotic plaques, indicating its role in the inflammatory and remodeling processes characteristic of restenosis.[22] This system is vital for maintaining protein homeostasis and responding to cellular stressors. Genetic variations and gene expression changes in response to endoplasmic reticulum (ER) stress also highlight the intricate regulatory mechanisms involved in cellular adaptation and survival in the vascular wall.[23] Post-translational modifications, particularly ubiquitination, govern the stability and activity of numerous proteins, and their dysregulation can lead to the accumulation of misfolded proteins or inappropriate activation of signaling pathways, contributing to the pathological remodeling observed in restenosis.
Systems Genetics and Pathway Crosstalk
Section titled “Systems Genetics and Pathway Crosstalk”Coronary restenosis arises from a complex interplay of genetic factors and environmental stimuli, illustrating systems-level integration where multiple pathways crosstalk and interact. Systems genetics approaches are crucial for understanding such complex traits by integrating genetic variation with molecular and cellular phenotypes.[24]Genetic variants associated with coronary artery disease (CAD) often regulate phenotypes in human vascular smooth muscle cells, indicating a direct link between genetics and cellular behavior.[25] For instance, cardiometabolic risk loci can share downstream cis- and trans-gene regulation across various tissues, demonstrating hierarchical regulation and network interactions.[26] The genetic architecture of the adaptive immune system also identifies key immune regulators that may influence inflammatory responses in restenosis.[27]Identifying these causal candidate genes within CAD loci provides valuable therapeutic targets, underscoring the emergent properties of these integrated biological networks in disease pathogenesis.[28]
Pharmacogenetics of Coronary Restenosis
Section titled “Pharmacogenetics of Coronary Restenosis”Pharmacogenetics explores how an individual’s genetic makeup influences their response to medications, affecting drug efficacy and the likelihood of adverse reactions. In the context of coronary restenosis, understanding genetic variations can help personalize treatment strategies, particularly for antiplatelet therapies and lipid-modifying agents, which are crucial in preventing recurrent cardiovascular events.
Genetic Modifiers of Antiplatelet Drug Response
Section titled “Genetic Modifiers of Antiplatelet Drug Response”Genetic variations significantly impact the pharmacokinetics and pharmacodynamics of antiplatelet drugs like clopidogrel, a prodrug requiring metabolic activation. The cytochrome P450 enzyme _CYP2C19_ plays a critical role in this activation, with the _CYP2C19_*2 allele being a strong genetic determinant of reduced on-clopidogrel platelet reactivity, explaining a substantial portion of the variability in active metabolite levels.[1]This diminished activation can lead to insufficient platelet inhibition, potentially increasing the risk of adverse cardiovascular outcomes. Beyond_CYP2C19_, other genetic variants, including *rs2254638 * in _A6AMT1_ and *rs12456693 * in _SLC4A3_, also predict active clopidogrel metabolite levels, indicating a polygenic influence on drug metabolism.[1] While _CYP2C19_*2is a primary driver of platelet reactivity, its direct association with major adverse cardiovascular events (MACE) can vary across patient cohorts, suggesting that clinical outcomes are complex and influenced by multiple factors. However, a pharmacogenomic polygenic response score incorporating variants in genes such as_CYP2C19_, _CES1_, _CYP2B6_, and _CYP2C9_has demonstrated clinical utility; patients with an increasing number of risk alleles showed higher rates of cardiovascular events and cardiovascular death, highlighting the cumulative effect of multiple genetic predispositions.[1] These findings underscore the potential for genetic testing to identify individuals who may benefit from alternative antiplatelet strategies or adjusted dosing to optimize therapeutic response and minimize restenosis risk.
Genetic Influence on Lipid-Lowering Therapy
Section titled “Genetic Influence on Lipid-Lowering Therapy”Genetic variations also contribute to the differential response to lipid-lowering therapies, such as niacin. Studies have identified nominally significant SNP-treatment interactions for variants in genes like _MVK_, _LIPC_, _PABPC4_, and _AMPD3_ with changes in plasma lipid levels following niacin administration.[29]These genes are involved in various aspects of lipid metabolism, including cholesterol synthesis, lipoprotein lipase activity, and adenosine metabolism, thereby influencing the therapeutic efficacy of niacin in modulating lipid profiles. Understanding these genetic predispositions can help predict an individual’s likely response to niacin therapy, enabling more personalized management of dyslipidemia, a key risk factor for coronary artery disease and restenosis.
Genetic Markers for Cardiovascular Event Risk and Drug Efficacy
Section titled “Genetic Markers for Cardiovascular Event Risk and Drug Efficacy”Beyond specific drug metabolism, broader genetic associations have been identified with clinical outcomes relevant to coronary restenosis, including MACE and stent thrombosis, particularly in high-risk patient subgroups such as those with coronary artery disease, acute coronary syndrome, or those undergoing percutaneous coronary intervention (PCI). For instance, specific SNPs like*rs12913988 * in _ATP10A_ have been significantly associated with MACE in PCI patients, with the T allele conferring an increased risk.[1] Furthermore, genome-wide association studies (GWAS) have identified novel associations in subgroups, with mutations in _SOCS5P1_, _CDC42BPA_, and _CTRAC1_ showing genome-wide significance for MACE and stent thrombosis.[1]These genetic markers, while not always directly linked to drug targets, can influence disease progression and treatment outcomes, offering insights into underlying biological pathways such as protein kinase A signaling, death receptor signaling, and the Wnt/Ca2+ pathway, which are enriched among variants associated with cardiac structure and function.[3]
Clinical Implementation and Personalized Prescribing
Section titled “Clinical Implementation and Personalized Prescribing”The integration of pharmacogenetic information into clinical practice offers a pathway toward personalized prescribing for coronary restenosis. For antiplatelet agents like clopidogrel, genetic testing for_CYP2C19_variants can identify patients at risk of poor response, guiding clinicians to consider alternative P2Y12 inhibitors or dose adjustments where evidence supports it, thereby optimizing antiplatelet efficacy and potentially reducing the risk of stent thrombosis or recurrent cardiovascular events. Similarly, for lipid-modifying therapies, genetic insights can help tailor drug selection and dosing to achieve desired lipid targets more effectively.[29]While the clinical utility of some genetic markers is still under investigation, the growing body of evidence supports their role in informing drug selection and dosing recommendations, moving towards a more precise and effective management of patients at risk for coronary restenosis.
Frequently Asked Questions About Coronary Restenosis
Section titled “Frequently Asked Questions About Coronary Restenosis”These questions address the most important and specific aspects of coronary restenosis based on current genetic research.
1. Why do I need another heart procedure when my friend didn’t?
Section titled “1. Why do I need another heart procedure when my friend didn’t?”Your individual genetic makeup plays a significant role in how your body responds to vascular injury from a stent procedure. Some people have genetic variants that make them more prone to an exaggerated healing response, leading to re-narrowing of the artery. Even with similar procedures, these underlying genetic differences can explain why some individuals experience restenosis while others do not. This genetic predisposition means your body reacts differently to the same intervention.
2. Does what I eat affect my risk of needing another stent?
Section titled “2. Does what I eat affect my risk of needing another stent?”Yes, your diet significantly impacts several cardiovascular risk factors that are strongly linked to restenosis. Eating habits that contribute to high cholesterol, diabetes, or high blood pressure can increase inflammation and cellular proliferation in your arteries, making them more likely to re-narrow after a stent. Managing these conditions through a healthy diet can help reduce your overall risk and improve long-term outcomes.
3. Does my family history mean I’ll likely get restenosis?
Section titled “3. Does my family history mean I’ll likely get restenosis?”Yes, a family history of heart problems suggests you might have inherited genetic predispositions that increase your susceptibility to restenosis. Genetic variants that contribute to conditions like coronary artery disease or myocardial infarction can also influence how your arteries heal after a procedure. While not a guarantee, your family’s history highlights a potential genetic vulnerability that’s important to consider.
4. Do my other health problems increase my restenosis risk?
Section titled “4. Do my other health problems increase my restenosis risk?”Absolutely, conditions like high blood pressure, high cholesterol, and diabetes are major risk factors for restenosis. These health issues create an environment in your body that promotes inflammation and abnormal cell growth in the arteries, making them more prone to re-narrowing after a stent. Effectively managing these conditions is crucial for reducing your risk of needing repeat procedures.
5. Does quitting smoking really help prevent another stent?
Section titled “5. Does quitting smoking really help prevent another stent?”Yes, quitting smoking is one of the most impactful steps you can take to reduce your risk of needing another stent. Smoking significantly contributes to inflammation and vascular damage, which are key drivers of restenosis. By eliminating this risk factor, you improve your body’s ability to heal properly and significantly lower the chances of your artery re-narrowing.
6. Can regular exercise stop my artery from re-narrowing?
Section titled “6. Can regular exercise stop my artery from re-narrowing?”While exercise isn’t a guaranteed “stop” button, regular physical activity is vital for overall cardiovascular health and can indirectly help reduce your restenosis risk. It improves blood flow, reduces inflammation, and helps manage other risk factors like high blood pressure and diabetes. By maintaining a healthy lifestyle including exercise, you create a more favorable environment for your arteries to stay open.
7. Could a DNA test predict if my stent will fail?
Section titled “7. Could a DNA test predict if my stent will fail?”Genetic tests can identify variants associated with an increased susceptibility to restenosis, but they don’t offer a definitive prediction of stent failure for everyone. While research has identified specific genetic markers linked to the process, the full picture is complex and involves many genes and environmental factors. Current tests might offer insights into your general genetic predisposition, but more widespread validation is needed for precise individual predictions.
8. Does my ancestry affect my restenosis risk?
Section titled “8. Does my ancestry affect my restenosis risk?”Yes, your ancestral background can influence your genetic risk for restenosis. Research has shown that genetic findings from one population may not directly apply to others, meaning different ethnic groups might have unique genetic variants or risk profiles. This highlights the importance of personalized risk assessment that considers your specific ancestry to understand your potential susceptibility.
9. Why do some people recover better after a stent?
Section titled “9. Why do some people recover better after a stent?”Individual recovery after a stent can vary significantly due to a combination of genetic and lifestyle factors. Your unique genetic makeup influences how your body responds to the vascular injury from the procedure, affecting inflammation and cell growth in the artery. These genetic differences, combined with factors like managing other health conditions, contribute to why some individuals experience a smoother recovery than others.
10. Even if I’m careful, can my artery still re-narrow?
Section titled “10. Even if I’m careful, can my artery still re-narrow?”Yes, even with diligent care and lifestyle management, your artery can still re-narrow due to underlying genetic predispositions. While managing risk factors is crucial, your individual genetic makeup plays a significant role in how your body heals and responds to the stent. These inherent genetic factors can increase your susceptibility to restenosis regardless of how careful you are with your lifestyle choices.
This FAQ was automatically generated based on current genetic research and may be updated as new information becomes available.
Disclaimer: This information is for educational purposes only and should not be used as a substitute for professional medical advice. Always consult with a healthcare provider for personalized medical guidance.
References
Section titled “References”[1] Verma SS, et al. “Genome-wide association study of platelet reactivity and cardiovascular response in patients treated with clopidogrel: a study by the International Clopidogrel Pharmacogenomics Consortium (ICPC).”Clin Pharmacol Ther, 2020.
[2] Erdmann J, et al. “New susceptibility locus for coronary artery disease on chromosome 3q22.3.”Nat Genet, vol. 41, no. 2, 2009, pp. 280-82.
[3] Wild PS, et al. “Large-scale genome-wide analysis identifies genetic variants associated with cardiac structure and function.” J Clin Invest, 2017.
[4] Holliday EG. “Common variants at 6p21.1 are associated with large artery atherosclerotic stroke.”Nat Genet, vol. 44, no. 10, 2012, pp. 1105-11.
[5] Benjamin, E. J., et al. “Genome-wide association with select biomarker traits in the Framingham Heart Study.” BMC Medical Genetics, vol. 8, no. 1, 2007, p. 55.
[6] Hicks AA et al. “Genetic determinants of circulating sphingolipid concentrations in European populations.” PLoS Genet, vol. 5, no. 9, 2009, e1000672.
[7] Martinelli N et al. “FADS genotypes and desaturase activity estimated by the ratio of arachidonic acid to linoleic acid are associated with inflammation and coronary artery disease.”Am J Clin Nutr, vol. 88, no. 4, 2008, pp. 941–949.
[8] Holland WL, Summers SA. “Sphingolipids, insulin resistance, and metabolic disease: new insights from in vivo manipulation of sphingolipid metabolism.”Endocr Rev, vol. 29, no. 3, 2008, pp. 381–402.
[9] Bielawska AE et al. “Ceramide is involved in triggering of cardiomyocyte apoptosis induced by ischemia and reperfusion.” Am J Pathol, vol. 151, no. 5, 1997, pp. 1257-63.
[10] Aung, N., et al. “Genome-Wide Analysis of Left Ventricular Image-Derived Phenotypes Identifies Fourteen Loci Associated with Cardiac Morphogenesis and Heart Failure Development.”Circulation, vol. 140, no. 14, 2019, pp. 1118–1131.
[11] Pu X et al. “ADAMTS7 cleavage and vascular smooth muscle cell migration is affected by a coronary-artery-disease-associated variant.”Am J Hum Genet, vol. 92, no. 3, 2013, pp. 366–374.
[12] Rusanescu G, Weissleder R & Aikawa E. “Notch signaling in cardiovascular disease and calcification.”Curr Cardiol Rev, vol. 4, no. 2, 2008, pp. 148–56.
[13] Zheng Z et al. “Inhibition of JAK2/STAT3-mediated VEGF upregulation under high glucose conditions by PEDF through a mitochondrial ROS pathway in vitro.”Invest Ophthalmol Vis Sci, vol. 51, no. 1, 2010, pp. 64–71.
[14] Krause MD et al. “Genetic variant at coronary artery disease and ischemic stroke locus 1p32.2 regulates endothelial responses to hemodynamics.”Proc Natl Acad Sci USA, vol. 115, no. 49, 2018, pp. E11349–E11358.
[15] Grunstein M. “Histone acetylation in chromatin structure and transcription.” Nature, vol. 389, no. 6649, 1997, pp. 349–52.
[16] Malhotra R et al. “HDAC9 is implicated in atherosclerotic aortic calcification and affects vascular smooth muscle cell phenotype.”Nat Genet, vol. 51, no. 12, 2019, pp. 1760–1770.
[17] Lobera M et al. “Selective class IIa histone deacetylase inhibition via a nonchelating zinc-binding group.” Nat Chem Biol, vol. 9, no. 5, 2013, pp. 319–25.
[18] Cheng S et al. “Metabolite profiling identifies pathways associated with metabolic risk in humans.” Circulation, vol. 125, no. 18, 2012, pp. 2222–31.
[19] Wang Y et al. “Kynurenine is an endothelium-derived relaxing factor produced during inflammation.”Nat Med, vol. 16, no. 3, 2010, pp. 279–85.
[20] Sallée M et al. “The aryl hydrocarbon receptor-activating effect of uremic toxins from tryptophan metabolism: a new concept to understand cardiovascular complications of chronic kidney disease.”Toxins, vol. 6, no. 3, 2014, pp. 934–49.
[21] Kraus WE et al. “Metabolomic Quantitative Trait Loci (mQTL) Mapping Implicates the Ubiquitin Proteasome System in Cardiovascular Disease Pathogenesis.”PLoS Genet, vol. 11, no. 11, 2015, e1005523.
[22] Marfella R et al. “Increased activity of the ubiquitin-proteasome system in patients with symptomatic carotid disease is associated with enhanced inflammation and may destabilize the atherosclerotic plaque: effects of rosiglitazone treatment.”Journal of the American College of Cardiology, vol. 52, no. 12, 2008, pp. 998-1005. (Journal name was found via search based on context)
[23] Dombroski BA et al. “Gene expression and genetic variation in response to endoplasmic reticulum stress in human cells.” American Journal of Human Genetics, vol. 77, no. 6, 2005, pp. 1018-32.
[24] Civelek M, Lusis AJ. “Systems genetics approaches to understand complex traits.” Nat Rev Genet, vol. 15, no. 1, 2014, pp. 34–48.
[25] Aherrahrou R et al. “Genetic Regulation of Atherosclerosis-Relevant Phenotypes in Human Vascular Smooth Muscle Cells.”Circ Res, vol. 127, no. 10, 2020, pp. 1321–1334.
[26] Franzen O et al. “Cardiometabolic risk loci share downstream cis- and trans-gene regulation across tissues and diseases.” Science, vol. 353, no. 6301, 2016, pp. 827–30.
[27] Lagou V et al. “Genetic Architecture of Adaptive Immune System Identifies Key Immune Regulators.” Cell Reports, vol. 25, no. 3, 2018, pp. 798–810.e6.
[28] Braenne I et al. “Prediction of Causal Candidate Genes in Coronary Artery Disease Loci.”Arterioscler Thromb Vasc Biol, vol. 35, no. 10, 2015, pp. 2207–17.
[29] Tuteja S, et al. “Genetic Variants Associated With Plasma Lipids Are Associated With the Lipid Response to Niacin.” J Am Heart Assoc, 2018.