ArticlePDF Available

X Mechanism PBD Reduces ISR

Authors:
  • Universitas Advent Indonesia (Indonesian Adventist University)

Abstract

Despite the advances in stent design, coating drugs, polymers, the use of intravascular imaging, special devices (such as intravascular lithotripsy, ultra-high-pressure non-compliant balloons, and drug-coated balloons), and improved operator skills and techniques, the problem of in-stent restenosis (ISR) and stent thrombosis (ST) remains a challenge for interventional cardiologists. Atherosclerotic lesions, ISR, and ST, are systemic metabolic chronic inflammatory illnesses with a local manifestation in the coronary vessels. Therefore, we propose managing these systemic illnesses systemically through plant-based diet (PBD) intervention. We explain how PBD intervention can help mitigate atherosclerosis, ISR, and ST. Thus, PBD intervention can overcome biological and epigenetic problems in managing ISR and ST similarly to reversing atherosclerosis. Our team is pioneering the implementation of PBD for cardiology patients who have undergone coronary intervention. So far, the results have been very promising. We hope that in the future, many research studies will be performed to investigate further the role of PBD in decreasing the incidence of ISR and ST.
Volume 9 | Issue 1 | 1
Cardio Open, 2024
Research Article
How a Plant-Based Diet (PBD) Reduces In-Stent Restenosis (ISR) and Stent
rombosis (ST)
Dasaad Mulijono1,2,3*, Albert M Hutapea2,4, I Nyoman E Lister2,5

Department of Cardiology, Bethsaida Hospital,
Tangerang-Indonesia
Indonesia College of Lifestyle Medicine, Indonesia
Department of Cardiology, Faculty of Medicine, Prima
University, Medan-Indonesia
Department of Pharmacy, Faculty of Life Sciences, Advent
University, Bandung-Indonesia
Department of Biomolecular and Physiology, Faculty of
Medicine, Prima University, Medan-Indonesia
Department of Epidemiology, Faculty of Public Health,
University of Indonesia, Jakarta-Indonesia
Department of Community Nutrition, Faculty of Dentistry,
Yarsi University, Jakarta-Indonesia
Citation: Mulijono, D., Hutapea, A. M., Lister, I. N. E., Sudaryo, M. K., Umniyati, H. (2024). How a Plant-Based Diet
(PBD) Reduces In-Stent Restenosis (ISR) and Stent Thrombosis (ST). Cardio Open, 9(1) , 01-15.
Abstract
Despite the advances in stent design, coating drugs, polymers, the use of intravascular imaging, special devices (such as
intravascular lithotripsy, ultra-high-pressure non-compliant balloons, and drug-coated balloons), and improved operator
skills and techniques, the problem of in-stent restenosis (ISR) and stent thrombosis (ST) remains a challenge for interventional


    
overcome biological and epigenetic problems in managing ISR and ST similarly to reversing atherosclerosis. Our team is


PBD in decreasing the incidence of ISR and ST.
 Plant-Based, Supplements, In-Stent Restenosis, Stent Thrombosis
1. Introduction
The American Heart Association reports that roughly 600,000
coronary stent procedures are conducted annually in the United
States [1], with more than three million stents utilized globally.
Despite progress in cardiovascular health and interventional
cardiology, coronary ISR and ST continue to present a
challenge to interventional cardiologists. Despite the signicant
advancements in interventional techniques and intracoronary
imaging, there is no universally accepted approach to managing
either condition. The lack of a denitive strategy is a testament
to the underlying mechanisms' complexity [2].
Balloon dilation and stent implantation may result in vascular
injury, which can lead to restenosis. Mechanical stretch,
endothelial denudation, and subintimal hemorrhage may
trigger an inammatory response, which subsequently induces
a proliferative process. The activation of vascular smooth
muscle cells (VSMC), including proliferation, migration, and
dierentiation, as well as matrix metalloproteinase activation,
DNA, and extracellular matrix synthesis, can all contribute to
neointimal hyperplasia formation [3,4]. Drug-eluting stents
(DES) have anti-inammatory, immunomodulatory, and
antiproliferative properties, which help the coronary vessel heal
properly. Nonetheless, if the suitable dose of the medication
does not elute at the appropriate moment, in cases of drug
resistance or if persistent systemic inammation exists, or if the
patient exhibits an allergy to the stent platform, stent polymer,
or in the formation of neoatherosclerosis, all of these factors can
contribute to the development of ISR and ST.
*Corresponding Author
Prof. Dasaad Mulijono, Department of Cardiology, Bethsaida Hospital,
Tangerang-Indonesia; Indonesia College of Lifestyle Medicine, Indonesia;
Department of Cardiology, Faculty of Medicine, Prima University, Medan-
Indonesia.
Submitted: 2024, Mar 29; Accepted: 2024, Apr 23 ; Published: 2024, Apr 29
ISSN: 2476-230X
Cardiology: Open Access
https://doi.org/10.33140/COA.09.01.05
Volume 9 | Issue 1 | 2
Cardio Open, 2024
Various factors may be implicated in the development of DES
restenosis, including patient factors, lesion factors, procedural
factors, biological, genetic (epi-genetic), mechanical, and
technical elements. In the context of this paper, we will focus on the
patient, biological, and epigenetic factors that can be eectively
managed with the help of a PBD. The biological mechanisms
that could play a role in the emergence of ISR and ST involve
inammatory/ immunity processes, heightened sensitivity, drug
resistance, the development of neoatherosclerosis, and defective
re-endothelialization within the coronary artery where the stent
was placed [5] (Please refer to Figure 1). Consequently, either
the vessel wall heals or neointimal hyperplasia and/or neo-
atherosclerosis develops. Identifying the core molecular and
signaling pathways responsible for inammatory, immune,
hypersensitivity, vascular healing, and thrombotic processes is
crucial for developing eective therapeutic approaches aimed at
overcoming the primary challenge to the success of percutaneous
coronary intervention (PCI). This is necessary to prevent the
occurrence of ISR and ST as eectively as possible [6-8].
*Red highlights are the ones that PBD has a role
Figure 1: Risk Factors and Mechanisms of DES ISR
Numerous studies have underscored the critical signicance of
PBDs in managing chronic inammatory diseases, including
atherosclerosis, hyperlipidemia, obesity, non-insulin-dependent
diabetes mellitus (NIDDM), and hypertension. It has been
widely acknowledged that PBD may not only contribute to the
prevention of atherosclerosis but may also cause regression of
coronary plaques that have occurred [9-15]. To our knowledge,
no published work has explored the role of PBDs in mitigating
ISR and ST. The role of PBDs as an anti-inammatory,
immunomodulatory, and assisting vascular healing has been
recognized and published as benecial in chronic inammatory
diseases, especially atherosclerosis. Consuming PBD in
conjunction with anti-platelet drugs may aid in the mitigation of
thrombosis processes, ultimately reducing the risk of developing
acute coronary syndrome (ACS) and subacute thrombosis [16-
22].
Atherosclerosis is a systemic disease resulting from metabolic
dysfunction and persistent inammation rather than a localized
process. Consequently, it is classied as a metabolic chronic
 condition. Patients who have experienced
coronary obstruction due to the atherosclerosis process have
typically exhibited a state of metabolic disorder and systemic
inammation for a prolonged period. Cardiologists who
exclusively employ interventions such as stent implantation to
manage systemic diseases are likely to be disappointed if they do
not address these systemic issues. Following stent implantation,
the artery is disturbed locally by the balloon, and foreign body
implantation (stent) will undergo a healing process for the local
injury. Nevertheless, the systemic environment will signicantly
inuence the local healing process. At the very least, the
atherosclerosis process, which is a systemic process, will still
be developing. Even if patients do not experience ISR or ST,
there may still be an atherosclerosis process occurring that could
cause coronary stenosis in new vessels dierent from the vessels
that have been treated with coronary interventions. Therefore, it
is not judicious to manage ISR and ST solely by addressing the
local vessel issue.
Our cardiac center, Bethsaida Hospital in Indonesia, conducted
interventional studies on approximately 1000 cardiology
patients. These patients implemented PBDs months before they
underwent their PCI procedures using various second and third-
generation DES. Our research has been ongoing for nearly ve
3
*Red highlights are the ones that PBD has a role
Fig. 1. Risk factors and mechanisms of DES ISR
Numerous studies have underscored the critical significance of PBDs in managing chronic
inflammatory diseases, including atherosclerosis, hyperlipidemia, obesity, non-insulin-dependent
diabetes mellitus (NIDDM), and hypertension. It has been widely acknowledged that PBD may not only
contribute to the prevention of atherosclerosis but may also cause regression of coronary plaques that
have occurred [9-15]. To our knowledge, no published work has explored the role of PBDs in mitigating
ISR and ST. The role of PBDs as an anti-inflammatory, immunomodulatory, and assisting vascular healing
has been recognized and published as beneficial in chronic inflammatory diseases, especially
atherosclerosis. Consuming PBD in conjunction with anti-platelet drugs may aid in the mitigation of
thrombosis processes, ultimately reducing the risk of developing acute coronary syndrome (ACS) and
subacute thrombosis [16-22].
Atherosclerosis is a systemic disease resulting from metabolic dysfunction and persistent
inflammation rather than a localized process. Consequently, it is classified as a metabolic chronic
inflammatory condition. Patients who have experienced coronary obstruction due to the atherosclerosis
process have typically exhibited a state of metabolic disorder and systemic inflammation for a prolonged
period. Cardiologists who exclusively employ interventions such as stent implantation to manage
systemic diseases are likely to be disappointed if they do not address these systemic issues. Following
stent implantation, the artery is disturbed locally by the balloon, and foreign body implantation (stent)
will undergo a healing process for the local injury. Nevertheless, the systemic environment will
significantly influence the local healing process. At the very least, the atherosclerosis process, which is a
Volume 9 | Issue 1 | 3
Cardio Open, 2024
years, making our center the only one in Indonesia to pioneer
PBDs for patients who receive PCI procedures. Our data
suggests that only a small percentage of individuals who adhere
to PBD experience ISR in their subsequent annual coronary
angiography follow-up. Our research ndings suggest that the
occurrence of ISR is signicantly lower in patients who adhere
to our PBD interventions, at approximately 2-3%, as compared
to those who do not follow our interventions, the omnivorous
patients, who have an ISR rate of 10-20%; similar with the
results of other studies [23,24]. We did not observe any early ST
among the participants in our PBD intervention within the rst
year following their stent implantations. In contrast, the rate of
ST in the NPBD group was 1% [25].
It would undoubtedly be more prudent to initiate anti-
atherosclerosis, anti-ISR, and anti-ST management as early as
feasible in relation to the development of ISR and ST. It is the
responsibility of the interventionalist to manage the patient's
metabolic and inammatory condition well in advance of their
scheduled intervention. In the acute setting, for instance, when
performing an intervention on patients with acute coronary
syndrome, similar issues arise. Sadly, interventionists often
underestimate the importance of managing the patient's
metabolic and chronic inammatory condition post-intervention
to minimize the occurrence of ISR and ST in the long run.
This paper emphasizes the signicance of launching metabolic
interventions as soon as feasible, with the aim of rectifying
metabolic abnormalities, restoring endothelial function,
reducing systemic inammation, preventing allergic reactions,
enhancing the availability of nitric oxide (NO), fostering a
healthy microbiota, correcting mitochondrial dysfunction, and
improving telomere as part of epigenetic adjustments. By doing
so, a solid foundation can be established prior to the occurrence
of injuries resulting from coronary interventions.
Given the promising outcomes that our cardiac center has
achieved, additional research is warranted to assess the ecacy
of PBDs in reducing the incidence of ISR and ST before the
broader application of this intervention in the cardiology
community, particularly among interventional cardiologists.
This would ultimately benet numerous patients in the future
and elevate the reputation of the interventional cardiology
society as a pioneer in promoting PBDs, thereby surpassing
other methods (optimal medical therapy (OMT) and bypass
surgery) in achieving optimal coronary intervention results.
2. Mechanism PBD in Reducing ISR and ST

the Risk of Atherosclerosis, ISR, and ST
The key mechanism by which a healthy PBD can reduce the
probability of atherosclerosis, ISR, and ST is by changing one's
diet to PBD, thereby eliminating the consumption of foods that
contribute to coronary heart disease in the rst place. Adhering to
an omnivorous diet and consuming foods such as sugary products,
processed meat, processed snacks, red meats, poultry, dairy
products, and eggs signicantly increases the risk of developing
atherosclerosis and the likelihood of ISR and ST. These
unhealthy eating behaviors have been linked to dyslipidemia,
insulin resistance, hypertension, glucose intolerance, endothelial
dysfunction, chronic systemic inammation, increased oxidative
stress, elevated trimethylamine N-oxide (TMAO), low NO,
gut dysbiosis, mitochondrial damage, and shortened telomeres
(accelerating the aging process) [26-30]. All of these are widely
accepted as risk factors for the development of atherosclerosis.
2.2. Mechanism of Nitric Oxide (NO) in Reducing
Atherosclerosis, ISR, and ST
The endothelium exhibits anti-atherosclerotic properties, which
can be ascribed to the presence of NO. NO prevents monocytes
and leukocytes from adhering to the endothelium, inhibits
platelet-vessel wall interactions, and suppresses the proliferation
of VSMC, which are important processes in developing ISR and
ST. Additionally, NO promotes coronary dilation and increases
coronary blood ow [31]. Furthermore, NO plays a vital role in
endothelial regeneration, which is crucial for the healing of the
coronary artery after stent placement [5].
Conditions such as obesity, hypertension, hyperlipidemia, insulin
resistance, and glucose intolerance, which are associated with
atherosclerosis risk factors, can decrease NO release into the
coronary wall due to impaired synthesis or excessive degradation
[32]. PBD has been found to improve chronic inammatory
diseases, including obesity, hypertension, hyperlipidemia, insulin
resistance, and elevated glucose levels, and it may also reverse
atherosclerosis [8-15]. Notably, NO production decreases with
age, which is signicant since atherosclerosis primarily aects
older populations [33]. To measure NO, we may use a salivary
strip, which has a 96% accuracy rate [34].
We can enhance our patients' NO availability by altering their
diet and lifestyle. The process of converting nitrate-rich foods,
such as green leafy vegetables, into NO is illustrated in Figure
2. Please note that it is crucial to consider that certain vegetables
that are high in nitrates can have their nitrate levels reduced
through cooking. Food preparation is also a signicant factor
in determining the eectiveness or benecial eects of the food
[35]. Many studies of PBDs have not given much consideration
to the quality of the food consumed by their participants,
including the selection, quantity, and processing of the food.
This lack of attention may fail to maximize the benets of PBDs.
Another important factor is the potential   
vitamins, minerals, and micronutrients often present in PBD.
Therefore, it is crucial to ensure that these important nutrients
are adequately supplemented, as a deciency in these nutrients
may not only decrease the ecacy of a PBD but can also be
potentially harmful [36,37].
Volume 9 | Issue 1 | 4
Cardio Open, 2024
Figure 2: The Entero-Salivary Circulation of Nitrate in Human
Recent progress has been made in applying NO coatings on stents,
demonstrating the potential for preventing ISR and ST [38,39].
NO strategies promote normal endothelial cell growth, prevent
neointimal hyperplasia formation, inhibit the proliferation of
VSMCs, increase vasodilation, and decrease platelet activation
and aggregation. This research should enlighten those who
advocate for administering PBD to their patients, as PBD can
potentially increase circulating levels of NO, which may be
more eective than local NO administration. This is because the
pathogenesis of atherosclerosis and the occurrence of ISR and
ST are driven by systemic processes rather than a local reaction
post-coronary intervention.
       
Crucial Factor in Repairing Endothelial Dysfunction
In 1998, a small randomized controlled trial known as the
Lifestyle Heart Trial was conducted by Ornish et al. The study
demonstrated that intensive lifestyle changes with PBD could
reverse coronary atherosclerosis. The most important mechanism
of this process is repairing endothelial dysfunction [40]. In 2014,
Esselstyn et al. presented a famous image, as depicted in Figure
3, which illustrated that strict PBD (without oil) can regress
coronary stenosis [41].
7
Figure 3. Reversal of coronary artery disease
It has been a decade since the remarkable research studies on atherosclerosis have garnered
significant praise and acceptance within the PBD community and among practitioners who recommend
PBD. Despite this, the widespread adoption of recommending PBD to patients has not been extensively
embraced within the interventional cardiology community. As an interventional cardiologist myself, I
initially had reservations about these findings. However, after being diagnosed with coronary stenosis
on my coronary multi-slice CT angiography, I decided to follow the advice of these PBD experts. To my
surprise, after three years, my LAD stenosis of 50% had regressed to 20%, and my CT calcium reading
had decreased by 30%. Moreover, I found that this method effectively regressed atherosclerosis and
reduced the incidence of my patient’s ISR and ST.
Eating poor-quality food that is high in sugar (refined carbohydrate), devoid of fiber-phytonutrients,
highly processed, and contains saturated and trans fats, cholesterol, and chemicals that promote chronic
inflammation is a major contributor to the development of atherosclerosis. The consumption of these
unhealthy foods has been demonstrated to increase levels of LDL cholesterol, triglycerides,
apolipoprotein (a), apolipoprotein (b), C-reactive protein (CRP), pro-inflammatory mediators, pro-
inflammatory chemokines/ cytokines, Trimethylamine N-oxide (TMAO), persistent organic pollutants
(POPs), oxidative stress, tumor promotion, and cell proliferation, among other factors. All of these
factors play significant role in the development of atherosclerosis, ISR, and ST [42,43]. On the contrary,
eating healthy PBD with adequate supplementations such as vitamins B12 and D and minerals will
enhance our body to fight against inflammation. Healthy foods such as vegetables, fruits, and legumes
contain carotenoids, isoflavones, phytoestrogens, and phytosterols, which have been shown to prevent
atherosclerosis. These polyphenols and phytochemicals' role in molecular signaling are anti-
inflammatory, antiplatelet aggregation, inhibitor to VSMCs proliferation and migration, and safeguard
for lipid oxidation (ox-LDL) [44,45]. Oxidized LDL, in addition to its infamous role in causing
atherosclerosis, also plays a significant part in the progression of ISR [46]. Healthy PBD will also help to
restore endothelial function and enhance the process of endothelialization post-traumatic balloon
inflation and stent insertion during coronary intervention.
Figure 3: Reversal of coronary artery disease
Volume 9 | Issue 1 | 5
Cardio Open, 2024
It has been a decade since the remarkable research studies on
atherosclerosis have garnered signicant praise and acceptance
within the PBD community and among practitioners who
recommend PBD. Despite this, the widespread adoption of
recommending PBD to patients has not been extensively
embraced within the interventional cardiology community. As
an interventional cardiologist myself, I initially had reservations
about these ndings. However, after being diagnosed with
coronary stenosis on my coronary multi-slice CT angiography,
I decided to follow the advice of these PBD experts. To my
surprise, after three years, my LAD stenosis of 50% had
regressed to 30%, and my CT calcium reading had decreased by
30%. Moreover, I found that this method eectively regressed
atherosclerosis and reduced the incidence of my patient’s ISR
and ST.
Eating poor-quality food that is high in sugar (rened
carbohydrate), devoid of ber-phytonutrients, highly processed,
and contains saturated and trans fats, cholesterol, and chemicals
that promote chronic inammation is a major contributor to
the development of atherosclerosis. The consumption of these
unhealthy foods has been demonstrated to increase levels of LDL
cholesterol, triglycerides, apolipoprotein (a), apolipoprotein (b),
C-reactive protein (CRP), pro-inammatory mediators, pro-
inammatory chemokines/ cytokines, Trimethylamine N-oxide
(TMAO), persistent organic pollutants (POPs), oxidative stress,
tumor promotion, and cell proliferation, among other factors.
All of these factors play signicant role in the development of
atherosclerosis, ISR, and ST [42,43]. On the contrary, eating
healthy PBD with adequate supplementations such as vitamins
B12 and D and minerals will enhance our body to ght against
inammation. Healthy foods such as vegetables, fruits, and
legumes contain carotenoids, isoavones, phytoestrogens, and
phytosterols, which have been shown to prevent atherosclerosis.
These polyphenols and phytochemicals' role in molecular
signaling are anti-inammatory, antiplatelet aggregation,
inhibitor to VSMCs proliferation and migration, and safeguard
for lipid oxidation (ox-LDL) [44,45]. Oxidized LDL, in addition
to its infamous role in causing atherosclerosis, also plays a
signicant part in the progression of ISR [46]. Healthy PBD will
also help to restore endothelial function and enhance the process
of endothelialization post-traumatic balloon ination and stent
insertion during coronary intervention.
Paclitaxel and sirolimus, which are used in the coating of DES,
possess therapeutic properties that can reduce intimal hyperplasia
and promote re-endothelialization, thereby decreasing the
incidence of ISR in bare-metal stents from 30-50% to 10-20%
in DES.
The formation of an excessive amount of reactive oxygen species
(ROS) results in oxidative stress, a signicant contributing
factor to the emergence and progression of atherosclerosis
[47]. Robust evidence indicates that ROS, through their pro-
atherogenic eects, play a crucial role in several pathological
processes, including inammation, endothelial dysfunction,
and dysregulated lipid metabolism. Moreover, ROS has been
demonstrated to impair mitochondrial function, which is critical
to ensure eective healing following coronary interventions.
Tissue damage resulting from coronary intervention and stent
implantation triggers increased production of ROS, which plays
a role in the initial proliferation, migration, and apoptosis of
VSMC, contributing to the development of ISR [48]. Vascular
damage during coronary intervention often results in the
overproduction of ROS, leading to platelet dysfunction and
subsequent abnormal activation and aggregation, which can
contribute to the formation of blood clots and ST [49].
Carotenoids, prominently present in foods like pumpkin, carrots,
tomatoes, green leafy vegetables, broccoli, and bell peppers, are
lipophilic antioxidants that can mitigate the detrimental eects
of ROS. Citrus fruits, bell peppers, strawberries, kiwis, broccoli,
green leafy vegetables, grains, and legumes are rich in vitamin
C and the B-complex group. Both vitamins function as potent
antioxidants, helping combat ROS's negative eects. Adopting
healthy dietary habits in conjunction with PBD can not only
guard against the onset of atherosclerosis but can also protect
against the potential complications of coronary interventions
such as ISR or ST. It is important to note that individuals who
have adopted healthy and correct PBDs may never need coronary
intervention in their lifetimes. Consuming foods that elevate
the dietary inammatory index (DII) is advisable to counteract
the inammation and ROS implications in the progression of
atherosclerosis and the development of ISR and ST, as depicted
in Table 1.
Table 1: List of foods that Reduce and Increase DII
Foods that reduce DII Foods that increase DII
Red meat (steak and hamburgers)
Animal products (including eggs and dairy products)
Processed meat
Commercial baked goods
White our (bread and noodles), white rice
Deep-fried foods
Sugary products
Products with trans-fats
Saturated fats (especially animal fats)
Cholesterol (red meats, processed meats, eggs, fried
foods and dairy products)
Plant-based proteins (beans, lentils, chickpeas, edamame,
hemp seeds, tofu, tempeh, and nuts)
Whole grains (oatmeal, buckwheat, quinoa, pigmented rice)
Starchy vegetables (sweet potatoes and beets)
Seeds (axseeds and chia seeds)
Green leafy vegetables (raw)
Colorful vegetables (raw)
Fruits (berries, apples, grapes, oranges, peaches, gs,
bananas, and kiwi)
Spices and herbs (turmeric, ginger, cumin, peppermint,
cinnamon, chili, parsley, bay leaf, and basil)
Volume 9 | Issue 1 | 6
Cardio Open, 2024
2.4. PBD Decreases Systemic Allergic Reactions and May
Prevent Stent Hypersensitivity/ Allergic Reaction
In contrast to the omnivorous diet, which contains many pro-
inammatory nutrients. PBDs are enriched with micronutrients
and dietary avonoids that are associated with not only anti-
inammatory but also anti-allergy eects.
There have been suggestions that ISR and ST may be linked to
allergies or hypersensitivity to stent materials, including metal,
polymer, and eluting drugs. Identifying whether individuals who
experience ISR or ST are allergic to these materials is a complex
challenge, and it has only recently been raised as a potential
cause. In certain circumstances, an allergic patch test may be
administered; however, even if the test returns a negative result,
the likelihood of the patient being allergic to the stent materials
cannot be entirely eliminated. The most common allergic
reaction to stent materials is type 4 hypersensitivity, which is
T-helper cell-mediated [50]. This type of allergic reaction can
be inhibited by avonoids, which are found in abundance in
PBDs. Flavonoids can inhibit the activation of T cells, reduce
the production of inammatory biomarkers, decrease IgE
production and binding, and prevent degranulation of mast cells
that release histamine, IL-4, IL-5, IL-6, IL-13, and MCP-1 [51].
Given that it is challenging to predict which patients will develop
hypersensitivity to the stent we employ, it is prudent to adopt
a diet that generally possesses anti-allergic properties. This
approach helps minimize the likelihood of patients developing
ISR and ST due to allergic reactions [51,52]. Moreover, using
drugs to eliminate allergies, as experienced in the past, normally
carries side eects and will reduce patients' compliance to
adhere.

The gut microbiota is a complex community of microorganisms
that inhabit the gastrointestinal tract and play a crucial role
in regulating various physiological processes, including
metabolism, inammation, and immunity. Recent research
has revealed that the bacteria in atherosclerotic plaques share
DNA similarities with gut bacteria. One of the metabolites
produced by the gut microbiota, TMAO, is primarily derived
from dietary sources such as choline, betaine, and L-carnitine
found in an omnivorous diet. TMAO has been linked to the
development and progression of atherosclerosis due to its pro-
inammatory properties and inhibition of reverse cholesterol
transport. Additionally, elevated levels of plasma TMAO have
been associated with neoatherosclerosis, ruptured plaque,
and thrombosis, which may contribute to ISR and ST [53,54].
With a high intake of red meat, Omnivore's risk for developing
atherosclerosis is much higher compared to those who
consume PBDs [55]. Switching to a PBD may protect against
atherosclerosis by promoting endothelial protective mechanisms
and lowering TMAO, a pro-atherosclerotic metabolite.
Atherosclerosis patients often exhibit elevated production
of TMAO-producing microbiomes and reduced short-chain
fatty acid (SCFA)- producing species. SCFAs, a class of anti-
inammatory metabolites, are generated through a ber-rich
diet. By increasing the expression of anti-inammatory factors,
improving the integrity of the intestinal barrier, and inhibiting pro-
inammatory cytokines, SCFAs help to reduce inammation, a
key risk factor for coronary heart disease (CHD). An imbalance
between SCFAs and TMAOs can contribute to inammation and
exacerbate the risk of CHD.
Plant-based dieters have been shown to possess a microbiome
capable of producing a signicant amount of SCFAs, including
butyrate, and increased Acetyl-CoA and X4-aminobutyrate-
succinate pathways. Additionally, PBDs have been linked to
a lowered post-prandial glycemic response (PPGR), lowered
inammatory markers, improved cardio-metabolic health, low
T-cell repertoire diversity, and low IgE expression levels. These
benecial microbiome characteristics may help protect against
the development of CHD and other chronic inammatory
conditions like obesity, hypertension, hyperlipidemia, glucose
intolerance, insulin resistance, allergic reaction, and the
likelihood of ISR and ST.
Changes in gut microbiota have the potential to impact gut
permeability, which can lead to the translocation of bacterial
DNA, lipopolysaccharides (LPS), and proinammatory
cytokines into circulation. This phenomenon, known as leaky
gut syndrome, can result in the absorption of metabolites and
endotoxins into the bloodstream. Leaky gut syndrome has
been linked to the development of atherosclerosis and acute
coronary syndrome [56]. SCFAs can reduce gut permeability
by decreasing nuclear factor-kappa B (NF-kB) activation and
reducing proinammatory cytokines such as IL-1b, IL-6, IL-
8, and TNF-α [57]. Low consumption of plant-based foods
may lead to increased penetration of the intestinal barrier, as a
low-ber diet triggers a shift from ber-degrading to mucus-
degrading bacteria. This could promote a hyperactive immune
response, conceivably with the production of pro-inammatory
metabolites that fuel the disease process.
A healthy diet includes non-digestible carbohydrates, such as
resistant starch, soluble and insoluble dietary ber, and plant
wall polysaccharides and oligosaccharides, fermented by
benecial gut bacteria to produce butyrate and other SCFAs.
These SCFAs possess anti-inammatory properties and have
the ability to strengthen the intestinal barrier, thereby promoting
optimal gut health. Moreover, SCFAs can inhibit the formation
of foam cells by stimulating the expression of interleukin-10
(IL-10) and decreasing the production of pro-inammatory
cytokines by the endothelium. This contributes to the recovery
of endothelial damage post-coronary intervention and reduces
the occurrence of atherosclerosis, ISR, and ST.
Foods high in ber, including barley, wheat bran, brown rice,
and other whole grains, legumes, fruits, and vegetables, as
well as prebiotics like fructo-oligosaccharides, are commonly
consumed by individuals following a PBD diet. Plant-based
foods also contain polyphenols such as lignans, isoavones,
anthocyanins, and avonols, as well as other phytochemicals
like carotenoids and phytosterols, which are metabolized into
bioactive compounds by benecial microbes, conferring health
Volume 9 | Issue 1 | 7
Cardio Open, 2024
benets and exhibiting anti-inammatory and antioxidant
activity. Phytochemicals have been shown to increase the
populations of benecial bacteria such as Lactobacillus and
, which are the primary species found in
probiotic supplements that are taken to improve gut health. In
addition, ber-rich plant foods like nuts (including walnuts,
almonds, and pistachios) have been found to have prebiotic
eects, leading to increases in butyrate-producing microbes and
other benecial microbes. Overall, the composition of the gut
microbiome is greatly inuenced by dietary ber, polyphenols,
and other phytochemicals and their metabolites, consumed in
greater quantities by those following PBDs [58]. Certainly, an
omnivorous diet that is low in ber, devoid of polyphenols and
phytochemicals, and decient in SCFAs while being high in
TMAO can contribute to the development of new atherosclerotic
plaques and increase the incidence of ISR and ST in patients
who have undergone coronary interventions. Thus, the gut and
the heart interaction is known as the gut-heart axis [59,60].

2.6. Caloric Restriction is an Essential Factor in Combating

In 2022, 43% of adults aged 18 years or older were considered
overweight, and 16% were diagnosed with obesity [61].
Overeating is a pervasive issue in contemporary society,
inuenced by various factors, including the availability of
copious amounts of food, stress, emotional eating, and social
tradition. While partaking in occasional feasts is an ingrained
aspect of human nature, persistent overeating can severely aect
an individual's health and society. Based on our analysis of
over 10,000 patients who have visited our cardiology clinic, we
have discovered that a signicant majority, or 90%, would be
classied as overweight using the BMI cuto point of 21 kg/
m2. This nding is not surprising, given the prevalence of CHD
among this group. Our clinic oers a lifestyle program to help
patients maintain their ideal weight by restricting their calorie
intake.
Caloric Restriction (CR), which involves reducing the intake of
calories without depriving essential nutrients, has consistently
demonstrated anti-aging eects across a broad range of
organisms. Furthermore, it has been shown to protect against
age-related diseases such as cardiovascular disease, diabetes,
hypertension, hypercholesterolemia, and cancer. CR achieves
this by reducing oxidative stress and inammation while
enhancing the production and activity of antioxidant and anti-
inammatory substances, thereby improving the body's overall
balance. Research has also shown that CR can improve overall
health and well-being, optimize energy metabolism, enhance
cellular protection, improve insulin sensitivity and glucose
regulation, induce functional changes in the neuroendocrine
systems, reduce oxidative damage and inammation, and even
shape the gut microbiota [62].
Implementing CR may improve cardiovascular health. By
promoting the activity of endothelial nitric oxide synthase
(eNOS) and sirtuin 1 (SIRT1), CR can enhance endothelial
function, leading to vasodilation, regulation of blood pressure,
and improved blood ow. Additionally, CR may also reduce the
development of atherosclerosis [63]. Eating an unhealthy diet
with calorie excess will certainly promote the development of
atherosclerosis, ISR, and ST [64]. CR protects DNA methylation,
histone modication, and non-coding RNA (nc-RNA), which
prevent VSMC proliferation, migration, and inammation,
which play an important role in developing atherosclerosis
and ISR [65,66]. Most individuals who follow PBDs generally
do not necessitate CR since these diets inherently promote a
sense of satiety. Additionally, the majority of items on PBD
menus are low in calories. In stark contrast, most omnivorous
individuals frequently experience feelings of hunger due to the
malfunctioning of their hormonal regulators, typically caused by
consuming unhealthy foods with minimal nutrients and empty
calories.
2.7. The Role of PBD as Mitochondrial Protectors
As we age, the powerhouses of our cells, called mitochondria,
undergo changes that lead to a decline in their function. This
decline is caused by the accumulation of oxidative damage and
mutations that induce ROS. As a result, the volume, integrity, and
functionality of mitochondrial DNA (mtDNA) decrease. In older
1
Volume 9 | Issue 1 | 8
Cardio Open, 2024
adults, mitochondria are characterized by signicant increases in
ROS and decreased antioxidant defense, which lead to impaired
functions. These include decreased ATP production, lowered
oxidative capacity, and reduced oxidative phosphorylation.
Additionally, with aging, mitochondrial biogenesis will decline
due to inhibition of mitophagy and alterations in mitochondrial
dynamics (ssion and fusion). Mitophagy, an autophagy
process that eliminates defective mitochondria, also deteriorates
with aging. An excessive generation of ROS characterizes
acute and chronic inammatory diseases, which can cause
damage to mtDNA, mitochondrial proteins, and lipids. This
negatively aects normal mitochondrial function and dynamics.
Inammation is generated by various mitochondrial products
called damage-associated molecular patterns (DAMPs) and is
released into the cytosol or extracellular environment. Protective
measures are in place to prevent mitochondria from triggering
harmful inammatory responses, such as disposing of damaged
mitochondria through mitophagy. However, if these mechanisms
are overwhelmed or not functioning correctly, inammatory
reactions instigated by mitochondria can become problematic,
contribute to developing disorders, and impede healing [67].
Atherosclerosis arises due to the malfunction of endothelial
cells and the accumulation of lipids. Mitochondrial malfunction
can have deleterious eects on various cells in the arterial wall,
such as endothelial cells, smooth muscle cells, macrophages,
monocytes, and lymphocytes. This can result in increased levels
of ROS, chronic inammation, oxidative stress, and intracellular
lipid deposition. Also, mitochondrial dysfunction is crucial
in chronic inammatory diseases, including hypertension,
obesity, hyperlipidemia, insulin resistance, and atherosclerosis.
Mitochondria are essential for cellular metabolism, energy
production, and cell survival. When these mechanisms
are impaired, it can lead to cellular dysfunction, excessive
ROS production, cellular damage, and the initiation of the
inammatory response. Mitochondria have been implicated in
the pathogenesis of cardiovascular diseases [68].
Normal mitochondrial functioning is vital for cell survival, and
it is determined by the equilibrium between mitophagy (the
autophagic degradation of mitochondria) and mitochondria
biogenesis (fusion and ssion). Mitophagy is crucial
in eliminating damaged mitochondria, preventing their
accumulation and associated cell malfunction and subsequent
apoptosis. Mitophagy acts as a barrier against ROS accumulation
in damaged mitochondria. Inadequate mitophagy has been linked
to the development of atherosclerosis, which contributes to
progressive cell death, cell stress, and ROS accumulation. This
results in the formation of a necrotic core and the destabilization
of the atherosclerosis plaque. The mitochondrial dysfunction
resulting in increased ROS generation causes damage to
mtDNA, which is more susceptible to mutagenesis than
nuclear DNA due to dierences in DNA packaging and repair
mechanisms. The accumulation of specic mtDNA mutations
further contributes to mitochondrial dysfunction and the
progression of atherosclerotic lesions and is involved in plaque
destabilization processes. Mutations in mtDNA can decrease the
synthesis of respiratory complexes and weaken mitochondrial
respiration in VSMC and macrophages. ROS plays a crucial
role in damaging mtDNA, decreasing the amount of coding
mtDNA, impairing mitochondrial protein synthesis, altering
mitochondrial membrane potential, and decreasing total ATP
production in smooth muscle and endothelial cells, all of which
are important factors in the development of atherosclerosis.
Mitochondrial genome changes, such as an increase in mtDNA
copy number, mtDNA methylation, and the appearance of
mutations (insertions, deletions, and insertions), are associated
with the development of atherosclerosis. Damaged mtDNA was
detected before the appearance of other atherosclerotic signs,
suggesting that mtDNA damage may be the primary event
that induces excessive ROS generation, violates mitochondrial
membrane potential, and causes mitochondrial dysfunction,
followed by the release of cytochrome C and the activation
of apoptotic pathways. Furthermore, damaged mtDNA can be
recognized by the body as an endogenous DAMP, which triggers
the inammatory response [69].
Vascular endothelial cells play a crucial role in regulating
apoptosis and NO production. They are also important in
modulating cell signaling and the cellular response to stress,
which this type of cell is particularly sensitive to. In addition
to their barrier function, endothelial cells are involved in
regulating vascular tone, the transport of blood plasma
molecules, hemostasis, inammation, and lipid metabolism. A
healthy endothelial barrier prevents the inltration of circulating
cells, such as monocytes/macrophages, into the vascular
wall. Endothelial dysfunction is one of the earliest signs of
mitochondrial disorder [70].
Mitochondrial dysfunction was observed in VSMC isolated
from human atherosclerotic plaques. These cells exhibited
the presence of mtDNA mutations, reduced mitochondrial
mass, and defects in ATP synthase. Impaired mitochondrial
dynamics in SMC from atherosclerotic plaques can contribute
to their proliferation. The morphology of the mitochondria is
determined by fusion and ssion processes, which also control
the eectiveness of ATP synthesis, oxygen consumption, and
the potential of the mitochondrial membrane. These processes
are regulated by mitochondria-associated small GTPases,
including the mitochondrial fusion protein mitofusin 2 (Mfn2),
which reduces proliferation and promotes apoptosis of SMCs.
Mitochondrial malfunctioning could potentially exacerbate the
activation of Poly (ADP-ribose) polymerase-1 (PARP-1), a
critical component in the pathogenesis of atherosclerosis.
Mitochondrial oxidative stress associated with atherosclerosis
contributes to inammation activation through the NF-kB-
mediated pathway in macrophages. This process is characterized
by a system of pro-inammatory cytokines, adhesion molecules,
and growth factors that, in turn, can trigger inammatory
signaling [71].
Chronic metabolic-inammatory conditions can impact
mitochondrial functionality and energy production capacity.
Addressing these conditions, including obesity, hypertension,
glucose intolerance, hyperlipidemia, and atherosclerosis, can
Volume 9 | Issue 1 | 9
Cardio Open, 2024
aid in mitochondrial regeneration. The relationship between
mitochondria and metabolic-inammatory disorders is reciprocal
in that both conditions can exacerbate one another. Metabolic-
inammatory disorders can lead to mitochondrial dysfunction,
and conversely, mitochondrial dysfunction can contribute to the
development of these disorders, as illustrated in Fig. 5. Thus,
the term secondary mitochondrial dysfunction is used since,
typically, it is acquired during a person's lifetime and is often
associated with chronic diseases and age-related changes.
           
Mechanisms of Mitochondrial Dysfunction Induce Atherosclerosis, ISR, and ST
Promoting healthy mitochondrial function can be achieved
through various lifestyle adjustments, including following a
balanced diet, engaging in regular physical activity, and taking
supplements containing vitamins (such as vitamin C, E, and
biotin), minerals (zinc), or nutraceuticals (ubiquinone or CoQ10,
NMN-precursor of NAD+, Quercetin, Astaxanthin, Resveratrol)
[72]. The concept of Mitochondrial nutrients has gained
momentum in recent years and refers to the essential nutrients
necessary to maintain optimal mitochondrial function. Recent
investigations have demonstrated that a Mediterranean diet,
characterized by the consumption of plant-derived compounds
such as vegetables, fruits, and nuts, which are rich in polyphenols
(isoavones, phytoestrogens) and other phytochemicals
(phytosterols, carotenoids, luteolin, organosulfur, terpenes,
saponins) as well as polyunsaturated fatty acids from axseeds
and sunower seeds and can enhance mitochondrial function
[73]. Polyphenols have been demonstrated to hinder the
activity of PARP-1, a crucial component in the pathogenesis of
atherosclerosis [74].
Enhancing mitochondrial function can potentially expedite the
healing process after coronary interventions, such as ballooning
and stent implantation. As illustrated in Fig. 5, this could
substantially decrease the incidence of new atherosclerosis, ISR,
and ST.
      
Atherosclerosis, ISR, and ST Utilizing PBD
Aging is a progressive deterioration of biological functions that
results in cellular malfunction and can lead to diseases such
as hypertension, CHD, and stroke. Numerous physiological
changes occur as time passes, including genomic instability,
epigenetic irregularities, loss of proteostasis, altered
intercellular communication, abnormal nutrient absorption,
altered mitochondrial function, depletion of stem cells, cellular
senescence, and shortened telomeres. Telomeres are the protective
caps at the ends of chromosomes, consisting of non-coding DNA
sequences that prevent chromosomal breaks and clustering. With
each cell division, telomeres become shorter, resulting in faster
15
Figure 5. Bidirectional: metabolic- chronic inflammatory diseases cause mitochondrial
dysfunction and vice-versa. Mechanisms of mitochondrial dysfunction induce
atherosclerosis, ISR, and ST
Promoting healthy mitochondrial function can be achieved through various lifestyle adjustments,
including following a balanced diet, engaging in regular physical activity, and taking supplements
containing vitamins (such as vitamin C, E, and biotin), minerals (zinc), or nutraceuticals (ubiquinone or
CoQ10, NMN-precursor of NAD+, Quercetin, Astaxanthin, Resveratrol) [72]. The concept of
Mitochondrial nutrients has gained momentum in recent years and refers to the essential nutrients
necessary to maintain optimal mitochondrial function. Recent investigations have demonstrated that a
Mediterranean diet, characterized by the consumption of plant-derived compounds such as vegetables,
fruits, and nuts, which are rich in polyphenols (isoflavones, phytoestrogens) and other phytochemicals
Volume 9 | Issue 1 | 10
Cardio Open, 2024
aging. Telomerase, a ribonucleoprotein enzyme, can synthesize
telomeric repeats at the ends of chromosomes, slowing down
the shortening of telomeres. By increasing telomerase enzyme
activity, the rate of telomere shortening (TeS) can be slowed
[75].
Various external factors, including lifestyle, nutrition, genetics,
and heredity, can inuence aging. Research on twin subjects
has revealed that only 20-30% of an individual's lifespan is
determined by genetic factors, while a signicant portion
depends on behavior and environmental factors. Some age-
related diseases, such as hypertension, obesity, hyperlipidemia,
insulin resistance, and atherosclerosis, are associated with
increased inammation, oxidative stress, decreased telomerase
activity, and TeS. Certain lifestyle habits, such as leading a
sedentary lifestyle, smoking, experiencing emotional stress,
being exposed to pollution, and engaging in unhealthy eating
behaviors, can signicantly increase oxidative stress and TeS,
accelerating the aging and disease process [76].
Several longitudinal studies have demonstrated that TeS is
strongly associated with mortality and age-related diseases such
as acute myocardial infarction (AMI), atherosclerosis, stroke,
hypertension, and type 2 diabetes mellitus. In one population-
based study, individuals with the longest telomeres had the
lowest risk of dying during a 10-year follow-up period. In
another study, participants with TeS had the lowest survival
rates. Large-scale studies have also found that subjects with
the shortest telomeres had a 17-66% increase in mortality risk
compared to those with the longest telomeres. Thus, TeS are
associated with early death, most likely due to cardiovascular
risk factors, including obesity, CHD, AMI, hyperlipidemia,
hypertension, and diabetes mellitus. Consequently, individuals
with the longest telomeres had the lowest risk of developing
CHD, AMI, stroke, and vascular death [77].
Oxidative stress and persistent inammation have the potential
to lead to TeS, dysregulation of genes associated with telomeres,
and a decrease in telomerase activity. The relationship between
inammation and TeS is reciprocal. Inammatory stimuli can
accelerate telomere attrition and result in telomere dysfunction
and shortening. Conversely, TeS can contribute to low-grade
inammation [78].
Eating edible plants can be advantageous because they are
abundant in compounds with antioxidant and anti-inammatory
properties. These compounds can counteract the negative impact
of oxidative stress and persistent inammation, contributing
to the TeS. According to observational studies, consuming
polyphenol/ phytochemicals-rich diets, such as vegetables,
fruits, nuts, seeds, and their derivatives, can prevent TeS. This,
in turn, can result in improved overall health and longevity. [79].
Elizabeth Blackburn, a Nobel laureate, discovered that adopting
a vegan diet can activate over 500 genes within a three-month
period. This diet has the ability to activate genes that help prevent
diseases and deactivate genes that cause chronic inammatory
diseases. There are various methods for strengthening telomeres,
such as regular exercise, avoiding smoking, and consuming a
diet rich in plant-based foods that protect telomeres. Dean Ornish
and Elizabeth Blackburn conducted a study demonstrating
how a vegan diet can increase telomerase activity, the enzyme
responsible for maintaining the length of telomeres. The ability
to lengthen telomeres is critical for longevity. Although we
cannot reverse our chronological age, we can reverse our
biological age, which can help us reverse chronic illnesses in
our patients. Furthermore, reducing our patients' biological age
by one or two decades will automatically decrease their risk of
developing atherosclerosis, ISR, and ST [80].
Figure 6 demonstrates the advantages of a healthy lifestyle,
specically adopting a well-balanced diet, in preventing TeS. By
doing so, the aging process is slowed down or halted, thereby
inhibiting the progression of atherosclerosis. Moreover, this
approach can prevent the onset of ISR and the incidence of ST.
Although our current capabilities do not permit altering our
patients' genetics through the manipulation of mitochondria
and telomeres, there is potential for modifying epigenetic
markers to activate healthy genes and deactivate unhealthy
ones. Epigenetics can inuence the body's interpretation of
specic DNA sequences. As research progresses, the possibility
of manipulating mitochondria and telomeres and even altering
genes may become a reality.
Volume 9 | Issue 1 | 11
Cardio Open, 2024

3. Conclusions
The signicance of the relationship between nutrition and health
should be acknowledged. Research has demonstrated that an
unhealthy diet can result in a heightened risk of developing
metabolic chronic inammatory diseases, such as hypertension,
hyperlipidemia, obesity, insulin resistance, and atherosclerosis.
To promote optimal health and prevent these conditions, it is
essential to adopt a healthy lifestyle that includes balanced-
proper nutrition, regular exercise, abstinence from harmful
substances (such as smoking, alcohol, and unhealthy foods),
stress management, adequate sleep, and social support.
Managing atherosclerosis and preventing ISR and ST solely by
concentrating on the local coronary vessel is no longer justiable
in modern medical practice. Trials from Courage (2007) to
Ischemia (2019) have demonstrated that optimal medical
therapies are as eective as performing coronary intervention in
stable coronary disease patients with chronic coronary syndrome
18
Figure 6. Mechanism of how preventing telomeres shortening will prevent
new atherosclerosis, ISR, and ST
Volume 9 | Issue 1 | 12
Cardio Open, 2024
[81-83]. Thus, the prerequisites for conducting coronary
interventions have become progressively more stringent [84].
Providing optimal medical treatment (OMT) prior to performing
an intervention in stable patients is a reasonable choice. Given that
most interventional cardiologists have traditionally concentrated
on the local concerns of their patients rather than addressing
the metabolic-chronic inammation issues. The ACC/AHA
guidelines recommend that patients with coronary artery disease
should primarily consume PBDs (COR 1 and LOE B-R), but it is
essential to note that the majority of patients who have received
coronary interventions have not adhered to this recommendation.
Furthermore, many interventional cardiologists do not follow
this nutrient recommendation themselves. Although medication
is crucial for addressing systemic issues, neglecting a patient's
dietary habits may not be considered ethical or noble.
Unhealthy diets signicantly contribute to the development of
atherosclerosis and the processes of ISR and ST.
Last year, Rathod et al. from St. Bartholomew's Hospital,
London, presented the NITRATE-OCT ndings, an ISR study
at TCT 2023, demonstrating that those who consumed a daily
serving of beet juice experienced signicantly less late lumen
loss compared to those who received a nitrate-depleted placebo.
Additionally, there was a trend towards fewer major adverse
cardiovascular events at two years. After six months, the median
stent late lumen loss in the nitrate-depleted beet juice group
was 0.244 mm, while it was 0.117 mm in the natural beet juice
group (P = .0165). Similarly, the mean segment late lumen loss
favored the natural beet juice group (0.269 vs. 0.050 mm; P
= .0011). Over the 24-month follow-up period, there were 18
major adverse cardiovascular events in the control group and 9
in the group randomized to dietary nitrate (P = .0718). Although
no in-stent thromboses were observed in either group, death,
myocardial infarction, and target-vessel revascularization were
all numerically lower in the group receiving dietary nitrate.
The results of the study have not yet been disclosed in a nal
report. However, these ndings are of great signicance, as
they indicate that a daily serving of beet juice may signicantly
decrease the likelihood of experiencing ISR. Compared to
a limited serving of beet juice, the whole food form of PBD
contains more antioxidants, anti-inammatory compounds, and
other benecial components, providing even greater protection
against ISR.
Elucidating the pathways by which benecial nutrients exert a
substantial impact on the progression of coronary artery disease
and its role in mitigating the occurrence of ISR and ST is
paramount. Acquiring this knowledge will facilitate larger-scale
studies and encourage the adoption of healthy lifestyle choices
within the interventional cardiac community.
Author Contributions: Conceptualization, D.M.; Writing-
original draft, D.M.; Writing-review and editing, D.M., A.M.H.,
I.N.E.L., M.K.S., and H.U. All authors have read and agreed to
the published version of the manuscript.
Funding: This research received no external funding.
Data Availability Statement: Data sharing is not applicable to
this article.
The author declares no conict of interest.
References
1. Mohamad, T., Jyotsna, F. N. U., Farooq, U., Fatima,
A., Kar, I., Khuwaja, S., ... & Muzammil, M. A. (2023).
Individualizing medicinal therapy post heart stent
implantation: tailoring for patient factors. Cureus, 15(8).
2. Klein, L. W., Nathan, S., Maehara, A., Messenger, J., Mintz,
G. S., Ali, Z. A., ... & Lot, A. (2023). SCAI expert consensus
statement on management of in-stent restenosis and stent
thrombosis. Journal of the Society for Cardiovascular
Angiography & Interventions, 2(4), 100971.
3. Pelliccia, F., Zimarino, M., Niccoli, G., Morrone, D., De
Luca, G., Miraldi, F., & De Caterina, R. (2023). In-stent
restenosis after percutaneous coronary intervention:
Emerging knowledge on biological pathways. European
Heart Journal Open, 3(5), oead083.
4. Wolf, D., & Ley, K. (2019). Immunity and inammation in
atherosclerosis. Circulation research, 124(2), 315-327.
5. Aoki, J., & Tanabe, K. (2021). Mechanisms of drug-
eluting stent restenosis. Cardiovascular Intervention and
Therapeutics, 36, 23-29.
6. Zheng, P. G., Chen, P., Wang, L. J., & Zhang, N. (2023).
The association of the systemic immune-inammation
index and stent thrombosis in myocardial infarction patients
after coronary stent implantation—a retrospectively study.
Journal of Thoracic Disease, 15(4), 1726.
7. Condello, F., Spaccarotella, C., Sorrentino, S., Indol, C.,
Stefanini, G. G., & Polimeni, A. (2023). Stent thrombosis
and restenosis with contemporary drug-eluting stents:
predictors and current evidence. Journal of Clinical
Medicine, 12(3), 1238.
8. Khalid, W., Arshad, M. S., Ranjha, M. M. A. N., Różańska,
M. B., Irfan, S., Shaque, B., ... & Kowalczewski, P. Ł.
(2022). Functional constituents of plant-based foods boost
immunity against acute and chronic disorders. Open Life
Sciences, 17(1), 1075-1093.
9. Peña-Jorquera, H., Cid-Jofré, V., Landaeta-Díaz, L.,
Petermann-Rocha, F., Martorell, M., Zbinden-Foncea,
H., ... & Cristi-Montero, C. (2023). Plant-based nutrition:
Exploring health benets for atherosclerosis, chronic
diseases, and metabolic syndrome—A comprehensive
review. Nutrients, 15(14), 3244.
10. Salehin, S., Rasmussen, P., Mai, S., Mushtaq, M., Agarwal,
M., Hasan, S. M., ... & Khalife, W. I. (2023). Plant based
diet and its eect on cardiovascular disease. International
journal of environmental research and public health, 20(4),
3337.
11. Koutentakis, M., Surma, S., Rogula, S., Filipiak, K.
J., & Gąsecka, A. (2023). The eect of a vegan diet on
the cardiovascular system. Journal of cardiovascular
development and disease, 10(3), 94.
12. Tucci, M., Marino, M., Martini, D., Porrini, M., Riso, P.,
& Del Bo’, C. (2022). Plant-based foods and vascular
function: a systematic review of dietary intervention trials
Volume 9 | Issue 1 | 13
Cardio Open, 2024
in older subjects and hypothesized mechanisms of action.
Nutrients, 14(13), 2615.
13. Islam, S. U., Ahmed, M. B., Ahsan, H., & Lee, Y. S. (2021).
Recent molecular mechanisms and benecial eects of
phytochemicals and plant-based whole foods in reducing
LDL-C and preventing cardiovascular disease. Antioxidants,
10(5), 784.
14. Mehta, P., Tawfeeq, S., Padte, S., Sunasra, R., Desai, H.,
Surani, S., & Kashyap, R. (2023). Plant-based diet and its
eect on coronary artery disease: A narrative review. World
Journal of Clinical Cases, 11(20), 4752.
15. Bruns, A., Greupner, T., Nebl, J., & Hahn, A. (2024). Plant-
based diets and cardiovascular risk factors: a comparison
of exitarians, vegans and omnivores in a cross-sectional
study. BMC nutrition, 10(1), 29.
16. Di Sotto, A., Vitalone, A., & Di Giacomo, S. (2020). Plant-
derived nutraceuticals and immune system modulation: An
evidence-based overview. Vaccines, 8(3), 468.
17. Xu, K., Al-Ani, M. K., Pan, X., Chi, Q., Dong, N., & Qiu,
X. (2018). Plant-derived products for treatment of vascular
intima hyperplasia selectively inhibit vascular smooth
muscle cell functions. Evidence-Based Complementary and
Alternative Medicine, 2018.
18. Monsalve, B., Concha-Meyer, A., Palomo, I., & Fuentes,
E. (2017). Mechanisms of endothelial protection by natural
bioactive compounds from fruit and vegetables. Anais da
Academia Brasileira de Ciências, 89, 615-633.
19. Aquila, G., Marracino, L., Martino, V., Calabria, D., Campo,
G., Caliceti, C., & Rizzo, P. (2019). The use of nutraceuticals
to counteract atherosclerosis: the role of the notch pathway.
Oxidative medicine and cellular longevity, 2019.
20. Moss, J. W., & Ramji, D. P. (2016). Nutraceutical therapies
for atherosclerosis.(9), 513-
532.
21. Wei, T., Liu, J., Zhang, D., Wang, X., Li, G., Ma, R., ...
& Guo, X. (2021). The relationship between nutrition
and atherosclerosis. Frontiers in bioengineering and
biotechnology, 9, 269.
22. Mitu, O., Cirneala, I. A., Lupsan, A. I., Iurciuc, M., Mitu,
I., Dimitriu, D. C., ... & Costache, I. I. (2020). The eect
of vitamin supplementation on subclinical atherosclerosis
in patients without manifest cardiovascular diseases: never-
ending hope or underestimated eect?. Molecules, 25(7),
1717.
23. Li, M., Hou, J., Gu, X., Weng, R., Zhong, Z., & Liu, S.
(2022). Incidence and risk factors of in-stent restenosis
after percutaneous coronary intervention in patients from
southern China. European Journal of Medical Research,
27(1), 12.
24. Rohman, M. S., Waranugraha, Y., Masbuchin, A. N.,
Baskoro, S. S., Sishartami, L. W., & Pratiwi, B. B. (2023).
Coronary In-Stent Restenosis Predictors Following Drug-
Eluting Stent Implantation: A Meta-Analysis Study. Journal
of Vascular Diseases, 2(3), 266-281.
25. Saleh, A., Hammoudeh, A., Tabbalat, R., Al-haddad, I., Al-
Mousa, E., Jarrah, M., ... & Khadder, Y. (2016). Incidence
and prognosis of stent thrombosis following percutaneous
coronary intervention in Middle Eastern patients: The First
Jordanian Percutaneous Coronary Intervention Registry
(JoPCR1). Annals of Saudi medicine, 36(1), 17-22.
26. Marchio, P., Guerra-Ojeda, S., Vila, J. M., Aldasoro, M.,
Victor, V. M., & Mauricio, M. D. (2019). Targeting early
atherosclerosis: a focus on oxidative stress and inammation.
Oxidative medicine and cellular longevity, 2019.
27. Ray, A., Maharana, K. C., Meenakshi, S., & Singh, S.
(2023). Endothelial dysfunction and its relation in dierent
disorders: Recent update. 100084.
28. Almeida, C., Barata, P., & Fernandes, R. (2021). The
inuence of gut microbiota in cardiovascular diseases—a
brief review. Porto Biomedical Journal, 6(1), e106.
29. Pollicino, F., Veronese, N., Dominguez, L. J., & Barbagallo,
M. (2023). Mediterranean diet and mitochondria: New
ndings. Experimental Gerontology, 176, 112165.
30. Cinegaglia, N., Antoniazzi, L., Rosa, D., Miranda, D.,
Acosta-Navarro, J., Bortolotto, L., ... & Sandrim, V.
(2019). Shortening telomere is associated with subclinical
atherosclerosis biomarker in omnivorous but not in
vegetarian healthy men. Aging (Albany NY), 11(14), 5070.
31. Napoli, C., de Nigris, F., Williams-Ignarro, S., Pignalosa,
O., Sica, V., & Ignarro, L. J. (2006). Nitric oxide and
atherosclerosis: an update. Nitric oxide, 15(4), 265-279.
32. Naseem, K. M. (2005). The role of nitric oxide in
cardiovascular diseases. Molecular aspects of medicine,
26(1-2), 33-65.
33. Torregrossa, A. C., Aranke, M., & Bryan, N. S. (2011).
Nitric oxide and geriatrics: Implications in diagnostics and
treatment of the elderly. Journal of geriatric cardiology:
JGC, 8(4), 230.
34. Babateen, A. M., Shannon, O. M., Mathers, J. C., & Siervo,
M. (2019). Validity and reliability of test strips for the
measurement of salivary nitrite concentration with and
without the use of mouthwash in healthy adults. Nitric
Oxide, 91, 15-22.
35. Salehzadeh, H., Maleki, A., Rezaee, R., Shahmoradi, B., &
Ponnet, K. (2020). The nitrate content of fresh and cooked
vegetables and their health-related risks. PloS one, 15(1),
e0227551.
36. Neungerl, N., & Eilander, A. (2021). Nutrient intake and
status in adults consuming plant-based diets compared to
meat-eaters: a systematic review. Nutrients, 14(1), 29.
37. Rajendran, S., Shen, X., Glawe, J., Kolluru, G. K., & Kevil,
C. G. (2019). Nitric oxide and hydrogen sulde regulation of
ischemic vascular growth and remodeling. Comprehensive
Physiology, 9(3), 1213.
38. Rao, J., Pan Bei, H., Yang, Y., Liu, Y., Lin, H., & Zhao, X.
(2020). Nitric oxide-producing cardiovascular stent coatings
for prevention of thrombosis and restenosis. Frontiers in
Bioengineering and Biotechnology, 8, 578.
39. Kabirian, F., Milan, P. B., Zamanian, A., Heying, R., &
Mozafari, M. (2019). Nitric oxide-releasing vascular grafts:
A therapeutic strategy to promote angiogenic activity and
endothelium regeneration. Acta biomaterialia, 92, 82-91.
40. Ornish, D., Scherwitz, L. W., Billings, J. H., Gould, K. L.,
Merritt, T. A., Sparler, S., ... & Brand, R. J. (1998). Intensive
lifestyle changes for reversal of coronary heart disease.
Jama, 280(23), 2001-2007.
Volume 9 | Issue 1 | 14
Cardio Open, 2024
41. Gendy, G., Doyle, J., Golubic, M., & Roizen, M. F. (2014).
A way to reverse CAD. J Fam Pract, 63(7), 356-364b.
42. Rose, S., & Strombom, A. (2018). A Comprehensive Review
of the Prevention and Treatment of Heart Disease with a
Plant-Based Diet. Journal of Cardiology & Cardiovascular
Therapy, 12(5), 110-121.
43. Bilal, M., Ashraf, S., & Zhao, X. (2022). Dietary component-
induced inammation and its amelioration by prebiotics,
probiotics, and synbiotics. Frontiers in Nutrition, 9, 931458.
44. Upadhyay, S., & Dixit, M. (2015). Role of polyphenols and
other phytochemicals on molecular signaling. Oxidative
medicine and cellular longevity, 2015.
45. Wu, Y. T., Chen, L., Tan, Z. B., Fan, H. J., Xie, L. P., Zhang,
W. T., ... & Zhou, Y. C. (2018). Luteolin inhibits vascular
smooth muscle cell proliferation and migration by inhibiting
TGFBR1 signaling. Frontiers in Pharmacology, 9, 1059.
46. Naruko, T., Ueda, M., Ehara, S., Itoh, A., Haze, K., Shirai, N.,
... & Becker, A. E. (2006). Persistent high levels of plasma
oxidized low-density lipoprotein after acute myocardial
infarction predict stent restenosis. Arteriosclerosis,
thrombosis, and vascular biology, 26(4), 877-883.
47. Batty, M., Bennett, M. R., & Yu, E. (2022). The role of
oxidative stress in atherosclerosis. Cells, 11(23), 3843.
48. Ganjali, S., Mansouri, A., Abbasifard, M., Moallem, S. A.,
Tayarani-Najaran, Z., & Sahebkar, A. (2022). Association
between oxidative burden and restenosis: A case-control
study. Oxidative Medicine and Cellular Longevity, 2022.
49. Li, P., Ma, X., & Huang, G. (2024). Understanding
thrombosis: the critical role of oxidative stress. Hematology,
29(1), 2301633.
50. Chioncel, V., Andrei, C. L., Brezeanu, R., Sinescu, C.,
Avram, A., & Tatu, A. L. (2021). Some perspectives on
hypersensitivity to coronary stents. International Journal of
General Medicine, 4327-4336.
51. Rakha, A., Umar, N., Rabail, R., Butt, M. S., Kieliszek, M.,
Hassoun, A., & Aadil, R. M. (2022). Anti-inammatory
and anti-allergic potential of dietary avonoids: A review.
Biomedicine & Pharmacotherapy, 156, 113945.
52. Zhang, P. (2023). The Role of Diet and Nutrition in Allergic
Diseases. Nutrients, 15(17), 3683.
53. Mehta, P., Tawfeeq, S., Padte, S., Sunasra, R., Desai, H.,
Surani, S., & Kashyap, R. (2023). Plant-based diet and its
eect on coronary artery disease: A narrative review. World
Journal of Clinical Cases, 11(20), 4752.
54. Tan, Y., Zhou, J., Liu, C., Zhou, P., Sheng, Z., Li, J., ... &
Yan, H. (2020). Association between plasma trimethylamine
N-oxide and neoatherosclerosis in patients with very late
stent thrombosis. Canadian Journal of Cardiology, 36(8),
1252-1260.
55. Kumar, A., Chidambaram, V., & Mehta, J. L. (2022).
Vegetarianism, microbiota, and cardiovascular health:
looking back, and forward. European Journal of Preventive
Cardiology, 29(14), 1895-1910.
56. Shen, X., Li, L., Sun, Z., Zang, G., Zhang, L., Shao, C.,
& Wang, Z. (2021). Gut microbiota and atherosclerosis—
focusing on the plaque stability. Frontiers in Cardiovascular
Medicine, 8, 668532.
57. Peña-Jorquera, H., Cid-Jofré, V., Landaeta-Díaz, L.,
Petermann-Rocha, F., Martorell, M., Zbinden-Foncea,
H., ... & Cristi-Montero, C. (2023). Plant-based nutrition:
Exploring health benets for atherosclerosis, chronic
diseases, and metabolic syndrome—A comprehensive
review. Nutrients, 15(14), 3244.
58. Craig, W. J., Mangels, A. R., Fresán, U., Marsh, K., Miles,
F. L., Saunders, A. V., ... & Orlich, M. (2021). The safe and
eective use of plant-based diets with guidelines for health
professionals. Nutrients, 13(11), 4144.
59. Desai, D., Desai, A., Jamil, A., Csendes, D., Gutlapalli, S.
D., Prakash, K., ... & Manoharan, M. P. (2023). Re-dening
the Gut Heart Axis: A Systematic Review of the Literature
on the Role of Gut Microbial Dysbiosis in Patients With
Heart Failure. Cureus, 15(2).
60. Bui, T. V. A., Hwangbo, H., Lai, Y., Hong, S. B., Choi, Y. J.,
Park, H. J., & Ban, K. (2023). The gut-heart axis: updated
review for the roles of microbiome in cardiovascular health.
Korean Circulation Journal, 53(8), 499.
61. Sørensen, T. I., Martinez, A. R., & Jørgensen, T. S. H. (2022).
Epidemiology of obesity. In From Obesity to Diabetes (pp.
3-27). Cham: Springer International Publishing.
62. Kökten, T., Hansmannel, F., Ndiaye, N. C., Heba, A. C.,
Quilliot, D., Dreumont, N., ... & Peyrin-Biroulet, L. (2021).
Calorie restriction as a new treatment of inammatory
diseases. Advances in Nutrition, 12(4), 1558-1570.
63. Perry, C. A., & Gadde, K. M. (2022). The role of calorie
restriction in the prevention of cardiovascular disease.
Current Atherosclerosis Reports, 24(4), 235-242.
64. Samad, F., & Ruf, W. (2013). Inammation, obesity, and
thrombosis. Blood, The Journal of the American Society of
Hematology, 122(20), 3415-3422.
65. Yang, X., Yang, Y., Guo, J., Meng, Y., Li, M., Yang, P., ... &
Li, Y. (2021). Targeting the epigenome in in-stent restenosis:
from mechanisms to therapy. Molecular Therapy-Nucleic
Acids, 23, 1136-1160.
66. Zhai, J., Kongsberg, W. H., Pan, Y., Hao, C., Wang, X., &
Sun, J. (2023). Caloric restriction induced epigenetic eects
on aging. Frontiers in Cell and Developmental Biology, 10,
1079920.
67. Marchi, S., Guilbaud, E., Tait, S. W., Yamazaki, T., &
Galluzzi, L. (2023). Mitochondrial control of inammation.
(3), 159-173.
68. Ciccarelli, G., Conte, S., Cimmino, G., Maiorano, P.,
Morrione, A., & Giordano, A. (2023). Mitochondrial
dysfunction: the hidden player in the pathogenesis of
atherosclerosis?. International Journal of Molecular
Sciences, 24(2), 1086.
69. Shemiakova, T., Ivanova, E., Wu, W. K., Kirichenko,
T. V., Starodubova, A. V., & Orekhov, A. N. (2021).
Atherosclerosis as mitochondriopathy: Repositioning
the disease to help nding new therapies. Frontiers in
Cardiovascular Medicine, 8, 660473.
70. Qu, K., Yan, F., Qin, X., Zhang, K., He, W., Dong, M.,
& Wu, G. (2022). Mitochondrial dysfunction in vascular
endothelial cells and its role in atherosclerosis. Frontiers in
Physiology, 13, 1084604.
71. Suarez-Rivero, J. M., Pastor-Maldonado, C. J., Povea-
Cabello, S., Álvarez-Córdoba, M., Villalón-García, I.,
Volume 9 | Issue 1 | 15
Cardio Open, 2024
Copyright: ©2024 Dasaad Mulijono, et al. This is an open-access article
distributed under the terms of the Creative Commons Attribution License,
        
medium, provided the original author and source are credited.
https://opastpublishers.com
Talaverón-Rey, M., ... & Sánchez-Alcázar, J. A. (2021).
From mitochondria to atherosclerosis: the inammation
path. Biomedicines, 9(3), 258.
72. Niyazov, D. M., Kahler, S. G., & Frye, R. E. (2016).
Primary mitochondrial disease and secondary mitochondrial
dysfunction: importance of distinction for diagnosis and
treatment. Molecular syndromology, 7(3), 122-137.
73. Pollicino, F., Veronese, N., Dominguez, L. J., & Barbagallo,
M. (2023). Mediterranean diet and mitochondria: New
ndings. Experimental Gerontology, 176, 112165.
74. Khalil, M., Shanmugam, H., Abdallah, H., John Britto,
J. S., Galerati, I., Gómez-Ambrosi, J., ... & Portincasa, P.
(2022). The potential of the Mediterranean diet to improve
mitochondrial function in experimental models of obesity
and metabolic syndrome. Nutrients, 14(15), 3112.
75. Maluchenko, N. V., Feofanov, A. V., & Studitsky, V.
M. (2021). PARP-1-associated pathological processes:
inhibition by natural polyphenols. International Journal of
Molecular Sciences, 22(21), 11441.
76. D’Angelo, S. (2023). Diet and Aging: The Role of
Polyphenol-Rich Diets in Slow Down the Shortening of
Telomeres: A Review. Antioxidants, 12(12), 2086.
77. Herrmann, W., & Herrmann, M. (2020). The importance
of telomere shortening for atherosclerosis and mortality.
Journal of Cardiovascular Development and Disease, 7(3),
29.
78. Liu, S., Nong, W., Ji, L., Zhuge, X., Wei, H., Luo, M., ... &
Huang, H. (2023). The regulatory feedback of inammatory
signaling and telomere/telomerase complex dysfunction in
chronic inammatory diseases. Experimental Gerontology,
174, 112132.
79. Crous-Bou, M., Molinuevo, J. L., & Sala-Vila, A. (2019).
Plant-rich dietary patterns, plant foods and nutrients, and
telomere length. Advances in Nutrition, 10, S296-S303.
80. Dwaraka, V. B., Aronica, L., Carerras-Gallo, N., Robinson,
J. L., Hennings, T., Lin, A., ... & Gardner, C. D. (2023).
Unveiling the Epigenetic Impact of Vegan vs. Omnivorous
Diets on Aging: Insights from the Twins Nutrition Study
(TwiNS). medRxiv, 2023-12.
81. Boden, W. E. (2009). Interpreting the results of the
COURAGE trial: a non-interventionalist perspective.
 S34-44.
82. Lopez-Sendon, J., Moreno, R., & Tamargo, J. (2021).
Ischemia trial: Key questions and answers. European

83. Pham, V., Moroni, A., Gall, E., Benedetti, A., Zivelonghi,
C., & Picard, F. (2023). Revascularization and medical
therapy for chronic coronary syndromes: Lessons learnt
from recent trials, a literature review. Journal of Clinical
Medicine, 12(8), 2833.
84. Writing Committee Members, Virani, S. S., Newby,
L. K., Arnold, S. V., Bittner, V., Brewer, L. C., ... &
Williams, M. S. (2023). 2023 AHA/ACC/ACCP/ASPC/
NLA/PCNA guideline for the management of patients
with chronic coronary disease: a report of the American
Heart Association/American College of Cardiology Joint
Committee on Clinical Practice Guidelines. Journal of the
American College of Cardiology, 82(9), 833-955.
85. Rathod, K. S., Jones, D. A., Van-Eijl, T. J. A., Tsang, H.,
Warren, H., Hamshere, S. M., ... & Ahluwalia, A. (2016).
Randomised, double-blind, placebo-controlled study
investigating the eects of inorganic nitrate on vascular
function, platelet reactivity and restenosis in stable angina:
protocol of the NITRATE-OCT study. BMJ open, 6(12),
e012728.
... A PBD has significantly reduced the risk of atherosclerosis and restenosis through multiple physiological and biochemical pathways [3][4][5][6][7][56][57][58][59][60][61]. The following mechanisms elucidate its cardioprotective effects: ...
... Moreover, patients with moderate renal impairment have experienced normalization of their serum creatinine levels, while those with diabetes have demonstrated significant glycaemic control, with glycated haemoglobin (HbA1c) levels consistently below 6%. In numerous cases, many individuals have successfully minimized pharmacological interventions, including insulin cessation [56][57][58][59][60][61][62][63][64][65]. ...
Article
Full-text available
Despite advances in drug-coated balloon (DCB) technology, restenosis remains a persistent challenge in percutaneous coronary intervention (PCI). At Bethsaida Hospital in Indonesia, chaired by Prof. Dasaad Mulijono, we've reduced restenosis rates from 10-20% to just 2% through an integrated approach that combines precise lesion preparation, imaging-guided intervention, and a groundbreaking lifestyle strategy: the plant-based diet (PBD). This review challenges the conventional, drug-heavy paradigm by highlighting PBD as a potent, non-pharmacological tool for combating atherosclerosis and restenosis. Backed by mechanistic insights-ranging from nitric oxide (NO)-mediated vasodilation to gut microbiome modulation-PBD offers profound cardiovascular protection while enhancing glycaemic control, blood pressure, lipid profiles, and overall metabolic health. Moreover, our centre's deployment of artificial intelligence (AI) has revolutionized patient education and dietary adherence, accelerating real-world outcomes. Our data show that combining AI-driven personalization with lifestyle medicine is feasible and transformative. It's time to disrupt the status quo in cardiology. This review calls for a bold paradigm shift: integrate cutting-edge interventional techniques with evidence-based nutrition and digital innovation to redefine restenosis prevention and reshape the future of cardiovascular care.
... Bethsaida Hospital has implemented a comprehensive model integrating PBDs, interventional cardiology, and holistic care [38][39][40][41][42][43][44][45][46][47][48]. Key components include: ...
Article
Full-text available
In an era characterized by the escalation of chronic diseases and fragmented healthcare, Bethsaida Hospital, under the visionary leadership of Prof. Dasaad Mulijono, has forged an unprecedented integration of biblical nutrition, lifestyle medicine, and artificial intelligence (AI). This pioneering approach transcends conventional medical paradigms, positioning physicians not merely as healthcare providers but as faith-aligned educators fostering physical healing and spiritual renewal. Grounded in scriptural principles promoting plant-based diets (PBDs), simplicity, and stewardship of health, Bethsaida's model has achieved remarkable clinical outcomes, including the reversal of coronary artery disease (CAD), hypertension, type 2 diabetes mellitus (T2DM), obesity, dyslipidaemia, and early-stage chronic kidney disease. Notably, patients undergoing drug-coated balloon (DCB) angioplasty experience a restenosis rate of just 2%, significantly lower than national averages, directly attributed to rigorous lesion preparation combined with evidence-based lifestyle modification. Despite clear biblical mandates advocating plant-based nutrition (PBN), widespread resistance persists among faith-based communities, reflecting deep-seated cultural, economic, and theological misalignments. Prosperity theology, in particular, often redirects community values toward material affluence and expensive medical interventions, undermining preventive health strategies. Bethsaida Hospital challenges these prevailing norms by re-centring health discourse within a scriptural context, thereby promoting sustainable and spiritually coherent lifestyles. AI emerges as a transformative tool within this paradigm, offering ethically designed, unbiased platforms that deliver personalized nutritional and spiritual guidance aligned with biblical teachings. By interpreting sacred texts without political or financial distortions, AI empowers patients and healthcare professionals alike to bridge gaps between scientific evidence and theological practice. Ultimately, this integrative approach advocates a redefinition of the physician's role, emphasizing spiritual mentorship and preventive education as core components of medical professionalism. Bethsaida's innovative model offers not merely a medical solution but a redemptive framework-a holistic, technologically-enhanced pathway toward enduring health, spiritual integrity, and community transformation.
Article
Full-text available
Artificial Intelligence (AI) and robotics are no longer optional enhancements in healthcare-they are seismic forces redefining the future of medicine, reshaping clinical decision-making, surgical precision, patient outcomes, and economic structures. While the United States struggles with entrenched bureaucracy, fractured systems, and policy paralysis, China has launched a strategic, coordinated offensive that is fast elevating it as the new global epicentre of medical technology. With government-backed research and development (R&D), streamlined regulations, and aggressive AI implementation across radiology, surgery, and diagnostics, China is poised to eclipse traditional Western dominance within the next decade. Meanwhile, countries like Indonesia risk being left behind, paralyzed by infrastructural gaps, outdated medical training, and fierce resistance from entrenched interest groups. Yet within this stagnation, one institution-Bethsaida Hospital, led by Prof. Dasaad Mulijono-offers a powerful counter-narrative. As the first in Indonesia to integrate Drug-Coated Balloon (DCB) therapy with a whole-food plant-based diet (WFPBD), Bethsaida has not only achieved unprecedented clinical outcomes (including restenosis rates below 2%) but now emerges as a credible pioneer for AI and robotic healthcare integration. With the passage of the 2024 Omnibus Law aimed at reforming medical education, a narrow window of opportunity has opened. The question is whether Indonesia's healthcare community will seize or squander it through inaction and internal discord. This paper issues a call to arms for Indonesian medical professionals and policymakers: the age of AI-powered healthcare is no longer approaching-it is here. Failure to adapt will lead to irrelevance and dependency on more agile, technologically advanced nations. Only by embracing innovation, protecting reform, and scaling models like Bethsaida can Indonesia reclaim agency over its medical future and avoid becoming a passive consumer in the next healthcare revolution.
Article
Full-text available
In a world where the cruelty of discrimination and the silence of complicity still haunt the corridors of medical education, some stories break through the darkness-not with anger but with radiant love. This is the story of Prof. Dasaad Mulijono (DM), a minority physician in Indonesia who rose from the ashes of ethnic persecution and systemic rejection to become a beacon of healing, innovation, and faith-driven excellence. Born into a world that tried to erase his worth, Prof. DM endured bullying, exclusion, and institutional prejudice throughout his formative years. Yet rather than yielding to bitterness, he chose forgiveness; instead of retreating, he advanced with unstoppable courage. Guided by unshakable faith, Prof. DM turned every insult into fuel for his purpose, ultimately ascending to the highest academic honours and becoming a pioneer in interventional cardiology and lifestyle-based medicine. While many withdrew in fear at the height of the COVID-19 pandemic, Prof. DM stepped forward in love. With the bold integration of Whole Food Plant-Based Diets (WFPBD) and advanced cardiology, he saved thousands of lives, patients deemed untreatable by conventional standards. His leadership redefined clinical outcomes and embodied a profound spiritual truth: that suffering, when met with compassion and conviction, can become a sacred path to transformation. Prof. DM's story is a personal triumph, moral testimony, and call to action. It urges institutions to no longer see marginalized individuals through a deficit lens but to recognize the resilience forged in adversity. Most critically, it demands that discrimination and bullying be confronted and eradicated from all levels of medical training forever. This article bears witness to the hidden suffering of many and honours those whose silent tears never made headlines. For every life changed by Prof. DM, there are countless others whose potential was crushed under the weight of systemic injustice. Let this story stir hearts and awaken consciences to build a future where excellence is nurtured by love, not hindered by hate.
Article
Full-text available
The ends of human chromosomes are defended by DNA–protein complexes named telomeres, which inhibit the chromosomes from fusing with each other and from being known as a double-strand break by DNA reparation proteins. Telomere length is a marker of biological aging, and disfunction of telomeres is related to age-related syndromes. Telomere attrition has been shown to be accelerated by oxidative stress and inflammation. Telomere length has been proven to be positively linked with nutritional status in human and animal scientific research as several nutrients influence it through mechanisms that imitate their function in cellular roles including oxidative stress and inflammation. Data reported in this article support the idea that following a low-in-fat and rich-plant polyphenols food diet seems to be able to slow down the shortening of telomeres.
Article
Full-text available
Percutaneous coronary intervention (PCI) has evolved significantly over the past four decades. Since its inception, in-stent restenosis (ISR) – the progressive reduction in vessel lumen diameter after PCI – has emerged as the main complication of the procedure. Although the incidence of ISR has reduced from 30% at 6 months with bare-metal stents (BMS) to 7% at 4 years with drug-eluting stents (DES), its occurrence is relevant in absolute terms because of the dimensions of the population treated with PCI. The aim of this review is to summarize the emerging understanding of the biological pathways that underlie ISR. ISR is associated with several factors, including patient-related, genetic, anatomic, stent, lesion and procedural characteristics. Regardless of associated factors, there are common pathophysiologic pathways involving molecular phenomena triggered by the mechanical trauma caused by PCI. Such biologic pathways are responses to the denudation of the intima during balloon angioplasty and involve inflammation, hypersensitivity reactions, and stem cell mobilization in particular of endothelial progenitor cells (EPCs). The results of these processes are either vessel wall healing or neointimal hyperplasia and/or neoatherosclerosis. Unraveling the key molecular and signal pathways involved in ISR is crucial to identify appropriate therapeutic strategies aimed at abolishing the ‘Achille’s heel’ of PCI. With this regard, we discuss novel approaches to prevent DES restenosis. Indeed, available evidence suggests that EPCs-capturing stents promote a rapid stent re-endothelisation, which in turn has the potential to decrease the risk of stent thrombosis and allow the use of a shorter duration of dual antiplatelet therapy.
Article
Full-text available
The field of cardiovascular medicine is undergoing a transformative shift towards personalized medicinal therapy, particularly in the context of post stent implantation. This narrative review explores the significance, challenges, and future directions of individualized treatment strategies for patients with coronary stents. The review highlights the pivotal role of personalized approaches in optimizing treatment efficacy and minimizing adverse events. Real-world clinical studies and trials underscore the importance of tailoring antiplatelet therapy based on platelet function testing, genetic testing, and risk scoring. These studies reveal that personalized medicinal treatment improves clinical outcomes by balancing preventing thrombotic events and mitigating bleeding risks. Challenges, including cost, test availability, patient adherence, and ethical considerations, are discussed in depth, shedding light on the complexities of implementing personalized approaches. Technological advancements, including omics data integration, artificial intelligence, and big data analytics, shape the future of personalized medicinal therapy. These tools enable precise pharmacogenomic selection of medications and the development of integrated risk-scoring systems. Patient engagement and education are also central, with empowered patients and remote monitoring contributing to collaborative decision-making. In conclusion, the narrative review underscores that personalized medicinal therapy post stent implantation holds immense promise for revolutionizing cardiovascular care. By embracing a comprehensive approach that considers genetics, clinical factors, and patient preferences, healthcare providers can optimize treatment outcomes and improve patient quality of life. The evolving landscape of personalized medicine offers a glimpse into a future where tailored treatment strategies become the cornerstone of precision cardiovascular care.
Article
Full-text available
Cardiovascular diseases (CVDs), including coronary artery disease, stroke, heart failure, and hypertension, are the global leading causes of death, accounting for more than 30% of deaths worldwide. Although the risk factors of CVDs have been well understood and various treatment and preventive measures have been established, the mortality rate and the financial burden of CVDs are expected to grow exponentially over time due to the changes in lifestyles and increasing life expectancies of the present generation. Recent advancements in metagenomics and metabolomics analysis have identified gut microbiome and its associated metabolites as potential risk factors for CVDs, suggesting the possibility of developing more effective novel therapeutic strategies against CVD. In addition, increasing evidence has demonstrated the alterations in the ratio of Firmicutes to Bacteroidetes and the imbalance of microbial-dependent metabolites, including short-chain fatty acids and trimethylamine N-oxide, play a crucial role in the pathogenesis of CVD. However, the exact mechanism of action remains undefined to this day. In this review, we focus on the compositional changes in the gut microbiome and its related metabolites in various CVDs. Moreover, the potential treatment and preventive strategies targeting the gut microbiome and its metabolites are discussed.
Article
Full-text available
Numerous studies have investigated in-stent restenosis (ISR) predictors in first-generation drug-eluting stents (DESs), but only a few have investigated second-generation DESs. We aimed to investigate the ISR predictors following a successful DES implantation in coronary artery disease (CAD) patients. A systematic review and meta-analysis study was conducted. Diabetes mellitus (DM) (OR 1.47; 95% CI 1.19 to 1.83; p < 0.01), family history of CAD (OR 1.26; 95% CI 1.03 to 1.55; p 0.03), and smoking (OR 1.23; 95% CI 1.02 to 1.48; p 0.03) were the strong predictors for the DES-ISR. The DES-ISR was more common in DESs with smaller stent diameter (MD −0.12; 95% CI −0.16 to −0.08; p < 0.01) and longer stent length (MD 2.24; 95% CI 1.36 to 3.13; p < 0.01). Angiography characteristics, including multi-vessel disease (MVD) (OR 1.45; 95% CI 1.07 to 1.97; p 0.02), type B2/C lesions (OR 1.56; 95% CI 1.06 to 2.30; p 0.02), and type C lesion (OR 1.33; 95% CI 1.09 to 1.62; p < 0.01), were also associated with DES-ISR. We confirmed that DM, family history of CAD, smoking, MVD, smaller stent diameter, longer stent length, and type B2 or C lesions were proven to be ISR predictors following DES implantation.
Article
Full-text available
Stent failure remains the major drawback to the use of coronary stents as a revascularization strategy. Recent advances in imaging have substantially improved our understanding of the mechanisms underlying these occurrences, which have in common numerous clinical risk factors and mechanical elements at the time of stent implantation. In-stent restenosis remains a common clinical problem despite numerous improvements in-stent design and polymer coatings over the past 2 decades. It generates significant health care cost and is associated with an increased risk of death and rehospitalization. Stent thrombosis causes abrupt closure of the stented artery and therefore carries a high risk of myocardial infarction and death. This Society for Cardiovascular Angiography & Interventions (SCAI) Expert Consensus Statement suggests updated practical algorithmic approaches to in-stent restenosis and stent thrombosis. A pragmatic outline of assessment and management of patients presenting with stent failure is presented. A new SCAI classification that is time-sensitive with mechanistic implications of in-stent restenosis is proposed. Emphasis is placed on frequent use of intracoronary imaging and assessment of timing to determine the precise etiology because that information is crucial to guide selection of the best treatment option. SCAI recommends image-guided coronary stenting at the time of initial implantation to minimize the occurrence of stent failure. When in-stent restenosis and stent thrombosis are encountered, imaging should be strongly considered to optimize the subsequent approach.
Article
Full-text available
Background: A small number of patients can develop stent thrombosis after coronary stent implantation. Diabetes, malignant tumors, anemia, etc. have been identified as risk factors for stent thrombosis. A previous study confirmed that the systemic immune-inflammatory index is associated with venous thrombosis. However, there are no studies investigating the association between the systemic immune-inflammation index and stent thrombosis after coronary stent implantation, and thus, we designed this study. Methods: A total of 887 myocardial infarction patients admitted to the Wuhan University Hospital from January 2019 to June 2021 were included. All of the patients received coronary stent implantation and were followed up for 1 year by clinic visit. The patients were divided into a stent thrombosis group (n=27) and a control group (n=860) according to whether or not they suffered stent thrombosis. The clinical features of the two groups were observed, and the receiver operator characteristic (ROC) curve was used to analyze the predictive value of the systemic immune-inflammation index for stent thrombosis in patients with myocardial infarction after coronary artery stenting. Results: Compared with the control group, the proportion of stent number ≥4 in the stent thrombosis group was significantly higher (62.96% vs. 38.72%, P=0.011), and the proportion of patients with a systemic immune-inflammation index ≥636 was markedly increased (55.56% vs. 23.26%, P=0.000). The number of stents and the systemic immune-inflammation index were both valuable in predicting stent thrombosis, and the predictive value of the systemic immune-inflammation index was higher, with an area under the curve of 0.736 (95% confidence interval: 0.647-0.824, P=0.000), the best diagnostic value was 636, and the sensitivity and specificity were 0.556 and 0.767. The systemic immune-inflammation index ≥636 and the number of stents ≥4 were independent risk factors for stent thrombosis after coronary stent implantation (P<0.05). Compared with the control group, the incidence of recurrent myocardial infarction was notably increased in the stent thrombosis group (33.33% vs. 3.26%, P=0.000), and mortality was significantly higher in the stent thrombosis group (14.81% vs. 0.93%, P=0.000). Conclusions: The systemic immune-inflammation index was associated with the development of stent thrombosis in patients with myocardial infarction after coronary stent implantation.
Article
Full-text available
Stable coronary artery disease (CAD) has recently been replaced by a new entity described as chronic coronary syndrome (CCS). This new entity has been developed based on a better understanding of the pathogenesis, the clinical characteristics, and the morbi-mortality associated to this condition as part of the dynamic spectrum of CAD. This has significant implications in the clinical management of CCS patients, that ranges from lifestyle adaptation, medical therapy targeting all the elements contributing to CAD progression (i.e., platelet aggregation, coagulation, dyslipidaemia, and systemic inflammation), to invasive strategies (i.e., revascularization). CCS is the most frequent presentation of coronary artery disease which is the first cardiovascular disease worldwide. Medical therapy is the first line therapy for these patients; however, revascularization and especially percutaneous coronary intervention remains beneficial for some of them. European and American guidelines on myocardial revascularization were released in 2018 and 2021, respectively. These guidelines provide different scenarios to help physicians choose the optimal therapy for CCS patients. Recently, several trials focusing on CCS patients have been published. We sought to synthetize the place of revascularization in CCS patients according to the latest guidelines, the lessons learnt from recent trials on revascularization and medical therapy, and future perspectives.
Article
Ischemic vascular remodeling occurs in response to stenosis or arterial occlusion leading to a change in blood flow and tissue perfusion. Altered blood flow elicits a cascade of molecular and cellular physiological responses leading to vascular remodeling of the macro‐ and micro‐circulation. Although cellular mechanisms of vascular remodeling such as arteriogenesis and angiogenesis have been studied, therapeutic approaches in these areas have had limited success due to the complexity and heterogeneous constellation of molecular signaling events regulating these processes. Understanding central molecular players of vascular remodeling should lead to a deeper understanding of this response and aid in the development of novel therapeutic strategies. Hydrogen sulfide (H 2 S) and nitric oxide (NO) are gaseous signaling molecules that are critically involved in regulating fundamental biochemical and molecular responses necessary for vascular growth and remodeling. This review examines how NO and H 2 S regulate pathophysiological mechanisms of angiogenesis and arteriogenesis, along with important chemical and experimental considerations revealed thus far. The importance of NO and H 2 S bioavailability, their synthesis enzymes and cofactors, and genetic variations associated with cardiovascular risk factors suggest that they serve as pivotal regulators of vascular remodeling responses. © 2019 American Physiological Society. Compr Physiol 9:1213‐1247, 2019.
Article
Endothelial dysfunction is characterized by a reduction in nitric oxide generation or an unbalanced distribution of the relaxing and contracting pressures produced by endothelial cells. Endothelium was once assumed to be nothing more than a mechanical barrier. Endothelium is now recognized as a tissue that regulates tone, cell formation, leukocyte, thrombocyte, and artery wall contact. The pathophysiologic pathways behind endothelial dysfunction are of great interest, and researchers are working hard to figure out what they are and how they affect diabetes, psoriasis, Inflammatory Bowel Disease, hypertension, heart failure, renal failure, inflammation, stroke, atherosclerosis, and oxidative stress. Inflammatory activation and endothelial dysfunction are important processes in the progression and pathogenesis of atherosclerosis. Endothelial dysfunction in psoriasis patients with arterial hypertension is thought to be caused by a disturbance in the action of L-oxidative arginine's metabolism, as revealed by a decrease in NO bioavailability and a high degree of inactivation, oxidative stress, and an antioxidant state. The failing heart is characterised by irregular cardiac and vascular characteristics caused by abnormalities in NO bioavailability and oxidative stress. In such conditions the bioavailability of the NO can be increased by the various approaches like by using the NO generating compounds which can be done by either through inhalation of NO, use of the hybrid drugs like oral NO-NSAIDs combination or the use of inorganic nitrites. On other hand the endogenous NO levels can be increased by direct delivery of l‑arginine, l‑citrullineor Arginase inhibition. eNOS also can be activated by the use of statins, Corticosteroids, and Second generation β-adrenoceptor antagonists to increase NO level. This review will assist in understanding endothelial dysfunction, its pathogenesis, and its connection to a variety of disorders.