Skip to main content
Erschienen in: Journal of Translational Medicine 1/2024

Open Access 01.12.2024 | Review

Role of gut microbiota in doxorubicin-induced cardiotoxicity: from pathogenesis to related interventions

verfasst von: Chao Huang, Xiaoxia Li, Hanqing Li, Ruolan Chen, Zhaoqing Li, Daisong Li, Xiaojian Xu, Guoliang Zhang, Luning Qin, Bing Li, Xian-Ming Chu

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2024

Abstract

Doxorubicin (DOX) is a broad-spectrum and highly efficient anticancer agent, but its clinical implication is limited by lethal cardiotoxicity. Growing evidences have shown that alterations in intestinal microbial composition and function, namely dysbiosis, are closely linked to the progression of DOX-induced cardiotoxicity (DIC) through regulating the gut-microbiota-heart (GMH) axis. The role of gut microbiota and its metabolites in DIC, however, is largely unelucidated. Our review will focus on the potential mechanism between gut microbiota dysbiosis and DIC, so as to provide novel insights into the pathophysiology of DIC. Furthermore, we summarize the underlying interventions of microbial-targeted therapeutics in DIC, encompassing dietary interventions, fecal microbiota transplantation (FMT), probiotics, antibiotics, and natural phytochemicals. Given the emergence of microbial investigation in DIC, finally we aim to point out a novel direction for future research and clinical intervention of DIC, which may be helpful for the DIC patients.
Hinweise
Chao Huang, Xiaoxia Li and Hanqing Li contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AAA
Abdominal aortic aneurysm
Abs
Antibiotics
AKT
protein kinase B
ATP
adenosine triphosphate
AMPK
AMP-activated protein kinase
CTLA-4
cytotoxic T lymphocyte antigen-4
ETC
electron transport chain
FAB
phenylalanine-butyramide
FMT
Fecal microbiota transplantation
GF
germ-free
GLA
glabridin
H2S
hydrogen sulfide
IF
intermittent fasting
LPS
lipopolysaccharides
MI
myocardial infarction
NLRP3
nucleotide-binding domain-like receptor protein 3
NF-κB
nuclear factor-κB
OXPHOS
oxidative phosphorylation
PD-1
Programmed cell death 1
PD-L1
Programmed cell death ligand 1
PUFA
polyunsaturated fatty acids
ROS
reactive oxygen species
SCFAs
short-chain fatty acids
System Xc-
cystine-glutamate antiporter
TFEB
transcription factor EB
TMAVA
Trimethyl-5-aminovaleric acid
TMAO
trimethyl-amine N-oxide
TLRs
toll-like receptors
YWPC
yellow wine polyphenolic compound
ZnCM
Zn(ii)-curcumin

Introduction

   With the significant advancements in cancer treatment and detection technologies, the survival rate of cancer patients has greatly improved in the past half-century [1, 2]. Doxorubicin (DOX), an efficient anthracycline antibiotic, has become a cornerstone in the chemotherapy of various cancers, such as breast cancer, lymphoma, and leukemia [3]. However, approximately a quarter of the patients who receive this highly efficient treatment ultimately discontinue their regimen due to the potential DOX-induced cardiotoxicity (DIC), which greatly hampered the clinical application of DOX [4]. Despite the DIC has attracted widespread clinical attentions, clinically available drugs for the treatment of this condition are limited [5]. In recent years, mounting studies have verified the probable molecular mechanisms underlying DIC, and developed cardioprotective adjuvants for potential targets, such as antioxidants [6], AMP-activated protein kinase (AMPK) agonists [7, 8] and other natural compounds [9]. However, none of these strategies is unanimously recommended and many drug researches lack clinical data, emphasizing the need for further studies [10].
   Recent evidences indicated that gut microbiota have been closely linked to multiple cardiovascular diseases through regulating “gut-microbiota-heart axis” [1114]. It has been found that metabolites produced by the gut microbiota, short-chain fatty acids (SCFAs), are not only associated with energy metabolism [15] but also involved in the development of DIC [1618]. Importantly, a recent in vivo study demonstrated for the first time that exogenous supplementation of phenylalanine-butyramide (FAB) protected against DIC by preventing mitochondrial dysfunction [18]. In this review, we will concentrate on an array of studies investigating the role of gut microbiota in alleviating DIC. We will emphasize the supporting evidence for the cardioprotective effects of both gut microbiota and their metabolites. Finally, we summarize the potential interventions of microbiota-targeted therapeutics in DIC. We aim to offer novel perspectives that might contribute to developing potential therapeutic strategies to prevent DIC.

The molecular mechanism of DIC

   Multiple mechanisms have been proposed in DIC, including oxidative stress [19], mitochondrial dysfunction [20], autophagy [21], calcium dysregulation [22], cardiac metabolic alterations [23] and regulated cell death [24]. Nevertheless, the precise mechanism of DIC remains incompletely understood and subjects to debate. The prevailing belief among scholars is that inflammation plays a pivotal role in the development of DOX-induced cardiomyopathy [25, 26]. The nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome is as significant part of the innate immune system. It can be activated by toll-like receptors (TLRs), nuclear factor-κB (NF-κB), reactive oxygen species (ROS) and other signals [27]. DOX can also activate NLRP3 inflammasome. It was reported to stimulate the production of caspase-1 and the release of proinflammatory cytokines, such as IL-1 β and IL-18, which caused cardiomyocyte death and inflammatory response in cardiac tissue [28, 29]. Another in vivo study revealed that vitamin C improved cardiac structure and function in DIC by reducing oxidative/nitrosative stress, apoptosis, and inflammation. They found DOX could increase the secretion and release of NF-κB, which leads to inflammation reaction [30].
   It is notable that the immune cells play a dual role in cardiac homeostasis and disease [31]. On the one hand, a large number of immune cells are recruited to the heart to remove dying tissue and promote healing after myocardial infarction; on the other hand, they can also cause irreversible damage under some circumstances, which contributes to heart failure. Interestingly, recent studies have shown that immune cells and cytokines are involved in DOX-induced heart injury via activating inflammation [32]. For instance, a study has shown that monocytes and macrophage apoptosis is linked to acute inflammation brought by DOX [33]. Mechanistically, the TLR2/TLR9-MyD88 pathway mediated the innate immune’s perception of apoptotic cells, which was essential for the start of an acute inflammatory response to DOX. Although investigations have pointed out the importance of inflammation in causing cardiomyopathy, it is increasingly clear that DOX cardiotoxicity is triggered by multiple complex mechanisms. The mainstream established pathomechanisms of DIC are briefly summarized in Fig. 1, such as oxidative stress [6], ferroptosis [34], apoptosis [35], autophagy [3638], and energy metabolism disorders [39].
   With the growing understanding of DIC, the intervention of inflammation [40] and autophagy [41], are constantly being updated. Recent advances in molecular biology have significantly expanded the scope of research on DOX cardiotoxicity.

Gut microbiota and DOX-induced cardiotoxicity

Brief overview of gut microbiota

   Trillions of microorganisms inhabit in human guts, including bacteria, viruses, archaea and fungi [43, 44]. The caecum and proximal colon are the regions with the highest concentration of microbial biomass in the digestive system [45]. Extensive studies show that gut microbiota is crucial for the fermentation of dietary fibers and the synthesis of vitamins. Furthermore, they also exert a vital role in maintaining intestinal health and regulating the immune system [46]. Notably, the Bacteroidetes(B) and Firmicutes(F) are two dominant bacterial phyla in the healthy gut. A decrease in the F/B ratio is generally associated with a healthy gut microbiome, while a higher ratio may indicate a disrupted microbial balance, potentially related to certain diseases or health conditions [47]. A low F/B ratio is frequently attributed to a reduction in certain beneficial bacteria, which are subsequently replaced by potentially harmful bacteria within the same phyla, particularly Firmicutes. This alteration of the F/B ratio influenced by distinct lifestyle factors, such as dietary choices, sleep patterns, physical activity and medication usage.

Gut microbiota and DIC risk factors

   The connection between gut microbiota and cardiovascular risk factors, like obesity, advanced age, and hypertension, is complex and multifaceted. These risk factors can affect systemic conditions, including metabolic changes, chronic inflammation, and gut microbiota dysbiosis [48]. In turn, intestinal microbiota dysbiosis can lead to the worsening of these risk factors [49, 50]. For example, the progression of diabetes has been associated with a decrease in the abundance of certain bacterial groups, including Firmicutes and Bacteroidetes [51]. Similarly, studies have shown decline in the ratio of bacteria produced butyrate in individuals with diabetes [52]. Furthermore, multiple bacterial species were found to be linked to hypertension, including Lactobacillus, Klebsiella, Parabacteroides, Desulfovibrio, and Prevotella [53, 54]. Previous evidence showed that Lactobacillus could reduce hypertension involving in modulation of sympathetic activity, oxidative stress, vascular tone, and inflammatory response [55].
   Similarly, advanced age is linked to alterations in the composition and abundance of gut microbiota [56]. Older adults tend to have lower levels of core commensal bacteria like Bacteroides, Bifidobacterium, and Firmicutes, which can contribute to health deterioration [57, 58]. Additionally, cancer patients often experience gut microbiota dysbiosis, which can further complicate their cardiovascular risk factors [59].
   Notably, there is evidence suggesting a connection between cardiovascular risk factors and DIC [60]. For example, research findings indicated that there exists an elevated likelihood of cardiac dysfunction in individuals aged 65 years and above, as opposed to their younger counterparts [61]. Although there is evidence linking gut microbiota, DIC, and cardiovascular risk factors, further investigation is required to completely comprehend this interaction. Therefore, understanding the link between intestinal flora and cardiovascular risk factors and restoring gut microbiota dysbiosis in cancer patients before doxorubicin treatment could be a promising strategy to reduce DIC.

Gut-microbiota-heart (GMH) axis

   As raised above, the communication between the gut microbiota and the cardiovascular system is bidirectional [62]. This is because extensive research views the gut microbiota as a “endocrine organ” [63, 64]. On one hand, cardiovascular diseases can contribute to dysbiosis in the gut microbiota. On the other hand, the gut microbiota can also impact the cardiovascular system via its metabolites [11, 65]. For example, enhanced populations of Bacteroides/Prevotellain, Eubacterium rectale, and Fusobacterium prausnitzii, in patient with heart failure, as well as less abundant Coriobacteriaceae, Erysipelotrichaceae, and Ruminococcaceae [66, 67]. More recently, the notion of the gut-organ axis, such as the gut-brain axis [68], gut-heart axis [69], and gut-artery axis [70], has gained attention. This concept provides a solid foundation for understanding the relationship between the gut microbiota and multiple cardiovascular diseases [54, 7173].
   SCFAs, such as acetate, butyrate and propionate, are small molecular metabolites produced through the fermentation of dietary fiber. They perform a crucial function in the regulation of immunological equilibrium, bolstering the integrity of the intestinal barrier, and serving as a source of energy for the host [7476]. Study has shown that SCFAs, particularly butyrate, can support the failing heart by bypassing the enzyme CPT1 and serving as an alternative energy source [15]. Importantly, a decrease in SCFAs due to gut dysbiosis can disrupt the integrity of the intestinal barrier, resulting in the translocation of microbial metabolites and inflammation, thereby promoting the development of cardiovascular diseases. For instance, a cohort study identified that dysbiosis and reduced butyrate production were associated with the aggravation of abdominal aortic aneurysm (AAA) [77]. Another study found that propionate could alleviate vascular calcification induced by vitamin D3 deficiency by restoring the homeostasis of intestinal microorganisms, promoting SCFA production, preserving intestinal integrity, preventing “leakage,” and inhibiting inflammation [78]. Moreover, gut dysbiosis, characterized by reduced butyrate production, can increase intestinal permeability and systemic inflammation, which in turn promote atherosclerosis and heart failure.
   Another metabolite, called trimethylamine N-oxide (TMAO), has also been found closely linked to cardiovascular disease. Research conducted by Makrecka-Kuka et al. demonstrated that feeding mice with 120 mg/kg of TMAO for 8 weeks resulted in elevated plasma TMAO levels, which subsequently impacted cardiac energy metabolism and mitochondrial function. This influence on energy metabolism and mitochondrial function ultimately led to ventricular remodeling and the development of heart failure [79]. Furthermore, Savi et al. discovered that TMAO affected the contractile function and intracellular calcium processing in cardiomyocytes. This effect is believed to be due to reduced energy production caused by TMAO-induced mitochondrial dysfunction [80]. In addition to these findings, studies have shown that the gut microbiota can influence platelet hyperresponsiveness and the formation of blood clots through the production of TMAO [81].
   In addition to TMAO and SCFAs, recent investigation has revealed the involvement of a gut microbiota-derived metabolite called Trimethyl-5-aminovaleric acid (TMAVA) in cardiovascular health. This study suggests that TMAVA can reduce fatty acid oxidation (FAO) and increase lipid accumulation, leading to the inhibition of carnitine synthesis and uptake. As a result, it can accelerate cardiac hypertrophy, a condition characterized by an abnormal increase in the size and mass of the heart [82]. This dysbiosis, or imbalance in the gut microbiota composition, is thought to be associated with increased inflammation and oxidative stress, which can further contribute to the progression of acute heart failure. While there is growing evidence suggesting a role of dysbiosis in the pathogenesis of DIC, the definitive role of gut microbiota dysbiosis in this condition remains unclear. Additional investigation is required in order to have a comprehensive understanding of the causes and consequences of gut microbiota-derived metabolites in relation to cardiovascular diseases and their association with DIC.

The potential pathogenesis involved in gut microbiota in DIC

   Drugs also have significant effects on the composition and function of intestinal microbiota [83]. The interactions between drugs and microbiota can influence bacterial metabolism, as well as the activity and effectiveness of the drugs [84]. Previous studies have found that doxorubicin can cause substantial changes in the intestinal flora [85, 86]. Several studies have shown that mice treated with doxorubicin exhibited gut dysbiosis compared to the control group [8789]. For instance, certain bacteria like Faecalibaculum, Dubosiella, and Lachnospiraceae were found to be increased in mice treated with DOX, while others such as Allobaculum, Muribaculum, and Lachnoclostridium were decreased [88]. Additionally, a recent study indicated that doxorubicin could reduce its toxicity to a model species called Caenorhabditis elegans through the presence of Raoultella panticola [90]. Notably, Lactic acid bacteria (LAB) and Bifidobacteria play an important role in the gut microbiota balance of the host health. However, study showed that most LAB and Bifidobacteria were easily inhibited by DOX [85]. Lin and colleagues showed that DOX treatment significantly altered the gut microbiota composition in rats, leading to a reduction in Bacteroidetes and Verrucomicrobia, and an increase Proteobacteria, and Epsilonbacteraeota [91]. Further investigations revealed that this dysbiosis was associated with cardiac inflammation and mitochondrial dysfunction, contributing to the development of cardiotoxicity. Notably, the transplantation of fecal microbiota from DOX-treated mice to germ-free (GF) mice induced cardiotoxicity, indicating a key role of gut microbiota dysbiosis in DIC [87]. Moreover, it was shown that Proteobacteria exhibited a considerably high prevalence in rats treated with DOX, suggesting that it might possibly serve as a sensitive indicator to distinguish between rats with and without cardiotoxicity [91]. With the advancement of metabolomics and 16s rDNA gene sequencing technique nowadays, more biomarkers may be used as a diagnostic in diseases. For instance, recent study has shown that an increased abundance of Proteobacteria is a potential diagnostic signature for dysbiosis and disease risk [92]. Similarly, a multicenter clinical trial indicated that special microbiota (e.g. Enterococcus spp) can be used as potential biomarkers for patient to monitor during intensive care unit stay [93]. Furthermore, a systematic review demonstrated that gut microbiota-generated metabolite TMAO was clearly associated with cardiovascular risk and mortality, and may be a potential biomarker [94]. Despite it remains challenging to identify specific gut microbiota dysbiosis in drug-induced cardiotoxicity, but it shows promise as a diagnostic tool.
   Although the gut microbiota dysbiosis caused by DOX has been reported, the exact mechanism by which this dysbiosis affects drug-induced cardiotoxicity is not fully understood. A more comprehensive understanding of this relationship could potentially contribute to the development of effective cardioprotective adjuvants. Currently, several possible mechanisms have been proposed, including intestinal barrier dysfunction, inflammatory response and immune activation, alteration of SCFAs and Hydrogen sulfide (H2S).

Intestinal barrier dysfunction

   The intestinal barrier is primarily made up of three components: the mucus layer, an epithelial barrier, and a gut vascular barrier. These components play a crucial role in maintaining overall health and preventing disease by facilitating nutrient absorption and simultaneously preventing the entry of pathogens [95]. Actually, the intestinal barrier is constantly exposed to various immunological and microbiological factors. When the barrier function is compromised, pathogens and bacterial metabolites such as lipopolysaccharides (LPS) can enter the bloodstream, leading to damage in distant organs, including the heart [96]. Previous studies in mice have indicated that the presence of gut microbiota can lead to intestinal mucosal damage when exposed to DOX [97]. These damages included the decrease in crypt number, crypt proliferation, and villus height. Besides, DOX administration also resulted in significant increases in apoptosis in jejunal epithelium. Immunostaining for MUC2 and lysozyme indicated the changes of goblet cells, Paneth cells or dual stained intermediate cells [97].
   The epithelial barrier function relies on the presence of a continuous layer of cells and tight junctions that seal the space between these cells, known as the paracellular space. The interecellular junctions consist of tight junction (zonula occludens), the adherens junction (zonula adherens), and the desmosome. Among them, the tight junction is the primary determinant of paracellular permeability. Molecularly, claudins are the most important of the transmembrane tight junction proteins [98]. The ZO family proteins, including ZO-1, ZO-2, and ZO-3, serve as scaffolding proteins that directly interact with transmembrane tight junction proteins like claudins and the tight junction-associated Marvel protein (TAMP) family, which includes occludin. These interactions help to regulate tight junction assembly and stability, thereby maintaining the integrity of the epithelial barrier [99]. A recent study has demonstrated that treatment with DOX can disrupt the organization of tight junction proteins, such as CLAUDIN-2, leading to an increase in the passage of small macromolecules, including bacterial products, across the epithelial barrier. This occurs due to alterations in the expression of tight junctional components and the loosening of cellular tight junctions, ultimately compromising the integrity of the barrier [100]. Consequently, the development of DIC has been linked to impaired intestinal barrier function. Moreover, a clinical study also found that increased gut permeability was associated with the deterioration of chronic heart failure [101]. Recently, abundant studies have taken the intestinal barrier as a potential therapeutic target in many diseases [102105]. Regrettably, there is limited research available regarding the relationship between DIC and intestinal barrier homeostasis. Additional research is required to investigate whether targeting the intestinal barrier could be a promising therapeutic approach for the treatment of DIC.

Inflammatory response and immune activation

   Gut dysbiosis leads to the translocation of bacterial products (especially LPS) from the gut into the bloodstream, causing immune dyshomeostasis and systemic inflammation. A study by Zhao et al. showed that intestinal bacteria translocation influences myocardial ischemia/reperfusion (I/R) injury by amplifying the inflammatory response, and they proposed a novel concept known as the heart-gut-microbiome-immune axis [106]. Additionally, Tian et al. discovered obvious dysbiosis of gut microbiota and changes in metabolites in patients with AAA through the use of 16s rRNA gene sequencing and metabolomics. Further investigation in AAA mice demonstrated that R. intestinalis and its metabolite butyrate significantly reduce neutrophil infiltration and NOX2-dependent neutrophil extracellular trap formation, thus alleviating inflammation and markedly reducing AAA development [77]. These findings suggest that inflammation caused by gut microbial invasion and translocation play an important role in the progress of cardiovascular disease.
   Gut microbial dysbiosis and immune response activation can contribute to DIC [89]. For instance, several studies have shown that DOX can induce intestinal mucositis and damage [107]. As a result, LPS can enter the bloodstream through the compromised intestinal barrier, triggering the expression of various downstream inflammatory products such as tumor necrosis factor (TNF), IL-1, and IL-6, via the TLR4 pattern recognition receptor. Systemic inflammation, in turn, contributes to DIC by activating proinflammatory signaling pathways in the heart. Additionally, TMAO, a signaling molecule derived from bacterial species like Firmicutes and Actinobacteria, has been implicated in inflammatory modulation by promoting the release of IL-18 and IL-1β, and it has been closely associated with cardiovascular diseases such as atherosclerosis, hypertension, and heart failure [63, 108110]. Zhang et al. reported that TMAO promoted vascular calcification through the activation of NLRP3 inflammasome and NF-κB signals [111]. Similarly, Li et al. demonstrated that TMAO exacerbates DOX-induced cardiac fibrosis by upregulating the expression of TLR4 and activating the NLRP3 inflammasome [112]. These findings indicate that alterations in gut microbiota may contribute to the development of doxorubicin-induced cardiomyopathy, involving interactions with the host immune system.

Alteration of SCFAs

   As mentioned before, SCFAs participate in anti-inflammatory responses and immunomodulation in the host. There is evidence suggesting that a decrease in SCFAs can lead to mitochondrial dysfunction, oxidative stress, and inflammation in the heart, contributing to DIC [18, 113, 114]. Chen et al. conducted a study that revealed a mouse model induced by programmed cell death 1(PD-1) or its ligand 1(PD-L1) inhibitors exhibited gut microbial dysbiosis. This dysbiosis was characterized by a notable decrease in SCFA-producing bacteria such as Prevotellaceae and Rikenellaceae, resulting in lower production of butyrate, an important SCFAs. The oral administration of butyrate-producing bacteria, such as Prevotellaloescheii, or butyrate itself has been shown to alleviate cardiotoxicity [115]. This effect was achieved by reducing the expression of pro-inflammatory cytokines like IL-1β and TNF-α and promoting the polarization of M2 macrophages in the colon. As a result, inflammatory responses were reduced, leading to a mitigation of cardiotoxicity.
   Similarly, a recent and promising study has demonstrated, for the first time, that oral administration of FAB, a novel synthetic derivative of butyrate, can protect the heart from DIC. This protective effect was mediated by reducing cardiac fibrosis and apoptosis, as well as decreasing levels of nitrosative and oxidative stress mediators, thereby improving mitochondrial dysfunction [18]. Recently, Nataly and colleagues have demonstrated that both butyrate and AN-7 (a butyric acid prodrug) could protect against DIC by reducing inflammatory factors [17]. Furthermore, oral administration of butyrate was shown to reduce cardiomyocyte apoptosis and induce an anti-inflammatory effect by promoting the polarization of anti-inflammatory M2 macrophages in the colon [89]. Another study found that butyrate could not only enhance the anticancer activity of doxorubicin but also protect cardiomyocytes from the side effects induced by DOX [16]. However, the specific mechanism of action still requires further exploration.

Alteration of hydrogen sulfide (H2S)

   Hydrogen sulfide (H2S) is an essential endogenous gaseous molecule that exert an important role in modulating numerous physiological and pathological processes, including cardiovascular diseases [116]. Studies have demonstrated that H2S can protect cardiomyocytes from DIC through its anti-inflammatory, anti-apoptotic, and antioxidant properties [117]. Moreover, emerging study suggests that H2S donors and prodrugs have dual activity, which not only synergizes with the anticancer effects of DOX but also protects cardiomyocytes from doxorubicin-induced damage [118120]. These findings indicate that H2S holds great promise as a preclinical myocardial protective agent in the context of DIC.
   The majority of existing research has been concentrated on endogenously produced H2S by the host. However, it is worth noting that gut microbiota also produces H2S, which has the potential to impact human homeostasis. Some studies have reported a close association between gut microbiota-derived H2S and conditions like hypertension and atherosclerosis [121123]. Furthermore, several studies have identified a correlation between the dysbiosis of gut microbiota in GF mice and the reduction of cystathionine gamma-lyase (CSE) activity in several organs, including the heart. This suggests that gut microbiota may also impact the expression of CSE in cardiomyocytes [124]. However, further investigation is needed to determine whether H2S derived from the gut microbiota is involved in the onset and progression of DIC.
   In summary, gut microbiota dysbiosis appears to exacerbate the development of DIC, including inflammatory responses and immune regulation (Fig. 2). Although the precise mechanism linking gut microbiota and DIC remains incompletely understood, the aforementioned research suggests that targeting the gut microbiota could be an encouraging strategy for ameliorating DIC.

The potential interventions of microbiota-targeted therapeutics in DIC

   The occurrence of DIC varies widely and is influenced by factors, such as lifetime cumulative dose and underlying comorbidities in patients. Evidence suggests that patients with risk factors like obesity, diabetes, and hyperlipidemia are more susceptible to DIC due to metabolic remodeling [48]. Recent studies have also demonstrated the beneficial role of gut microbiota in reducing cholesterol levels in the host, providing a strong foundation for the development of novel probiotics for the prevention and treatment of cardiovascular diseases [125]. Additionally, numerous animal studies and a limited number of human studies have shown promising potential of gut microbiota in preventing and treating cardiovascular diseases and metabolic syndrome, generating widespread interest among researchers [126128]. While few studies have directly linked the gut microbiota to the risk factors associated with doxorubicin-induced cardiotoxicity and improving these risk factors to mitigate cardiotoxicity, this direction holds promise as a novel area for further investigation.
In recent years, there has been an increasing scholarly focus on the interaction between microbiota and anticancer medications, along with the exploration of therapies aimed at shaping the microbiota to enhance therapeutic effectiveness and mitigate adverse reactions [129]. Numerous studies have demonstrated that gut microbiota not only influence chemotherapy effectiveness but also modulate the side effects of anticancer drugs, as previously reviewed [130]. A new field called Pharmacomicrobiomics has emerged to explore the impact of variations in microbiome composition and function on drug efficacy, toxicity, and pharmacokinetics [130]. The diversity of gut microbiota grants it an extraordinary metabolic capacity surpassing that of the host [131, 132]. Gut microbiota can produce a range of metabolic responses to drugs and xenobiotics, leading to both direct effects on drug metabolism and toxicity. These responses can ultimately influence individual variability in drug response [133]. Thus far, several enterobacteria capable of inactivating DOX, by deglycosylation to 7-deoxydoxorubicinol and 7-deoxydoxorubicinolone, have been identified, such as Raoultella_planticola [90]. Microbial detoxification of DOX could impact its therapeutic concentration in cancer patients, significantly mitigating its off-target toxicity [134]. In this context, the simultaneous administration of probiotics, symbiotics, postbiotics, or antibiotics with anticancer therapy has been proposed to restore balance to the gut microbiota [135]. For instance, Wang et al. indicated that cytotoxic T lymphocyte antigen-4 (CTLA-4) related-colitis was ameliorated by giving probiotic Bifidobacterium in mice [136]. Similarly, supplement of probiotics could reduce chemo/radiation therapy toxicity in many cancers’ treatment as well [137]. This section will describe the potential interventions of microbe-targeted therapeutics in DIC (Fig. 3).

Dietary interventions

   Gut microbiota is closely linked to daily diets, and even a short-term adjustment in the diet, as short as 5 days, is sufficient to alter the composition of gut microbiota and induce corresponding changes for adaptation [138]. Stewart et al. discovered that long-term ingestion of milk or kefir (a milk containing lactic acid bacteria) could be beneficial for rats treated with doxorubicin [139]. Additionally, it has been demonstrated that the administration of phenylalanine-butyramide (a novel synthetic derivative of butyric acid) can have a positive effect on DIC [18]. A recent study has shown that supplementation of dietary Zn(ii)-curcumin (ZnCM) can attenuate gut dysbiosis during DOX-induced cardiomyopathy in vivo [140]. Furthermore, recent investigation has indicated that intermittent fasting (IF) has the potential to modulate the composition of gut microbiota, contributing to improvements in obesity and host energy metabolism [141]. Interestingly, a recent significant study found that alternate-day fasting (ADF) can aggravate DIC through transcription factor EB (TFEB)-mediated autophagy [41]. However, whether ADF affects the occurrence of DIC by altering gut microbiota is an area that deserves further exploration. Overall, dietary interventions have the potential to influence gut microbiota and assist in the prevention of DIC, but additional trials are needed to establish their efficacy.

Probiotic therapy

   Probiotics are live microorganisms that provide health benefits to the host when administered in sufficient amounts [142]. They are the most extensively studied microbiota-targeting therapies for cardiovascular diseases [143]. Probiotics are known to alter the intestinal microbiota, create substances that can protect against harmful microbes, increase the integrity of the intestinal barrier, and reduce intestinal inflammation, however the exact mechanism by which they work is yet unknown [144]. Additionally, it was show that probiotics also had anti-oxidant and anti-inflammatory activities [145]. A study conducted by Zhao et al. has demonstrated that Lactobacillus supplementation can prevent cisplatin-induced cardiotoxicity possibly by inhibiting inflammation [146]. Similarly, recent animal studies have shown that the consumption of probiotics may have potent anti-cytotoxic effects [147], and a diet containing yogurt components can improve membrane integrity and cardiac contractility in a rat model of DOX-induced cardiomyopathy [148]. This probiotic treatment restored the composition and function of gut microbiota, reduced oxidative stress and inflammation, and improved DOX-induced cardiomyopathy [148]. Amaretti et al. observed an increase in reactive oxygen species metabolites in the plasma of rats treated with DOX, which was alleviated after supplementation with Bifidobacterium [149]. Based on the above research findings, supplementation with probiotics after DIC occurrence can alleviate myocardial cell damage and improve cardiac function. The application of probiotics to patients with DIC could serve as one of the auxiliary treatment options for clinical physicians. Additionally, probiotics may offer benefits by addressing microbiota dysbiosis-associated risk factors for DIC, such as obesity, aging, and hypertension. Although it has been reported that probiotics could protect against DIC, further work is needed to clarify the possible mechanisms and specific beneficial strains.

Fecal microbiota transplantation

   Fecal microbiota transplantation (FMT) is a strategy to improve gut microbiota dysbiosis via also transferring feces from a healthy donor to the intestines of another patient [150]. Preclinical studies have demonstrated that FMT has encouraging outcomes in alleviating heart failure. Chen et al. recently demonstrated that recolonization with Prevotella loescheii and supplementation with butyrate reduced cardiotoxicity associated with PD-1/PD-L1 inhibitors [115]. Another study indicated that FMT improved DOX-induced cardiac dysfunction by altering the composition of the gut microbiota [87]. Whereas there have been reports on the adverse effects and complications of FMT therapy, such as the introduction of viral communities [151], this treatment approach is gaining momentum in scientific research and clinical practice. One promising development is the exploration of alternative forms of FMT, such as pills derived from human feces, which offer a less invasive and more standardized approach. It is imperative to acknowledge that further investigation is required so as to comprehensively understand the underlying benefits, risks, and optimal applications of FMT in the context of cardiotoxicity and other conditions.

Natural phytochemicals

   Natural drugs primarily refer to pharmacologically active natural products found in nature, such as plants [152]. Increasing evidence suggests that natural phytochemicals have the ability to influence both gut microbiota and cardiac function [153]. Lin et al. conducted a study demonstrating that a polyphenolic compound from yellow wine (YWPC) effectively alleviated inflammation induced by DOX and improved mitochondrial function. This effect was attributed to the modulation of gut microbial ecology and related metabolites [91]. Similarly, another study showed that the flavonoid Glabridin (GLA) regulated DOX-induced gut microbiota dysbiosis, which contributed to the prevention of cardiotoxicity, potentially involving the Desulfovibrio genus [89]. Additionally, Wu et al. observed that polyphenols from Arctium Lappa L. (ALPP) improved gut microbiota dysbiosis and ameliorated heart failure in mice treated with doxorubicin. ALPP increased the abundance of Lactobacillaceae, Muribaculaceae, and Ruminococcaceae, while decreasing the abundance of Proteobacteria, Enterobacteriaceae, and the Escherichia_Shigella group compared to the DOX-treated group [154]. These results demonstrate the potential of natural compounds in modulating the gut microbiota and mitigating cardiotoxicity. However, to completely understand the potential mechanisms of action and to maximize their therapeutic application, more study is necessary.

Antibiotics

   Antibiotics(Abs) possess both positive and negative effects [155]. The improper utilization of antibiotics has the potential to disturb an individual’s microbiome, resulting in adverse outcomes. Nevertheless, in specific circumstances, antibiotics can prove advantageous, particularly when adverse events are triggered by microbial translocation. One example is the utilization of Rifaximin as a therapeutic intervention for the management of microbiota toxicity and translocation. This medication has anti-inflammatory properties and facilitates the proliferation of advantageous microbial species, such as Bifidobacteria and Lactobacillus [156]. Similarly, a study by Shen et al. showed that mechanical hyperalgesia induced by oxaliplatin was alleviated in germ-free (GF) mice and mice treated with antibiotics, compared to the control group [157]. Huang et al. conducted an experiment in which they depleted the gut microbiota using antibiotics and found that this intervention could alleviate DIC [88]. Furthermore, a recent study demonstrated that minocycline could improve DIC by reducing inflammation and oxidative stress [158]. However, some studies have indicated that Abs-induced gut microbiota dysbiosis exacerbates the disease [159, 160]. For example, recent findings demonstrated that mice treated with antibiotics before myocardial infarction (MI) exhibited significantly increased mortality compared to MI mice models not administered antibiotics. Further investigation revealed an association between MI and gut microbiota dysbiosis, particularly affecting Lactobacillus [161]. Additionally, a rat study showed that broad-spectrum antibiotic treatment worsened DIC. Subsequent 16s rDNA gene sequencing revealed significantly higher abundance of Klebsiella, Providencia, and Parasutterella in the DOX + Abs group, whereas the abundance of Muribaculaceae_unclassified, Lactobacillus, and Firmicutes_unclassified was significantly lower compared to the DOX group [91]. While there is limited research on antibiotics-based treatments for DIC, antibiotics show promise as a microbial therapeutic approach for this condition. Notably, before considering antibiotics as a viable therapeutic option, further research is necessary to elucidate their precise mechanisms of action on gut microbiota and potential benefits in DIC. Otherwise, indiscriminate use may result in nonspecific depletion of the microbiota. The representative study of gut microbiota in the treatment of DIC is summarized in Table 1.
Table 1
Representative research on gut microbiota for the treatment of DIC
Interventions
Model
Microbiota changes
Main findings
Ref
FBA
Mice
N/A
Reduced oxidation stress,
mitochondrial dysfunction.
[18]
FMT
Mice
Prevotellaceae_UGG-001,
Alloprevotella, Rilennellaceae_RC9
Increased ZO-1 expression,
improved cardiac function,
altering microbiota composition.
[87]
Antibiotics
Mice
Depletion of gut microbiota
Alleviated DOX-induced myocardial injury and cardiomyocyte apoptosis.
[88]
GLA
Mice
Lactobacillus genus
Desulfovibrio genus
Prevented DIC, altered gut microbiota and colonic macrophage phenotype.
[89]
YWPC
Rat
Alloprevotella, Eubacteriaceae,
Negativibacillus;
Proteobacteria, Escherichia-Shigellagroup, Gammaproteobacteria
Alleviated inflammation and mitochondrial dysfunction,
modulated gut microbial community;
[91]
ZnCM
Rat
Firmicutes
Bacteroidetes
Alleviated gut dysbiosis, improved heart function; reduced cardiomyocyte apoptosis;
[140]
ALPP
Mice
Lactobacillaceae, Muribaculaceae, Ruminococcaceeae;
Proteobacteria, Enterobacteriaee, Escherichia_Shigella;
Reduced heart failure; improved gut microbiota composition;
[154]
Abbreviations DIC, doxorubicin-induced cardiotoxicity; SCFA, short chain fatty acid; FBA, phenylalanine-butyramide; YWPC, yellow wine polyphenolic compound; ZnCM, Zn(ii)-curcumin; FMT, Fecal microbiota transplantation; ALPP, Arctium Lappa L; GLA, Glabridin; N/A, not applicable

Conclusions and future perspectives

   The gut microbiota exerts a crucial role in the regulation of cardiovascular diseases, and emerging study suggests that changes in the gut microbiota and its metabolites may potentially contribute to the onset of DIC. Targeting the gut microbiota presents a potential avenue for novel DIC prevention strategies. Currently, various microbial-targeted interventions, such as dietary changes, FMT, probiotics, antibiotics, and natural phytochemicals, have been explored for DIC prevention. However, several important challenges need to be addressed to further advance the field:
(1) Mechanistic understanding: The extent and specific mechanisms by which alterations in the gut microbiota led to DIC are still not fully understood. (2) Establishing causal relationships: Determining the causal relationship between the gut microbiota and DIC poses a challenge that requires further investigation. (3) Identifying probiotic candidates: Identifying specific microbiota strains that can be considered as probiotics for DIC treatment is an ongoing research area. (4) Translating findings to clinical practice: While most studies on gut microbiota and DIC are conducted on animal models, it is crucial to achieve the translation of these findings to human clinical settings considering the differences between human and animal gut microbiota.
   Addressing these challenges will be essential for the development of effective microbial-based therapeutics for DIC prevention and treatment. To address these issues, several steps can be taken. Firstly, it is crucial to enhance interdisciplinary collaboration and carry out large-scale prospective studies to gather more comprehensive information. This can facilitate a better understanding of the relationship between gut microbiota and DIC. Secondly, more preclinical and clinical studies are needed to employ advanced techniques like high-throughput sequencing and multi-omics approaches. These methods can help identify the specific pathogenic and beneficial strains associated with DIC. Thirdly, the utilization of innovative tools such as engineered strains and isotope tracing can shed light on the underlying pathways involving bacteria and their metabolites.
   Currently, there is a lack of studies in this area, thus further research can be conducted from two perspectives: (1) Elucidating the functional roles of specific bacterial strains and their metabolites in the development and prevention of DIC. (2) Investigating the clinical translation of FMT from animal models to human patients, considering the differences in gut microbiota between species. By addressing these aspects, we can make significant progress in understanding the role of gut microbiota in DIC and developing effective microbiota-based therapies for the condition.
   In addition to the previously mentioned approaches, there are emerging strategies targeting energy sources and metabolic pathways that have shown potential benefits in DIC. These include intermittent fasting [41], ketogenic diet [162], and exercise [163]. These approaches have been found to have positive effects on DIC management. It is important to note that the gut microbiota is highly susceptible to external factors, such as diet. The composition of gut microbiota can vary significantly between individuals, even among healthy individuals. Therefore, future studies should aim to investigate and clarify the effects of various confounding factors on gut microbiota.
   The emerging field of pharmacomicrobiomics is paving the way for innovative therapeutic approaches to various diseases, including cardiovascular disease [133, 164]. This approach involves initially reviewing the gut microbiota profile before treatment and then analyzing the patient’s microbiota after treatment. Moreover, exploring the potential for personalized treatment based on each individual’s microbiota and their ability to metabolize drugs correctly holds great promise. It is noteworthy that some patients with specific microbiota diversity may not metabolize drugs like DOX correctly, leading to potential harm instead of a beneficial intervention.
   Understanding the impact of external factors on gut microbiota composition can provide valuable insights into the potential influence of dietary interventions, such as intermittent fasting and ketogenic diet, on DIC prevention and treatment. Additionally, exploring the role of exercise in modulating gut microbiota and its potential benefits in DIC management can contribute to the development of comprehensive treatment strategies. Further studies focusing on these areas will help unravel the intricacies of the gut microbiota-DIC relationship and pave the way for personalized interventions targeting the gut microbiota for DIC prevention and management.

Acknowledgements

Thank you for the help of the members from the Department of Cardiology at the Affiliated Hospital of Qingdao University.

Declarations

Not applicable.
Not applicable.

Competing interests

There are no conflicts of interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Miller KD, Nogueira L, Devasia T, et al. Cancer treatment and survivorship statistics, 2022. CA Cancer J Clin. 2022;72(5):409–36.PubMedCrossRef Miller KD, Nogueira L, Devasia T, et al. Cancer treatment and survivorship statistics, 2022. CA Cancer J Clin. 2022;72(5):409–36.PubMedCrossRef
2.
Zurück zum Zitat Kocarnik JM, Compton K, Dean FE, et al. Cancer Incidence, Mortality, Years of Life Lost, Years lived with disability, and disability-adjusted life years for 29 Cancer groups from 2010 to 2019: a systematic analysis for the global burden of Disease Study 2019. JAMA Oncol. 2022;8(3):420–44.PubMedCrossRef Kocarnik JM, Compton K, Dean FE, et al. Cancer Incidence, Mortality, Years of Life Lost, Years lived with disability, and disability-adjusted life years for 29 Cancer groups from 2010 to 2019: a systematic analysis for the global burden of Disease Study 2019. JAMA Oncol. 2022;8(3):420–44.PubMedCrossRef
3.
Zurück zum Zitat Curigliano G, Cardinale D, Dent S, et al. Cardiotoxicity of anticancer treatments: Epidemiology, detection, and management. CA Cancer J Clin. 2016;66(4):309–25.PubMedCrossRef Curigliano G, Cardinale D, Dent S, et al. Cardiotoxicity of anticancer treatments: Epidemiology, detection, and management. CA Cancer J Clin. 2016;66(4):309–25.PubMedCrossRef
4.
Zurück zum Zitat Zamorano JL, Lancellotti P, Rodriguez Munoz D, et al. 2016 ESC position paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines: the Task Force for cancer treatments and cardiovascular toxicity of the European Society of Cardiology (ESC). Eur Heart J. 2016;37(36):2768–801. Zamorano JL, Lancellotti P, Rodriguez Munoz D, et al. 2016 ESC position paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines: the Task Force for cancer treatments and cardiovascular toxicity of the European Society of Cardiology (ESC). Eur Heart J. 2016;37(36):2768–801.
5.
Zurück zum Zitat Von Hoff DD, Layard MW, Basa P, et al. Risk factors for doxorubicin-induced congestive heart failure. Ann Intern Med. 1979;91(5):710–7.CrossRef Von Hoff DD, Layard MW, Basa P, et al. Risk factors for doxorubicin-induced congestive heart failure. Ann Intern Med. 1979;91(5):710–7.CrossRef
6.
Zurück zum Zitat Stěrba M, Popelová O, Vávrová A, et al. Oxidative stress, redox signaling, and metal chelation in anthracycline cardiotoxicity and pharmacological cardioprotection. Antioxid Redox Signal. 2013;18(8):899–929.PubMedPubMedCentralCrossRef Stěrba M, Popelová O, Vávrová A, et al. Oxidative stress, redox signaling, and metal chelation in anthracycline cardiotoxicity and pharmacological cardioprotection. Antioxid Redox Signal. 2013;18(8):899–929.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Gratia S, Kay L, Potenza L, et al. Inhibition of AMPK signalling by doxorubicin: at the crossroads of the cardiac responses to energetic, oxidative, and genotoxic stress. Cardiovasc Res. 2012;95(3):290–9.PubMedCrossRef Gratia S, Kay L, Potenza L, et al. Inhibition of AMPK signalling by doxorubicin: at the crossroads of the cardiac responses to energetic, oxidative, and genotoxic stress. Cardiovasc Res. 2012;95(3):290–9.PubMedCrossRef
8.
Zurück zum Zitat Singh M, Nicol AT, DelPozzo J, et al. Demystifying the Relationship between Metformin, AMPK, and Doxorubicin Cardiotoxicity. Front Cardiovasc Med. 2022;9:839644.PubMedPubMedCentralCrossRef Singh M, Nicol AT, DelPozzo J, et al. Demystifying the Relationship between Metformin, AMPK, and Doxorubicin Cardiotoxicity. Front Cardiovasc Med. 2022;9:839644.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Liu C, Ma X, Zhuang J, Liu L, Sun C. Cardiotoxicity of doxorubicin-based cancer treatment: what is the protective cognition that phytochemicals provide us? Pharmacol Res. 2020;160:105062.PubMedCrossRef Liu C, Ma X, Zhuang J, Liu L, Sun C. Cardiotoxicity of doxorubicin-based cancer treatment: what is the protective cognition that phytochemicals provide us? Pharmacol Res. 2020;160:105062.PubMedCrossRef
10.
Zurück zum Zitat Chen Y, Shi S, Dai Y. Research progress of therapeutic drugs for doxorubicin-induced cardiomyopathy. Biomed Pharmacother. 2022;156:113903.PubMedCrossRef Chen Y, Shi S, Dai Y. Research progress of therapeutic drugs for doxorubicin-induced cardiomyopathy. Biomed Pharmacother. 2022;156:113903.PubMedCrossRef
12.
Zurück zum Zitat Du Z, Wang J, Lu Y, et al. The cardiac protection of Baoyuan decoction via gut-heart axis metabolic pathway. Phytomedicine. 2020;79:153322.PubMedCrossRef Du Z, Wang J, Lu Y, et al. The cardiac protection of Baoyuan decoction via gut-heart axis metabolic pathway. Phytomedicine. 2020;79:153322.PubMedCrossRef
13.
Zurück zum Zitat Ge J, Shelby SL, Wang Y, et al. Cardioprotective properties of quercetin in fescue toxicosis-induced cardiotoxicity via heart-gut axis in lambs (Ovis Aries). J Hazard Mater. 2023;458:131843.PubMedCrossRef Ge J, Shelby SL, Wang Y, et al. Cardioprotective properties of quercetin in fescue toxicosis-induced cardiotoxicity via heart-gut axis in lambs (Ovis Aries). J Hazard Mater. 2023;458:131843.PubMedCrossRef
14.
Zurück zum Zitat Zhao J, Zhang Q, Cheng W, et al. Heart-gut microbiota communication determines the severity of cardiac injury after myocardial ischaemia/reperfusion. Cardiovasc Res. 2023;119(6):1390–402.PubMedPubMedCentralCrossRef Zhao J, Zhang Q, Cheng W, et al. Heart-gut microbiota communication determines the severity of cardiac injury after myocardial ischaemia/reperfusion. Cardiovasc Res. 2023;119(6):1390–402.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Carley AN, Maurya SK, Fasano M, et al. Short-chain fatty acids outpace Ketone Oxidation in the failing heart. Circulation. 2021;143(18):1797–808.PubMedPubMedCentralCrossRef Carley AN, Maurya SK, Fasano M, et al. Short-chain fatty acids outpace Ketone Oxidation in the failing heart. Circulation. 2021;143(18):1797–808.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Rephaeli A, Waks-Yona S, Nudelman A, et al. Anticancer prodrugs of butyric acid and formaldehyde protect against doxorubicin-induced cardiotoxicity. Br J Cancer. 2007;96(11):1667–74.PubMedPubMedCentralCrossRef Rephaeli A, Waks-Yona S, Nudelman A, et al. Anticancer prodrugs of butyric acid and formaldehyde protect against doxorubicin-induced cardiotoxicity. Br J Cancer. 2007;96(11):1667–74.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Tarasenko N, Kessler-Icekson G, Boer P, et al. The histone deacetylase inhibitor butyroyloxymethyl diethylphosphate (AN-7) protects normal cells against toxicity of anticancer agents while augmenting their anticancer activity. Invest New Drugs. 2012;30(1):130–43.PubMedCrossRef Tarasenko N, Kessler-Icekson G, Boer P, et al. The histone deacetylase inhibitor butyroyloxymethyl diethylphosphate (AN-7) protects normal cells against toxicity of anticancer agents while augmenting their anticancer activity. Invest New Drugs. 2012;30(1):130–43.PubMedCrossRef
18.
Zurück zum Zitat Russo M, Guida F, Paparo L, et al. The novel butyrate derivative phenylalanine-butyramide protects from doxorubicin-induced cardiotoxicity. Eur J Heart Fail. 2019;21(4):519–28.PubMedCrossRef Russo M, Guida F, Paparo L, et al. The novel butyrate derivative phenylalanine-butyramide protects from doxorubicin-induced cardiotoxicity. Eur J Heart Fail. 2019;21(4):519–28.PubMedCrossRef
19.
Zurück zum Zitat Kong CY, Guo Z, Song P, et al. Underlying the mechanisms of Doxorubicin-Induced Acute Cardiotoxicity: oxidative stress and cell death. Int J Biol Sci. 2022;18(2):760–70.PubMedPubMedCentralCrossRef Kong CY, Guo Z, Song P, et al. Underlying the mechanisms of Doxorubicin-Induced Acute Cardiotoxicity: oxidative stress and cell death. Int J Biol Sci. 2022;18(2):760–70.PubMedPubMedCentralCrossRef
20.
21.
Zurück zum Zitat Bartlett JJ, Trivedi PC, Pulinilkunnil T. Autophagic dysregulation in doxorubicin cardiomyopathy. J Mol Cell Cardiol. 2017;104:1–8.PubMedCrossRef Bartlett JJ, Trivedi PC, Pulinilkunnil T. Autophagic dysregulation in doxorubicin cardiomyopathy. J Mol Cell Cardiol. 2017;104:1–8.PubMedCrossRef
22.
Zurück zum Zitat Sala V, Della Sala A, Hirsch E, Ghigo A. Signaling pathways underlying Anthracycline Cardiotoxicity. Antioxid Redox Signal 2020 32(15):1098–114. Sala V, Della Sala A, Hirsch E, Ghigo A. Signaling pathways underlying Anthracycline Cardiotoxicity. Antioxid Redox Signal 2020 32(15):1098–114.
23.
Zurück zum Zitat Russo M, Della Sala A, Tocchetti CG, Porporato PE, Ghigo A. Metabolic aspects of Anthracycline Cardiotoxicity. Curr Treat Options Oncol. 2021;22(2):18.PubMedPubMedCentralCrossRef Russo M, Della Sala A, Tocchetti CG, Porporato PE, Ghigo A. Metabolic aspects of Anthracycline Cardiotoxicity. Curr Treat Options Oncol. 2021;22(2):18.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Riad A, Bien S, Gratz M, et al. Toll-like receptor-4 deficiency attenuates doxorubicin-induced cardiomyopathy in mice. Eur J Heart Fail. 2008;10(3):233–43.PubMedCrossRef Riad A, Bien S, Gratz M, et al. Toll-like receptor-4 deficiency attenuates doxorubicin-induced cardiomyopathy in mice. Eur J Heart Fail. 2008;10(3):233–43.PubMedCrossRef
26.
Zurück zum Zitat Yarmohammadi F, Karbasforooshan H, Hayes AW, Karimi G. Inflammation suppression in doxorubicin-induced cardiotoxicity: natural compounds as therapeutic options. Naunyn Schmiedebergs Arch Pharmacol. 2021;394(10):2003–11.PubMedCrossRef Yarmohammadi F, Karbasforooshan H, Hayes AW, Karimi G. Inflammation suppression in doxorubicin-induced cardiotoxicity: natural compounds as therapeutic options. Naunyn Schmiedebergs Arch Pharmacol. 2021;394(10):2003–11.PubMedCrossRef
27.
Zurück zum Zitat Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int J Mol Sci 2019 20(13). Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int J Mol Sci 2019 20(13).
28.
Zurück zum Zitat Zeng C, Duan F, Hu J, et al. NLRP3 inflammasome-mediated pyroptosis contributes to the pathogenesis of non-ischemic dilated cardiomyopathy. Redox Biol. 2020;34:101523.PubMedPubMedCentralCrossRef Zeng C, Duan F, Hu J, et al. NLRP3 inflammasome-mediated pyroptosis contributes to the pathogenesis of non-ischemic dilated cardiomyopathy. Redox Biol. 2020;34:101523.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Mishra SR, Mahapatra KK, Behera BP, et al. Mitochondrial dysfunction as a driver of NLRP3 inflammasome activation and its modulation through mitophagy for potential therapeutics. Int J Biochem Cell Biol. 2021;136:106013.PubMedCrossRef Mishra SR, Mahapatra KK, Behera BP, et al. Mitochondrial dysfunction as a driver of NLRP3 inflammasome activation and its modulation through mitophagy for potential therapeutics. Int J Biochem Cell Biol. 2021;136:106013.PubMedCrossRef
30.
Zurück zum Zitat Akolkar G, da Silva Dias D, Ayyappan P, et al. Vitamin C mitigates oxidative/nitrosative stress and inflammation in doxorubicin-induced cardiomyopathy. Am J Physiol Heart Circ Physiol. 2017;313(4):H795–809.PubMedCrossRef Akolkar G, da Silva Dias D, Ayyappan P, et al. Vitamin C mitigates oxidative/nitrosative stress and inflammation in doxorubicin-induced cardiomyopathy. Am J Physiol Heart Circ Physiol. 2017;313(4):H795–809.PubMedCrossRef
31.
Zurück zum Zitat Swirski FK, Nahrendorf M. Cardioimmunology: the immune system in cardiac homeostasis and disease. Nat Rev Immunol. 2018;18(12):733–44.PubMedCrossRef Swirski FK, Nahrendorf M. Cardioimmunology: the immune system in cardiac homeostasis and disease. Nat Rev Immunol. 2018;18(12):733–44.PubMedCrossRef
32.
Zurück zum Zitat Bhagat A, Shrestha P, Kleinerman ES. The Innate Immune System in Cardiovascular diseases and its role in Doxorubicin-Induced Cardiotoxicity. Int J Mol Sci 2022. 23(23). Bhagat A, Shrestha P, Kleinerman ES. The Innate Immune System in Cardiovascular diseases and its role in Doxorubicin-Induced Cardiotoxicity. Int J Mol Sci 2022. 23(23).
33.
Zurück zum Zitat Krysko DV, Kaczmarek A, Krysko O, et al. TLR-2 and TLR-9 are sensors of apoptosis in a mouse model of doxorubicin-induced acute inflammation. Cell Death Differ. 2011;18(8):1316–25.PubMedPubMedCentralCrossRef Krysko DV, Kaczmarek A, Krysko O, et al. TLR-2 and TLR-9 are sensors of apoptosis in a mouse model of doxorubicin-induced acute inflammation. Cell Death Differ. 2011;18(8):1316–25.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Kitakata H, Endo J, Ikura H et al. Therapeutic targets for DOX-Induced Cardiomyopathy: role of apoptosis vs. Ferroptosis. Int J Mol Sci 2022. 23(3). Kitakata H, Endo J, Ikura H et al. Therapeutic targets for DOX-Induced Cardiomyopathy: role of apoptosis vs. Ferroptosis. Int J Mol Sci 2022. 23(3).
35.
Zurück zum Zitat Sangweni NF, Gabuza K, Huisamen B, et al. Molecular insights into the pathophysiology of doxorubicin-induced cardiotoxicity: a graphical representation. Arch Toxicol. 2022;96(6):1541–50.PubMedPubMedCentralCrossRef Sangweni NF, Gabuza K, Huisamen B, et al. Molecular insights into the pathophysiology of doxorubicin-induced cardiotoxicity: a graphical representation. Arch Toxicol. 2022;96(6):1541–50.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Bartlett JJ, Trivedi PC, Yeung P, Kienesberger PC, Pulinilkunnil T. Doxorubicin impairs cardiomyocyte viability by suppressing transcription factor EB expression and disrupting autophagy. Biochem J. 2016;473(21):3769–89.PubMedCrossRef Bartlett JJ, Trivedi PC, Yeung P, Kienesberger PC, Pulinilkunnil T. Doxorubicin impairs cardiomyocyte viability by suppressing transcription factor EB expression and disrupting autophagy. Biochem J. 2016;473(21):3769–89.PubMedCrossRef
37.
Zurück zum Zitat Jiao Y, Li Y, Zhang J et al. RRM2 alleviates Doxorubicin-Induced Cardiotoxicity through the AKT/mTOR signaling pathway. Biomolecules 2022. 12(2). Jiao Y, Li Y, Zhang J et al. RRM2 alleviates Doxorubicin-Induced Cardiotoxicity through the AKT/mTOR signaling pathway. Biomolecules 2022. 12(2).
38.
Zurück zum Zitat Gu J, Hu W, Song ZP, et al. Resveratrol-induced autophagy promotes survival and attenuates doxorubicin-induced cardiotoxicity. Int Immunopharmacol. 2016;32:1–7.PubMedCrossRef Gu J, Hu W, Song ZP, et al. Resveratrol-induced autophagy promotes survival and attenuates doxorubicin-induced cardiotoxicity. Int Immunopharmacol. 2016;32:1–7.PubMedCrossRef
39.
Zurück zum Zitat Carvalho FS, Burgeiro A, Garcia R, et al. Doxorubicin-induced cardiotoxicity: from bioenergetic failure and cell death to cardiomyopathy. Med Res Rev. 2014;34(1):106–35.PubMedCrossRef Carvalho FS, Burgeiro A, Garcia R, et al. Doxorubicin-induced cardiotoxicity: from bioenergetic failure and cell death to cardiomyopathy. Med Res Rev. 2014;34(1):106–35.PubMedCrossRef
40.
Zurück zum Zitat Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L. Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev. 2004;56(2):185–229.PubMedCrossRef Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L. Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev. 2004;56(2):185–229.PubMedCrossRef
41.
Zurück zum Zitat Ozcan M, Guo Z, Valenzuela Ripoll C, et al. Sustained alternate-day fasting potentiates doxorubicin cardiotoxicity. Cell Metab. 2023;35(6):928–42. e924.PubMedCrossRef Ozcan M, Guo Z, Valenzuela Ripoll C, et al. Sustained alternate-day fasting potentiates doxorubicin cardiotoxicity. Cell Metab. 2023;35(6):928–42. e924.PubMedCrossRef
42.
Zurück zum Zitat Wu L, Wang L, Du Y, Zhang Y, Ren J. Mitochondrial quality control mechanisms as therapeutic targets in doxorubicin-induced cardiotoxicity. Trends Pharmacol Sci. 2023;44(1):34–49.PubMedCrossRef Wu L, Wang L, Du Y, Zhang Y, Ren J. Mitochondrial quality control mechanisms as therapeutic targets in doxorubicin-induced cardiotoxicity. Trends Pharmacol Sci. 2023;44(1):34–49.PubMedCrossRef
43.
Zurück zum Zitat Bäckhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Sci 2005 307(5717):1915–20. Bäckhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Sci 2005 307(5717):1915–20.
44.
Zurück zum Zitat Aggarwal N, Kitano S, Puah GRY, et al. Microbiome and Human Health: current understanding, Engineering, and Enabling technologies. Chem Rev. 2023;123(1):31–72.PubMedCrossRef Aggarwal N, Kitano S, Puah GRY, et al. Microbiome and Human Health: current understanding, Engineering, and Enabling technologies. Chem Rev. 2023;123(1):31–72.PubMedCrossRef
45.
Zurück zum Zitat Donaldson GP, Lee SM, Mazmanian SK. Gut biogeography of the bacterial microbiota. Nat Rev Microbiol. 2016;14(1):20–32.PubMedCrossRef Donaldson GP, Lee SM, Mazmanian SK. Gut biogeography of the bacterial microbiota. Nat Rev Microbiol. 2016;14(1):20–32.PubMedCrossRef
47.
Zurück zum Zitat Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021;19(1):55–71.PubMedCrossRef Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021;19(1):55–71.PubMedCrossRef
48.
Zurück zum Zitat Bertero E, Maack C. Metabolic remodelling in heart failure. Nat Rev Cardiol. 2018;15(8):457–70.PubMedCrossRef Bertero E, Maack C. Metabolic remodelling in heart failure. Nat Rev Cardiol. 2018;15(8):457–70.PubMedCrossRef
49.
Zurück zum Zitat Gabriel CL, Ferguson JF. Gut microbiota and Microbial Metabolism in Early Risk of Cardiometabolic Disease. Circ Res. 2023;132(12):1674–91.PubMedCrossRef Gabriel CL, Ferguson JF. Gut microbiota and Microbial Metabolism in Early Risk of Cardiometabolic Disease. Circ Res. 2023;132(12):1674–91.PubMedCrossRef
50.
Zurück zum Zitat Kim M, Huda MN, Bennett BJ. Sequence meets function-microbiota and cardiovascular disease. Cardiovasc Res. 2022;118(2):399–412.PubMedCrossRef Kim M, Huda MN, Bennett BJ. Sequence meets function-microbiota and cardiovascular disease. Cardiovasc Res. 2022;118(2):399–412.PubMedCrossRef
51.
Zurück zum Zitat Giongo A, Gano KA, Crabb DB, et al. Toward defining the autoimmune microbiome for type 1 diabetes. Isme j. 2011;5(1):82–91.PubMedCrossRef Giongo A, Gano KA, Crabb DB, et al. Toward defining the autoimmune microbiome for type 1 diabetes. Isme j. 2011;5(1):82–91.PubMedCrossRef
52.
Zurück zum Zitat Wu H, Tremaroli V, Schmidt C, et al. The gut microbiota in Prediabetes and Diabetes: a Population-based cross-sectional study. Cell Metab. 2020;32(3):379–90. e373.PubMedCrossRef Wu H, Tremaroli V, Schmidt C, et al. The gut microbiota in Prediabetes and Diabetes: a Population-based cross-sectional study. Cell Metab. 2020;32(3):379–90. e373.PubMedCrossRef
53.
Zurück zum Zitat Khalesi S, Sun J, Buys N, Jayasinghe R. Effect of probiotics on blood pressure: a systematic review and meta-analysis of randomized, controlled trials. Hypertension. 2014;64(4):897–903.PubMedCrossRef Khalesi S, Sun J, Buys N, Jayasinghe R. Effect of probiotics on blood pressure: a systematic review and meta-analysis of randomized, controlled trials. Hypertension. 2014;64(4):897–903.PubMedCrossRef
54.
Zurück zum Zitat O’Donnell JA, Zheng T, Meric G, Marques FZ. The gut microbiome and hypertension. Nat Rev Nephrol. 2023;19(3):153–67.PubMedCrossRef O’Donnell JA, Zheng T, Meric G, Marques FZ. The gut microbiome and hypertension. Nat Rev Nephrol. 2023;19(3):153–67.PubMedCrossRef
55.
Zurück zum Zitat de Assis Gadelha DD, de Brito Alves JL, da Costa PCT et al. Lactobacillus group and arterial hypertension: a broad review on effects and proposed mechanisms. Crit Rev Food Sci Nutr. 2022:1–22. de Assis Gadelha DD, de Brito Alves JL, da Costa PCT et al. Lactobacillus group and arterial hypertension: a broad review on effects and proposed mechanisms. Crit Rev Food Sci Nutr. 2022:1–22.
56.
Zurück zum Zitat O’Toole PW, Jeffery IB. Gut microbiota and aging. Sci 2015 350(6265):1214–5. O’Toole PW, Jeffery IB. Gut microbiota and aging. Sci 2015 350(6265):1214–5.
57.
Zurück zum Zitat Santoro A, Ostan R, Candela M, et al. Gut microbiota changes in the extreme decades of human life: a focus on centenarians. Cell Mol Life Sci. 2018;75(1):129–48.PubMedCrossRef Santoro A, Ostan R, Candela M, et al. Gut microbiota changes in the extreme decades of human life: a focus on centenarians. Cell Mol Life Sci. 2018;75(1):129–48.PubMedCrossRef
58.
Zurück zum Zitat Conway J, N AD. Ageing of the gut microbiome: potential influences on immune senescence and inflammageing. Ageing Res Rev. 2021;68:101323.PubMedCrossRef Conway J, N AD. Ageing of the gut microbiome: potential influences on immune senescence and inflammageing. Ageing Res Rev. 2021;68:101323.PubMedCrossRef
59.
Zurück zum Zitat Fernandes MR, Aggarwal P, Costa RGF, Cole AM, Trinchieri G. Targeting the gut microbiota for cancer therapy. Nat Rev Cancer 2022 22(12):703–22. Fernandes MR, Aggarwal P, Costa RGF, Cole AM, Trinchieri G. Targeting the gut microbiota for cancer therapy. Nat Rev Cancer 2022 22(12):703–22.
60.
Zurück zum Zitat Sawicki KT, Sala V, Prever L, et al. Preventing and treating Anthracycline Cardiotoxicity: New insights. Annu Rev Pharmacol Toxicol. 2021;61:309–32.PubMedCrossRef Sawicki KT, Sala V, Prever L, et al. Preventing and treating Anthracycline Cardiotoxicity: New insights. Annu Rev Pharmacol Toxicol. 2021;61:309–32.PubMedCrossRef
61.
Zurück zum Zitat Swain SM, Whaley FS, Ewer MS. Congestive heart failure in patients treated with doxorubicin: a retrospective analysis of three trials. Cancer. 2003;97(11):2869–79.PubMedCrossRef Swain SM, Whaley FS, Ewer MS. Congestive heart failure in patients treated with doxorubicin: a retrospective analysis of three trials. Cancer. 2003;97(11):2869–79.PubMedCrossRef
62.
Zurück zum Zitat Troseid M, Andersen GO, Broch K, Hov JR. The gut microbiome in coronary artery disease and heart failure: current knowledge and future directions. EBioMedicine. 2020;52:102649.PubMedPubMedCentralCrossRef Troseid M, Andersen GO, Broch K, Hov JR. The gut microbiome in coronary artery disease and heart failure: current knowledge and future directions. EBioMedicine. 2020;52:102649.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Gomes AC, Hoffmann C, Mota JF. The human gut microbiota: metabolism and perspective in obesity. Gut Microbes 2018 9(4):308–25. Gomes AC, Hoffmann C, Mota JF. The human gut microbiota: metabolism and perspective in obesity. Gut Microbes 2018 9(4):308–25.
65.
Zurück zum Zitat Jin L, Shi X, Yang J, et al. Gut microbes in cardiovascular diseases and their potential therapeutic applications. Protein Cell. 2021;12(5):346–59.PubMedCrossRef Jin L, Shi X, Yang J, et al. Gut microbes in cardiovascular diseases and their potential therapeutic applications. Protein Cell. 2021;12(5):346–59.PubMedCrossRef
66.
Zurück zum Zitat Sandek A, Bauditz J, Swidsinski A, et al. Altered intestinal function in patients with chronic heart failure. J Am Coll Cardiol. 2007;50(16):1561–9.PubMedCrossRef Sandek A, Bauditz J, Swidsinski A, et al. Altered intestinal function in patients with chronic heart failure. J Am Coll Cardiol. 2007;50(16):1561–9.PubMedCrossRef
67.
Zurück zum Zitat Luedde M, Winkler T, Heinsen FA, et al. Heart failure is associated with depletion of core intestinal microbiota. ESC Heart Fail. 2017;4(3):282–90.PubMedPubMedCentralCrossRef Luedde M, Winkler T, Heinsen FA, et al. Heart failure is associated with depletion of core intestinal microbiota. ESC Heart Fail. 2017;4(3):282–90.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Morais LH, Schreiber HLt, Mazmanian SK. The gut microbiota-brain axis in behaviour and brain disorders. Nat Rev Microbiol. 2021;19(4):241–55.PubMedCrossRef Morais LH, Schreiber HLt, Mazmanian SK. The gut microbiota-brain axis in behaviour and brain disorders. Nat Rev Microbiol. 2021;19(4):241–55.PubMedCrossRef
69.
Zurück zum Zitat Li N, Wang L, Li L, et al. The correlation between gut microbiome and atrial fibrillation: pathophysiology and therapeutic perspectives. Mil Med Res. 2023;10(1):51.PubMedPubMedCentral Li N, Wang L, Li L, et al. The correlation between gut microbiome and atrial fibrillation: pathophysiology and therapeutic perspectives. Mil Med Res. 2023;10(1):51.PubMedPubMedCentral
70.
Zurück zum Zitat Zhang Q, Zhang L, Chen C, Li P, Lu B. The gut microbiota-artery axis: a bridge between dietary lipids and atherosclerosis? Prog Lipid Res. 2023;89:101209.PubMedCrossRef Zhang Q, Zhang L, Chen C, Li P, Lu B. The gut microbiota-artery axis: a bridge between dietary lipids and atherosclerosis? Prog Lipid Res. 2023;89:101209.PubMedCrossRef
71.
Zurück zum Zitat Jonsson AL, Bäckhed F. Role of gut microbiota in atherosclerosis. Nat Rev Cardiol. 2017;14(2):79–87.PubMedCrossRef Jonsson AL, Bäckhed F. Role of gut microbiota in atherosclerosis. Nat Rev Cardiol. 2017;14(2):79–87.PubMedCrossRef
72.
Zurück zum Zitat Kazemian N, Mahmoudi M, Halperin F, Wu JC, Pakpour S. Gut microbiota and cardiovascular disease: opportunities and challenges. Microbiome. 2020;8(1):36.PubMedPubMedCentralCrossRef Kazemian N, Mahmoudi M, Halperin F, Wu JC, Pakpour S. Gut microbiota and cardiovascular disease: opportunities and challenges. Microbiome. 2020;8(1):36.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Li XS, Obeid S, Klingenberg R, et al. Gut microbiota-dependent trimethylamine N-oxide in acute coronary syndromes: a prognostic marker for incident cardiovascular events beyond traditional risk factors. Eur Heart J. 2017;38(11):814–24.PubMedPubMedCentral Li XS, Obeid S, Klingenberg R, et al. Gut microbiota-dependent trimethylamine N-oxide in acute coronary syndromes: a prognostic marker for incident cardiovascular events beyond traditional risk factors. Eur Heart J. 2017;38(11):814–24.PubMedPubMedCentral
74.
Zurück zum Zitat van der Hee B, Wells JM. Microbial regulation of host physiology by short-chain fatty acids. Trends Microbiol. 2021;29(8):700–12.PubMedCrossRef van der Hee B, Wells JM. Microbial regulation of host physiology by short-chain fatty acids. Trends Microbiol. 2021;29(8):700–12.PubMedCrossRef
75.
Zurück zum Zitat Miller TLW. Pathways of acetate, Propionate, and butyrate formation by the human fecal Microbial Flora. Appl Environ Microbiol. 1996;62:1589–92.PubMedPubMedCentralCrossRef Miller TLW. Pathways of acetate, Propionate, and butyrate formation by the human fecal Microbial Flora. Appl Environ Microbiol. 1996;62:1589–92.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Amabebe E, Robert FO, Agbalalah T, Orubu ESF. Microbial dysbiosis-induced obesity: role of gut microbiota in homoeostasis of energy metabolism. Br J Nutr. 2020;123(10):1127–37.PubMedCrossRef Amabebe E, Robert FO, Agbalalah T, Orubu ESF. Microbial dysbiosis-induced obesity: role of gut microbiota in homoeostasis of energy metabolism. Br J Nutr. 2020;123(10):1127–37.PubMedCrossRef
77.
Zurück zum Zitat Tian Z, Zhang Y, Zheng Z, et al. Gut microbiome dysbiosis contributes to abdominal aortic aneurysm by promoting neutrophil extracellular trap formation. Cell Host Microbe. 2022;30(10):1450–e14631458.PubMedCrossRef Tian Z, Zhang Y, Zheng Z, et al. Gut microbiome dysbiosis contributes to abdominal aortic aneurysm by promoting neutrophil extracellular trap formation. Cell Host Microbe. 2022;30(10):1450–e14631458.PubMedCrossRef
78.
Zurück zum Zitat Yan J, Pan Y, Shao W, et al. Beneficial effect of the short-chain fatty acid propionate on vascular calcification through intestinal microbiota remodelling. Microbiome. 2022;10(1):195.PubMedPubMedCentralCrossRef Yan J, Pan Y, Shao W, et al. Beneficial effect of the short-chain fatty acid propionate on vascular calcification through intestinal microbiota remodelling. Microbiome. 2022;10(1):195.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Makrecka-Kuka M, Volska K, Antone U, et al. Trimethylamine N-oxide impairs pyruvate and fatty acid oxidation in cardiac mitochondria. Toxicol Lett. 2017;267:32–8.PubMedCrossRef Makrecka-Kuka M, Volska K, Antone U, et al. Trimethylamine N-oxide impairs pyruvate and fatty acid oxidation in cardiac mitochondria. Toxicol Lett. 2017;267:32–8.PubMedCrossRef
80.
Zurück zum Zitat Savi M, Bocchi L, Bresciani L et al. Trimethylamine-N-Oxide (TMAO)-Induced impairment of cardiomyocyte function and the protective role of Urolithin B-Glucuronide. Molecules. 2018. 23(3). Savi M, Bocchi L, Bresciani L et al. Trimethylamine-N-Oxide (TMAO)-Induced impairment of cardiomyocyte function and the protective role of Urolithin B-Glucuronide. Molecules. 2018. 23(3).
81.
Zurück zum Zitat Zhu W, Gregory JC, Org E, et al. Gut Microbial Metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell. 2016;165(1):111–24.PubMedPubMedCentralCrossRef Zhu W, Gregory JC, Org E, et al. Gut Microbial Metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell. 2016;165(1):111–24.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Zhao M, Wei H, Li C, et al. Gut microbiota production of trimethyl-5-aminovaleric acid reduces fatty acid oxidation and accelerates cardiac hypertrophy. Nat Commun. 2022;13(1):1757.PubMedPubMedCentralCrossRef Zhao M, Wei H, Li C, et al. Gut microbiota production of trimethyl-5-aminovaleric acid reduces fatty acid oxidation and accelerates cardiac hypertrophy. Nat Commun. 2022;13(1):1757.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Weersma RK, Zhernakova A, Fu J. Interaction between drugs and the gut microbiome. Gut. 2020;69(8):1510–9.PubMedCrossRef Weersma RK, Zhernakova A, Fu J. Interaction between drugs and the gut microbiome. Gut. 2020;69(8):1510–9.PubMedCrossRef
84.
Zurück zum Zitat Lindell AE, Zimmermann-Kogadeeva M, Patil KR. Multimodal interactions of drugs, natural compounds and pollutants with the gut microbiota. Nat Rev Microbiol. 2022;20(7):431–43.PubMedPubMedCentralCrossRef Lindell AE, Zimmermann-Kogadeeva M, Patil KR. Multimodal interactions of drugs, natural compounds and pollutants with the gut microbiota. Nat Rev Microbiol. 2022;20(7):431–43.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Flórez AB, Sierra M, Ruas-Madiedo P, Mayo B. Susceptibility of lactic acid bacteria, bifidobacteria and other bacteria of intestinal origin to chemotherapeutic agents. Int J Antimicrob Agents. 2016;48(5):547–50.PubMedCrossRef Flórez AB, Sierra M, Ruas-Madiedo P, Mayo B. Susceptibility of lactic acid bacteria, bifidobacteria and other bacteria of intestinal origin to chemotherapeutic agents. Int J Antimicrob Agents. 2016;48(5):547–50.PubMedCrossRef
86.
Zurück zum Zitat Rawls JF, Mahowald MA, Ley RE, Gordon JI. Reciprocal gut microbiota transplants from zebrafish and mice to germ-free recipients reveal host habitat selection. Cell. 2006;127(2):423–33. Rawls JF, Mahowald MA, Ley RE, Gordon JI. Reciprocal gut microbiota transplants from zebrafish and mice to germ-free recipients reveal host habitat selection. Cell. 2006;127(2):423–33.
87.
Zurück zum Zitat An L, Wuri J, Zheng Z, Li W, Yan T. Microbiota modulate doxorubicin induced cardiotoxicity. Eur J Pharm Sci. 2021;166:105977.PubMedCrossRef An L, Wuri J, Zheng Z, Li W, Yan T. Microbiota modulate doxorubicin induced cardiotoxicity. Eur J Pharm Sci. 2021;166:105977.PubMedCrossRef
88.
Zurück zum Zitat Huang J, Wei S, Jiang C, et al. Involvement of abnormal gut microbiota composition and function in Doxorubicin-Induced Cardiotoxicity. Front Cell Infect Microbiol. 2022;12:808837.PubMedPubMedCentralCrossRef Huang J, Wei S, Jiang C, et al. Involvement of abnormal gut microbiota composition and function in Doxorubicin-Induced Cardiotoxicity. Front Cell Infect Microbiol. 2022;12:808837.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Huang K, Liu Y, Tang H, et al. Glabridin prevents Doxorubicin-Induced Cardiotoxicity through Gut Microbiota Modulation and Colonic Macrophage polarization in mice. Front Pharmacol. 2019;10:107.PubMedPubMedCentralCrossRef Huang K, Liu Y, Tang H, et al. Glabridin prevents Doxorubicin-Induced Cardiotoxicity through Gut Microbiota Modulation and Colonic Macrophage polarization in mice. Front Pharmacol. 2019;10:107.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Yan A, Culp E, Perry J, et al. Transformation of the Anticancer Drug Doxorubicin in the human gut Microbiome. ACS Infect Dis. 2018;4(1):68–76.PubMedCrossRef Yan A, Culp E, Perry J, et al. Transformation of the Anticancer Drug Doxorubicin in the human gut Microbiome. ACS Infect Dis. 2018;4(1):68–76.PubMedCrossRef
91.
Zurück zum Zitat Lin H, Meng L, Sun Z, et al. Yellow wine polyphenolic compound protects against Doxorubicin-Induced cardiotoxicity by modulating the composition and metabolic function of the gut microbiota. Circ Heart Fail. 2021;14(10):e008220.PubMedCrossRef Lin H, Meng L, Sun Z, et al. Yellow wine polyphenolic compound protects against Doxorubicin-Induced cardiotoxicity by modulating the composition and metabolic function of the gut microbiota. Circ Heart Fail. 2021;14(10):e008220.PubMedCrossRef
92.
Zurück zum Zitat Shin NR, Whon TW, Bae JW. Proteobacteria: microbial signature of dysbiosis in gut microbiota. Trends Biotechnol. 2015;33(9):496–503.PubMedCrossRef Shin NR, Whon TW, Bae JW. Proteobacteria: microbial signature of dysbiosis in gut microbiota. Trends Biotechnol. 2015;33(9):496–503.PubMedCrossRef
93.
Zurück zum Zitat Agudelo-Ochoa GM, Valdés-Duque BE, Giraldo-Giraldo NA, et al. Gut microbiota profiles in critically ill patients, potential biomarkers and risk variables for sepsis. Gut Microbes. 2020;12(1):1707610.PubMedPubMedCentralCrossRef Agudelo-Ochoa GM, Valdés-Duque BE, Giraldo-Giraldo NA, et al. Gut microbiota profiles in critically ill patients, potential biomarkers and risk variables for sepsis. Gut Microbes. 2020;12(1):1707610.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Schiattarella GG, Sannino A, Toscano E, et al. Gut microbe-generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: a systematic review and dose-response meta-analysis. Eur Heart J. 2017;38(39):2948–56.PubMedCrossRef Schiattarella GG, Sannino A, Toscano E, et al. Gut microbe-generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: a systematic review and dose-response meta-analysis. Eur Heart J. 2017;38(39):2948–56.PubMedCrossRef
95.
Zurück zum Zitat Turner JR. Intestinal mucosal barrier function in health and disease. Nat Rev Immunol. 2009;9(11):799–809.PubMedCrossRef Turner JR. Intestinal mucosal barrier function in health and disease. Nat Rev Immunol. 2009;9(11):799–809.PubMedCrossRef
96.
Zurück zum Zitat Lewis CV, Taylor WR. Intestinal barrier dysfunction as a therapeutic target for cardiovascular disease. Am J Physiol Heart Circ Physiol. 2020;319(6):H1227–33.PubMedPubMedCentralCrossRef Lewis CV, Taylor WR. Intestinal barrier dysfunction as a therapeutic target for cardiovascular disease. Am J Physiol Heart Circ Physiol. 2020;319(6):H1227–33.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Rigby RJ, Carr J, Orgel K, et al. Intestinal bacteria are necessary for doxorubicin-induced intestinal damage but not for doxorubicin-induced apoptosis. Gut Microbes. 2016;7(5):414–23.PubMedPubMedCentralCrossRef Rigby RJ, Carr J, Orgel K, et al. Intestinal bacteria are necessary for doxorubicin-induced intestinal damage but not for doxorubicin-induced apoptosis. Gut Microbes. 2016;7(5):414–23.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Horowitz A, Chanez-Paredes SD, Haest X, Turner JR. Paracellular permeability and tight junction regulation in gut health and disease. Nat Rev Gastroenterol Hepatol. 2023;20(7):417–32.PubMedCrossRef Horowitz A, Chanez-Paredes SD, Haest X, Turner JR. Paracellular permeability and tight junction regulation in gut health and disease. Nat Rev Gastroenterol Hepatol. 2023;20(7):417–32.PubMedCrossRef
99.
Zurück zum Zitat Beutel O, Maraspini R, Pombo-García K, Martin-Lemaitre C, Honigmann A. Phase separation of Zonula Occludens Proteins drives formation of tight junctions. Cell 2019 179(4):923–e936911. Beutel O, Maraspini R, Pombo-García K, Martin-Lemaitre C, Honigmann A. Phase separation of Zonula Occludens Proteins drives formation of tight junctions. Cell 2019 179(4):923–e936911.
100.
Zurück zum Zitat Cray P, Sheahan BJ, Cortes JE, Dekaney CM. Doxorubicin increases permeability of murine small intestinal epithelium and cultured T84 monolayers. Sci Rep. 2020;10(1):21486.PubMedPubMedCentralCrossRef Cray P, Sheahan BJ, Cortes JE, Dekaney CM. Doxorubicin increases permeability of murine small intestinal epithelium and cultured T84 monolayers. Sci Rep. 2020;10(1):21486.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Pasini E, Aquilani R, Testa C, et al. Pathogenic gut Flora in patients with Chronic Heart failure. JACC Heart Fail. 2016;4(3):220–7.PubMedCrossRef Pasini E, Aquilani R, Testa C, et al. Pathogenic gut Flora in patients with Chronic Heart failure. JACC Heart Fail. 2016;4(3):220–7.PubMedCrossRef
102.
Zurück zum Zitat Pellegrini C, Fornai M, D’Antongiovanni V, et al. The intestinal barrier in disorders of the central nervous system. Lancet Gastroenterol Hepatol. 2023;8(1):66–80.PubMedCrossRef Pellegrini C, Fornai M, D’Antongiovanni V, et al. The intestinal barrier in disorders of the central nervous system. Lancet Gastroenterol Hepatol. 2023;8(1):66–80.PubMedCrossRef
103.
Zurück zum Zitat Odenwald MA, Turner JR. The intestinal epithelial barrier: a therapeutic target? Nat Rev Gastroenterol Hepatol. 2017;14(1):9–21.PubMedCrossRef Odenwald MA, Turner JR. The intestinal epithelial barrier: a therapeutic target? Nat Rev Gastroenterol Hepatol. 2017;14(1):9–21.PubMedCrossRef
104.
Zurück zum Zitat Camara-Lemarroy CR, Metz L, Meddings JB, Sharkey KA, Wee Yong V. The intestinal barrier in multiple sclerosis: implications for pathophysiology and therapeutics. Brain 2018 141(7):1900–16. Camara-Lemarroy CR, Metz L, Meddings JB, Sharkey KA, Wee Yong V. The intestinal barrier in multiple sclerosis: implications for pathophysiology and therapeutics. Brain 2018 141(7):1900–16.
105.
Zurück zum Zitat Matei DE, Menon M, Alber DG, et al. Intestinal barrier dysfunction plays an integral role in arthritis pathology and can be targeted to ameliorate disease. Med. 2021;2(7):864–e883869.PubMedCrossRef Matei DE, Menon M, Alber DG, et al. Intestinal barrier dysfunction plays an integral role in arthritis pathology and can be targeted to ameliorate disease. Med. 2021;2(7):864–e883869.PubMedCrossRef
106.
Zurück zum Zitat Zhao J, Zhang Q, Cheng W et al. Heart-gut-microbiota communication determines the severity of cardiac injury after myocardial ischemia/reperfusion. Cardiovasc Res. 2023. Zhao J, Zhang Q, Cheng W et al. Heart-gut-microbiota communication determines the severity of cardiac injury after myocardial ischemia/reperfusion. Cardiovasc Res. 2023.
107.
Zurück zum Zitat Kaczmarek A, Brinkman BM, Heyndrickx L, Vandenabeele P, Krysko DV. Severity of doxorubicin-induced small intestinal mucositis is regulated by the TLR-2 and TLR-9 pathways. J Pathol. 2012;226(4):598–608.PubMedCrossRef Kaczmarek A, Brinkman BM, Heyndrickx L, Vandenabeele P, Krysko DV. Severity of doxorubicin-induced small intestinal mucositis is regulated by the TLR-2 and TLR-9 pathways. J Pathol. 2012;226(4):598–608.PubMedCrossRef
108.
Zurück zum Zitat O’Donnell JA, Zheng T, Meric G, Marques FZ. The gut microbiome and hypertension. Nat Rev Nephrol. 2023. O’Donnell JA, Zheng T, Meric G, Marques FZ. The gut microbiome and hypertension. Nat Rev Nephrol. 2023.
109.
Zurück zum Zitat Bjorkegren JLM, Lusis AJ, Atherosclerosis. Recent developments. Cell 2022 185(10):1630–45. Bjorkegren JLM, Lusis AJ, Atherosclerosis. Recent developments. Cell 2022 185(10):1630–45.
110.
Zurück zum Zitat Zhang Y, Wang Y, Ke B, Du J. TMAO: how gut microbiota contributes to heart failure. Transl Res. 2021;228:109–25.PubMedCrossRef Zhang Y, Wang Y, Ke B, Du J. TMAO: how gut microbiota contributes to heart failure. Transl Res. 2021;228:109–25.PubMedCrossRef
111.
Zurück zum Zitat Zhang X, Li Y, Yang P, et al. Trimethylamine-N-Oxide promotes vascular calcification through activation of NLRP3 (nucleotide-Binding domain, leucine-rich-containing family, pyrin domain-Containing-3) inflammasome and NF-kappaB (nuclear factor kappaB) signals. Arterioscler Thromb Vasc Biol. 2020;40(3):751–65.PubMedCrossRef Zhang X, Li Y, Yang P, et al. Trimethylamine-N-Oxide promotes vascular calcification through activation of NLRP3 (nucleotide-Binding domain, leucine-rich-containing family, pyrin domain-Containing-3) inflammasome and NF-kappaB (nuclear factor kappaB) signals. Arterioscler Thromb Vasc Biol. 2020;40(3):751–65.PubMedCrossRef
112.
Zurück zum Zitat Li X, Geng J, Zhao J, et al. Trimethylamine N-Oxide exacerbates Cardiac Fibrosis via activating the NLRP3 inflammasome. Front Physiol. 2019;10:866.PubMedPubMedCentralCrossRef Li X, Geng J, Zhao J, et al. Trimethylamine N-Oxide exacerbates Cardiac Fibrosis via activating the NLRP3 inflammasome. Front Physiol. 2019;10:866.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Gonzalez-Bosch C, Boorman E, Zunszain PA, Mann GE. Short-chain fatty acids as modulators of redox signaling in health and disease. Redox Biol. 2021;47:102165.PubMedPubMedCentralCrossRef Gonzalez-Bosch C, Boorman E, Zunszain PA, Mann GE. Short-chain fatty acids as modulators of redox signaling in health and disease. Redox Biol. 2021;47:102165.PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Al-Qadami GH, Secombe KR, Subramaniam CB, Wardill HR, Bowen JM. Gut microbiota-derived short-chain fatty acids: impact on Cancer Treatment Response and Toxicities. Microorganisms 2022. 10(10). Al-Qadami GH, Secombe KR, Subramaniam CB, Wardill HR, Bowen JM. Gut microbiota-derived short-chain fatty acids: impact on Cancer Treatment Response and Toxicities. Microorganisms 2022. 10(10).
115.
Zurück zum Zitat Chen Y, Liu Y, Wang Y, et al. Prevotellaceae produces butyrate to alleviate PD-1/PD-L1 inhibitor-related cardiotoxicity via PPARalpha-CYP4X1 axis in colonic macrophages. J Exp Clin Cancer Res. 2022;41(1):1.PubMedPubMedCentralCrossRef Chen Y, Liu Y, Wang Y, et al. Prevotellaceae produces butyrate to alleviate PD-1/PD-L1 inhibitor-related cardiotoxicity via PPARalpha-CYP4X1 axis in colonic macrophages. J Exp Clin Cancer Res. 2022;41(1):1.PubMedPubMedCentralCrossRef
116.
Zurück zum Zitat Kolluru GK, Shackelford RE, Shen X, Dominic P, Kevil CG. Sulfide regulation of cardiovascular function in health and disease. Nat Rev Cardiol. 2023;20(2):109–25.PubMedCrossRef Kolluru GK, Shackelford RE, Shen X, Dominic P, Kevil CG. Sulfide regulation of cardiovascular function in health and disease. Nat Rev Cardiol. 2023;20(2):109–25.PubMedCrossRef
117.
118.
Zurück zum Zitat Hu Q, Yammani RD, Brown-Harding H, et al. Mitigation of doxorubicin-induced cardiotoxicity with an H(2)O(2)-Activated, H(2)S-Donating hybrid prodrug. Redox Biol. 2022;53:102338.PubMedPubMedCentralCrossRef Hu Q, Yammani RD, Brown-Harding H, et al. Mitigation of doxorubicin-induced cardiotoxicity with an H(2)O(2)-Activated, H(2)S-Donating hybrid prodrug. Redox Biol. 2022;53:102338.PubMedPubMedCentralCrossRef
119.
Zurück zum Zitat Chegaev K, Rolando B, Cortese D, et al. H2S-Donating doxorubicins may overcome cardiotoxicity and Multidrug Resistance. J Med Chem. 2016;59(10):4881–9.PubMedCrossRef Chegaev K, Rolando B, Cortese D, et al. H2S-Donating doxorubicins may overcome cardiotoxicity and Multidrug Resistance. J Med Chem. 2016;59(10):4881–9.PubMedCrossRef
120.
Zurück zum Zitat Tesei A, Brigliadori G, Carloni S, et al. Organosulfur derivatives of the HDAC inhibitor valproic acid sensitize human lung cancer cell lines to apoptosis and to cisplatin cytotoxicity. J Cell Physiol. 2012;227(10):3389–96.PubMedCrossRef Tesei A, Brigliadori G, Carloni S, et al. Organosulfur derivatives of the HDAC inhibitor valproic acid sensitize human lung cancer cell lines to apoptosis and to cisplatin cytotoxicity. J Cell Physiol. 2012;227(10):3389–96.PubMedCrossRef
121.
Zurück zum Zitat Donertas Ayaz B, Zubcevic J. Gut microbiota and neuroinflammation in pathogenesis of hypertension: a potential role for hydrogen sulfide. Pharmacol Res. 2020;153:104677.PubMedPubMedCentralCrossRef Donertas Ayaz B, Zubcevic J. Gut microbiota and neuroinflammation in pathogenesis of hypertension: a potential role for hydrogen sulfide. Pharmacol Res. 2020;153:104677.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Xiong Q, Wang Z, Yu Y, et al. Hydrogen sulfide stabilizes atherosclerotic plaques in apolipoprotein E knockout mice. Pharmacol Res. 2019;144:90–8.PubMedCrossRef Xiong Q, Wang Z, Yu Y, et al. Hydrogen sulfide stabilizes atherosclerotic plaques in apolipoprotein E knockout mice. Pharmacol Res. 2019;144:90–8.PubMedCrossRef
123.
Zurück zum Zitat Zhang H, Guo C, Wu D, et al. Hydrogen sulfide inhibits the development of atherosclerosis with suppressing CX3CR1 and CX3CL1 expression. PLoS ONE. 2012;7(7):e41147.PubMedPubMedCentralCrossRef Zhang H, Guo C, Wu D, et al. Hydrogen sulfide inhibits the development of atherosclerosis with suppressing CX3CR1 and CX3CL1 expression. PLoS ONE. 2012;7(7):e41147.PubMedPubMedCentralCrossRef
124.
Zurück zum Zitat Shen X, Carlström M, Borniquel S, et al. Microbial regulation of host hydrogen sulfide bioavailability and metabolism. Free Radic Biol Med. 2013;60:195–200.PubMedPubMedCentralCrossRef Shen X, Carlström M, Borniquel S, et al. Microbial regulation of host hydrogen sulfide bioavailability and metabolism. Free Radic Biol Med. 2013;60:195–200.PubMedPubMedCentralCrossRef
125.
Zurück zum Zitat Jia B, Zou Y, Han X, Bae JW, Jeon CO. Gut microbiome-mediated mechanisms for reducing cholesterol levels: implications for ameliorating cardiovascular disease. Trends Microbiol. 2023;31(1):76–91.PubMedCrossRef Jia B, Zou Y, Han X, Bae JW, Jeon CO. Gut microbiome-mediated mechanisms for reducing cholesterol levels: implications for ameliorating cardiovascular disease. Trends Microbiol. 2023;31(1):76–91.PubMedCrossRef
126.
Zurück zum Zitat Wang Y, Ai Z, Xing X et al. The ameliorative effect of probiotics on diet-induced lipid metabolism disorders: a review. Crit Rev Food Sci Nutr. 2022:1–17. Wang Y, Ai Z, Xing X et al. The ameliorative effect of probiotics on diet-induced lipid metabolism disorders: a review. Crit Rev Food Sci Nutr. 2022:1–17.
127.
Zurück zum Zitat Ramirez-Macias I, Orenes-Pinero E, Camelo-Castillo A, et al. Novel insights in the relationship of gut microbiota and coronary artery diseases. Crit Rev Food Sci Nutr. 2022;62(14):3738–50.PubMedCrossRef Ramirez-Macias I, Orenes-Pinero E, Camelo-Castillo A, et al. Novel insights in the relationship of gut microbiota and coronary artery diseases. Crit Rev Food Sci Nutr. 2022;62(14):3738–50.PubMedCrossRef
128.
Zurück zum Zitat Yang G, Wei J, Liu P, et al. Role of the gut microbiota in type 2 diabetes and related diseases. Metabolism. 2021;117:154712.PubMedCrossRef Yang G, Wei J, Liu P, et al. Role of the gut microbiota in type 2 diabetes and related diseases. Metabolism. 2021;117:154712.PubMedCrossRef
129.
Zurück zum Zitat Alexander JL, Wilson ID, Teare J, et al. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat Rev Gastroenterol Hepatol. 2017;14(6):356–65.PubMedCrossRef Alexander JL, Wilson ID, Teare J, et al. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat Rev Gastroenterol Hepatol. 2017;14(6):356–65.PubMedCrossRef
130.
Zurück zum Zitat Panebianco C, Andriulli A, Pazienza V. Pharmacomicrobiomics: exploiting the drug-microbiota interactions in anticancer therapies. Microbiome. 2018;6(1):92.PubMedPubMedCentralCrossRef Panebianco C, Andriulli A, Pazienza V. Pharmacomicrobiomics: exploiting the drug-microbiota interactions in anticancer therapies. Microbiome. 2018;6(1):92.PubMedPubMedCentralCrossRef
131.
Zurück zum Zitat Gutierrez Lopez DE, Lashinger LM, Weinstock GM, Bray MS. Circadian rhythms and the gut microbiome synchronize the host’s metabolic response to diet. Cell Metab. 2021;33(5):873–87.PubMedCrossRef Gutierrez Lopez DE, Lashinger LM, Weinstock GM, Bray MS. Circadian rhythms and the gut microbiome synchronize the host’s metabolic response to diet. Cell Metab. 2021;33(5):873–87.PubMedCrossRef
132.
Zurück zum Zitat Xue MY, Sun HZ, Wu XH, Liu JX, Guan LL. Multi-omics reveals that the rumen microbiome and its metabolome together with the host metabolome contribute to individualized dairy cow performance. Microbiome. 2020;8(1):64.PubMedPubMedCentralCrossRef Xue MY, Sun HZ, Wu XH, Liu JX, Guan LL. Multi-omics reveals that the rumen microbiome and its metabolome together with the host metabolome contribute to individualized dairy cow performance. Microbiome. 2020;8(1):64.PubMedPubMedCentralCrossRef
133.
Zurück zum Zitat Zhao Q, Chen Y, Huang W, Zhou H, Zhang W. Drug-microbiota interactions: an emerging priority for precision medicine. Signal Transduct Target Ther. 2023;8(1):386.PubMedPubMedCentralCrossRef Zhao Q, Chen Y, Huang W, Zhou H, Zhang W. Drug-microbiota interactions: an emerging priority for precision medicine. Signal Transduct Target Ther. 2023;8(1):386.PubMedPubMedCentralCrossRef
134.
Zurück zum Zitat Conti G, D’Amico F, Fabbrini M et al. Pharmacomicrobiomics in Anticancer therapies: why the gut microbiota should be pointed out. Genes (Basel). 2022. 14(1). Conti G, D’Amico F, Fabbrini M et al. Pharmacomicrobiomics in Anticancer therapies: why the gut microbiota should be pointed out. Genes (Basel). 2022. 14(1).
135.
Zurück zum Zitat Zitvogel L, Galluzzi L, Viaud S, et al. Cancer and the gut microbiota: an unexpected link. Sci Transl Med. 2015;7(271):271ps271.CrossRef Zitvogel L, Galluzzi L, Viaud S, et al. Cancer and the gut microbiota: an unexpected link. Sci Transl Med. 2015;7(271):271ps271.CrossRef
136.
Zurück zum Zitat Wang F, Yin Q, Chen L, Davis MM. Bifidobacterium can mitigate intestinal immunopathology in the context of CTLA-4 blockade. Proc Natl Acad Sci U S A. 2018;115(1):157–61.PubMedCrossRef Wang F, Yin Q, Chen L, Davis MM. Bifidobacterium can mitigate intestinal immunopathology in the context of CTLA-4 blockade. Proc Natl Acad Sci U S A. 2018;115(1):157–61.PubMedCrossRef
137.
Zurück zum Zitat Badgeley A, Anwar H, Modi K, Murphy P, Lakshmikuttyamma A. Effect of probiotics and gut microbiota on anti-cancer drugs: mechanistic perspectives. Biochim Biophys Acta Rev Cancer. 2021;1875(1):188494.PubMedCrossRef Badgeley A, Anwar H, Modi K, Murphy P, Lakshmikuttyamma A. Effect of probiotics and gut microbiota on anti-cancer drugs: mechanistic perspectives. Biochim Biophys Acta Rev Cancer. 2021;1875(1):188494.PubMedCrossRef
138.
Zurück zum Zitat David LA, Maurice CF, Carmody RN et al. Diet rapidly and reproducibly alters the human gut microbiome. Nat 2014 505(7484):559–63. David LA, Maurice CF, Carmody RN et al. Diet rapidly and reproducibly alters the human gut microbiome. Nat 2014 505(7484):559–63.
139.
Zurück zum Zitat Stewart LK, Smoak P, Hydock DS, et al. Milk and kefir maintain aspects of health during doxorubicin treatment in rats. J Dairy Sci. 2019;102(3):1910–7.PubMedCrossRef Stewart LK, Smoak P, Hydock DS, et al. Milk and kefir maintain aspects of health during doxorubicin treatment in rats. J Dairy Sci. 2019;102(3):1910–7.PubMedCrossRef
140.
Zurück zum Zitat Wu R, Mei X, Wang J, et al. Zn(ii)-Curcumin supplementation alleviates gut dysbiosis and zinc dyshomeostasis during doxorubicin-induced cardiotoxicity in rats. Food Funct. 2019;10(9):5587–604.PubMedCrossRef Wu R, Mei X, Wang J, et al. Zn(ii)-Curcumin supplementation alleviates gut dysbiosis and zinc dyshomeostasis during doxorubicin-induced cardiotoxicity in rats. Food Funct. 2019;10(9):5587–604.PubMedCrossRef
141.
Zurück zum Zitat Hu X, Xia K, Dai M, et al. Intermittent fasting modulates the intestinal microbiota and improves obesity and host energy metabolism. NPJ Biofilms Microbiomes. 2023;9(1):19.PubMedPubMedCentralCrossRef Hu X, Xia K, Dai M, et al. Intermittent fasting modulates the intestinal microbiota and improves obesity and host energy metabolism. NPJ Biofilms Microbiomes. 2023;9(1):19.PubMedPubMedCentralCrossRef
142.
Zurück zum Zitat Suez J, Zmora N, Segal E, Elinav E. The pros, cons, and many unknowns of probiotics. Nat Med. 2019;25(5):716–29.PubMedCrossRef Suez J, Zmora N, Segal E, Elinav E. The pros, cons, and many unknowns of probiotics. Nat Med. 2019;25(5):716–29.PubMedCrossRef
143.
Zurück zum Zitat Companys J, Pla-Pagà L, Calderón-Pérez L, et al. Fermented dairy products, probiotic supplementation, and Cardiometabolic diseases: a systematic review and Meta-analysis. Adv Nutr. 2020;11(4):834–63.PubMedPubMedCentralCrossRef Companys J, Pla-Pagà L, Calderón-Pérez L, et al. Fermented dairy products, probiotic supplementation, and Cardiometabolic diseases: a systematic review and Meta-analysis. Adv Nutr. 2020;11(4):834–63.PubMedPubMedCentralCrossRef
144.
Zurück zum Zitat Celiberto LS, Bedani R, Rossi EA, Cavallini DC, Probiotics. The scientific evidence in the context of inflammatory bowel disease. Crit Rev Food Sci Nutr. 2017;57(9):1759–68.PubMed Celiberto LS, Bedani R, Rossi EA, Cavallini DC, Probiotics. The scientific evidence in the context of inflammatory bowel disease. Crit Rev Food Sci Nutr. 2017;57(9):1759–68.PubMed
145.
Zurück zum Zitat Cavalcanti Neto MP, Aquino JS, Romão da Silva LF, et al. Gut microbiota and probiotics intervention: a potential therapeutic target for management of cardiometabolic disorders and chronic kidney disease? Pharmacol Res. 2018;130:152–63.PubMedCrossRef Cavalcanti Neto MP, Aquino JS, Romão da Silva LF, et al. Gut microbiota and probiotics intervention: a potential therapeutic target for management of cardiometabolic disorders and chronic kidney disease? Pharmacol Res. 2018;130:152–63.PubMedCrossRef
146.
Zurück zum Zitat Zhao L, Xing C, Sun W, et al. Lactobacillus supplementation prevents cisplatin-induced cardiotoxicity possibly by inflammation inhibition. Cancer Chemother Pharmacol. 2018;82(6):999–1008.PubMedCrossRef Zhao L, Xing C, Sun W, et al. Lactobacillus supplementation prevents cisplatin-induced cardiotoxicity possibly by inflammation inhibition. Cancer Chemother Pharmacol. 2018;82(6):999–1008.PubMedCrossRef
147.
Zurück zum Zitat Sharma S, Singh RL, Kakkar P. Modulation of Bax/Bcl-2 and caspases by probiotics during acetaminophen induced apoptosis in primary hepatocytes. Food Chem Toxicol. 2011;49(4):770–9.PubMedCrossRef Sharma S, Singh RL, Kakkar P. Modulation of Bax/Bcl-2 and caspases by probiotics during acetaminophen induced apoptosis in primary hepatocytes. Food Chem Toxicol. 2011;49(4):770–9.PubMedCrossRef
148.
Zurück zum Zitat Abu-Elsaad NM, Abd Elhameed AG, El-Karef A, Ibrahim TM. Yogurt containing the Probacteria Lactobacillus acidophilus combined with natural antioxidants mitigates Doxorubicin-Induced Cardiomyopathy in rats. J Med Food. 2015;18(9):950–9.PubMedCrossRef Abu-Elsaad NM, Abd Elhameed AG, El-Karef A, Ibrahim TM. Yogurt containing the Probacteria Lactobacillus acidophilus combined with natural antioxidants mitigates Doxorubicin-Induced Cardiomyopathy in rats. J Med Food. 2015;18(9):950–9.PubMedCrossRef
149.
Zurück zum Zitat Amaretti A, di Nunzio M, Pompei A, et al. Antioxidant properties of potentially probiotic bacteria: in vitro and in vivo activities. Appl Microbiol Biotechnol. 2013;97(2):809–17.PubMedCrossRef Amaretti A, di Nunzio M, Pompei A, et al. Antioxidant properties of potentially probiotic bacteria: in vitro and in vivo activities. Appl Microbiol Biotechnol. 2013;97(2):809–17.PubMedCrossRef
151.
Zurück zum Zitat Chehoud C, Dryga A, Hwang Y, et al. Transfer of viral communities between Human individuals during fecal microbiota transplantation. mBio. 2016;7(2):e00322.PubMedPubMedCentralCrossRef Chehoud C, Dryga A, Hwang Y, et al. Transfer of viral communities between Human individuals during fecal microbiota transplantation. mBio. 2016;7(2):e00322.PubMedPubMedCentralCrossRef
152.
Zurück zum Zitat Zhou D, Zhang H, Xue X, et al. Safety Evaluation of Natural Drugs in chronic skeletal disorders: a literature review of clinical trials in the past 20 years. Front Pharmacol. 2021;12:801287.PubMedCrossRef Zhou D, Zhang H, Xue X, et al. Safety Evaluation of Natural Drugs in chronic skeletal disorders: a literature review of clinical trials in the past 20 years. Front Pharmacol. 2021;12:801287.PubMedCrossRef
153.
Zurück zum Zitat Anhê FF, Choi BSY, Dyck JRB, Schertzer JD, Marette A. Host-microbe interplay in the Cardiometabolic benefits of Dietary Polyphenols. Trends Endocrinol Metab. 2019;30(6):384–95.PubMedCrossRef Anhê FF, Choi BSY, Dyck JRB, Schertzer JD, Marette A. Host-microbe interplay in the Cardiometabolic benefits of Dietary Polyphenols. Trends Endocrinol Metab. 2019;30(6):384–95.PubMedCrossRef
154.
Zurück zum Zitat Wu X, Zhang N, Kan J, et al. Polyphenols from Arctium lappa L ameliorate doxorubicin-induced heart failure and improve gut microbiota composition in mice. J Food Biochem. 2022;46(3):e13731.PubMedCrossRef Wu X, Zhang N, Kan J, et al. Polyphenols from Arctium lappa L ameliorate doxorubicin-induced heart failure and improve gut microbiota composition in mice. J Food Biochem. 2022;46(3):e13731.PubMedCrossRef
155.
Zurück zum Zitat Ianiro G, Tilg H, Gasbarrini A. Antibiotics as deep modulators of gut microbiota: between good and evil. Gut 2016 65(11):1906–15. Ianiro G, Tilg H, Gasbarrini A. Antibiotics as deep modulators of gut microbiota: between good and evil. Gut 2016 65(11):1906–15.
156.
Zurück zum Zitat Awoyemi A, Mayerhofer C, Felix AS, et al. Rifaximin or Saccharomyces boulardii in heart failure with reduced ejection fraction: results from the randomized GutHeart trial. EBioMedicine. 2021;70:103511.PubMedPubMedCentralCrossRef Awoyemi A, Mayerhofer C, Felix AS, et al. Rifaximin or Saccharomyces boulardii in heart failure with reduced ejection fraction: results from the randomized GutHeart trial. EBioMedicine. 2021;70:103511.PubMedPubMedCentralCrossRef
157.
158.
Zurück zum Zitat Naderi Y, Khosraviani S, Nasiri S, et al. Cardioprotective effects of minocycline against doxorubicin-induced cardiotoxicity. Biomed Pharmacother. 2023;158:114055.PubMedCrossRef Naderi Y, Khosraviani S, Nasiri S, et al. Cardioprotective effects of minocycline against doxorubicin-induced cardiotoxicity. Biomed Pharmacother. 2023;158:114055.PubMedCrossRef
159.
Zurück zum Zitat Kappel BA, De Angelis L, Puetz A, et al. Antibiotic-induced gut microbiota depletion exacerbates host hypercholesterolemia. Pharmacol Res. 2023;187:106570.PubMedCrossRef Kappel BA, De Angelis L, Puetz A, et al. Antibiotic-induced gut microbiota depletion exacerbates host hypercholesterolemia. Pharmacol Res. 2023;187:106570.PubMedCrossRef
160.
161.
Zurück zum Zitat Tang TWH, Chen HC, Chen CY, et al. Loss of Gut Microbiota alters Immune System Composition and cripples Postinfarction Cardiac Repair. Circulation. 2019;139(5):647–59.PubMedCrossRef Tang TWH, Chen HC, Chen CY, et al. Loss of Gut Microbiota alters Immune System Composition and cripples Postinfarction Cardiac Repair. Circulation. 2019;139(5):647–59.PubMedCrossRef
163.
Zurück zum Zitat Marques-Aleixo I, Santos-Alves E, Oliveira PJ, et al. The beneficial role of exercise in mitigating doxorubicin-induced Mitochondrionopathy. Biochim Biophys Acta Rev Cancer. 2018;1869(2):189–99.PubMedCrossRef Marques-Aleixo I, Santos-Alves E, Oliveira PJ, et al. The beneficial role of exercise in mitigating doxorubicin-induced Mitochondrionopathy. Biochim Biophys Acta Rev Cancer. 2018;1869(2):189–99.PubMedCrossRef
164.
Zurück zum Zitat Ting NL, Lau HC, Yu J. Cancer pharmacomicrobiomics: targeting microbiota to optimise cancer therapy outcomes. Gut 2022 71(7):1412–25. Ting NL, Lau HC, Yu J. Cancer pharmacomicrobiomics: targeting microbiota to optimise cancer therapy outcomes. Gut 2022 71(7):1412–25.
Metadaten
Titel
Role of gut microbiota in doxorubicin-induced cardiotoxicity: from pathogenesis to related interventions
verfasst von
Chao Huang
Xiaoxia Li
Hanqing Li
Ruolan Chen
Zhaoqing Li
Daisong Li
Xiaojian Xu
Guoliang Zhang
Luning Qin
Bing Li
Xian-Ming Chu
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2024
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-024-05232-5

Weitere Artikel der Ausgabe 1/2024

Journal of Translational Medicine 1/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.