scispace - formally typeset
Search or ask a question
Author

Yanyang Li

Bio: Yanyang Li is an academic researcher from Tianjin Medical University Cancer Institute and Hospital. The author has contributed to research in topics: Cardiotoxicity & Heart failure. The author has an hindex of 4, co-authored 8 publications receiving 76 citations.

Papers
More filters
Journal ArticleDOI
TL;DR: It is clarified that intestinal barrier damage and bacterial translocation induced inflammation and immune response aggravated heart failure, and altered intestinal microflora affected various metabolic pathways including trimethylamine/TMAO, SCFA, and Bile acid pathway leads to heart failure.
Abstract: Although the mechanism of the occurrence and development of heart failure has been continuously explored in the past ten years, the mortality and readmission rate of heart failure is still very high. Modern studies have shown that gut microbiota is associated with a variety of cardiovascular diseases, among which the study of gut microbiota and heart failure attracts particular attention. Therefore, understanding the role of gut microbiota in the occurrence and development of heart failure will help us further understand the pathogenesis of heart failure and provide new ideas for its treatment. This paper introduced intestinal flora and its metabolites, summarized the changes of intestinal flora in patients with heart failure, clarified that intestinal barrier damage and bacterial translocation induced inflammation and immune response aggravated heart failure, and altered intestinal microflora affected various metabolic pathways including trimethylamine/TMAO, SCFA, and Bile acid pathway leads to heart failure. At the same time, regulating intestinal microflora through diet, probiotics, antibiotics, fecal transplantation and microbial enzyme inhibitors has grown up to be a potential treatment for many metabolic disorders.

54 citations

Journal ArticleDOI
TL;DR: The target and mechanism of traditional Chinese medicine treating heart failure by acting gut microbiota will be specified, and ideas for the treatment of heart failure are provided.

49 citations

Journal ArticleDOI
TL;DR: The relevant mechanisms of the miR-30 in VR induced by various diseases will be reviewed, including autophagy, apoptosis, oxidative stress, and inflammation.
Abstract: Ventricular remodeling (VR) is a complex pathological process of cardiomyocyte apoptosis, cardiac hypertrophy, and myocardial fibrosis, which is often caused by various cardiovascular diseases (CVDs) such as hypertension, acute myocardial infarction, heart failure (HF), etc. It is also an independent risk factor for a variety of CVDs, which will eventually to damage the heart function, promote cardiovascular events, and lead to an increase in mortality. MicroRNAs (miRNAs) can participate in a variety of CVDs through post-transcriptional regulation of target gene proteins. Among them, microRNA-30 (miR-30) is one of the most abundant miRNAs in the heart. In recent years, the study found that the miR-30 family can participate in VR through a variety of mechanisms, including autophagy, apoptosis, oxidative stress, and inflammation. VR is commonly found in ischemic heart disease (IHD), hypertensive heart disease (HHD), diabetic cardiomyopathy (DCM), antineoplastic drug cardiotoxicity (CTX), and other CVDs. Therefore, we will review the relevant mechanisms of the miR-30 in VR induced by various diseases.

43 citations

Journal ArticleDOI
TL;DR: In this paper, the authors collected and collated the latest researches and mainly discussed the following four parts: Gut microbiota, TMA-producing bacteria, and TMAO.
Abstract: Hypertension is the most prevalent chronic disease and a risk factor for various diseases. Although its mechanisms and therapies are constantly being updated and developed, they are still not fully clarified. In recent years, novel gut microbiota and its metabolites have attracted widespread attention. It is strongly linked with physiological and pathological systems, especially TMA and TMAO. TMA is formed by intestinal microbial metabolism of choline and L-carnitine and converted into TMAO by FMO3. This paper collected and collated the latest researches and mainly discussed the following four parts. It introduced gut microbiota; provided a focus on TMA, TMA-producing bacteria, and TMAO; summarized the alternations in hypertensive patients and animals; discussed the mechanisms of TMAO with two respects; and summarized the regulatory factors may be as new interventions and therapies of hypertension. And, more relevant studies are still prospected to be accomplished between hypertension and TMA/TMAO for further clinical services.

25 citations

Journal ArticleDOI
TL;DR: The role of biomarkers in myocardial fibrosis from the perspective of collagen is discussed, which shows that collagen plays a key role in the impairment of cardiac function.
Abstract: Myocardial fibrosis is observed in various cardiovascular diseases and plays a key role in the impairment of cardiac function. Endomyocardial biopsy, as the gold standard for the diagnosis of myocardial fibrosis, has limitations in terms of clinical application. Therefore, biomarkers have been recommended for noninvasive assessment of myocardial fibrosis. This review discusses the role of biomarkers in myocardial fibrosis from the perspective of collagen.

20 citations


Cited by
More filters
Journal ArticleDOI
24 Dec 2004-Science

1,949 citations

15 Sep 2015
TL;DR: It is found that residence in the small intestine, rather than bacterial identity, dictated induction of specific IgA, and a specialized function of the B1b lineage in TI mucosal IgA responses is revealed.
Abstract: Immunoglobulin A (IgA) is prominently secreted at mucosal surfaces and coats a fraction of the intestinal microbiota However, the commensal bacteria bound by IgA are poorly characterized and the type of humoral immunity they elicit remains elusive We used bacterial flow cytometry coupled with 16S rRNA gene sequencing (IgA-Seq) in murine models of immunodeficiency to identify IgA-bound bacteria and elucidate mechanisms of commensal IgA targeting We found that residence in the small intestine, rather than bacterial identity, dictated induction of specific IgA Most commensals elicited strong T-independent (TI) responses that originated from the orphan B1b lineage and from B2 cells, but excluded natural antibacterial B1a specificities Atypical commensals including segmented filamentous bacteria and Mucispirillum evaded TI responses but elicited T-dependent IgA These data demonstrate exquisite targeting of distinct commensal bacteria by multiple layers of humoral immunity and reveal a specialized function of the B1b lineage in TI mucosal IgA responses

295 citations

Journal Article
TL;DR: In this article, the authors investigated the intestinal microbiome of 20 patients with chronic heart failure with reduced ejection fraction due to dilated or ischemic or dilated cardiomyopathy.
Abstract: Introduction: Despite current medical treatment approaches, mortality of chronic heart failure (HF) remains high Novel treatment modalities are thus urgently needed A recent theory proposes a possible impact of the intestinal microbiome on the incidence and clinical course of HF This study sought to investigate, if there are specific changes of the intestinal microbiome in HF patients Methods and Results: The intestinal microbiome of 20 patients with HF with reduced ejection fraction due to ischemic or dilated cardiomyopathy was investigated applying high-throughput sequencing of the bacterial 16S rRNA gene Microbial profiles were compared to those of healthy matched controls (n=20) According to the Shannon diversity index (measuring the intra-individual alpha-diversity) based on the distribution of operational taxonomic units, HF cases showed a nominally significantly lower diversity index compared to controls (Pnom=001), testing for genera abundance showed a tendency towards a decreased alpha di

78 citations

12 Sep 2017
TL;DR: This review will focus mainly on the recent advances in the molecular mechanisms of cardiotoxicity related to common cancer therapies while introducing the concept of cardio-oncology service.
Abstract: Current oncologic treatments have brought a strong reduction in mortality in cancer patients. However, the cancer therapy-related cardiovascular complications, in particular chemo-therapy and radiation therapy-induced cardiotoxicities are a major cause of morbidity and mortality in people living with or surviving cancer. The simple fact is that all antineoplastic agents and radiation therapy target tumor cells but also result in collateral damage to other tissues including the cardiovascular system. The commonly used anthracycline chemotherapy agents can induce cardiomyopathy and congestive heart failure. Targeted therapies with human epidermal growth factor antibodies, tyrosine kinase inhibitors or vascular endothelial growth factor antibodies, and the antimetabolites also have shown to induce cardiomyopathy and myocardial ischemia. Cardiac arrhythmias and hypertension have been well described with the use of tyrosine kinase inhibitors and antimicrotubule agents. Pericarditis can happen with the use of cyclophosphamide or cytarabine. Mediastinal radiation can cause constrictive pericarditis, myocardial fibrosis, valvular lesions, and coronary artery disease. Despite significant progresses in the understanding of the molecular and pathophysiologic mechanisms behind the cardiovascular toxicity of cancer therapy, there is still lack of evidence-based approach for the monitoring and management of patients. This review will focus mainly on the recent advances in the molecular mechanisms of cardiotoxicity related to common cancer therapies while introducing the concept of cardio-oncology service. Applying the general principles of multi-disciplinary approaches toward the diagnosis, prevention, monitoring, and treatment of cancer therapy-induced cardiomyopathy and heart failure will also be discussed.

76 citations

Journal ArticleDOI
TL;DR: This review introduced and summarized the results from recent studies dealing with the relationship between gut microbiota and cardiac disorders, highlighting the role of programmed cell death and hopes to shed light on microbiota-targeted therapeutic strategies in CVD management.
Abstract: Emerging evidence has identified the association between gut microbiota and various diseases, including cardiovascular diseases (CVDs). Altered intestinal flora composition has been described in detail in CVDs, such as hypertension, atherosclerosis, myocardial infarction, heart failure, and arrhythmia. In contrast, the importance of fermentation metabolites, such as trimethylamine N-oxide (TMAO), short-chain fatty acids (SCFAs), and secondary bile acid (BA), has also been implicated in CVD development, prevention, treatment, and prognosis. The potential mechanisms are conventionally thought to involve immune regulation, host energy metabolism, and oxidative stress. However, numerous types of programmed cell death, including apoptosis, autophagy, pyroptosis, ferroptosis, and clockophagy, also serve as a key link in microbiome-host cross talk. In this review, we introduced and summarized the results from recent studies dealing with the relationship between gut microbiota and cardiac disorders, highlighting the role of programmed cell death. We hope to shed light on microbiota-targeted therapeutic strategies in CVD management.

61 citations