1
|
Liu T, Zhuang XX, Qin XJ, Wei LB, Gao JR. Alteration of N6-methyladenosine epitranscriptome profile in lipopolysaccharide-induced mouse mesangial cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:445-458. [PMID: 35119478 DOI: 10.1007/s00210-022-02208-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/18/2022] [Indexed: 11/30/2022]
Abstract
N6-Methyladenosine (m6A) is the most prevalent internal modification of messenger RNA (mRNA) in eukaryotes. The underlying molecular mechanisms of m6A modification in chronic glomerulonephritis (CGN) remain unexplored. Here, we performed methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA sequencing (RNA-seq) analyses to assess the alterations of epitranscriptome-wide m6A profile in lipopolysaccharide (LPS)-induced mouse mesangial cells (MMC). The results of our data showed 2153 significantly differential m6A peaks and 358 significantly differentially expressed genes. Furthermore, integrated analysis from MeRIP-seq and RNA-seq identified a total of 64 genes with differential m6A modification and expressed levels, of which 5 genes displayed hypermethylation and upregulation, 42 genes displayed hypermethylation and downregulation, 11 genes displayed hypomethylation and upregulation, and 8 genes displayed hypomethylation and downregulation. Many of them (including Fosl1, Sorbs1, Ambp, Fgfr3, Nedd9, Fgg, Trim13, Fgf22, Mylk, and Muc6) are implicated in the regulation of the immune and inflammatory response. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analysis found that differential 64 genes were mainly enriched in fatty acid oxidation, apoptosis signaling pathway, complement and coagulation cascades, and PPAR signaling pathway. Together, our study provided a new perspective on the understanding of molecular features of m6A modification in CGN pathogenic pathogenesis.
Collapse
Affiliation(s)
- Tao Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230011, Anhui, China
| | - Xing Xing Zhuang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230011, Anhui, China.,Department of Pharmacy, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, Anhui, China
| | - Xiu Juan Qin
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Liang Bing Wei
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Jia Rong Gao
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China. .,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230031, Anhui, China.
| |
Collapse
|
2
|
Alharthi A, Alhazmi S, Alburae N, Bahieldin A. The Human Gut Microbiome as a Potential Factor in Autism Spectrum Disorder. Int J Mol Sci 2022; 23:ijms23031363. [PMID: 35163286 PMCID: PMC8835713 DOI: 10.3390/ijms23031363] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/18/2022] [Accepted: 01/24/2022] [Indexed: 11/16/2022] Open
Abstract
The high prevalence of gastrointestinal (GI) disorders among autism spectrum disorder (ASD) patients has prompted scientists to look into the gut microbiota as a putative trigger in ASD pathogenesis. Thus, many studies have linked the gut microbial dysbiosis that is frequently observed in ASD patients with the modulation of brain function and social behavior, but little is known about this connection and its contribution to the etiology of ASD. This present review highlights the potential role of the microbiota–gut–brain axis in autism. In particular, it focuses on how gut microbiota dysbiosis may impact gut permeability, immune function, and the microbial metabolites in autistic people. We further discuss recent findings supporting the possible role of the gut microbiome in initiating epigenetic modifications and consider the potential role of this pathway in influencing the severity of ASD. Lastly, we summarize recent updates in microbiota-targeted therapies such as probiotics, prebiotics, dietary supplements, fecal microbiota transplantation, and microbiota transfer therapy. The findings of this paper reveal new insights into possible therapeutic interventions that may be used to reduce and cure ASD-related symptoms. However, well-designed research studies using large sample sizes are still required in this area of study.
Collapse
Affiliation(s)
- Amani Alharthi
- Department of Biology, Faculty of Science, Majmaah University, Al Zulfi 11932, Saudi Arabia
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.); (N.A.)
- Correspondence: (A.A.); (A.B.)
| | - Safiah Alhazmi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.); (N.A.)
| | - Najla Alburae
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.); (N.A.)
| | - Ahmed Bahieldin
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.); (N.A.)
- Correspondence: (A.A.); (A.B.)
| |
Collapse
|
3
|
Soranno DE, Baker P, Kirkbride-Romeo L, Wennersten SA, Ding K, Keith B, Cavasin MA, Altmann C, Bagchi RA, Haefner KR, Montford J, Gist KM, Vergnes L, Reue K, He Z, Elajaili H, Okamura K, Nozik E, McKinsey TA, Faubel S. Female and male mice have differential longterm cardiorenal outcomes following a matched degree of ischemia-reperfusion acute kidney injury. Sci Rep 2022; 12:643. [PMID: 35022484 PMCID: PMC8755805 DOI: 10.1038/s41598-021-04701-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/29/2021] [Indexed: 12/13/2022] Open
Abstract
Acute kidney injury (AKI) is common in patients, causes systemic sequelae, and predisposes patients to long-term cardiovascular disease. To date, studies of the effects of AKI on cardiovascular outcomes have only been performed in male mice. We recently demonstrated that male mice developed diastolic dysfunction, hypertension and reduced cardiac ATP levels versus sham 1 year after AKI. The effects of female sex on long-term cardiac outcomes after AKI are unknown. Therefore, we examined the 1-year cardiorenal outcomes following a single episode of bilateral renal ischemia-reperfusion injury in female C57BL/6 mice using a model with similar severity of AKI and performed concomitantly to recently published male cohorts. To match the severity of AKI between male and female mice, females received 34 min of ischemia time compared to 25 min in males. Serial renal function, echocardiograms and blood pressure assessments were performed throughout the 1-year study. Renal histology, and cardiac and plasma metabolomics and mitochondrial function in the heart and kidney were evaluated at 1 year. Measured glomerular filtration rates (GFR) were similar between male and female mice throughout the 1-year study period. One year after AKI, female mice had preserved diastolic function, normal blood pressure, and preserved levels of cardiac ATP. Compared to males, females demonstrated pathway enrichment in arginine metabolism and amino acid related energy production in both the heart and plasma, and glutathione in the plasma. Cardiac mitochondrial respiration in Complex I of the electron transport chain demonstrated improved mitochondrial function in females compared to males, regardless of AKI or sham. This is the first study to examine the long-term cardiac effects of AKI on female mice and indicate that there are important sex-related cardiorenal differences. The role of female sex in cardiovascular outcomes after AKI merits further investigation.
Collapse
Affiliation(s)
- Danielle E Soranno
- Department of Pediatrics, Pediatric Nephrology, University of Colorado, 13123 E. 16th Ave, Box #328, Aurora, CO, 80045, USA.
- Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado, Aurora, CO, USA.
- Consortium for Fibrosis Research & Translation, University of Colorado, Aurora, CO, USA.
- Department of Bioengineering, University of Colorado, Aurora, CO, USA.
| | - Peter Baker
- Department of Pediatrics, Clinical Genetics & Metabolism, University of Colorado, Aurora, CO, USA
| | - Lara Kirkbride-Romeo
- Department of Pediatrics, Pediatric Nephrology, University of Colorado, 13123 E. 16th Ave, Box #328, Aurora, CO, 80045, USA
| | - Sara A Wennersten
- Consortium for Fibrosis Research & Translation, University of Colorado, Aurora, CO, USA
- Division of Cardiology, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Kathy Ding
- School of Medicine, University of Colorado, Aurora, CO, USA
| | - Brysen Keith
- Department of Bioengineering, University of Colorado, Aurora, CO, USA
| | - Maria A Cavasin
- Consortium for Fibrosis Research & Translation, University of Colorado, Aurora, CO, USA
- Division of Cardiology, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Christopher Altmann
- Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Rushita A Bagchi
- Consortium for Fibrosis Research & Translation, University of Colorado, Aurora, CO, USA
- Division of Cardiology, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Korey R Haefner
- Consortium for Fibrosis Research & Translation, University of Colorado, Aurora, CO, USA
- Division of Cardiology, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - John Montford
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, USA
| | - Katja M Gist
- Department of Pediatrics, Pediatric Cardiology, Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, OH, USA
| | - Laurent Vergnes
- Department of Human Genetics and Metabolism Theme Area, University of California Los Angeles, Los Angeles, CA, USA
| | - Karen Reue
- Department of Human Genetics and Metabolism Theme Area, University of California Los Angeles, Los Angeles, CA, USA
| | - Zhibin He
- Department of Pediatrics, Pediatric Nephrology, University of Colorado, 13123 E. 16th Ave, Box #328, Aurora, CO, 80045, USA
| | - Hanan Elajaili
- Department of Pediatrics, Critical Care Medicine, University of Colorado, Aurora, CO, USA
| | - Kayo Okamura
- Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Eva Nozik
- Department of Pediatrics, Critical Care Medicine, University of Colorado, Aurora, CO, USA
| | - Timothy A McKinsey
- Consortium for Fibrosis Research & Translation, University of Colorado, Aurora, CO, USA
- Division of Cardiology, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Sarah Faubel
- Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado, Aurora, CO, USA
| |
Collapse
|
4
|
Exploring the Pleiotropic Genes and Therapeutic Targets Associated with Heart Failure and Chronic Kidney Disease by Integrating metaCCA and SGLT2 Inhibitors' Target Prediction. BIOMED RESEARCH INTERNATIONAL 2021; 2021:4229194. [PMID: 34540994 PMCID: PMC8443964 DOI: 10.1155/2021/4229194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/08/2021] [Accepted: 08/11/2021] [Indexed: 11/18/2022]
Abstract
Background Previous studies have shown that heart failure (HF) and chronic kidney disease (CKD) have common genetic mechanisms, overlapping pathophysiological pathways, and therapeutic drug—sodium-glucose cotransporter 2 (SGLT2) inhibitors. Methods The genetic pleiotropy metaCCA method was applied on summary statistics data from two independent meta-analyses of GWAS comprising more than 1 million people to identify shared variants and pleiotropic effects between HF and CKD. Targets of SGLT2 inhibitors were predicted by SwissTargetPrediction and DrugBank databases. To refine all genes, we performed using versatile gene-based association study 2 (VEGAS2) and transcriptome-wide association studies (TWAS) for HF and CKD, respectively. Gene enrichment and KEGG pathway analyses were used to explore the potential functional significance of the identified genes and targets. Results After metaCCA analysis, 4,624 SNPs and 1,745 genes were identified to be potentially pleiotropic in the univariate and multivariate SNP-multivariate phenotype analyses, respectively. 21 common genes were detected in both metaCCA and SGLT2 inhibitors' target prediction. In addition, 169 putative pleiotropic genes were identified, which met the significance threshold both in metaCCA analysis and in the VEGAS2 or TWAS analysis for at least one disease. Conclusion We identified novel variants associated with HF and CKD using effectively incorporating information from different GWAS datasets. Our analysis may provide new insights into HF and CKD therapeutic approaches based on the pleiotropic genes, common targets, and mechanisms by integrating the metaCCA method, TWAS and VEGAS2 analyses, and target prediction of SGLT2 inhibitors.
Collapse
|
5
|
Epigenetic Alterations in Pediatric Sleep Apnea. Int J Mol Sci 2021; 22:ijms22179523. [PMID: 34502428 PMCID: PMC8430725 DOI: 10.3390/ijms22179523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 12/03/2022] Open
Abstract
Pediatric obstructive sleep apnea has significant negative effects on health and behavior in childhood including depression, failure to thrive, neurocognitive impairment, and behavioral issues. It is strongly associated with an increased risk for chronic adult disease such as obesity and diabetes, accelerated atherosclerosis, and endothelial dysfunction. Accumulating evidence suggests that adult-onset non-communicable diseases may originate from early life through a process by which an insult applied at a critical developmental window causes long-term effects on the structure or function of an organism. In recent years, there has been increased interest in the role of epigenetic mechanisms in the pathogenesis of adult disease susceptibility. Epigenetic mechanisms that influence adaptive variability include histone modifications, non-coding RNAs, and DNA methylation. This review will highlight what is currently known about the phenotypic associations of epigenetic modifications in pediatric obstructive sleep apnea and will emphasize the importance of epigenetic changes as both modulators of chronic disease and potential therapeutic targets.
Collapse
|
6
|
Soranno DE, Kirkbride-Romeo L, Wennersten SA, Ding K, Cavasin MA, Baker P, Altmann C, Bagchi RA, Haefner KR, Steinkühler C, Montford JR, Keith B, Gist KM, McKinsey TA, Faubel S. Acute Kidney Injury Results in Long-Term Diastolic Dysfunction That Is Prevented by Histone Deacetylase Inhibition. ACTA ACUST UNITED AC 2021; 6:119-133. [PMID: 33665513 PMCID: PMC7907538 DOI: 10.1016/j.jacbts.2020.11.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/19/2020] [Accepted: 11/19/2020] [Indexed: 01/06/2023]
Abstract
This is the first long-term (1-year) study to evaluate both the kidney and systemic sequelae of acute kidney injury in mice. Serial kidney function was measured via transcutaneous glomerular filtration rate. AKI resulted in diastolic dysfunction, followed by hypertension. Ejection fraction was preserved. One year after AKI, cardiac ATP levels were reduced compared with sham controls. Mice treated with the histone deacetylase inhibitor, ITF2357, maintained normal diastolic function normal blood pressure, and normal cardiac ATP after AKI. Metabolomics data suggest that treatment with ITF2357 preserves pathways related to energy metabolism.
Growing epidemiological data demonstrate that acute kidney injury (AKI) is associated with long-term cardiovascular morbidity and mortality. Here, the authors present a 1-year study of cardiorenal outcomes following bilateral ischemia-reperfusion injury in male mice. These data suggest that AKI causes long-term dysfunction in the cardiac metabolome, which is associated with diastolic dysfunction and hypertension. Mice treated with the histone deacetylase inhibitor, ITF2357, had preservation of cardiac function and remained normotensive throughout the study. ITF2357 did not protect against the development of kidney fibrosis after AKI.
Collapse
Affiliation(s)
- Danielle E Soranno
- Department of Pediatrics, Pediatric Nephrology, University of Colorado, Aurora, Colorado, USA.,Department of Medicine, Division of Renal Disease and Hypertension, University of Colorado, Aurora, Colorado, USA.,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lara Kirkbride-Romeo
- Department of Pediatrics, Pediatric Nephrology, University of Colorado, Aurora, Colorado, USA
| | - Sara A Wennersten
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Medicine, Division of Cardiology, University of Colorado, Aurora, Colorado, USA
| | - Kathy Ding
- Department of Pediatrics, Pediatric Nephrology, University of Colorado, Aurora, Colorado, USA
| | - Maria A Cavasin
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Medicine, Division of Cardiology, University of Colorado, Aurora, Colorado, USA
| | - Peter Baker
- Department of Pediatrics, Clinical Genetics and Metabolism, University of Colorado, Aurora, Colorado, USA
| | - Christopher Altmann
- Department of Medicine, Division of Renal Disease and Hypertension, University of Colorado, Aurora, Colorado, USA
| | - Rushita A Bagchi
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Medicine, Division of Cardiology, University of Colorado, Aurora, Colorado, USA
| | - Korey R Haefner
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Medicine, Division of Cardiology, University of Colorado, Aurora, Colorado, USA
| | | | - John R Montford
- Department of Medicine, Division of Renal Disease and Hypertension, University of Colorado, Aurora, Colorado, USA.,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, USA
| | - Brysen Keith
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Katja M Gist
- Department of Pediatrics, Pediatric Cardiology, University of Colorado, Aurora, Colorado, USA
| | - Timothy A McKinsey
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Medicine, Division of Cardiology, University of Colorado, Aurora, Colorado, USA
| | - Sarah Faubel
- Department of Medicine, Division of Renal Disease and Hypertension, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
7
|
Epigenetics, HIV, and Cardiovascular Disease Risk. Curr Probl Cardiol 2020; 46:100615. [PMID: 32507271 DOI: 10.1016/j.cpcardiol.2020.100615] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022]
Abstract
Human immunodeficiency virus (HIV) is currently considered a risk factor for cardiovascular disease (CVD). With the advent of antiretroviral treatment and prevention, HIV-related morbidity and mortality rates have decreased significantly. Prolonged life expectancy heralded higher prevalence of diseases of aging, including CVD-associated morbidity and mortality, having an earlier onset in people living with HIV (PLHIV) compared to their noninfected counterparts. Several epigenetic biomarkers are now available as predictors of health and disease, with DNA methylation being one of the most widely studied. Epigenetic biomarkers are changes in gene expression without alterations to the intrinsic DNA sequence, with the potential to predict risk of future CVD, as well as the outcome and response to therapy among PLHIV. We sought to review the available literature referencing epigenetic markers to determine underlying biomechanism predisposing high-risk PLHIV to CVD, elucidating areas of possible intervention.
Collapse
|
8
|
Guo Y, Wang W, Chen Y, Sun Y, Li Y, Guan F, Shen Q, Guo Y, Zhang W. Continuous gibberellin A3 exposure from weaning to sexual maturity induces ovarian granulosa cell apoptosis by activating Fas-mediated death receptor signaling pathways and changing methylation patterns on caspase-3 gene promoters. Toxicol Lett 2020; 319:175-186. [PMID: 31733319 DOI: 10.1016/j.toxlet.2019.11.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 12/20/2022]
Abstract
Information on the effects of gibberellic acid (gibberellin A3, GA3) on ovarian follicle development is limited. In our present study, 21-day-old female Wistar rats were exposed to GA3 by gavage (25, 50, and 100 mg/kg body weight, once per day) for eight weeks to evaluate the influence of GA3 on ovarian follicle development. After treatment, significant (P < 0.05) increases (to 40.17 % and 44.5 %, respectively) in atretic follicle proportions and significant decreases (to 19.49 % and 17.86 %, respectively) in corpus luteum proportions were observed in the 50 and 100 mg/kg treatment groups compared to the control group. Significant (P < 0.05) increases (to 31.3 % and 42.0 %, respectively) in follicle apoptosis were observed in the 50 and 100 mg/kg treatment groups by transmission electron microscopy and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assays. Significantly increased expression of caspase-3, caspase-8, caspase-9 and Fas was observed by real-time PCR and Western blotting. Bisulfite sequencing PCR (BSP) revealed obviously decreased total methylation percentages of the caspase-3 promoter region in the two treatment groups. Real-time quantitative PCR also showed significantly decreased mRNA expression of DNA methyltransferase (Dnmt) 3a and Dnmt3b. Further in vitro studies showed that a DNA methylation inhibitor could enhance the GA3-induced increase in the mRNA expression of caspase-3. Overall, our present study indicates that GA3 administration from weaning until sexual maturity can affect ovarian follicle development by inducing apoptosis and suggests that signaling through the Fas-mediated apoptotic pathway may be an important underlying mechanism of this apoptosis. In addition, GA3-induced aberrant DNA methylation patterns might be partly responsible for upregulation of caspase-3 gene expression.
Collapse
Affiliation(s)
- Yiwei Guo
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China; Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Wenxiang Wang
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China; Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China.
| | - Yiqin Chen
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China; Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Yan Sun
- Center for Reproductive Medicine, Teaching Hospital of Fujian Medical University, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
| | - Yuchen Li
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Fangyuan Guan
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Qi Shen
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Yiruo Guo
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Wenchang Zhang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
9
|
MALAT1: a therapeutic candidate for a broad spectrum of vascular and cardiorenal complications. Hypertens Res 2019; 43:372-379. [PMID: 31853043 DOI: 10.1038/s41440-019-0378-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/15/2019] [Accepted: 11/15/2019] [Indexed: 01/26/2023]
Abstract
Cardiovascular and renal complications cover a wide array of diseases. The most commonly known overlapping complications include cardiac and renal fibrosis, cardiomyopathy, cardiac hypertrophy, hypertension, and cardiorenal failure. The known or reported causes for the abovementioned complications include injury, ischemia, infection, and metabolic stress. To date, various targets have been reported and investigated in detail that are considered to be the cause of these complications. In the past 5 years, the role of noncoding RNAs has emerged in the area of cardiovascular and renal research, especially in relation to metabolic stress. The long noncoding RNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) has shown immense promise among the long noncoding RNA targets for treating cardiorenal complications. In this review, we shed light on the role of MALAT1 as a primary and novel target in treating cardiovascular and renal diseases as a whole.
Collapse
|
10
|
Clementi A, Virzì G, Battaglia G, Ronco C. Neurohormonal, Endocrine, and Immune Dysregulation and Inflammation in Cardiorenal Syndrome. Cardiorenal Med 2019; 9:265-273. [DOI: 10.1159/000500715] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 04/30/2019] [Indexed: 11/19/2022] Open
Abstract
“Organ crosstalk” is the complex physiological communication between different body systems, and it is necessary for the optimal equilibrium and functioning of the organism. In particular, heart and kidney function is tightly connected, and interplay between these two organs occurs through a vast array of dynamic and bidirectional mechanisms. The term cardiorenal syndrome (CRS) indicates an interaction between the heart and kidneys in acute and chronic disease settings. In all types of CRS, multiple pathophysiological processes are implicated in the initiation and progression of organ injury. In addition to hemodynamic parameters, endothelial injury, immunological imbalance, cell death, inflammatory cascades, oxidative stress, neutrophil migration, leukocyte trafficking, caspase-mediated apoptosis, extracellular vesicles, small noncoding RNAs, and epigenetics play pivotal roles in the development of CRS. In this review, we will focus on neurohormonal, endocrine, and immune dysregulation and inflammation as mechanisms involved in the pathogenesis of CRS.
Collapse
|
11
|
Epigenetics: A Potential Mechanism Involved in the Pathogenesis of Various Adverse Consequences of Obstructive Sleep Apnea. Int J Mol Sci 2019; 20:ijms20122937. [PMID: 31208080 PMCID: PMC6627863 DOI: 10.3390/ijms20122937] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/09/2019] [Accepted: 06/13/2019] [Indexed: 12/14/2022] Open
Abstract
Epigenetics is defined as the heritable phenotypic changes which do not involve alterations in the DNA sequence, including histone modifications, non-coding RNAs, and DNA methylation. Recently, much attention has been paid to the role of hypoxia-mediated epigenetic regulation in cancer, pulmonary hypertension, adaptation to high altitude, and cardiorenal disease. In contrast to sustained hypoxia, chronic intermittent hypoxia with re-oxygenation (IHR) plays a major role in the pathogenesis of various adverse consequences of obstructive sleep apnea (OSA), resembling ischemia re-perfusion injury. Nevertheless, the role of epigenetics in the pathogenesis of OSA is currently underexplored. This review proposes that epigenetic processes are involved in the development of various adverse consequences of OSA by influencing adaptive potential and phenotypic variability under conditions of chronic IHR. Improved understanding of the interaction between genetic and environmental factors through epigenetic regulations holds great value to give deeper insight into the mechanisms underlying IHR-related low-grade inflammation, oxidative stress, and sympathetic hyperactivity, and clarify their implications for biomedical research.
Collapse
|
12
|
Fernández-Colino A, Iop L, Ventura Ferreira MS, Mela P. Fibrosis in tissue engineering and regenerative medicine: treat or trigger? Adv Drug Deliv Rev 2019; 146:17-36. [PMID: 31295523 DOI: 10.1016/j.addr.2019.07.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 05/11/2019] [Accepted: 07/04/2019] [Indexed: 02/07/2023]
Abstract
Fibrosis is a life-threatening pathological condition resulting from a dysfunctional tissue repair process. There is no efficient treatment and organ transplantation is in many cases the only therapeutic option. Here we review tissue engineering and regenerative medicine (TERM) approaches to address fibrosis in the cardiovascular system, the kidney, the lung and the liver. These strategies have great potential to achieve repair or replacement of diseased organs by cell- and material-based therapies. However, paradoxically, they might also trigger fibrosis. Cases of TERM interventions with adverse outcome are also included in this review. Furthermore, we emphasize the fact that, although organ engineering is still in its infancy, the advances in the field are leading to biomedically relevant in vitro models with tremendous potential for disease recapitulation and development of therapies. These human tissue models might have increased predictive power for human drug responses thereby reducing the need for animal testing.
Collapse
|
13
|
Novel Molecular Targets Participating in Myocardial Ischemia-Reperfusion Injury and Cardioprotection. Cardiol Res Pract 2019; 2019:6935147. [PMID: 31275641 PMCID: PMC6558612 DOI: 10.1155/2019/6935147] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 03/28/2019] [Indexed: 12/11/2022] Open
Abstract
Worldwide morbidity and mortality from acute myocardial infarction (AMI) and related heart failure remain high. While effective early reperfusion of the criminal coronary artery after a confirmed AMI is the typical treatment at present, collateral myocardial ischemia-reperfusion injury (MIRI) and pertinent cardioprotection are still challenging to address and have inadequately understood mechanisms. Therefore, unveiling the related novel molecular targets and networks participating in triggering and resisting the pathobiology of MIRI is a promising and valuable frontier. The present study specifically focuses on the recent MIRI advances that are supported by sophisticated bio-methodology in order to bring the poorly understood interrelationship among pro- and anti-MIRI participant molecules up to date, as well as to identify findings that may facilitate the further investigation of novel targets.
Collapse
|
14
|
Lipopolysaccharide in systemic circulation induces activation of inflammatory response and oxidative stress in cardiorenal syndrome type 1. J Nephrol 2019; 32:803-810. [DOI: 10.1007/s40620-019-00613-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/15/2019] [Indexed: 01/21/2023]
|
15
|
Virzì G, Clementi A, Battaglia G, Ronco C. Multi-Omics Approach: New Potential Key Mechanisms Implicated in Cardiorenal Syndromes. Cardiorenal Med 2019; 9:201-211. [DOI: 10.1159/000497748] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/31/2019] [Indexed: 11/19/2022] Open
Abstract
Cardiorenal syndromes (CRS) include a scenario of clinical interactions characterized by the heart and kidney dysfunction. The crosstalk between cardiac and renal systems is clearly evidenced but not completely understood. Multi-factorial mechanisms leading to CRS do not involve only hemodynamic parameters. In fact, in recent works on organ crosstalk endothelial injury, the alteration of normal immunologic balance, cell death, inflammatory cascades, cell adhesion molecules, cytokine and chemokine overexpression, neutrophil migration, leukocyte trafficking, caspase-mediated induction of apoptotic mechanisms and oxidative stress has been demonstrated to induce distant organ dysfunction. Furthermore, new alternative mechanisms using the multi-omics approach may be implicated in the pathogenesis of cardiorenal crosstalk. The study of “omics” modifications in the setting of cardiovascular and renal disease represents an emerging area of research. Over the last years, indeed, many studies have elucidated the exact mechanisms involved in gene expression and regulation, cellular communication and organ crosstalk. In this review, we analyze epigenetics, gene expression, small non-coding RNAs, extracellular vesicles, proteomics, and metabolomics in the setting of CRS.
Collapse
|
16
|
Liu S. Heart-kidney interactions: mechanistic insights from animal models. Am J Physiol Renal Physiol 2019; 316:F974-F985. [PMID: 30838876 DOI: 10.1152/ajprenal.00624.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pathological changes in the heart or kidney can instigate the release of a cascade of cardiorenal mediators that promote injury in the other organ. Combined dysfunction of heart and kidney is referred to as cardiorenal syndrome (CRS) and has gained considerable attention. CRS has been classified into five distinct entities, each with different major pathophysiological changes. Despite the magnitude of the public health problem of CRS, the underlying mechanisms are incompletely understood, and effective intervention is unavailable. Animal models have allowed us to discover pathogenic molecular changes to clarify the pathophysiological mechanisms responsible for heart-kidney interactions and to enable more accurate risk stratification and effective intervention. Here, this article focuses on the use of currently available animal models to elucidate mechanistic insights in the clinical cardiorenal phenotype arising from primary cardiac injury, primary renal disease with special emphasis of chronic kidney disease-specific risk factors, and simultaneous cardiorenal/renocardiac dysfunction. The development of novel animal models that recapitulate more closely the cardiorenal phenotype in a clinical scenario and discover the molecular basis of this condition will be of great benefit.
Collapse
Affiliation(s)
- Shan Liu
- School of Medicine, South China University of Technology , Guangzhou , China
| |
Collapse
|
17
|
Unveiling the Role of DNA Methylation in Kidney Transplantation: Novel Perspectives toward Biomarker Identification. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1602539. [PMID: 30766879 PMCID: PMC6350635 DOI: 10.1155/2019/1602539] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 12/30/2018] [Indexed: 12/13/2022]
Abstract
The burden of chronic kidney disease is dramatically rising, making it a major public health concern worldwide. Kidney transplantation is now the best treatment for patients with end-stage renal disease. Although kidney transplantation may improve survival and quality of life, its long-term results are hampered by immune- and/or non-immune-mediated complications. Thus, the identification of transplanted patients with a higher risk of posttransplant complications has become a big challenge for public health. However, current biomarkers of posttransplant complications have a poor predictive value, rising the need to explore novel approaches for the management of transplant patient. In this review we summarize the emerging literature about DNA methylation in kidney transplant complications, in order to highlight its perspectives toward biomarker identification. In the forthcoming future the monitoring of DNA methylation in kidney transplant patients could become a plausible strategy toward the prevention and/or treatment of kidney transplant complications.
Collapse
|
18
|
Eshraghi RS, Deth RC, Mittal R, Aranke M, Kay SIS, Moshiree B, Eshraghi AA. Early Disruption of the Microbiome Leading to Decreased Antioxidant Capacity and Epigenetic Changes: Implications for the Rise in Autism. Front Cell Neurosci 2018; 12:256. [PMID: 30158857 PMCID: PMC6104136 DOI: 10.3389/fncel.2018.00256] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 07/27/2018] [Indexed: 12/20/2022] Open
Abstract
Currently, 1 out of every 59 children in the United States is diagnosed with autism. While initial research to find the possible causes for autism were mostly focused on the genome, more recent studies indicate a significant role for epigenetic regulation of gene expression and the microbiome. In this review article, we examine the connections between early disruption of the developing microbiome and gastrointestinal tract function, with particular regard to susceptibility to autism. The biological mechanisms that accompany individuals with autism are reviewed in this manuscript including immune system dysregulation, inflammation, oxidative stress, metabolic and methylation abnormalities as well as gastrointestinal distress. We propose that these autism-associated biological mechanisms may be caused and/or sustained by dysbiosis, an alteration to the composition of resident commensal communities relative to the community found in healthy individuals and its redox and epigenetic consequences, changes that in part can be due to early use and over-use of antibiotics across generations. Further studies are warranted to clarify the contribution of oxidative stress and gut microbiome in the pathophysiology of autism. A better understanding of the microbiome and gastrointestinal tract in relation to autism will provide promising new opportunities to develop novel treatment modalities.
Collapse
Affiliation(s)
- Rebecca S. Eshraghi
- Division of Gastroenterology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Richard C. Deth
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Rahul Mittal
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Mayank Aranke
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Sae-In S. Kay
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Baharak Moshiree
- Division of Gastroenterology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Adrien A. Eshraghi
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
19
|
Breglia A, Virzì GM, Pastori S, Brocca A, de Cal M, Bolin C, Vescovo G, Ronco C. Determinants of Monocyte Apoptosis in Cardiorenal Syndrome Type 1. Cardiorenal Med 2018; 8:208-216. [PMID: 29847820 DOI: 10.1159/000488949] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 03/22/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Cardiorenal syndrome type 1 (CRS type 1) is characterized by a rapid worsening of cardiac function leading to acute kidney injury (AKI). Its pathophysiology is complex and not completely understood. In this study, we examined the role of apoptosis and the caspase pathways involved. MATERIAL AND METHODS We enrolled 40 acute heart failure (AHF) patients, 11 of whom developed AKI characterizing CRS type 1. We exposed the human cell line U937 to plasma from the CRS type 1 and AHF groups and then we evaluated apoptotic activity by annexin-V evaluation, determination of caspase-3, -8 and -9 levels, and BAX, BAD, and FAS gene expression. RESULTS We observed significant upregulation of apoptosis in monocytes exposed to CRS type 1 plasma compared to AHF, with increased levels of caspase-3 (p < 0.01), caspase-9 (p < 0.01), and caspase-8 (p < 0.03) showing activation of both intrinsic and extrinsic pathways. Furthermore, monocytes exposed to CRS type 1 plasma had increased gene expression of BAX and BAD (intrinsic pathways) (p = 0.010 for both). Furthermore, strong significant correlations between the caspase-9 levels and BAD and BAX gene expression were observed (Spearman ρ = - 0.76, p = 0.011, and ρ = - 0.72, p = 0.011). CONCLUSION CRS type 1 induces dual apoptotic pathway activation in monocytes; the two pathways converged on caspase-3. Many factors may induce activation of both intrinsic and extrinsic apoptotic pathways in CRS type 1 patients, such as upregulation of proinflammatory cytokines and hypoxia/ischemia. Further investigations are necessary to corroborate the present findings, and to better understand the pathophysiological mechanism and consequent therapeutic and prognostic implications for CRS type 1.
Collapse
Affiliation(s)
- Andrea Breglia
- Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, Vicenza, Italy.,IRRIV - International Renal Research Institute Vicenza, Vicenza, Italy.,Department of Internal Medicine, University of Trieste, Trieste, Italy
| | - Grazia Maria Virzì
- Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, Vicenza, Italy.,IRRIV - International Renal Research Institute Vicenza, Vicenza, Italy
| | - Silvia Pastori
- Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, Vicenza, Italy.,IRRIV - International Renal Research Institute Vicenza, Vicenza, Italy
| | - Alessandra Brocca
- Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, Vicenza, Italy.,IRRIV - International Renal Research Institute Vicenza, Vicenza, Italy.,Department of Medicine DIMED, University of Padua Medical School, Padua, Italy
| | - Massimo de Cal
- Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, Vicenza, Italy.,IRRIV - International Renal Research Institute Vicenza, Vicenza, Italy
| | - Chiara Bolin
- Internal Medicine, San Bortolo Hospital, Vicenza, Italy
| | - Giorgio Vescovo
- Internal Medicine, San Bortolo Hospital, Vicenza, Italy.,Internal Medicine Unit, Sant'Antonio Hospital, Padua, Italy
| | - Claudio Ronco
- Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, Vicenza, Italy.,IRRIV - International Renal Research Institute Vicenza, Vicenza, Italy
| |
Collapse
|
20
|
|