1
|
Wang X, Kang X, Li B, Chen C, Chen L, Liu D. High Glucose Treatment Induces Nuclei Aggregation of Microvascular Endothelial Cells via the foxo1a- klf2a Pathway. Arterioscler Thromb Vasc Biol 2025; 45:398-411. [PMID: 39882604 PMCID: PMC11856006 DOI: 10.1161/atvbaha.124.321719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Hyperglycemia is a major contributor to endothelial dysfunction and blood vessel damage, leading to severe diabetic microvascular complications. Despite the growing body of research on the underlying mechanisms of endothelial cell (EC) dysfunction, the available drugs based on current knowledge fall short of effectively alleviating these complications. Therefore, our endeavor to explore novel insights into the cellular and molecular mechanisms of endothelial dysfunction is crucial for the field. METHODS In this study, we performed a high-resolution imaging and time-lapse imaging analysis of the behavior of ECs in Tg(kdrl:ras-mCherry::fli1a:nGFP) zebrafish embryos upon high glucose treatment. Genetic manipulation and chemical biology approaches were utilized to analyze the underlying mechanism of high glucose-induced nuclei aggregation and aberrant migration of zebrafish ECs and cultured human ECs. Bioinformatical analysis of single-cell RNA-sequencing data and molecular biological techniques was performed to identify the target genes of foxo1a. RESULTS In this study, we observed that the high glucose treatment resulted in nuclei aggregation of ECs in zebrafish intersegmental vessels. Additionally, the aberrant migration of microvascular ECs in high glucose-treated embryos, which might be a cause of nuclei aggregation, was discovered. High glucose induced aggregation of vascular endothelial nuclei via foxo1a downregulation in zebrafish embryos. Then, we revealed that high glucose resulted in the downregulation of foxo1a expression and increased the expression of its direct downstream effector, klf2a, through which the aberrant migration and aggregation of vascular endothelial nuclei were caused. CONCLUSIONS High glucose treatment caused the nuclei of ECs to aggregate in vivo, which resembles the crowded nuclei of ECs in microaneurysms. High glucose suppresses foxo1a expression and increases the expression of its downstream effector, klf2a, thereby causing the aberrant migration and aggregation of vascular endothelial nuclei. Our findings provide a novel insight into the mechanism of microvascular complications in hyperglycemia.
Collapse
Affiliation(s)
- Xiaoning Wang
- Research Center of Clinical Medicine, Affiliated Hospital (X.W., D.L.), Nantong University, China
| | - Xinyi Kang
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Nantong University, China (X.K., L.C., D.L.)
| | - Bowen Li
- School of Life Science, Nantong Laboratory of Development and Diseases, The Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration (B.L., C.C., D.L.), Nantong University, China
| | - Changsheng Chen
- School of Life Science, Nantong Laboratory of Development and Diseases, The Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration (B.L., C.C., D.L.), Nantong University, China
| | - Liping Chen
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Nantong University, China (X.K., L.C., D.L.)
| | - Dong Liu
- Research Center of Clinical Medicine, Affiliated Hospital (X.W., D.L.), Nantong University, China
- School of Life Science, Nantong Laboratory of Development and Diseases, The Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration (B.L., C.C., D.L.), Nantong University, China
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Nantong University, China (X.K., L.C., D.L.)
| |
Collapse
|
2
|
Tajima H, Morikita N, Mukohda M, Nakamura S, Seki M, Imai R, Saito F, Mizuno R, Ozaki H. Enhanced vascular contraction induced by exposure to angiotensin II mediated by endothelin-1 biosynthesis following PKCβ activation. Am J Physiol Heart Circ Physiol 2025; 328:H484-H495. [PMID: 39887322 DOI: 10.1152/ajpheart.00541.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 08/23/2024] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Protein kinase C (PKC) reportedly plays a role in the pathogenesis of many vascular dysfunction-related conditions. In this study, we investigated whether PKCβ is associated with vascular contractile changes induced by angiotensin II (Ang II) exposure. Long-term (24 h) treatment of rat aortae and mesenteric arteries in Ang II-containing culture medium enhanced 5-hydroxytryptamine (5-HT)-induced vascular contraction in a dose-dependent manner, in association with enhanced phosphorylation of PKCβ S660. Increased contraction induced by Ang II treatment was also observed in endothelium-denuded aorta. Enhanced contraction induced by Ang II was markedly diminished by the knockout of the PKCβ gene or treatment with LY333531 and CGP53353 (PKCβ inhibitors). Cycloheximide (protein synthesis inhibitor) blocked the Ang II-induced enhanced contraction. Gene expression profiling and real-time PCR analyses showed marked upregulation of endothelin-1 (ET-1) expression in Ang II-treated aorta but was not observed in PKCβ-knockout aorta. Ang II increased the secretion of ET-1 peptide in endothelium-intact and -denuded aortae. Ang II-induced enhancement of vascular contraction was diminished by BQ-123 (ETAR blocker). In vivo treatment with Ang II (250 ng/kg/min) for 7 days increased phosphorylation of PKCβ S660 in rat vascular tissue and increased the in vitro contractile responses to 5-HT and in vivo systolic blood pressure, but these changes were largely absent in PKCβ-knockout experiments. These data suggest that long-term exposure to Ang II increases vascular smooth muscle contraction and blood pressure elevation, mediated by activation of PKCβ and subsequent biosynthesis of ET-1 in vascular smooth muscle cells.NEW & NOTEWORTHY We studied the role of PKCβ in Ang II-induced hypertension using a rat model. Our study showed that PKCβ plays a key role in Ang II-induced vascular hypercontractility. Our results also suggest that transcriptional activation-mediated ET-1 expression in vascular smooth muscle is responsible for this vascular change as a downstream pathway of PKCβ activation, which leads to blood pressure elevation. This Ang II-PKCβ-ET-1 mechanism may affect vascular homeostasis in hypertension.
Collapse
MESH Headings
- Animals
- Angiotensin II/pharmacology
- Endothelin-1/metabolism
- Protein Kinase C beta/metabolism
- Protein Kinase C beta/genetics
- Vasoconstriction/drug effects
- Male
- Rats, Sprague-Dawley
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/enzymology
- Rats
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/metabolism
- Mesenteric Arteries/enzymology
- Mesenteric Arteries/physiopathology
- Phosphorylation
- Serotonin/metabolism
- Serotonin/pharmacology
- Enzyme Activation
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/enzymology
- Aorta, Thoracic/physiopathology
- Signal Transduction
- Vasoconstrictor Agents/pharmacology
Collapse
Affiliation(s)
- Hirotaka Tajima
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Nayu Morikita
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Masashi Mukohda
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Sho Nakamura
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Mihiro Seki
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Ryuya Imai
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Fumiyo Saito
- Department of Toxicology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Risuke Mizuno
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Hiroshi Ozaki
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| |
Collapse
|
3
|
Kleeberg A, Luft T, Golkowski D, Purrucker JC. Endothelial dysfunction in acute ischemic stroke: a review. J Neurol 2025; 272:143. [PMID: 39812851 PMCID: PMC11735568 DOI: 10.1007/s00415-025-12888-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND AND PURPOSE Endothelial dysfunction is considered an emerging therapeutic target to prevent complications during acute stroke and to prevent recurrent stroke. This review aims to provide an overview of the current knowledge on endothelial dysfunction, outline the diagnostic methods used to measure it and highlight the drugs currently being investigated for the treatment of endothelial dysfunction in acute ischemic stroke. METHODS The PubMed® and ClinicalTrials.gov electronic databases were searched for eligible articles/studies dealing with endothelial dysfunction and stroke. The references of the articles were screened to identify additional sources. The data were abstracted and summarized. FINDINGS AND DISCUSSION Endothelial dysfunction can be measured by serum biomarkers as well as by ultrasound or plethysmography techniques. Drugs targeting endothelial dysfunction include widely used agents such as angiotensin-converting enzyme inhibitors or isosorbide mononitrate, but also experimental therapies such as endothelial progenitor cells. CONCLUSION The role of endothelial dysfunction in acute ischemic stroke has been studied increasingly in recent years. It has been shown that there is a correlation between endothelial dysfunction and parenchymal hematoma after endovascular thrombectomy. Also, early clinical trials are conducted investigating, e.g., endothelial progenitor cells in the treatment of endothelial dysfunction in ischemic stroke. Current research focuses on the integration of novel markers of endothelial dysfunction into routine clinical practice to support decision making in the treatment of acute ischemic stroke.
Collapse
Affiliation(s)
- Antonia Kleeberg
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Thomas Luft
- Department of Oncology and Hematology, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel Golkowski
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jan C Purrucker
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
4
|
Wang T, Wu H, Shi X, Dai M, Liu Y. Aminoadipic acid aggravates atherosclerotic vascular inflammation through ROS/TXNIP/NLRP3 pathway, a harmful microbial metabolite reduced by paeonol. Int J Biochem Cell Biol 2024; 177:106678. [PMID: 39490917 DOI: 10.1016/j.biocel.2024.106678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/26/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
AIM Our previous study has found a differential microbial metabolite in atherosclerosis (AS) mice, aminoadipic acid (AAA), which was considered as a potential harmful metabolite. However, whether it can promote AS vascular inflammation and its mechanisms remain unclear. Paeonol (Pae) plays an anti-AS role by regulating the metabolic profile, but whether Pae exerts its antiatherogenic effect by reducing serum AAA levels is unknown. RESULTS The clinical trial results showed that the AS patients' serum AAA levels were higher than those healthy people'. Besides, AAA supplementation could increase aortic plaque size, serum inflammatory cytokines levels and liver malondialdehyde, superoxide dismutase levels in AS mice. Moreover, after AAA stimulation, the ROS levels and ASC, TXNIP, NLRP3 and caspase-1 proteins levels were increased in HUVECs, which could be reversed by antioxidant NAC and NLRP3 inhibitor. Pae significantly reduced the plaque size in the aorta, improved blood lipid levels and decreased serum inflammation factor levels in AS mice. Simultaneously, Pae could reduce the serum AAA levels of AS mice through the gut microbiota transmission. Finally, Pae inhibited NLRP3 inflammasome activation in aortas of AS mice. Broad-spectrum antibiotics could weaken the inhibitory effect of Pae on NLRP3 inflammasome. CONCLUSION Our study clarified that AAA could promote AS vascular inflammation via activating the ROS/TXNIP/NLRP3 pathway. Pae could inhibit AS development by reducing serum AAA levels in a microbiota-dependent manner. Taken together, we proposed that AAA could be served as a potential biomarker for AS clinical diagnosis and provided a new treatment strategy for AS.
Collapse
Affiliation(s)
- Tian Wang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei Anhui, 230012, China
| | - Hongfei Wu
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei Anhui, 230012, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei Anhui, 230012, China
| | - Xiaoyan Shi
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei Anhui, 230012, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei Anhui, 230012, China
| | - Min Dai
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei Anhui, 230012, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei Anhui, 230012, China
| | - Yarong Liu
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei Anhui, 230012, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei Anhui, 230012, China.
| |
Collapse
|
5
|
Datta S, Pasham S, Inavolu S, Boini KM, Koka S. Role of Gut Microbial Metabolites in Cardiovascular Diseases-Current Insights and the Road Ahead. Int J Mol Sci 2024; 25:10208. [PMID: 39337693 PMCID: PMC11432476 DOI: 10.3390/ijms251810208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of premature morbidity and mortality globally. The identification of novel risk factors contributing to CVD onset and progression has enabled an improved understanding of CVD pathophysiology. In addition to the conventional risk factors like high blood pressure, diabetes, obesity and smoking, the role of gut microbiome and intestinal microbe-derived metabolites in maintaining cardiovascular health has gained recent attention in the field of CVD pathophysiology. The human gastrointestinal tract caters to a highly diverse spectrum of microbes recognized as the gut microbiota, which are central to several physiologically significant cascades such as metabolism, nutrient absorption, and energy balance. The manipulation of the gut microbial subtleties potentially contributes to CVD, inflammation, neurodegeneration, obesity, and diabetic onset. The existing paradigm of studies suggests that the disruption of the gut microbial dynamics contributes towards CVD incidence. However, the exact mechanistic understanding of such a correlation from a signaling perspective remains elusive. This review has focused upon an in-depth characterization of gut microbial metabolites and their role in varied pathophysiological conditions, and highlights the potential molecular and signaling mechanisms governing the gut microbial metabolites in CVDs. In addition, it summarizes the existing courses of therapy in modulating the gut microbiome and its metabolites, limitations and scientific gaps in our current understanding, as well as future directions of studies involving the modulation of the gut microbiome and its metabolites, which can be undertaken to develop CVD-associated treatment options. Clarity in the understanding of the molecular interaction(s) and associations governing the gut microbiome and CVD shall potentially enable the development of novel druggable targets to ameliorate CVD in the years to come.
Collapse
Affiliation(s)
- Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Sindhura Pasham
- Department of Pharmaceutical Sciences, Irma Lerma College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| | - Sriram Inavolu
- Department of Pharmaceutical Sciences, Irma Lerma College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| | - Krishna M Boini
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Saisudha Koka
- Department of Pharmaceutical Sciences, Irma Lerma College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| |
Collapse
|
6
|
Tian B, Zhao C, Liang JL, Zhang HT, Xu YF, Zheng HL, Zhou J, Gong JN, Lu ST, Zeng ZS. Glymphatic function and its influencing factors in different glucose metabolism states. World J Diabetes 2024; 15:1537-1550. [PMID: 39099805 PMCID: PMC11292332 DOI: 10.4239/wjd.v15.i7.1537] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/13/2024] [Accepted: 06/11/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Dysfunction of the glymphatic system in the brain in different stages of altered glucose metabolism and its influencing factors are not well characterized. AIM To investigate the function of the glymphatic system and its clinical correlates in patients with different glucose metabolism states, the present study employed diffusion tensor imaging along the perivascular space (DTI-ALPS) index. METHODS Sample size was calculated using the pwr package in R software. This cross-sectional study enrolled 22 patients with normal glucose metabolism (NGM), 20 patients with prediabetes, and 22 patients with type 2 diabetes mellitus (T2DM). A 3.0T magnetic resonance imaging was used to evaluate the function of the glymphatic system. The mini-mental state examination (MMSE) was used to assess general cognitive function. The DTI-ALPS index of bilateral basal ganglia and the mean DTI-ALPS index was calculated. Further, the correlation between DTI-ALPS and clinical features was assessed. RESULTS The left-side, right-side, and mean DTI-ALPS index in the T2DM group were significantly lower than that in the NGM group. The right-side DTI-ALPS and mean DTI-ALPS index in the T2DM group were significantly lower than those in the prediabetes group. DTI-ALPS index lateralization was not observed. The MMSE score in the T2DM group was significantly lower than that in the NGM and prediabetes group. After controlling for sex, the left-side DTI-ALPS and mean DTI-ALPS index in the prediabetes group were positively correlated with 2-hour postprandial blood glucose level; the left-side DTI-ALPS index was negatively correlated with total cholesterol and low-density lipoprotein level. The right-side DTI-ALPS and mean DTI-ALPS index were negatively correlated with the glycosylated hemoglobin level and waist-to-hip ratio in the prediabetes group. The left-side, right-side, and mean DTI-ALPS index in the T2DM group were positively correlated with height. The left-side and mean DTI-ALPS index in the T2DM group were negatively correlated with high-density lipoprotein levels. CONCLUSION Cerebral glymphatic system dysfunction may mainly occur in the T2DM stage. Various clinical variables were found to affect the DTI-ALPS index in different glucose metabolism states. This study enhances our understanding of the pathophysiology of diabetic brain damage and provides some potential biological evidence for its early diagnosis.
Collapse
Affiliation(s)
- Bin Tian
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Chen Zhao
- Magnetic Resonance Research Collaboration, Siemens Healthineers, Guangzhou 510620, Guangdong Province, China
| | - Jia-Li Liang
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Hui-Ting Zhang
- Magnetic Resonance Research Collaboration, Siemens Healthineers Ltd., Wuhan 430071, Hubei Province, China
| | - Yi-Fan Xu
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Hui-Lei Zheng
- Department of Health Management, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Jia Zhou
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Jiang-Nian Gong
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Shu-Ting Lu
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Zi-San Zeng
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
7
|
Desai DA, Baby A, Ananthamohan K, Green LC, Arif M, Duncan BC, Kumar M, Singh RR, Koch SE, Natesan S, Rubinstein J, Jegga AG, Sadayappan S. Roles of cMyBP-C phosphorylation on cardiac contractile dysfunction in db/db mice. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 8:100075. [PMID: 38957358 PMCID: PMC11218625 DOI: 10.1016/j.jmccpl.2024.100075] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disease and comorbidity associated with several conditions, including cardiac dysfunction leading to heart failure with preserved ejection fraction (HFpEF), in turn resulting in T2DM-induced cardiomyopathy (T2DM-CM). However, the molecular mechanisms underlying the development of T2DM-CM are poorly understood. It is hypothesized that molecular alterations in myopathic genes induced by diabetes promote the development of HFpEF, whereas cardiac myosin inhibitors can rescue the resultant T2DM-mediated cardiomyopathy. To test this hypothesis, a Leptin receptor-deficient db/db homozygous (Lepr db/db) mouse model was used to define the pathogenesis of T2DM-CM. Echocardiographic studies at 4 and 6 months revealed that Lepr db/db hearts started developing cardiac dysfunction by four months, and left ventricular hypertrophy with diastolic dysfunction was evident at 6 months. RNA-seq data analysis, followed by functional enrichment, revealed the differential regulation of genes related to cardiac dysfunction in Lepr db/db heart tissues. Strikingly, the level of cardiac myosin binding protein-C phosphorylation was significantly increased in Lepr db/db mouse hearts. Finally, using isolated skinned papillary muscles and freshly isolated cardiomyocytes, CAMZYOS ® (mavacamten, MYK-461), a prescription heart medicine used for symptomatic obstructive hypertrophic cardiomyopathy treatment, was tested for its ability to rescue T2DM-CM. Compared with controls, MYK-461 significantly reduced force generation in papillary muscle fibers and cardiomyocyte contractility in the db/db group. This line of evidence shows that 1) T2DM-CM is associated with hyperphosphorylation of cardiac myosin binding protein-C and 2) MYK-461 significantly lessened disease progression in vitro, suggesting its promise as a treatment for HFpEF.
Collapse
Affiliation(s)
- Darshini A. Desai
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Akhil Baby
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai 625021, India
| | - Kalyani Ananthamohan
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Lisa C. Green
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mohammed Arif
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Brittany C. Duncan
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mohit Kumar
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Rohit R. Singh
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Sheryl E. Koch
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Sankar Natesan
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai 625021, India
| | - Jack Rubinstein
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Anil G. Jegga
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Sakthivel Sadayappan
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
8
|
Anees S, Ahmad M, Ashraf S, Bhat AH, Hamid R, Ganie SA. Bioactive fractions from Allium humile alleviate the risk of high fat diet induced atherosclerosis in albino Wistar rats by inhibiting protein kinase C. Fitoterapia 2024; 172:105775. [PMID: 38097019 DOI: 10.1016/j.fitote.2023.105775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Atherosclerosis is a global concern that worsens with age, and plants that are effective medicinal herbs can give a viable alternative. PKC is a key factor in cardiovascular and other disorders; targeting it can reduce the risk of these diseases. We evaluated Allium humile for PKC inhibition and therapeutic efficacy against atherosclerosis. Soxhlet extraction was done to obtain extracts (hexane, ethyl acetate, methanol, ethanol and aqueous) and then tested for DPPH radical scavenging and PKC inhibitory activity. The methanolic extract was more active than the other extracts, so it was subjected to column chromatography, and seventeen fractions were obtained. Only 11, 12, and 15 showed good activity against PKC. Wistar rats were divided into six groups and each group received high fat diet for 30 days. Then the three potent fractions (10 mg/kg) were administered for 15 days along with high fat diet. Fraction II had the highest effectiveness (P < 0.0001) in decreasing lipid levels, lipid peroxidation, reducing IL-6 and TNF-α expression, and raising nitric oxide. This also demonstrated a decrease in PKC activity, as well as a decrease in the formation of the lipoidal layer in the aorta wall and rupture of the intima and media as validated by histological analysis. The two compounds, phytol acetate and cyanidin 3-(6″-o-malonyllaminaribioside) were characterised in fraction II by NMR and HRMS and cyanidin 3-(6″-o-malonyllaminaribioside) inhibited PKC more efficiently. Thus, Allium humile has strong anti-atherogenic activity as well as the ability to inhibit PKC both in vitro and in vivo.
Collapse
Affiliation(s)
- Suhail Anees
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, India
| | - Muzaffar Ahmad
- Department of Biochemistry, University of Kashmir, Srinagar, India
| | - Suhail Ashraf
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, India
| | | | - Rabia Hamid
- Department of Nanotechnology, University of Kashmir, Srinagar, India.
| | - Showkat Ahmad Ganie
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, India.
| |
Collapse
|
9
|
Tang J, Li T, Xiong X, Yang Q, Su Z, Zheng M, Chen Q. Colchicine delivered by a novel nanoparticle platform alleviates atherosclerosis by targeted inhibition of NF-κB/NLRP3 pathways in inflammatory endothelial cells. J Nanobiotechnology 2023; 21:460. [PMID: 38037046 PMCID: PMC10690998 DOI: 10.1186/s12951-023-02228-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023] Open
Abstract
Atherosclerosis, a chronic inflammatory disease characterized by arterial plaque formation, is one of the most prominent causes of cardiovascular diseases. However, the current treatments often do not adequately compromise the chronic inflammation-mediated plaque accumulation and the disease progression. Therefore, a new and effective strategy that blocks atherosclerosis-associated inflammation is urgently needed to further reduce the risk. Colchicine, a potent anti-inflammatory medication, has shown great potential in the treatment of atherosclerosis, but its adverse effects have hampered its clinical application. Herein, we developed a novel delivery nanosystem encapsulated with colchicine (VHPK-PLGA@COL), which exhibited improved biosafety and sustained drug release along with the gradual degradation of PLGA and PEG as confirmed both in vitro and in vivo. Surface modification of the nanoparticles with the VHPK peptide ensured its capability to specifically target inflammatory endothelial cells and alleviate atherosclerotic plaque accumulation. In the ApoE - / - atherosclerotic mouse model, both colchicine and VHPK-PLGA@COL treatment significantly decreased the plaque area and enhanced plaque stability by blocking the NF-κB/NLRP3 pathways, while VHPK-PLGA@COL exhibited enhanced therapeutic effects due to its unique ability to target inflammatory endothelial cells without obvious long-term safety concerns. In summary, VHPK-PLGA@COL has the potential to overcome the key translational barriers of colchicine and open new avenues to repurpose this drug for anti-atherosclerotic therapy.
Collapse
Affiliation(s)
- Juan Tang
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Department of Endocrinology, The First People's Hospital of Ziyang, Sichuan, 641300, China
| | - Tao Li
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Department of Ophthalmology, The First People's Hospital of Ziyang, Sichuan, 641300, China
| | - Xiaojing Xiong
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Qiaoyun Yang
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Zedazhong Su
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Minming Zheng
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| | - Qingwei Chen
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
10
|
Qin P, He C, Ye P, Li Q, Cai C, Li Y. PKCδ regulates the vascular biology in diabetic atherosclerosis. Cell Commun Signal 2023; 21:330. [PMID: 37974282 PMCID: PMC10652453 DOI: 10.1186/s12964-023-01361-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023] Open
Abstract
Diabetes mellitus, known for its complications, especially vascular complications, is becoming a globally serious social problem. Atherosclerosis has been recognized as a common vascular complication mechanism in diabetes. The diacylglycerol (DAG)-protein kinase C (PKC) pathway plays an important role in atherosclerosis. PKCs can be divided into three subgroups: conventional PKCs (cPKCs), novel PKCs (nPKCs), and atypical PKCs (aPKCs). The aim of this review is to provide a comprehensive overview of the role of the PKCδ pathway, an isoform of nPKC, in regulating the function of endothelial cells, vascular smooth muscle cells, and macrophages in diabetic atherosclerosis. In addition, potential therapeutic targets regarding the PKCδ pathway are summarized. Video Abstract.
Collapse
Affiliation(s)
- Peiliang Qin
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Changhuai He
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Pin Ye
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qin Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chuanqi Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
11
|
Zhu J, Lyu J, Zhao R, Liu G, Wang S. Gut macrobiotic and its metabolic pathways modulate cardiovascular disease. Front Microbiol 2023; 14:1272479. [PMID: 37822750 PMCID: PMC10562559 DOI: 10.3389/fmicb.2023.1272479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/12/2023] [Indexed: 10/13/2023] Open
Abstract
Thousands of microorganisms reside in the human gut, and extensive research has demonstrated the crucial role of the gut microbiota in overall health and maintaining homeostasis. The disruption of microbial populations, known as dysbiosis, can impair the host's metabolism and contribute to the development of various diseases, including cardiovascular disease (CVD). Furthermore, a growing body of evidence indicates that metabolites produced by the gut microbiota play a significant role in the pathogenesis of cardiovascular disease. These bioactive metabolites, such as short-chain fatty acids (SCFAs), trimethylamine (TMA), trimethylamine N-oxide (TMAO), bile acids (BAs), and lipopolysaccharides (LPS), are implicated in conditions such as hypertension and atherosclerosis. These metabolites impact cardiovascular function through various pathways, such as altering the composition of the gut microbiota and activating specific signaling pathways. Targeting the gut microbiota and their metabolic pathways represents a promising approach for the prevention and treatment of cardiovascular diseases. Intervention strategies, such as probiotic drug delivery and fecal transplantation, can selectively modify the composition of the gut microbiota and enhance its beneficial metabolic functions, ultimately leading to improved cardiovascular outcomes. These interventions hold the potential to reshape the gut microbial community and restore its balance, thereby promoting cardiovascular health. Harnessing the potential of these microbial metabolites through targeted interventions offers a novel avenue for tackling cardiovascular health issues. This manuscript provides an in-depth review of the recent advances in gut microbiota research and its impact on cardiovascular health and offers a promising avenue for tackling cardiovascular health issues through gut microbiome-targeted therapies.
Collapse
Affiliation(s)
- Junwen Zhu
- Department of Cardiology, The Affiliated Wenling Hospital of Wenzhou Medical University (The First People’s Hospital of Wenling), Zhejiang, China
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China
| | - Jin Lyu
- Department of Pathology, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Ruochi Zhao
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Affiliated First Hospital of Ningbo University, Ningbo, China
| | - Gang Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China
| | - Shuangshuang Wang
- Department of Cardiology, The Affiliated Wenling Hospital of Wenzhou Medical University (The First People’s Hospital of Wenling), Zhejiang, China
| |
Collapse
|
12
|
An Y, Xu BT, Wan SR, Ma XM, Long Y, Xu Y, Jiang ZZ. The role of oxidative stress in diabetes mellitus-induced vascular endothelial dysfunction. Cardiovasc Diabetol 2023; 22:237. [PMID: 37660030 PMCID: PMC10475205 DOI: 10.1186/s12933-023-01965-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/14/2023] [Indexed: 09/04/2023] Open
Abstract
Diabetes mellitus is a metabolic disease characterized by long-term hyperglycaemia, which leads to microangiopathy and macroangiopathy and ultimately increases the mortality of diabetic patients. Endothelial dysfunction, which has been recognized as a key factor in the pathogenesis of diabetic microangiopathy and macroangiopathy, is characterized by a reduction in NO bioavailability. Oxidative stress, which is the main pathogenic factor in diabetes, is one of the major triggers of endothelial dysfunction through the reduction in NO. In this review, we summarize the four sources of ROS in the diabetic vasculature and the underlying molecular mechanisms by which the pathogenic factors hyperglycaemia, hyperlipidaemia, adipokines and insulin resistance induce oxidative stress in endothelial cells in the context of diabetes. In addition, we discuss oxidative stress-targeted interventions, including hypoglycaemic drugs, antioxidants and lifestyle interventions, and their effects on diabetes-induced endothelial dysfunction. In summary, our review provides comprehensive insight into the roles of oxidative stress in diabetes-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Ying An
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, 646000, China
| | - Bu-Tuo Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, 646000, China
| | - Sheng-Rong Wan
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, 646000, China
| | - Xiu-Mei Ma
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, 646000, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, China
| | - Yang Long
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, 646000, China
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, 646000, China.
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, 646000, China.
| | - Zong-Zhe Jiang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, 646000, China.
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
13
|
Xiao Q, Wang D, Li D, Huang J, Ma F, Zhang H, Sheng Y, Zhang C, Ha X. Protein kinase C: A potential therapeutic target for endothelial dysfunction in diabetes. J Diabetes Complications 2023; 37:108565. [PMID: 37540984 DOI: 10.1016/j.jdiacomp.2023.108565] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/13/2023] [Accepted: 07/22/2023] [Indexed: 08/06/2023]
Abstract
Protein kinase C (PKC) is a family of serine/threonine protein kinases that play an important role in many organs and systems and whose activation contributes significantly to endothelial dysfunction in diabetes. The increase in diacylglycerol (DAG) under high glucose conditions mediates PKC activation and synthesis, which stimulates oxidative stress and inflammation, resulting in impaired endothelial cell function. This article reviews the contribution of PKC to the development of diabetes-related endothelial dysfunction and summarizes the drugs that inhibit PKC activation, with the aim of exploring therapeutic modalities that may alleviate endothelial dysfunction in diabetic patients.
Collapse
Affiliation(s)
- Qian Xiao
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Dan Wang
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Danyang Li
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Jing Huang
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Feifei Ma
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, Gansu, China
| | - Haocheng Zhang
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; The Second School of Clinical Medicine, Lanzhou University, Lanzhou, 730030, Gansu, China
| | - Yingda Sheng
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Caimei Zhang
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Xiaoqin Ha
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China.
| |
Collapse
|
14
|
Zhu B, Niu Y, Niu L, Zhang X, Liu F. Exploring the application of sildenafil for high-fat diet-induced erectile dysfunction based on interleukin-18-mediated NLRP3/Caspase-1 signaling pathway. Sex Med 2023; 11:qfad044. [PMID: 37636019 PMCID: PMC10460117 DOI: 10.1093/sexmed/qfad044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 08/29/2023] Open
Abstract
Background Inflammation is a key risk factor for heart disease and has also been linked to erectile dysfunction (ED). Sildenafil is a phosphodiesterase type 5 inhibitor with a strong antioxidant effect. Interleukin (IL)-18 is a proinflammatory factor. Excessive production and release of IL-18 disrupt the balance between IL-18 and IL-18 binding proteins in certain inflammatory diseases, leading to the occurrence of pathological inflammation. Aim We evaluated the effects of sildenafil on erectile function in a rat model of high-fat diet-induced ED. Methods Male Sprague Dawley rats (6 weeks old) were divided into 5 groups: control, ED, sildenafil, IL-18, and IL-18 + sildenafil. Subsequently, intracavernous pressure and mean arterial pressure were used to assess the erectile function of these rats. The expression of endothelial nitric oxide synthase, pyroptosis factors, and the ratio of smooth muscle cells and collagen fibers were evaluated in the serum and corpora tissue. Outcomes Exploring the role and mechanism of sildenafil in ED through NLRP3-mediated pyroptosis pathway. Results In comparison to the ED and IL-18 groups, there were statistically significant increases in the ratio of intracavernous pressure to mean arterial pressure, endothelial nitric oxide synthase expression, and the ratio of smooth muscle cells to collagen fibers following sildenafil intervention (P < .05). The sildenafil group and IL-18 + sildenafil group also showed statistically significant decreases the expression of NLRP3, caspase-1, and gasdermin D (P < .05). Clinical Implications Sildenafil can improve erectile dysfunction by inhibiting inflammation. Strengths and Limitations Strengths are that the relationship between pyroptosis and ED has been verified through in vitro and in vivo experiments. The limitation is that the conclusions drawn from animal and cells experiments need to be confirmed in clinical research. Conclusion Sildenafil may reduce the effect of IL-18-induced inflammation in high-fat diet-induced ED rats through NLRP3/caspase-1 pyroptosis pathway.
Collapse
Affiliation(s)
- Bingbing Zhu
- Department of Human Anatomy, School of Basic Medical Science, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, 830011, China
| | - Yangjiu Niu
- Department of Human Anatomy, School of Basic Medical Science, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, 830011, China
| | - Lipan Niu
- Department of Human Anatomy, School of Basic Medical Science, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, 830011, China
| | - Xijia Zhang
- Department of Human Anatomy, School of Basic Medical Science, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, 830011, China
| | - Fengxia Liu
- Department of Human Anatomy, School of Basic Medical Science, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, 830011, China
| |
Collapse
|
15
|
Han S, Cai L, Chen P, Kuang W. A study of the correlation between stroke and gut microbiota over the last 20years: a bibliometric analysis. Front Microbiol 2023; 14:1191758. [PMID: 37350780 PMCID: PMC10282156 DOI: 10.3389/fmicb.2023.1191758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
Purpose This study intends to uncover a more thorough knowledge structure, research hotspots, and future trends in the field by presenting an overview of the relationship between stroke and gut microbiota in the past two decades. Method Studies on stroke and gut microbiota correlations published between 1st January 2002 and 31st December 2021 were retrieved from the Web of Science Core Collection and then visualized and scientometrically analyzed using CiteSpace V. Results A total of 660 papers were included in the study, among which the United States, the United Kingdom, and Germany were the leading research centers. Cleveland Clinic, Southern Medical University, and Chinese Academy of Science were the top three institutions. The NATURE was the most frequently co-cited journal. STANLEY L HAZEN was the most published author, and Tang WHW was the most cited one. The co-occurrence analysis revealed eight clusters (i.e., brain-gut microbiota axis, fecal microbiome transplantation, gut microbiota, hypertension, TMAO, ischemic stroke, neuroinflammation, atopobiosis). "gut microbiota," "Escherichia coli," "cardiovascular disease," "risk," "disease," "ischemic stroke," "stroke," "metabolism," "inflammation," and "phosphatidylcholine" were the most recent keyword explosions. Conclusion Findings suggest that in the next 10 years, the number of publications produced annually may increase significantly. Future research trends tend to concentrate on the mechanisms of stroke and gut microbiota, with the inflammation and immunological mechanisms, TMAO, and fecal transplantation as hotspots. And the relationship between these mechanisms and a particular cardiovascular illness may also be a future research trend.
Collapse
Affiliation(s)
- Shengnan Han
- Clinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Longhui Cai
- First School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peipei Chen
- School of Medical Technology, Qiqihar Medical College, Qiqihar, Heilongjiang, China
| | - Weihong Kuang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
| |
Collapse
|
16
|
Shanmugham M, Bellanger S, Leo CH. Gut-Derived Metabolite, Trimethylamine-N-oxide (TMAO) in Cardio-Metabolic Diseases: Detection, Mechanism, and Potential Therapeutics. Pharmaceuticals (Basel) 2023; 16:ph16040504. [PMID: 37111261 PMCID: PMC10142468 DOI: 10.3390/ph16040504] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/17/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Trimethylamine N-oxide (TMAO) is a biologically active gut microbiome-derived dietary metabolite. Recent studies have shown that high circulating plasma TMAO levels are closely associated with diseases such as atherosclerosis and hypertension, and metabolic disorders such as diabetes and hyperlipidemia, contributing to endothelial dysfunction. There is a growing interest to understand the mechanisms underlying TMAO-induced endothelial dysfunction in cardio-metabolic diseases. Endothelial dysfunction mediated by TMAO is mainly driven by inflammation and oxidative stress, which includes: (1) activation of foam cells; (2) upregulation of cytokines and adhesion molecules; (3) increased production of reactive oxygen species (ROS); (4) platelet hyperreactivity; and (5) reduced vascular tone. In this review, we summarize the potential roles of TMAO in inducing endothelial dysfunction and the mechanisms leading to the pathogenesis and progression of associated disease conditions. We also discuss the potential therapeutic strategies for the treatment of TMAO-induced endothelial dysfunction in cardio-metabolic diseases.
Collapse
Affiliation(s)
- Meyammai Shanmugham
- Science, Math & Technology, Singapore University of Technology & Design, 8 Somapah Road, Singapore 487372, Singapore
| | - Sophie Bellanger
- A*STAR Skin Research Labs, Agency for Science, Technology and Research, Singapore 138648, Singapore
| | - Chen Huei Leo
- Science, Math & Technology, Singapore University of Technology & Design, 8 Somapah Road, Singapore 487372, Singapore
- Correspondence: ; Tel.: +65-6434-8213
| |
Collapse
|
17
|
Al Samarraie A, Pichette M, Rousseau G. Role of the Gut Microbiome in the Development of Atherosclerotic Cardiovascular Disease. Int J Mol Sci 2023; 24:ijms24065420. [PMID: 36982492 PMCID: PMC10051145 DOI: 10.3390/ijms24065420] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the primary cause of death globally, with nine million deaths directly attributable to ischemic heart diseases in 2020. Since the last few decades, great effort has been put toward primary and secondary prevention strategies through identification and treatment of major cardiovascular risk factors, including hypertension, diabetes, dyslipidemia, smoking, and a sedentary lifestyle. Once labelled “the forgotten organ”, the gut microbiota has recently been rediscovered and has been found to play key functions in the incidence of ASCVD both directly by contributing to the development of atherosclerosis and indirectly by playing a part in the occurrence of fundamental cardiovascular risk factors. Essential gut metabolites, such as trimethylamine N-oxide (TMAO), secondary bile acids, lipopolysaccharides (LPS), and short-chain fatty acids (SCFAs), have been associated with the extent of ischemic heart diseases. This paper reviews the latest data on the impact of the gut microbiome in the incidence of ASCVD.
Collapse
Affiliation(s)
- Ahmad Al Samarraie
- Internal Medicine Department, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada
| | - Maxime Pichette
- Cardiology Department, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada
| | - Guy Rousseau
- Centre de Biomédecine, CIUSSS-NÎM/Hôpital du Sacré-Cœur, Montréal, QC H4J 1C5, Canada
- Correspondence:
| |
Collapse
|
18
|
Yu H, Li L, Deng Y, Zhang G, Jiang M, Huang H, Li C, Lv Z, Zhou Y, Liu X. The relationship between the number of stenotic coronary arteries and the gut microbiome in coronary heart disease patients. Front Cell Infect Microbiol 2022; 12:903828. [PMID: 36093192 PMCID: PMC9458979 DOI: 10.3389/fcimb.2022.903828] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/09/2022] [Indexed: 12/02/2022] Open
Abstract
An increasing number of studies have shown that the gut microbiome plays an important role in the development of coronary heart disease (CHD). However, there are no clear studies on the relationship between the gut microbiome and the number of stenotic coronary arteries. To clarify whether the gut microbiome is associated with the number of stenotic coronary arteries in CHD, we performed the 16S rRNA gene sequencing for the V3-V4 region in the gut microbiota from 9 healthy controls (C) and 36 CHD patients, which including 25 CHD patients with multivessel (MV) lesion and 11 CHD patients with single-vessel (SV) lesion. It showed that the abundance of the genus Escherichia-Shigella was significantly increased in the MV and SV groups compared with C group, while the abundance of the genera Subdoligranulum and Collinsella was significantly decreased. Biomarkers based on three gut microbiotas (Escherichia-Shigella, Subdoligranulum, and Collinsella) and three plasma metabolites(left atrial diameter (LA), low density lipoprotein (LDL), and total bile acids (TBA)) were able to distinguish CHD patients with different numbers of stenotic coronary arteries. Functional prediction of the gut microbiome was performed based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. The results showed that the gut microbial function of MV and SV group patients was richer than C group in betaine biosynthesis and unsaturated fatty acid biosynthesis, in the contrast less than C group in sphingolipid metabolism and primary bile acid biosynthesis. In summary, our study showed that the composition and function of the gut microbiome changed significantly from healthy controls to CHD patients with different numbers of coronary lesions.
Collapse
Affiliation(s)
- Hao Yu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Le Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yu Deng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Guolan Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Mimi Jiang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - He Huang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Cheng Li
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zhiyu Lv
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yingshun Zhou
- Department of Pathogen Biology, The public platform of the Pathogen Biology technology, School of Basic Medicine, Southwest Medical University, Luzhou, China
- *Correspondence: Xing Liu, ; Yingshun Zhou,
| | - Xing Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province; Southwest Medical University, Luzhou, China
- *Correspondence: Xing Liu, ; Yingshun Zhou,
| |
Collapse
|
19
|
Jubaidi FF, Zainalabidin S, Taib IS, Abdul Hamid Z, Mohamad Anuar NN, Jalil J, Mohd Nor NA, Budin SB. The Role of PKC-MAPK Signalling Pathways in the Development of Hyperglycemia-Induced Cardiovascular Complications. Int J Mol Sci 2022; 23:ijms23158582. [PMID: 35955714 PMCID: PMC9369123 DOI: 10.3390/ijms23158582] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/24/2022] [Accepted: 07/30/2022] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular disease is the most common cause of death among diabetic patients worldwide. Hence, cardiovascular wellbeing in diabetic patients requires utmost importance in disease management. Recent studies have demonstrated that protein kinase C activation plays a vital role in the development of cardiovascular complications via its activation of mitogen-activated protein kinase (MAPK) cascades, also known as PKC-MAPK pathways. In fact, persistent hyperglycaemia in diabetic conditions contribute to preserved PKC activation mediated by excessive production of diacylglycerol (DAG) and oxidative stress. PKC-MAPK pathways are involved in several cellular responses, including enhancing oxidative stress and activating signalling pathways that lead to uncontrolled cardiac and vascular remodelling and their subsequent dysfunction. In this review, we discuss the recent discovery on the role of PKC-MAPK pathways, the mechanisms involved in the development and progression of diabetic cardiovascular complications, and their potential as therapeutic targets for cardiovascular management in diabetic patients.
Collapse
Affiliation(s)
- Fatin Farhana Jubaidi
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
- Correspondence: (F.F.J.); (S.B.B.); Tel.: +603-9289-7645 (S.S.B.)
| | - Satirah Zainalabidin
- Center for Toxicology and Health Risk Research, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.Z.); (N.N.M.A.)
| | - Izatus Shima Taib
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
| | - Zariyantey Abdul Hamid
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
| | - Nur Najmi Mohamad Anuar
- Center for Toxicology and Health Risk Research, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.Z.); (N.N.M.A.)
| | - Juriyati Jalil
- Center for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Nor Anizah Mohd Nor
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
- Faculty of Health Sciences, University College MAIWP International, Kuala Lumpur 68100, Malaysia
| | - Siti Balkis Budin
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
- Correspondence: (F.F.J.); (S.B.B.); Tel.: +603-9289-7645 (S.S.B.)
| |
Collapse
|
20
|
Petersen C, Bharat D, Wankhade UD, Kim JS, Cutler BR, Denetso C, Gholami S, Nelson S, Bigley J, Johnson A, Chintapalli SV, Piccolo BD, Babu AKS, Paz HA, Shankar K, Symons JD, Babu PVA. Dietary Blueberry Ameliorates Vascular Complications in Diabetic Mice Possibly through NOX4 and Modulates Composition and Functional Diversity of Gut Microbes. Mol Nutr Food Res 2022; 66:e2100784. [PMID: 35120277 PMCID: PMC9132135 DOI: 10.1002/mnfr.202100784] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 01/27/2022] [Indexed: 11/05/2022]
Abstract
SCOPE In diabetes, endothelial inflammation and dysfunction play a pivotal role in the development of vascular disease. This study investigates the effect of dietary blueberries on vascular complications and gut microbiome in diabetic mice. METHODS AND RESULTS Seven-week-old diabetic db/db mice consume a standard diet (db/db) or a diet supplemented with 3.8% freeze-dried blueberry (db/db+BB) for 10 weeks. Control db/+ mice are fed a standard diet (db/+). Vascular inflammation is assessed by measuring monocyte binding to vasculature and inflammatory markers. Isometric tension procedures are used to assess mesenteric artery function. db/db mice exhibit enhanced vascular inflammation and reduced endothelial-dependent vasorelaxation as compared to db/+ mice, but these are improved in db/db+BB mice. Blueberry supplementation reduces the expression of NOX4 and IκKβ in the aortic vessel and vascular endothelial cells (ECs) isolated from db/db+BB compared to db/db mice. The blueberry metabolites serum reduces glucose and palmitate induced endothelial inflammation in mouse aortic ECs. Further, blueberry supplementation increases commensal microbes and modulates the functional potential of gut microbes in diabetic mice. CONCLUSION Dietary blueberry suppresses vascular inflammation, attenuates arterial endothelial dysfunction, and supports the growth of commensal microbes in diabetic mice. The endothelial-specific vascular benefits of blueberries are mediated through NOX4 signaling.
Collapse
Affiliation(s)
- Chrissa Petersen
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
| | - Divya Bharat
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
| | - Umesh D. Wankhade
- Arkansas Children’s Nutrition Center, Little Rock, AR 72205, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ji-Seok Kim
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
- Current address: Department of Physical Education & Research Institute of Pharmaceutical Sciences, Gyeongsang National University, South Korea
| | - Brett Ronald Cutler
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
| | - Christopher Denetso
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
| | - Samira Gholami
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
| | - Samantha Nelson
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
| | - Jessica Bigley
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
| | - Aspen Johnson
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
| | | | - Brian D. Piccolo
- Arkansas Children’s Nutrition Center, Little Rock, AR 72205, USA
| | | | - Henry A. Paz
- Arkansas Children’s Nutrition Center, Little Rock, AR 72205, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Kartik Shankar
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Current address: Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - J. David Symons
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
- Division of Endocrinology, Metabolism, and Diabetes; and Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Pon Velayutham Anandh Babu
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
| |
Collapse
|
21
|
Zhao L, Li Y, Xu T, Lv Q, Bi X, Liu X, Fu G, Zou Y, Ge J, Chen Z, Zhang W. Dendritic cell-mediated chronic low-grade inflammation is regulated by the RAGE-TLR4-PKCβ 1 signaling pathway in diabetic atherosclerosis. Mol Med 2022; 28:4. [PMID: 35062863 PMCID: PMC8780245 DOI: 10.1186/s10020-022-00431-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 01/03/2022] [Indexed: 11/16/2022] Open
Abstract
Background The unique mechanism of diabetic atherosclerosis has been a central research focus. Previous literature has reported that the inflammatory response mediated by dendritic cells (DCs) plays a vital role in the progression of atherosclerosis. The objective of the study was to explore the role of DCs in diabetes mellitus complicated by atherosclerosis. Methods ApoE−/− mice and bone marrow-derived DCs were used for in vivo and in vitro experiments, respectively. Masson’s staining and Oil-red-O staining were performed for atherosclerotic lesion assessment. The content of macrophages and DCs in plaque was visualized by immunohistochemistry. The expression of CD83 and CD86 were detected by flow cytometry. The fluctuations in the RNA levels of cytokines, chemokines, chemokine receptors and adhesions were analyzed by quantitative RT-PCR. The concentrations of IFN-γ and TNF-α were calculated using ELISA kits and the proteins were detected using western blot. Coimmunoprecipitation was used to detect protein–protein interactions. Results Compared with the ApoE−/− group, the volume of atherosclerotic plaques in the aortic root of diabetic ApoE−/− mice was significantly increased, numbers of macrophages and DCs were increased, and the collagen content in plaques decreased. The expression of CD83 and CD86 were significantly upregulated in splenic CD11c+ DCs derived from mice with hyperglycemia. Increased secretion of cytokines, chemokines, chemokine receptors, intercellular cell adhesion molecule (ICAM), and vascular cell adhesion molecule (VCAM) also were observed. The stimulation of advanced glycation end products plus oxidized low-density lipoprotein, in cultured BMDCs, further activated toll-like receptor 4, protein kinase C and receptor of AGEs, and induced immune maturation of DCs through the RAGE-TLR4-PKCβ1 signaling pathway that was bound together by intrinsic structures on the cell membrane. Administering LY333531 significantly increased the body weight of diabetic ApoE−/− mice, inhibited the immune maturation of spleen DCs, and reduced atherosclerotic plaques in diabetic ApoE−/− mice. Furthermore, the number of DCs and macrophages in atherosclerotic plaques was significantly reduced in the LY333531 group, and the collagen content was increased. Conclusions Diabetes mellitus aggravates chronic inflammation, and promotes atherosclerotic plaques in conjunction with hyperlipidemia, which at least in part through inducing the immune maturation of DCs, and its possible mechanism of action is through the RAGE-TLR4-pPKCβ1 signaling pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00431-6.
Collapse
Affiliation(s)
- Liding Zhao
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Ya Li
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Tian Xu
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Qingbo Lv
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Xukun Bi
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Xianglan Liu
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Guosheng Fu
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases of Zhongshan Hospital, Fudan University, Shanghai, China.,Institute of Biomedical Science, Fudan University, Shanghai, China
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases of Zhongshan Hospital, Fudan University, Shanghai, China.,Institute of Biomedical Science, Fudan University, Shanghai, China
| | - Zhaoyang Chen
- Heart Center of Fujian Province, Union Hospital, Fujian Medical University, 29 Xin-Quan Road, Fuzhou, 350001, People's Republic of China.
| | - Wenbin Zhang
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China. .,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
22
|
Ye S, Yousuf A, McVey D. Relationship between red meat metabolite trimethylamine N-oxide and cardiovascular disease. HEART AND MIND 2022. [DOI: 10.4103/hm.hm_8_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
23
|
Hasheminasabgorji E, Jha JC. Dyslipidemia, Diabetes and Atherosclerosis: Role of Inflammation and ROS-Redox-Sensitive Factors. Biomedicines 2021; 9:biomedicines9111602. [PMID: 34829831 PMCID: PMC8615779 DOI: 10.3390/biomedicines9111602] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 12/16/2022] Open
Abstract
The prevalence of diabetes is growing at an alarming rate with increased disability, morbidity, and often premature mortality because of the various complications of this disorder. Chronic hyperglycemia, dyslipidemia, and other metabolic alterations lead to the development and progression of macro- and microvascular complications of diabetes including cardiovascular, retinal and kidney disease. Despite advances in glucose and lipid lowering treatments, a large number of diabetic individuals develop one or more types of these complications, ultimately leading to end-organ damage over the time. Atherosclerosis is the major macro-vascular complications of diabetes and the primary underlying cause of cardiovascular disease (CVD) posing heavy burden on the health care system. In this review, we discuss the involvement of dyslipidemia in the progression of atherosclerosis by activating the pro-inflammatory cytokines and oxidative stress-related factors. In addition, we also provide information on various pharmacological agents that provides protection against diabetic atherosclerosis by reducing inflammation and oxidative stress.
Collapse
Affiliation(s)
- Elham Hasheminasabgorji
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari 4815733971, Iran;
| | - Jay C. Jha
- Department of Diabetes, Central Clinical School, Monash University, Melbourne 3004, Australia
- Correspondence:
| |
Collapse
|
24
|
Zheng Y, He JQ. Pathogenic Mechanisms of Trimethylamine N-Oxide-induced Atherosclerosis and Cardiomyopathy. Curr Vasc Pharmacol 2021; 20:29-36. [PMID: 34387163 DOI: 10.2174/1570161119666210812152802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/24/2021] [Accepted: 06/25/2021] [Indexed: 11/22/2022]
Abstract
Trimethylamine N-oxide (TMAO) is a gut microbiota metabolite derived from trimethylamine-containing nutrient precursors such as choline, L-carnitine, and betaine, which are rich in many vegetables, fruits, nuts, dairy products, and meats. An increasing number of clinical studies have demonstrated a strong relationship between elevated plasma TMAO levels and adverse cardiovascular events. It is commonly agreed that TMAO acts as both an independent risk factor and a prognostic index for patients with cardiovascular disease. Although most animal (mainly rodent) data support the clinical findings, the mechanisms by which TMAO modulates the cardiovascular system are still not well understood. In this context, we provide an overview of the potential mechanisms underlying TMAO-induced cardiovascular disease at the cellular and molecular levels, with a focus on atherosclerosis. We also address the direct effects of TMAO on cardiomyocytes (a new and under-researched area) and finally propose TMAO as a potential biomarker and/or therapeutic target for diagnosis and treatment of patients with cardiovascular disease.
Collapse
Affiliation(s)
- Youjing Zheng
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061. United States
| | - Jia-Qiang He
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061. United States
| |
Collapse
|
25
|
Zhang Y, Xu D, Huang P, Zhang Y, Li Q, Fan Z, Ren L. Essential role of protein kinase C βI in icariin-mediated protection against atherosclerosis. J Pharm Pharmacol 2021; 73:1169-1179. [PMID: 33822184 DOI: 10.1093/jpp/rgab040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 02/16/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVES This study aimed to clarify the superior beneficial effects of icariin on atherosclerosis, as well as to explore the possible underlying mechanisms for its effect via the modulation of protein kinase C βI. METHODS Lipid profiles were determined while dissected aortas were prepared of ApoE-/- mice. The expression of protein kinase C βI and phosphorylation of protein kinase C βI were determined by immunohistochemistry analysis. Human vascular smooth muscle cells were subjected to ox-LDL stimulation. MTS assay was conducted to detect cell proliferation. A transwell migration assay was performed to evaluate migration capacity. Flow cytometric analysis was used to determine cell cycle progression. Quantitative real-time PCR and western blot were performed to assess gene expression. RESULTS Icariin significantly alleviated atherogenesis, as well as protein levels of protein kinase C βI and phosphorylated protein kinase C βI in the aorta. Icariin effectively suppressed cell proliferation and migration. protein kinase C βI, cyclin D1 and matrix metalloproteinase-9 were modulated in response to treatment with icariin. Protein kinase C activator reversed the protective effect of icariin on human vascular smooth muscle cells against ox- low-density lipoprotein, protein kinase C β inhibitor augmented the inhibitory effect of icariin. CONCLUSIONS Our findings highlight the probable application of icariin in atherosclerotic therapy and reveal that protein kinase C βI acts as a crucial regulator in the anti-atherosclerotic action of icariin.
Collapse
MESH Headings
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Apolipoproteins E/metabolism
- Atherosclerosis/metabolism
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Cyclin D1/metabolism
- Epimedium/chemistry
- Flavonoids/pharmacology
- Humans
- Lipoproteins, LDL/metabolism
- Matrix Metalloproteinase 9/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Plant Extracts/pharmacology
- Protein Kinase C/metabolism
- Mice
Collapse
Affiliation(s)
- Yibing Zhang
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Dongsheng Xu
- Cancer Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Peng Huang
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, China
| | - Yang Zhang
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, China
| | - Qi Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, China
| | - Zhimin Fan
- Department of Breast Surgery, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Liqun Ren
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, China
| |
Collapse
|
26
|
Lien CF, Chen SJ, Tsai MC, Lin CS. Potential Role of Protein Kinase C in the Pathophysiology of Diabetes-Associated Atherosclerosis. Front Pharmacol 2021; 12:716332. [PMID: 34276388 PMCID: PMC8283198 DOI: 10.3389/fphar.2021.716332] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 06/22/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus is a metabolic syndrome that affects millions of people worldwide. Recent studies have demonstrated that protein kinase C (PKC) activation plays an important role in hyperglycemia-induced atherosclerosis. PKC activation is involved in several cellular responses such as the expression of various growth factors, activation of signaling pathways, and enhancement of oxidative stress in hyperglycemia. However, the role of PKC activation in pro-atherogenic and anti-atherogenic mechanisms remains controversial, especially under hyperglycemic condition. In this review, we discuss the role of different PKC isoforms in lipid regulation, oxidative stress, inflammatory response, and apoptosis. These intracellular events are linked to the pathogenesis of atherosclerosis in diabetes. PKC deletion or treatment with PKC inhibitors has been studied in the regulation of atherosclerotic plaque formation and evolution. Furthermore, some preclinical and clinical studies have indicated that PKCβ and PKCδ are potential targets for the treatment of diabetic vascular complications. The current review summarizes these multiple signaling pathways and cellular responses regulated by PKC activation and the potential therapeutic targets of PKC in diabetic complications.
Collapse
Affiliation(s)
- Chih-Feng Lien
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Sy-Jou Chen
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Min-Chien Tsai
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Chin-Sheng Lin
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
27
|
Sehgal P, Mathew S, Sivadas A, Ray A, Tanwar J, Vishwakarma S, Ranjan G, Shamsudheen KV, Bhoyar RC, Pateria A, Leonard E, Lalwani M, Vats A, Pappuru RR, Tyagi M, Jakati S, Sengupta S, B K B, Chakrabarti S, Kaur I, Motiani RK, Scaria V, Sivasubbu S. LncRNA VEAL2 regulates PRKCB2 to modulate endothelial permeability in diabetic retinopathy. EMBO J 2021; 40:e107134. [PMID: 34180064 PMCID: PMC8327952 DOI: 10.15252/embj.2020107134] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 05/16/2021] [Accepted: 05/21/2021] [Indexed: 12/29/2022] Open
Abstract
Long non‐coding RNAs (lncRNAs) are emerging as key regulators of endothelial cell function. Here, we investigated the role of a novel vascular endothelial‐associated lncRNA (VEAL2) in regulating endothelial permeability. Precise editing of veal2 loci in zebrafish (veal2gib005Δ8/+) induced cranial hemorrhage. In vitro and in vivo studies revealed that veal2 competes with diacylglycerol for interaction with protein kinase C beta‐b (Prkcbb) and regulates its kinase activity. Using PRKCB2 as bait, we identified functional ortholog of veal2 in humans from HUVECs and named it as VEAL2. Overexpression and knockdown of VEAL2 affected tubulogenesis and permeability in HUVECs. VEAL2 was differentially expressed in choroid tissue in eye and blood from patients with diabetic retinopathy, a disease where PRKCB2 is known to be hyperactivated. Further, VEAL2 could rescue the effects of PRKCB2‐mediated turnover of endothelial junctional proteins thus reducing hyperpermeability in hyperglycemic HUVEC model of diabetic retinopathy. Based on evidence from zebrafish and hyperglycemic HUVEC models and diabetic retinopathy patients, we report a hitherto unknown VEAL2 lncRNA‐mediated regulation of PRKCB2, for modulating junctional dynamics and maintenance of endothelial permeability.
Collapse
Affiliation(s)
- Paras Sehgal
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India.,Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Samatha Mathew
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India.,Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Ambily Sivadas
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India.,Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Arjun Ray
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India.,Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Jyoti Tanwar
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India.,Academy of Scientific and Innovative Research, Ghaziabad, India.,Laboratory of Calciomics and Systemic Pathophysiology, Regional Center for Biotechnology, Faridabad, India
| | - Sushma Vishwakarma
- Brien Holden Eye Research Centre, L V Prasad Eye Institute, Hyderabad, India
| | - Gyan Ranjan
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India.,Academy of Scientific and Innovative Research, Ghaziabad, India
| | - K V Shamsudheen
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India.,Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Rahul C Bhoyar
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Abhishek Pateria
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Elvin Leonard
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Mukesh Lalwani
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Archana Vats
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Rajeev R Pappuru
- Kannuri Santhamma Centre for Retina and Vitreous, L V Prasad Eye Institute, Hyderabad, India
| | - Mudit Tyagi
- Kannuri Santhamma Centre for Retina and Vitreous, L V Prasad Eye Institute, Hyderabad, India
| | - Saumya Jakati
- Brien Holden Eye Research Centre, L V Prasad Eye Institute, Hyderabad, India
| | - Shantanu Sengupta
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India.,Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Binukumar B K
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India.,Academy of Scientific and Innovative Research, Ghaziabad, India
| | | | - Inderjeet Kaur
- Brien Holden Eye Research Centre, L V Prasad Eye Institute, Hyderabad, India
| | - Rajender K Motiani
- Laboratory of Calciomics and Systemic Pathophysiology, Regional Center for Biotechnology, Faridabad, India
| | - Vinod Scaria
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India.,Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Sridhar Sivasubbu
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India.,Academy of Scientific and Innovative Research, Ghaziabad, India
| |
Collapse
|
28
|
Bahrami A, Sathyapalan T, Sahebkar A. The Role of Interleukin-18 in the Development and Progression of Atherosclerosis. Curr Med Chem 2021; 28:1757-1774. [PMID: 32338205 DOI: 10.2174/0929867327666200427095830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/14/2020] [Accepted: 04/28/2020] [Indexed: 11/22/2022]
Abstract
Atherosclerosis (AS), as a chronic inflammatory disorder of the cardiovascular system, is one of the leading causes of ischemic heart disease, stroke and peripheral vascular disease. There is growing evidence on the role of innate and adaptive immunity in the pathogenesis of atherosclerosis. Interleukin-18 is one of the novel proinflammatory cytokines involved in atherogenesis, atherosclerotic plaque instability and plaque rupture. In this review, we overview the findings of preclinical and clinical studies about the role and mechanism of action of IL-18 in the pathogenesis of AS, which could offer novel prognostic and therapeutic approaches.
Collapse
Affiliation(s)
- Afsane Bahrami
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, United Kingdom
| | | |
Collapse
|
29
|
Croteau L, Mercier C, Fafard-Couture É, Nadeau A, Robillard S, Breton V, Guay A, Lizotte F, Despatis MA, Geraldes P. Endothelial deletion of PKCδ prevents VEGF inhibition and restores blood flow reperfusion in diabetic ischemic limb. Diab Vasc Dis Res 2021; 18:1479164121999033. [PMID: 33722087 PMCID: PMC8481738 DOI: 10.1177/1479164121999033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
AIMS Peripheral artery disease is a complication of diabetes leading to critical hindlimb ischemia. Diabetes-induced inhibition of VEGF actions is associated with the activation of protein kinase Cδ (PKCδ). We aim to specifically investigate the role of PKCδ in endothelial cell (EC) function and VEGF signaling. METHODS Nondiabetic and diabetic mice, with (ec-Prkcd-/-) or without (ec-Prkcdf/f) endothelial deletion of PKCδ, underwent femoral artery ligation. Blood flow reperfusion was assessed up to 4 weeks post-surgery. Capillary density, EC apoptosis and VEGF signaling were evaluated in the ischemic muscle. Src homology region 2 domain-containing phosphatase-1 (SHP-1) phosphatase activity was assessed in vitro using primary ECs. RESULTS Ischemic muscle of diabetic ec-Prkcdf/f mice exhibited reduced blood flow reperfusion and capillary density while apoptosis increased as compared to nondiabetic ec-Prkcdf/f mice. In contrast, blood flow reperfusion and capillary density were significantly improved in diabetic ec-Prkcd-/- mice. VEGF signaling pathway was restored in diabetic ec-Prkcd-/- mice. The deletion of PKCδ in ECs prevented diabetes-induced VEGF unresponsiveness through a reduction of SHP-1 phosphatase activity. CONCLUSIONS Our data provide new highlights in mechanisms by which PKCδ activation in EC contributed to poor collateral vessel formation, thus, offering novel therapeutic targets to improve angiogenesis in the diabetic limb.
Collapse
Affiliation(s)
- Laura Croteau
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Clément Mercier
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Étienne Fafard-Couture
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Alexandre Nadeau
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Stéphanie Robillard
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Valérie Breton
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Andréanne Guay
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Farah Lizotte
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Marc-Antoine Despatis
- Department of Surgery of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Pedro Geraldes
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
- Department of Medicine, Division of Endocrinology, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
30
|
Endothelial response to glucose: dysfunction, metabolism, and transport. Biochem Soc Trans 2021; 49:313-325. [PMID: 33522573 DOI: 10.1042/bst20200611] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/23/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
The endothelial cell response to glucose plays an important role in both health and disease. Endothelial glucose-induced dysfunction was first studied in diabetic animal models and in cells cultured in hyperglycemia. Four classical dysfunction pathways were identified, which were later shown to result from the common mechanism of mitochondrial superoxide overproduction. More recently, non-coding RNA, extracellular vesicles, and sodium-glucose cotransporter-2 inhibitors were shown to affect glucose-induced endothelial dysfunction. Endothelial cells also metabolize glucose for their own energetic needs. Research over the past decade highlighted how manipulation of endothelial glycolysis can be used to control angiogenesis and microvascular permeability in diseases such as cancer. Finally, endothelial cells transport glucose to the cells of the blood vessel wall and to the parenchymal tissue. Increasing evidence from the blood-brain barrier and peripheral vasculature suggests that endothelial cells regulate glucose transport through glucose transporters that move glucose from the apical to the basolateral side of the cell. Future studies of endothelial glucose response should begin to integrate dysfunction, metabolism and transport into experimental and computational approaches that also consider endothelial heterogeneity, metabolic diversity, and parenchymal tissue interactions.
Collapse
|
31
|
Zhu H, Wang H, Jia Y, Cheng L, Cheng X. Increased serum calcium levels are associated with carotid atherosclerotic plaque in normocalcaemic individuals with type 2 diabetes. Ther Adv Endocrinol Metab 2021; 12:2042018821995369. [PMID: 33854752 PMCID: PMC8010831 DOI: 10.1177/2042018821995369] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 01/22/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Patients with type 2 diabetes mellitus (T2DM) have an elevated risk of atherosclerotic cardiovascular disease. Although previous data have suggested that serum calcium levels could be involved in T2DM and cardiovascular disease, whether this applies in T2DM patients with atherosclerosis remains unclear. This study therefore aimed to investigate the relationship between serum calcium levels within the physiological ranges and carotid atherosclerotic plaque in T2DM patients. METHODS A total of 594 normocalcaemic in-patients with T2DM were recruited, of whom 231 had carotid atherosclerotic plaque. Serum calcium levels were measured and carotid ultrasonography was performed. RESULTS Patients with plaque had significantly higher serum albumin-corrected calcium than those without plaque [9.02 (8.78-9.34) mg/dL versus 8.86 (8.66-9.06) mg/dL, p < 0.001]. As serum albumin-corrected calcium levels increased across tertiles, the percentage of plaque increased (27.6%, 35.5%, and 55.7%; p < 0.001). Logistic regression showed that serum albumin-corrected calcium levels were independently and positively correlated with the presence of plaque, but not parathyroid hormone levels. Compared with patients in the lowest serum calcium tertiles, the odds ratio for plaque in patients in the upper quartile was 2.47 (95% confidence interval 1.51-4.03, p < 0.001) after adjustment for potential confounders. CONCLUSION Serum albumin-corrected calcium levels are elevated in patients with T2DM and carotid atherosclerotic plaques.
Collapse
Affiliation(s)
- Huijing Zhu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Endocrinology & Metabolism, Heze Municipal Hospital, Heze, Shandong, China
| | - Huili Wang
- Department of Endocrinology & Metabolism, Heze Municipal Hospital, Heze, Shandong, China
| | | | - Lin Cheng
- Department of Endocrinology & Metabolism, Heze Municipal Hospital, Heze, Shandong, China
| | - Xingbo Cheng
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, Jiangsu 215006, China
| |
Collapse
|
32
|
Cai H, Wang P, Zhang B, Dong X. Expression of the NEK7/NLRP3 inflammasome pathway in patients with diabetic lower extremity arterial disease. BMJ Open Diabetes Res Care 2020; 8:8/2/e001808. [PMID: 33323459 PMCID: PMC7745315 DOI: 10.1136/bmjdrc-2020-001808] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION The NLRP3 inflammasome is closely related to diabetes and atherosclerosis. Recent studies suggest NIMA-related kinase 7 (NEK7) is necessary for NLRP3 inflammasome activation during potassium efflux. However, the expression of the NEK7/NLRP3 inflammasome pathway in diabetic lower extremity arterial disease (DLEAD) is unclear. The present study aimed to explore whether the NEK7/NLRP3 inflammasome pathway is involved in the pathogenesis of DLEAD. RESEARCH DESIGN AND METHODS The serum levels of interleukin-1β (IL-1β) and IL-18 in the control group (n=39), diabetes without lower extremity artery diseases group (n=39) and DLEAD group (n=85) were measured. H&E and Von Kossa staining were used to observe the vasculature of amputated feet from patients with diabetic foot. Furthermore, immunohistochemical staining, immunofluorescence and western blot were used to detect the expression of NEK7 and the NLRP3 inflammasome. RESULTS The serum IL-1β level in the DLEAD group was significantly increased compared with that in the control group and diabetes without lower extremity artery disease group. The serum IL-18 level was significantly higher in the DLEAD group and diabetes without lower extremity artery disease group than in the control group. H&E staining showed that the subintimal tissue of the arteries of patients with diabetic foot were highly thickened and exhibited irregular atherosclerotic plaques, and the arterial lumen was nearly occluded. Von Kossa staining showed dense brown-black calcium salt deposits in the vascular mesangium. Moreover, the expression of NEK7 and the NLRP3 inflammasome was significantly increased in the vascular cells of patients with diabetic foot, especially in vascular smooth muscle cells. CONCLUSION The NEK7/NLRP3 inflammasome pathway might be involved in the pathogenesis of DLEAD.
Collapse
Affiliation(s)
- HuaiQiu Cai
- Department of Ultrasonography, Fourth Hospital of Harbin Medical University, Harbin, China
| | - Peng Wang
- Department of Orthopeadic Surgery, Fourth Hospital of Harbin Medical University, Harbin, China
| | - Bo Zhang
- Department of Cardiology, Fourth Hospital of Harbin Medical University, Harbin, China
| | - XiaoQiu Dong
- Department of Ultrasonography, Fourth Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
33
|
Radujkovic A, Kordelas L, Bogdanov R, Müller-Tidow C, Beelen DW, Dreger P, Luft T. Interleukin-18 and Hematopoietic Recovery after Allogeneic Stem Cell Transplantation. Cancers (Basel) 2020; 12:cancers12102789. [PMID: 32998441 PMCID: PMC7601738 DOI: 10.3390/cancers12102789] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary We have previously shown that high pre-conditioning levels of Interleukin-18 were associated with worse survival after allogeneic stem cell transplantation due to increased non-relapse mortality. While no correlations with acute graft-versus-host disease were observed, interleukin-18-related excess mortality was mainly driven by fatal infectious complications. In multiple studies, delayed hematopoietic recovery and poor graft function following allogeneic stem cell transplantation has been demonstrated as a powerful predictor of non-relapse mortality. The present study links high interleukin-18 to delayed platelet recovery in allografted patients. Given the functions of interleukin-18 in regulating the quiescence of hematopoietic stem/progenitor cells, our findings may be explained by Interferon gamma-independent inhibitory effects of interleukin-18 on stem cell proliferation and hematopoietic reconstitution in allografted patients. Importantly, considering recent successful interleukin-18-neutralizing approaches in autoimmune disorders, our results provide a rationale to explore modulation of interleukin-18 for improving hematopoietic recovery and outcomes in allogeneic stem cell transplantation recipients. Abstract Interleukin-18 (IL-18) is an immunoregulatory cytokine and a context-dependent regulator of hematopoietic stem/progenitor cell (HSPC) quiescence in murine models. In a previous study, high pre-conditioning levels of IL-18 were associated with increased non-relapse mortality (NRM) after allogeneic stem cell transplantation (alloSCT). To investigate the clinical impact of IL-18 status on hematopoietic function, the associations of pre-conditioning and day 0–3 cytokine levels with platelet and neutrophil recovery were analyzed in a training cohort of 714 allografted patients. In adjusted logistic regression analyses, both increasing pre-conditioning and day 0–3 IL-18 levels had a significantly higher adjusted odds ratio (aOR) of delayed platelet and neutrophil recovery on day +28 post-transplant (aOR per two-fold increase: 1.6–2.0). The adverse impact of high pre-conditioning IL-18 on day +28 platelet recovery was verified in an independent cohort of 673 allografted patients (aOR per two-fold increase: 1.8 and 1.7 for total and free IL-18, respectively). In both cohorts, a platelet count ≤20/nL on day +28 was associated with a significantly increased hazard of NRM (hazard ratio 2.13 and 2.94, respectively). Our findings support the hypothesis that elevated peritransplant IL-18 levels affect post-transplant HSPC function and may provide a rationale to explore modulation of IL-18 for improving alloSCT outcomes.
Collapse
Affiliation(s)
- Aleksandar Radujkovic
- Department of Internal Medicine V, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.M.-T.); (P.D.); (T.L.)
- Correspondence:
| | - Lambros Kordelas
- Department of Bone Marrow Transplantation, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (L.K.); (R.B.); (D.W.B.)
| | - Rashit Bogdanov
- Department of Bone Marrow Transplantation, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (L.K.); (R.B.); (D.W.B.)
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.M.-T.); (P.D.); (T.L.)
| | - Dietrich W. Beelen
- Department of Bone Marrow Transplantation, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (L.K.); (R.B.); (D.W.B.)
| | - Peter Dreger
- Department of Internal Medicine V, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.M.-T.); (P.D.); (T.L.)
| | - Thomas Luft
- Department of Internal Medicine V, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.M.-T.); (P.D.); (T.L.)
| |
Collapse
|
34
|
Protective Effect of Astragaloside IV on High Glucose-Induced Endothelial Dysfunction via Inhibition of P2X7R Dependent P38 MAPK Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5070415. [PMID: 33014270 PMCID: PMC7512101 DOI: 10.1155/2020/5070415] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/10/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022]
Abstract
Vascular endothelial dysfunction is associated with increased mortality in patients with diabetes. Astragaloside IV (As-IV) is a bioactive saponin with therapeutic potential as an anti-inflammatory and antiendothelial dysfunction. However, the underlying mechanism for how As-IV ameliorated endothelial dysfunction is still unclear. Therefore, in this study, we examined the protective effect of As-IV against endothelial dysfunction and explored potential molecular biology mechanism. In vivo, rats were intraperitoneally injected with streptozotocin (STZ) at a dose of 65 mg/kg body weight to establish a diabetic model. In vitro studies, rat aortic endothelial cells (RAOEC) were pretreated with As-IV, SB203580 (p38 MAPK inhibitor) for 2 h prior to the addition of high glucose (33 mM glucose). Our findings indicated that As-IV improved impaired endothelium-dependent relaxation and increased the levels of endothelial NO synthase (eNOS) and nitric oxide (NO) both in vivo and in vitro. Besides, As-IV treatment inhibited the elevated inflammation and oxidative stress in diabetic model both in vivo and in vitro. Moreover, As-IV administration reversed the upregulated expression of P2X7R and p-p38 MAPK in vivo and in vitro. Additionally, the effects of both P2X7R siRNA and SB203580 on endothelial cells were similar to As-IV. Collectively, our study demonstrated that As-IV rescued endothelial dysfunction induced by high glucose via inhibition of P2X7R dependent p38 MAPK signaling pathway. This provides a theoretical basis for the further study of the vascular endothelial protective effects of As-IV.
Collapse
|
35
|
Pirri D, Fragiadaki M, Evans PC. Diabetic atherosclerosis: is there a role for the hypoxia-inducible factors? Biosci Rep 2020; 40:BSR20200026. [PMID: 32816039 PMCID: PMC7441368 DOI: 10.1042/bsr20200026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 07/28/2020] [Accepted: 08/07/2020] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis is a major cause of mortality worldwide and is driven by multiple risk factors, including diabetes. Diabetes is associated with either an insulin deficiency in its juvenile form or with insulin resistance and obesity in Type 2 diabetes mellitus, and the latter is clustered with other comorbidities to define the metabolic syndrome. Diabetes and metabolic syndrome are complex pathologies and are associated with cardiovascular risk via vascular inflammation and other mechanisms. Several transcription factors are activated upon diabetes-driven endothelial dysfunction and drive the progression of atherosclerosis. In particular, the hypoxia-inducible factor (HIF) transcription factor family is a master regulator of endothelial biology and is raising interest in the field of atherosclerosis. In this review, we will present an overview of studies contributing to the understanding of diabetes-driven atherosclerosis, integrating the role of HIF in this disease with the knowledge of its functions in metabolic syndrome and diabetic scenario.
Collapse
Affiliation(s)
- Daniela Pirri
- Department of Infection, Immunity and Cardiovascular disease, The University of Sheffield, U.K
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Maria Fragiadaki
- Department of Infection, Immunity and Cardiovascular disease, The University of Sheffield, U.K
| | - Paul C. Evans
- Department of Infection, Immunity and Cardiovascular disease, The University of Sheffield, U.K
| |
Collapse
|
36
|
Balzer MS, Helmke A, Ackermann M, Casper J, Dong L, Hiss M, Kiyan Y, Rong S, Timrott K, von Vietinghoff S, Wang L, Haller H, Shushakova N. Protein kinase C beta deficiency increases glucose-mediated peritoneal damage via M1 macrophage polarization and up-regulation of mesothelial protein kinase C alpha. Nephrol Dial Transplant 2020; 34:947-960. [PMID: 30247663 DOI: 10.1093/ndt/gfy282] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Peritoneal membrane (PM) damage during peritoneal dialysis (PD) is mediated largely by high glucose (HG)-induced pro-inflammatory and neo-angiogenic processes, resulting in PM fibrosis and ultrafiltration failure. We recently demonstrated a crucial role for protein kinase C (PKC) isoform α in mesothelial cells. METHODS In this study we investigate the role of PKCβ in PM damage in vitro using primary mouse peritoneal macrophages (MPMΦ), human macrophages (HMΦ) and immortalized mouse peritoneal mesothelial cells (MPMCs), as well as in vivo using a chronic PD mouse model. RESULTS We demonstrate that PKCβ is the predominant classical PKC isoform expressed in primary MPMΦ and its expression is up-regulated in vitro under HG conditions. After in vitro lipopolysaccharides stimulation PKCβ-/- MPMΦ demonstrates increased levels of interleukin 6 (IL-6), tumour necrosis factor α, and monocyte chemoattractant protein-1 and drastically decrease IL-10 release compared with wild-type (WT) cells. In vivo, catheter-delivered treatment with HG PD fluid for 5 weeks induces PKCβ up-regulation in omentum of WT mice and results in inflammatory response and PM damage characterized by fibrosis and neo-angiogenesis. In comparison to WT mice, all pathological changes are strongly aggravated in PKCβ-/- animals. Underlying molecular mechanisms involve a pro-inflammatory M1 polarization shift of MPMΦ and up-regulation of PKCα in MPMCs of PKCβ-/- mice. Finally, we demonstrate PKCβ involvement in HG-induced polarization processes in HMΦ. CONCLUSIONS PKCβ as the dominant PKC isoform in MPMΦ is up-regulated by HG PD fluid and exerts anti-inflammatory effects during PD through regulation of MPMΦ M1/M2 polarization and control of the dominant mesothelial PKC isoform α.
Collapse
Affiliation(s)
- Michael S Balzer
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Alexandra Helmke
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Martina Ackermann
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany.,Phenos, Hannover, Germany
| | - Janis Casper
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Lei Dong
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Marcus Hiss
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Yulia Kiyan
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Song Rong
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Kai Timrott
- Department for General, Abdominal and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | | | - Le Wang
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany.,Department of Nephrology, Tongji Medical College, Wuhan, China
| | - Hermann Haller
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Nelli Shushakova
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany.,Phenos, Hannover, Germany
| |
Collapse
|
37
|
Couplet medicines of leech and centipede granules improve erectile dysfunction via inactivation of the CaSR/PLC/PKC signaling in streptozotocin-induced diabetic rats. Biosci Rep 2020; 40:221835. [PMID: 31922200 PMCID: PMC7000366 DOI: 10.1042/bsr20193845] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/05/2020] [Accepted: 01/08/2020] [Indexed: 01/20/2023] Open
Abstract
Erectile dysfunction (ED) is one of the significant complications of diabetes mellitus (DM), and CASR plays an important role in cellular antiapoptosis and NO production in the vascular endothelium by activating PKC. The present study was aimed to investigate the efficacy of Leech and Centipede Granules (LCG) through the CaSR/PLC/PKC signaling. Fifty male Sprague-Dawley rats were treated with streptozotocin to induce the DM model. After 10 weeks, an apomorphine test was used to confirm DMED. Rats with DMED were administrated with LCG and U73122 for 4 weeks. Fasting blood glucose, body weight, insulin and glucagon levels were measured. Erectile function in rats was assessed by apomorphine. Serums were measured using enzyme-linked immunosorbent assay and flow cytometry, and penile tissues were harvested for histologic and the expression of related targets analyses. After treatment, fasting blood glucose, body weight, insulin, glucagon levels, and erectile function were significantly ameliorated in the LCG groups. The LOX-1, NOX, and EMPs concentrations were significantly decreased with LCG treatment. LCG also continuously increased NO and decreased ET-1 content in penile tissues. LCG and U73122 administration also improved penile fibrosis by significantly decreasing VCAM-1, ICAM-1, and CD62P. The data also showed that LCG reduced the apoptosis level in the penis. Furthermore, the inhibited activation of the CaSR/PLC/PKC pathway was observed in DMED rats with LCG treatment. Collectively, LCG significantly ameliorated erectile function of DMED rats via increased NO generation, inhibiting endothelial cells apoptosis and penile fibrosis, which might benefit from the suppression of CaSR/PLC/PKC pathway in DMED rats.
Collapse
|
38
|
Wolters TLC, van der Heijden CDCC, van Leeuwen N, Hijmans-Kersten BTP, Netea MG, Smit JWA, Thijssen DHJ, Hermus ARMM, Riksen NP, Netea-Maier RT. Persistent inflammation and endothelial dysfunction in patients with treated acromegaly. Endocr Connect 2019; 8:1553-1567. [PMID: 31751301 PMCID: PMC6933829 DOI: 10.1530/ec-19-0430] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 11/11/2019] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Acromegaly is characterized by an excess of growth hormone (GH) and insulin-like growth factor 1 (IGF1). Cardiovascular disease (CVD) risk factors are common in acromegaly and often persist after treatment. Both acute and long-lasting pro-inflammatory effects have been attributed to IGF1. Therefore, we hypothesized that inflammation persists in treated acromegaly and may contribute to CVD risk. METHODS In this cross-sectional study, we assessed cardiovascular structure and function, and inflammatory parameters in treated acromegaly patients. Immune cell populations and inflammatory markers were assessed in peripheral blood from 71 treated acromegaly patients (with controlled or uncontrolled disease) and 41 matched controls. Whole blood (WB) was stimulated with Toll-like receptor ligands. In a subgroup of 21 controls and 33 patients with controlled disease, vascular ultrasound measurements were performed. RESULTS Leukocyte counts were lower in patients with controlled acromegaly compared to patients with uncontrolled acromegaly and controls. Circulating IL18 concentrations were lower in patients; concentrations of other inflammatory mediators were comparable with controls. In stimulated WB, cytokine production was skewed toward inflammation in patients, most pronounced in those with uncontrolled disease. Vascular measurements in controlled patients showed endothelial dysfunction as indicated by a lower flow-mediated dilatation/nitroglycerine-mediated dilatation ratio. Surprisingly, pulse wave analysis and pulse wave velocity, both markers of endothelial dysfunction, were lower in patients, whereas intima-media thickness did not differ. CONCLUSIONS Despite treatment, acromegaly patients display persistent inflammatory changes and endothelial dysfunction, which may contribute to CVD risk and development of CVD.
Collapse
Affiliation(s)
- T L C Wolters
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - C D C C van der Heijden
- Division of Experimental Internal Medicine, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Division of Vascular Medicine, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - N van Leeuwen
- Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - B T P Hijmans-Kersten
- Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M G Netea
- Division of Experimental Internal Medicine, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - J W A Smit
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - D H J Thijssen
- Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - A R M M Hermus
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - N P Riksen
- Division of Vascular Medicine, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - R T Netea-Maier
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Correspondence should be addressed to R T Netea-Maier:
| |
Collapse
|
39
|
Liu B, Wang Y, Zhang Y, Yan B. Mechanisms of Protective Effects of SGLT2 Inhibitors in Cardiovascular Disease and Renal Dysfunction. Curr Top Med Chem 2019; 19:1818-1849. [PMID: 31456521 DOI: 10.2174/1568026619666190828161409] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/09/2019] [Accepted: 07/25/2019] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes mellitus is one of the most common forms of the disease worldwide. Hyperglycemia and insulin resistance play key roles in type 2 diabetes mellitus. Renal glucose reabsorption is an essential feature in glycaemic control. Kidneys filter 160 g of glucose daily in healthy subjects under euglycaemic conditions. The expanding epidemic of diabetes leads to a prevalence of diabetes-related cardiovascular disorders, in particular, heart failure and renal dysfunction. Cellular glucose uptake is a fundamental process for homeostasis, growth, and metabolism. In humans, three families of glucose transporters have been identified, including the glucose facilitators GLUTs, the sodium-glucose cotransporter SGLTs, and the recently identified SWEETs. Structures of the major isoforms of all three families were studied. Sodium-glucose cotransporter (SGLT2) provides most of the capacity for renal glucose reabsorption in the early proximal tubule. A number of cardiovascular outcome trials in patients with type 2 diabetes have been studied with SGLT2 inhibitors reducing cardiovascular morbidity and mortality. The current review article summarises these aspects and discusses possible mechanisms with SGLT2 inhibitors in protecting heart failure and renal dysfunction in diabetic patients. Through glucosuria, SGLT2 inhibitors reduce body weight and body fat, and shift substrate utilisation from carbohydrates to lipids and, possibly, ketone bodies. These pleiotropic effects of SGLT2 inhibitors are likely to have contributed to the results of the EMPA-REG OUTCOME trial in which the SGLT2 inhibitor, empagliflozin, slowed down the progression of chronic kidney disease and reduced major adverse cardiovascular events in high-risk individuals with type 2 diabetes. This review discusses the role of SGLT2 in the physiology and pathophysiology of renal glucose reabsorption and outlines the unexpected logic of inhibiting SGLT2 in the diabetic kidney.
Collapse
Affiliation(s)
- Ban Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuliang Wang
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Yangyang Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China.,Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Biao Yan
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,Eye Institute, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
40
|
Radujkovic A, Kordelas L, Dai H, Schult D, Majer-Lauterbach J, Beelen D, Müller-Tidow C, Dreger P, Luft T. Interleukin-18 and outcome after allogeneic stem cell transplantation: A retrospective cohort study. EBioMedicine 2019; 49:202-212. [PMID: 31680001 PMCID: PMC6945194 DOI: 10.1016/j.ebiom.2019.10.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/07/2019] [Accepted: 10/14/2019] [Indexed: 12/11/2022] Open
Abstract
Background Interleukin-18 (IL-18) is involved in endothelial activation and dysfunction, and in the pathogenesis and severity of acute graft-versus-host disease (aGVHD). Its relevance for patient outcome after allogeneic stem cell transplantation (alloSCT) has not yet been comprehensively addressed. Methods Pre-transplant serum levels of free IL-18 were retrospectively assessed in a cohort of 589 patients (training cohort). Results were validated in 688 patients allografted in a different centre. The primary endpoint was overall survival (OS). Secondary endpoints included incidences of non-relapse mortality (NRM), relapse, and aGVHD. Findings In the training cohort, higher pre-transplant levels of free IL-18 were significantly associated with worse OS (hazard ratio [HR] per 1-log2 increase, 1.25, P = 0.008) in multivariable models. This was due to a higher hazard of NRM (HR per 1-log2 increase, 1.39, P = 0.001), rather than relapse. The associations of pre-transplant free IL-18 with higher NRM (HR per 1-log2 increase, 1.24, P = 0.02) and shorter OS (HR per 1-log2 increase, 1.22, P = 0.006) were confirmed in the validation cohort. In both cohorts, the correlations of higher pre-transplant free IL-18 serum levels with increased NRM and worse OS were mainly driven by fatal infectious complications. No associations with incidence of aGVHD were observed. Interpretation Higher pre-transplant levels of free IL-18 were associated with non-relapse and overall mortality after alloSCT. Our results may provide a rationale for prospective studies evaluating IL-18 status and inhibition of IL-18 activity in patients undergoing allografting.
Collapse
Affiliation(s)
- Aleksandar Radujkovic
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany
| | - Lambros Kordelas
- Department of Bone Marrow Transplantation, University Hospital Essen, Germany
| | - Hao Dai
- Department of Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - David Schult
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany
| | - Joshua Majer-Lauterbach
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany
| | - Dietrich Beelen
- Department of Bone Marrow Transplantation, University Hospital Essen, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany
| | - Peter Dreger
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany
| | - Thomas Luft
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany.
| |
Collapse
|
41
|
Li JY, Chen RJ, Huang LT, Lee TY, Lu WJ, Lin KH. Embelin as a Novel Inhibitor of PKC in the Prevention of Platelet Activation and Thrombus Formation. J Clin Med 2019; 8:jcm8101724. [PMID: 31635287 PMCID: PMC6832570 DOI: 10.3390/jcm8101724] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/09/2019] [Accepted: 10/16/2019] [Indexed: 12/25/2022] Open
Abstract
Embelin is a quinone derivative and found in the fruits of Embelia ribes Burm.f. Embelin has been identified as a small molecular inhibitor of X-chromosome-linked inhibitor of apoptosis proteins, and has multiple biological activities, including antioxidation, anti-inflammation, and antitumor effects. However, the effect of embelin in platelets remains unclear. Thus, this study investigated the antiplatelet mechanism of embelin. Our data revealed that embelin could inhibit platelet aggregation induced by various agonists, including the protein kinase C (PKC) activator phorbol 12,13-dibutyrate (PDBu). Embelin, as well as the PKC inhibitor Ro 31-8220, markedly reduced PDBu-mediated phosphorylation of the PKC substrate, suggesting that embelin may be a PKC inhibitor for platelets. Embelin could block PKC downstream signaling and events, including the inhibition of protein kinase B and mitogen-activated protein kinase activation, granule release, and glycoprotein IIbIIIa activation. Moreover, embelin could delay thrombus formation in the mesenteric microvessels of mice, but did not significantly affect the tail bleeding time. In conclusion, we demonstrated that embelin is a PKC inhibitor and possesses antiplatelet and antithrombotic effects. The further analysis is necessary to more accurately determine clinical therapeutic potential of embelin in all clinical thromboembolic events with disturbance of thrombocyte function.
Collapse
Affiliation(s)
- Jiun Yi Li
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan.
- Department of Surgery, MacKay Memorial Hospital, Taipei 104, Taiwan.
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Ray Jade Chen
- Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Li Ting Huang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Tzu Yin Lee
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Wan Jung Lu
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan.
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan.
| | - Kuan Hung Lin
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 252, Taiwan.
| |
Collapse
|
42
|
EASIX and mortality after allogeneic stem cell transplantation. Bone Marrow Transplant 2019; 55:553-561. [PMID: 31558788 DOI: 10.1038/s41409-019-0703-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/12/2019] [Accepted: 09/13/2019] [Indexed: 01/05/2023]
Abstract
Allogeneic stem cell transplantation (alloSCT) is an effective immunotherapy in patients with hematological malignancies. Endothelial dysfunction was linked to major complications after alloSCT. We asked the question if the "Endothelial Activation and Stress Index" (EASIX; [(creatinine × LDH) ÷ thrombocytes]) can predict mortality after alloSCT. We performed a retrospective cohort analysis in five alloSCT centers in the USA and Germany. EASIX was assessed prior to conditioning (EASIX-pre) and correlated with mortality in 755 patients of a training cohort in multivariable models. The predictive model established in the training cohort was validated in 1267 adult allo-recipients. Increasing EASIX-pre predicted lower overall survival (OS) after alloSCT, and successful model validation was achieved for the validation cohort. We found that EASIX-pre predicts OS irrespective of established scores. Moreover, EASIX-pre was also a significant prognostic factor for transplant-associated microangiopathy. Finally, EASIX-pre correlated with biomarkers of endothelial homeostasis such as CXCL8, interleukin-18, and insulin-like-growth-factor-1 serum levels. This study establishes EASIX-pre based on a standard laboratory biomarker panel as a predictor of individual risk of mortality after alloSCT independently from established clinical criteria.
Collapse
|
43
|
Xiong R, Lu X, Song J, Li H, Wang S. Molecular mechanisms of hydrogen sulfide against uremic accelerated atherosclerosis through cPKCβII/Akt signal pathway. BMC Nephrol 2019; 20:358. [PMID: 31521120 PMCID: PMC6744675 DOI: 10.1186/s12882-019-1550-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/04/2019] [Indexed: 12/24/2022] Open
Abstract
Background Cardiovascular disease is the most common complication and leading cause of death in maintenance hemodialysis patients. The protection mechanism of hydrogen sulfide (H2S) and the specific role of conventional protein kinase C βII (cPKCβII)/Akt signaling pathway in the formation of atherosclerosis is still controversial. Methods 8-week-old male ApoE−/− mice were treated with 5/6 nephrectomy and high-fat diet to make uremia accelerated atherosclerosis (UAAS) model. Mice were divided into normal control group (control group), sham operation group (sham group), UAAS group, L-cysteine group (UAAS+L-cys group), sodium hydrosulfide group (UAAS+NaHS group), and propargylglycine group (UAAS+PPG group). Western blot was used to detect cPKCβII activation, Akt phosphorylation and endothelial nitric oxide synthase (eNOS) expression in mice aorta. Results The membrane translocation of cPKCβII in UAAS group was higher than sham group, and L-cys or NaHS injection could suppress the membrane translocation, but PPG treatment resulted in more membrane translocation of cPKCβII (P < 0.05, n = 6 per group). Akt phosphorylation and the eNOS expression in UAAS group was lower than sham group, and L-cys or NaHS injection could suppress the degradation of Akt phosphorylation and the eNOS expression, but PPG treatment resulted in more decrease in the Akt phosphorylation and the eNOS expression (P < 0.05, n = 6 per group). Conclusion Endogenous cystathionine-γ-lyase (CSE)/H2S system protected against the formation of UAAS via cPKCβII/Akt signal pathway. The imbalance of CSE/H2S system may participate in the formation of UAAS by affecting the expression of downstream molecule eNOS, which may be mediated by cPKCβII/Akt signaling pathway.
Collapse
Affiliation(s)
- Ruifang Xiong
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Xiangxue Lu
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Jinghong Song
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Han Li
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing, 100020, China.
| | - Shixiang Wang
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| |
Collapse
|
44
|
The protective effects of orexin a against high glucose-induced activation of NLRP3 inflammasome in human vascular endothelial cells. Arch Biochem Biophys 2019; 672:108052. [DOI: 10.1016/j.abb.2019.07.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/03/2019] [Accepted: 07/23/2019] [Indexed: 01/15/2023]
|
45
|
Effects of icariin on long noncoding RNA and mRNA expression profile in the aortas of apoE-deficient mice. Biosci Rep 2019; 39:BSR20190855. [PMID: 31296789 PMCID: PMC6658818 DOI: 10.1042/bsr20190855] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/26/2019] [Accepted: 07/10/2019] [Indexed: 12/21/2022] Open
Abstract
Objective: The beneficial effects of icariin (ICA) in ameliorating atherosclerosis (AS) are well known, but the underlying protective mechanism has not been fully elucidated. The present study aimed to investigate altered long noncosing RNA (lncRNA) and mRNA expression profiles in ApoE−/− mice after ICA treatment. Method: The atherosclerotic plaque area was evaluated on high-fat diet (HFD)-induced ApoE−/− mice treated with either ICA or vehicle. LncRNA and mRNA integrated microarrays was performed on aortic tissues. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were utilized to explore the significant function and pathway of the differentially expressed (DE) mRNAs, global signal transduction network were constructed to select key mRNAs, and lncRNA–mRNA co-expression network was built to find out the interactions between lncRNA and mRNA. Quantitative real-time PCR (qPCR) was used to further validate the expressions of selected lncRNAs and mRNAs. Results: Administration of ICA significantly reduced plaque size after 12 weeks (P<0.05). A total of 1512 DE lncRNAs and 2059 DE mRNAs were identified. The mRNAs: protein kinase C, β (Prkcb), Cyp2c65, Mapk10, Calmodulin 5 (Calm5), Calmodulin-like 3 (Calml3) and Camk4 were selected as hub mRNAs, the correlated lncRNAs in co-expression network were identified as important regulatory lncRNAs. The identified target pairs such as lncRNA-NONMMUT000659/Prkcb may play critical roles in AS development mediated by ICA. Conclusion: Taken together, our study highlights a panel of DE lncRNAs and mRNAs that could explain the molecular mechanism of ICA’s anti-atherosclerotic effects. The work lays a foundation for subsequent genes functional researches, which could contribute to provide new therapeutic targets for AS.
Collapse
|
46
|
Radujkovic A, Dai H, Kordelas L, Beelen D, Rachakonda SP, Müller-Tidow C, Kumar R, Dreger P, Luft T. Asymmetric dimethylarginine serum levels are associated with early mortality after allogeneic stem cell transplantation. Haematologica 2019; 104:827-834. [PMID: 30514796 PMCID: PMC6442976 DOI: 10.3324/haematol.2018.202267] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/23/2018] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence suggests that endothelial cell distress is associated with mortality after allogeneic stem cell transplantation and acute graft-versus-host disease. Asymmetric dimethylarginine is an endogenous nitric oxide synthase inhibitor that induces endothelial cell dysfunction. We analyzed the impact of pre-transplant serum levels of asymmetric dimethylarginine on outcome after allogeneic stem cell transplantation. Since acute graft-versus-host disease and its treatment are major contributors to post-transplant mortality, the effect of asymmetric dimethylarginine on outcome measures was also assessed after onset of acute graft-versus-host disease. A total of 938 patients allografted at two centers between 2002 and 2013 were included in the retrospective study. In multivariable models, higher pre-transplant asymmetric dimethylarginine levels were significantly associated with an increased risk of non-relapse mortality (hazard ratio 1.43 per 1-log2 increase, P=0.005) but not with relapse (hazard ratio 1.21, P=0.109) within the first year after transplantation. This translated into worse overall survival (hazard ratio 1.45, P<0.0001) and shorter progression-free survival (hazard ratio 1.30, P=0.002) in the first year after transplantation. Higher pre-transplant asymmetric dimethylarginine levels were also associated with shorter overall survival (hazard ratio 1.46, P=0.001) and progression-free survival (hazard ratio 1.32, P=0.010) and higher non-relapse mortality (hazard ratio 1.36, P=0.042) within 1 year after the onset of acute graft-versus-host disease. Taken together, our data indicate an association between pre-transplant asymmetric dimethylarginine status and early non-relapse mortality in allografted patients, both overall and after the onset of acute graft-versus-host disease. These findings underline the relevance of endothelial dysfunction for transplant complications.
Collapse
Affiliation(s)
| | - Hao Dai
- Department of Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg
| | - Lambros Kordelas
- Department of Bone Marrow Transplantation, University Hospital Essen, Germany
| | - Dietrich Beelen
- Department of Bone Marrow Transplantation, University Hospital Essen, Germany
| | - Sivaramakrishna P Rachakonda
- Department of Internal Medicine V, University Hospital Heidelberg
- Department of Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg
| | | | - Rajiv Kumar
- Department of Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg
| | - Peter Dreger
- Department of Internal Medicine V, University Hospital Heidelberg
| | - Thomas Luft
- Department of Internal Medicine V, University Hospital Heidelberg
| |
Collapse
|
47
|
Park S, Kang HJ, Jeon JH, Kim MJ, Lee IK. Recent advances in the pathogenesis of microvascular complications in diabetes. Arch Pharm Res 2019; 42:252-262. [PMID: 30771210 DOI: 10.1007/s12272-019-01130-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 01/31/2019] [Indexed: 12/29/2022]
Abstract
Millions of people worldwide have diabetes, which is diagnosed by fasting blood glucose levels exceeding 126 mg/dL. Regardless of the type of diabetes, prolonged hyperglycemia is damaging to several organs including eyes, kidneys, nerve, and/or heart. The damages are associated with a high risk of morbidity and mortality. Diabetes has been implicated in ischemia in the microvasculature of the target tissues, which occurs due to the insufficient perfusion of tissues. The resulting occlusion and pain affect the quality of life. Multiple therapeutic approaches have been proposed for a long time to overcome these vascular complications. Apart from systemically controlling high glucose levels, other therapeutic strategies are not well understood. In this review, we summarize the recent literature for biochemical/cellular targets that are being utilized for the treatment of diabetic microvascular diseases. These targets, which are closely associated with mitochondrial dysfunction, include the polyol and diacylglycerol-protein kinase C pathways, oxidative stress, non-enzymatic glycation and the formation of advanced glycation end products, and immune dysregulation/inflammation.
Collapse
Affiliation(s)
- Sungmi Park
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea.
| | - Hyeon-Ji Kang
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, South Korea
| | - Jae-Han Jeon
- Department of Internal Medicine, School of Medicine, Kyungpook National University Hospital, Kyungpook National University, Daegu, South Korea
| | - Min-Ji Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University Hospital, Kyungpook National University, Daegu, South Korea
| | - In-Kyu Lee
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea.
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, South Korea.
- Department of Internal Medicine, School of Medicine, Kyungpook National University Hospital, Kyungpook National University, Daegu, South Korea.
| |
Collapse
|
48
|
Suo F, Jiang F, Fang X, Ma A, Ma L. Contrast of diagnostic value between IL-17 combined with IL-18 and CT angiography in carotid atherosclerosis. Exp Ther Med 2019; 17:1400-1404. [PMID: 30680020 PMCID: PMC6327566 DOI: 10.3892/etm.2018.7088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 11/30/2018] [Indexed: 11/24/2022] Open
Abstract
Diagnostic value between IL-17 combined with IL-18 and CT angiography in carotid atherosclerosis was evaluated. A total of 158 patients with suspected carotid artery stenosis in People's Hospital of Dongying from March 2014 to April 2017 were selected as the subjects. One hundred and two patients with carotid atherosclerosis were selected as the atherosclerosis group, the other 56 patients with no obvious carotid artery abnormalities were selected as the disease control group. In addition, there were 100 healthy subjects selected as the healthy control group. The level of IL-17 and IL-18 in peripheral blood of all the subjects was detected by ELISA. The ROC curve was used to analyze the diagnostic value of IL-17 combined with IL-18 and CT angiography in atherosclerosis. The levels of IL-17 and IL-18 in the three groups were different (P<0.05). The level of IL-17 and IL-18 in the atherosclerosis and disease control groups was higher than that in the healthy control group, and the level of IL-17 and IL-18 in the atherosclerosis was higher than that in the disease control group. The sensitivity of IL-17 or IL-18 was less than the coincidence rate and sensitivity of IL-17 combined with IL-18 as diagnostic criteria, and AUC was also less than AUC of IL-17 combined with IL-18. The sensitivity and diagnostic accuracy of IL-17 combined with IL-18 in the diagnosis of carotid atherosclerosis were not significantly different from CT angiography (P>0.05), but the specificity was obviously lower than CT angiography (P<0.05). The diagnosis of atherosclerosis of IL-17 combined with IL-18 was more accurate, it can improve the diagnostic efficiency of atherosclerosis and be used as a routine method for screening the atherosclerosis.
Collapse
Affiliation(s)
- Feng Suo
- Department of Radiology, People's Hospital of Dongying, Dongying, Shandong 257091, P.R. China
| | - Fei Jiang
- Department of Radiology, People's Hospital of Dongying, Dongying, Shandong 257091, P.R. China
| | - Xiuhong Fang
- Department of Radiology, People's Hospital of Dongying, Dongying, Shandong 257091, P.R. China
| | - Aihua Ma
- Pharmacy Intravenous Admixture Services, Zhangqiu, Shandong 250200, P.R. China
| | - Liping Ma
- Department of Science and Education, The People's Hospital of Zhangqiu Area, Zhangqiu, Shandong 250200, P.R. China
| |
Collapse
|
49
|
Yuan T, Yang T, Chen H, Fu D, Hu Y, Wang J, Yuan Q, Yu H, Xu W, Xie X. New insights into oxidative stress and inflammation during diabetes mellitus-accelerated atherosclerosis. Redox Biol 2019; 20:247-260. [PMID: 30384259 PMCID: PMC6205410 DOI: 10.1016/j.redox.2018.09.025] [Citation(s) in RCA: 431] [Impact Index Per Article: 71.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/12/2018] [Accepted: 09/29/2018] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress and inflammation interact in the development of diabetic atherosclerosis. Intracellular hyperglycemia promotes production of mitochondrial reactive oxygen species (ROS), increased formation of intracellular advanced glycation end-products, activation of protein kinase C, and increased polyol pathway flux. ROS directly increase the expression of inflammatory and adhesion factors, formation of oxidized-low density lipoprotein, and insulin resistance. They activate the ubiquitin pathway, inhibit the activation of AMP-protein kinase and adiponectin, decrease endothelial nitric oxide synthase activity, all of which accelerate atherosclerosis. Changes in the composition of the gut microbiota and changes in microRNA expression that influence the regulation of target genes that occur in diabetes interact with increased ROS and inflammation to promote atherosclerosis. This review highlights the consequences of the sustained increase of ROS production and inflammation that influence the acceleration of atherosclerosis by diabetes. The potential contributions of changes in the gut microbiota and microRNA expression are discussed.
Collapse
Affiliation(s)
- Ting Yuan
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Ting Yang
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Huan Chen
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| | - Danli Fu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Yangyang Hu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Jing Wang
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Qing Yuan
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Hong Yu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Wenfeng Xu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Xiang Xie
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| |
Collapse
|
50
|
Mima A. Renal protection by sodium-glucose cotransporter 2 inhibitors and its underlying mechanisms in diabetic kidney disease. J Diabetes Complications 2018; 32:720-725. [PMID: 29880432 DOI: 10.1016/j.jdiacomp.2018.04.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 12/12/2022]
Abstract
AIM Diabetic kidney disease (DKD) is the most frequent cause of mortality and morbidity, leading a global health burden. This review will focus on the potential therapeutic interventions using Sodium-glucose cotransporter-2 (SGLT2) inhibitors that could prevent the development and progression of DKD. RESULTS SGLT2 inhibitors have been widely used as anti-diabetic drugs. Recent clinical studies have demonstrated that these drugs, which improve glycemic control and hypertension and decrease body weight, decrease the risk of renal function impairment and heart failure in patients with type 2 diabetes. With regard to long-term clinical outcomes, the Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes (EMPA-REG OUTCOME), the EMPA-REG Renal OUTCOME, and the CANagliflozin cardioVascular Assessment Study (CANVAS) program which have been integrated from CANVAS and CANVAS-Renal (CANVAS-R) trials reported significant risk reductions in primary combined major adverse cardiovascular events. Furthermore, regarding renal outcomes, the EMPA-REG Renal OUTCOME and CANVAS program clearly showed improvements in renal outcomes, including decreases in albuminuria and progression of nephropathy, doubling of serum creatinine levels, and initiation of renal replacement therapy. CONCLUSIONS Potential mechanisms of SGLT2 inhibitors related to renoprotection can be divided into two categories: hemodynamic actions and metabolic actions.
Collapse
Affiliation(s)
- Akira Mima
- Department of Nephrology, Kindai University Faculty of Medicine, Kindai University Nara Hospital, Nara, Japan.
| |
Collapse
|