51
|
Guo J, Wang H, Li Y, Zhu S, Hu H, Gu Z. Nanotechnology in coronary heart disease. Acta Biomater 2023; 171:37-67. [PMID: 37714246 DOI: 10.1016/j.actbio.2023.09.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/17/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023]
Abstract
Coronary heart disease (CHD) is one of the major causes of death and disability worldwide, especially in low- and middle-income countries and among older populations. Conventional diagnostic and therapeutic approaches have limitations such as low sensitivity, high cost and side effects. Nanotechnology offers promising alternative strategies for the diagnosis and treatment of CHD by exploiting the unique properties of nanomaterials. In this review, we use bibliometric analysis to identify research hotspots in the application of nanotechnology in CHD and provide a comprehensive overview of the current state of the art. Nanomaterials with enhanced imaging and biosensing capabilities can improve the early detection of CHD through advanced contrast agents and high-resolution imaging techniques. Moreover, nanomaterials can facilitate targeted drug delivery, tissue engineering and modulation of inflammation and oxidative stress, thus addressing multiple aspects of CHD pathophysiology. We discuss the application of nanotechnology in CHD diagnosis (imaging and sensors) and treatment (regulation of macrophages, cardiac repair, anti-oxidative stress), and provide insights into future research directions and clinical translation. This review serves as a valuable resource for researchers and clinicians seeking to harness the potential of nanotechnology in the management of CHD. STATEMENT OF SIGNIFICANCE: Coronary heart disease (CHD) is the one of leading cause of death and disability worldwide. Nanotechnology offers new strategies for diagnosing and treating CHD by exploiting the unique properties of nanomaterials. This review uses bibliometric analysis to uncover research trends in the use of nanotechnology for CHD. We discuss the potential of nanomaterials for early CHD detection through advanced imaging and biosensing, targeted drug delivery, tissue engineering, and modulation of inflammation and oxidative stress. We also offer insights into future research directions and potential clinical applications. This work aims to guide researchers and clinicians in leveraging nanotechnology to improve CHD patient outcomes and quality of life.
Collapse
Affiliation(s)
- Junsong Guo
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Hao Wang
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Ying Li
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Shuang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nano-safety, Institute of High Energy Physics, Beijing 100049, China; CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Houxiang Hu
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China.
| | - Zhanjun Gu
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nano-safety, Institute of High Energy Physics, Beijing 100049, China; Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
52
|
Liu Z, Daniels T, Campen MJ, Alvidrez RIM. Inflammatory atherosclerotic plaque identification by SPECT/CT imaging of LFA-1 using [ 111In] In-DANBIRT in a novel dyslipidemic rat model. Ann Nucl Med 2023; 37:635-643. [PMID: 37742306 DOI: 10.1007/s12149-023-01868-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/11/2023] [Indexed: 09/26/2023]
Abstract
INTRODUCTION Atherosclerosis is prevalent globally, closely associated with dyslipidemia and other metabolic dysfunction. Early diagnosis of atherosclerosis is challenging due to limited diagnostic capabilities that need to be expanded with animal models with enhanced vascular biology like rats. Our previous research showed [111In] In-DANBIRT has potential as a diagnostic tool for detecting atherosclerosis in mice. The primary aim of the present study is to evaluate [111In] In-DANBIRT in a novel atherosclerotic rat with early- and late-stage atherosclerosis and metabolic disease. METHODS We characterized metabolic and body composition differences in these novel dyslipidemic rats under different diets using serum chemistry and dual-energy X-ray absorptiometry (DEXA) scan, respectively. We performed 1-h post-injection in vivo molecular imaging of ApoE knockout, lean Zucker (LZ) male rats at baseline and followed them into 10 weeks of either normal or high-fat/cholesterol diet implementation (22 weeks of age). RESULTS We identified significant differences in body composition and metabolic changes in ApoE knockout rats compared to ApoE wildtype rats. Our findings indicate an increased uptake of [111In] In-DANBIRT in ApoE knockout, lean Zucker (LZ) rats, particularly in the descending aorta, a location where early-stage atherosclerosis is commonly found. Our findings, however, also revealed that the ApoE knockout, Zucker diabetic fatty (ZDF) model has high mortality rate, which may be attributed to alterations of critical enzymes involved in regulating metabolism and liver function. CONCLUSION Our results are highly encouraging as they demonstrated the potential of [111In] In-DANBIRT to detect early-stage atherosclerosis in rats that might otherwise go unnoticed by other methods, showcasing the high sensitivity of [111In] In-DANBIRT. Our future studies will aim to establish a viable T2D atherosclerosis model in rats with more advanced stages of the disease to further demonstrate the reliability of [111In] In-DANBIRT as a diagnostic tool for patients in all stages of atherosclerosis.
Collapse
Affiliation(s)
- Zeyu Liu
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Tamara Daniels
- Department of Radiopharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM, 87131, USA
- College of Pharmacy, Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA
- Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, 87106, USA
| | - Matthew J Campen
- College of Pharmacy, Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA
- Clinical and Translational Science Center, University of New Mexico, Albuquerque, NM, 87131, USA
- Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, 87106, USA
| | - Roberto Ivan Mota Alvidrez
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA.
- Department of Radiopharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM, 87131, USA.
- College of Pharmacy, Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA.
- Pittsburgh Liver Research Center Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Clinical and Translational Science Center, University of New Mexico, Albuquerque, NM, 87131, USA.
- Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, 87106, USA.
| |
Collapse
|
53
|
Allphin AJ, Mahzarnia A, Clark DP, Qi Y, Han ZY, Bhandari P, Ghaghada KB, Badea A, Badea CT. Advanced photon counting CT imaging pipeline for cardiac phenotyping of apolipoprotein E mouse models. PLoS One 2023; 18:e0291733. [PMID: 37796905 PMCID: PMC10553338 DOI: 10.1371/journal.pone.0291733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/01/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) is associated with the apolipoprotein E (APOE) gene and lipid metabolism. This study aimed to develop an imaging-based pipeline to comprehensively assess cardiac structure and function in mouse models expressing different APOE genotypes using photon-counting computed tomography (PCCT). METHODS 123 mice grouped based on APOE genotype (APOE2, APOE3, APOE4, APOE knockout (KO)), gender, human NOS2 factor, and diet (control or high fat) were used in this study. The pipeline included PCCT imaging on a custom-built system with contrast-enhanced in vivo imaging and intrinsic cardiac gating, spectral and temporal iterative reconstruction, spectral decomposition, and deep learning cardiac segmentation. Statistical analysis evaluated genotype, diet, sex, and body weight effects on cardiac measurements. RESULTS Our results showed that PCCT offered high quality imaging with reduced noise. Material decomposition enabled separation of calcified plaques from iodine enhanced blood in APOE KO mice. Deep learning-based segmentation showed good performance with Dice scores of 0.91 for CT-based segmentation and 0.89 for iodine map-based segmentation. Genotype-specific differences were observed in left ventricular volumes, heart rate, stroke volume, ejection fraction, and cardiac index. Statistically significant differences were found between control and high fat diets for APOE2 and APOE4 genotypes in heart rate and stroke volume. Sex and weight were also significant predictors of cardiac measurements. The inclusion of the human NOS2 gene modulated these effects. CONCLUSIONS This study demonstrates the potential of PCCT in assessing cardiac structure and function in mouse models of CVD which can help in understanding the interplay between genetic factors, diet, and cardiovascular health.
Collapse
Affiliation(s)
- Alex J. Allphin
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University Medical Center, Durham, NC, United States of America
| | - Ali Mahzarnia
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University Medical Center, Durham, NC, United States of America
| | - Darin P. Clark
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University Medical Center, Durham, NC, United States of America
| | - Yi Qi
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University Medical Center, Durham, NC, United States of America
| | - Zay Y. Han
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University Medical Center, Durham, NC, United States of America
| | - Prajwal Bhandari
- Department of Radiology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Radiology, Texas Children’s Hospital, Houston, Texas, United States of America
| | - Ketan B. Ghaghada
- Department of Radiology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Radiology, Texas Children’s Hospital, Houston, Texas, United States of America
| | - Alexandra Badea
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University Medical Center, Durham, NC, United States of America
- Department of Neurology, Duke University Medical Center, Durham, NC, United States of America
| | - Cristian T. Badea
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University Medical Center, Durham, NC, United States of America
| |
Collapse
|
54
|
Garger D, Meinel M, Dietl T, Hillig C, Garzorz‐Stark N, Eyerich K, de Angelis MH, Eyerich S, Menden MP. The impact of the cardiovascular component and somatic mutations on ageing. Aging Cell 2023; 22:e13957. [PMID: 37608601 PMCID: PMC10577550 DOI: 10.1111/acel.13957] [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: 03/06/2023] [Revised: 06/27/2023] [Accepted: 07/20/2023] [Indexed: 08/24/2023] Open
Abstract
Mechanistic insight into ageing may empower prolonging the lifespan of humans; however, a complete understanding of this process is still lacking despite a plethora of ageing theories. In order to address this, we investigated the association of lifespan with eight phenotypic traits, that is, litter size, body mass, female and male sexual maturity, somatic mutation, heart, respiratory, and metabolic rate. In support of the somatic mutation theory, we analysed 15 mammalian species and their whole-genome sequencing deriving somatic mutation rate, which displayed the strongest negative correlation with lifespan. All remaining phenotypic traits showed almost equivalent strong associations across this mammalian cohort, however, resting heart rate explained additional variance in lifespan. Integrating somatic mutation and resting heart rate boosted the prediction of lifespan, thus highlighting that resting heart rate may either directly influence lifespan, or represents an epiphenomenon for additional lower-level mechanisms, for example, metabolic rate, that are associated with lifespan.
Collapse
Affiliation(s)
- Daniel Garger
- Computational Health Center, Helmholtz MunichNeuherbergGermany
- Faculty of BiologyLudwig Maximilian UniversityMartinsriedGermany
| | - Martin Meinel
- Computational Health Center, Helmholtz MunichNeuherbergGermany
- Faculty of BiologyLudwig Maximilian UniversityMartinsriedGermany
- Department of Dermatology and AllergyTechnical University of MunichMunichGermany
| | - Tamina Dietl
- Computational Health Center, Helmholtz MunichNeuherbergGermany
- Faculty of BiologyLudwig Maximilian UniversityMartinsriedGermany
| | - Christina Hillig
- Computational Health Center, Helmholtz MunichNeuherbergGermany
- Department of MathematicsTechnical University of MunichMunichGermany
| | - Natalie Garzorz‐Stark
- Department of Dermatology and AllergyTechnical University of MunichMunichGermany
- Division of Dermatology and Venereology, Department of Medicine Solna, and Center for molecular medicineKarolinska InstitutetStockholmSweden
| | - Kilian Eyerich
- Division of Dermatology and Venereology, Department of Medicine Solna, and Center for molecular medicineKarolinska InstitutetStockholmSweden
- Department of Dermatology and Venerology, Medical SchoolUniversity of FreiburgFreiburgGermany
| | - Martin Hrabě de Angelis
- Institute of Experimental GeneticsHelmholtz MunichNeuherbergGermany
- Chair of Experimental Genetics, TUM School of Life SciencesTechnical University MunichFreisingGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| | - Stefanie Eyerich
- Center for Allergy and Environment (ZAUM)Technical University MunichMunichGermany
- Institute for Allergy ResearchHelmholtz Munich, NeuherbergNeuherbergGermany
| | - Michael P. Menden
- Computational Health Center, Helmholtz MunichNeuherbergGermany
- Faculty of BiologyLudwig Maximilian UniversityMartinsriedGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
- Department of Biochemistry and PharmacologyUniversity of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
55
|
Jiang MC, Ding HY, Huang YH, Cheng CK, Lau CW, Xia Y, Yao XQ, Wang L, Huang Y. Thioridazine protects against disturbed flow-induced atherosclerosis by inhibiting RhoA/YAP-mediated endothelial inflammation. Acta Pharmacol Sin 2023; 44:1977-1988. [PMID: 37217602 PMCID: PMC10545737 DOI: 10.1038/s41401-023-01102-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 04/27/2023] [Indexed: 05/24/2023]
Abstract
Atherosclerotic diseases remain the leading cause of adult mortality and impose heavy burdens on health systems globally. Our previous study found that disturbed flow enhanced YAP activity to provoke endothelial activation and atherosclerosis, and targeting YAP alleviated endothelial inflammation and atherogenesis. Therefore, we established a luciferase reporter assay-based drug screening platform to seek out new YAP inhibitors for anti-atherosclerotic treatment. By screening the FDA-approved drug library, we identified that an anti-psychotic drug thioridazine markedly suppressed YAP activity in human endothelial cells. Thioridazine inhibited disturbed flow-induced endothelial inflammatory response in vivo and in vitro. We verified that the anti-inflammatory effects of thioridazine were mediated by inhibition of YAP. Thioridazine regulated YAP activity via restraining RhoA. Moreover, administration of thioridazine attenuated partial carotid ligation- and western diet-induced atherosclerosis in two mouse models. Overall, this work opens up the possibility of repurposing thioridazine for intervention of atherosclerotic diseases. This study also shed light on the underlying mechanisms that thioridazine inhibited endothelial activation and atherogenesis via repression of RhoA-YAP axis. As a new YAP inhibitor, thioridazine might need further investigation and development for the treatment of atherosclerotic diseases in clinical practice.
Collapse
Affiliation(s)
- Min-Chun Jiang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Huan-Yu Ding
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu-Hong Huang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chak Kwong Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Chi Wai Lau
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yin Xia
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiao-Qiang Yao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yu Huang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
56
|
An C, Sun F, Liu C, Huang S, Xu T, Zhang C, Ge S. IQGAP1 promotes mitochondrial damage and activation of the mtDNA sensor cGAS-STING pathway to induce endothelial cell pyroptosis leading to atherosclerosis. Int Immunopharmacol 2023; 123:110795. [PMID: 37597406 DOI: 10.1016/j.intimp.2023.110795] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 08/21/2023]
Abstract
Atherosclerosis (AS) is the most common cardiovascular disease and has limited therapeutic options. IQ motif-containing GTPase-activating protein 1 (IQGAP1) is an important scaffolding protein regulating mitochondrial function influencing endothelial cell activity. Evidence suggests that mitochondrial damage can lead to leakage of mtDNA into the cytoplasm to activate the DNA sensor cGAS-STING to mediate pyroptosis. However, whether IQGAP1 induces NLRP3-mediated endothelial cell pyroptosis by regulating mitochondrial function and activating the DNA sensor cGAS-STING, and its underlying mechanisms remain unclear. In vivo, ApoE-/- C57BL/J and Ldlr-/- C57BL/J mice were pre-injected with adeno-associated virus (AAV) by the tail vein to specifically silence IQGAP1 expression and were fed a high-fat diet (HFD) for 12 weeks. IQGAP1 knockdown reduced mtDNA release and decreased the expression of DNA receptors and pyroptosis-related molecules as determined by immunohistochemistry and immunofluorescence. In vitro, palmitic acid (0.3 mmol/L) was incubated with human umbilical vein endothelial cells (HUVECs) for 24 h. Overexpression of IQGAP1 in HUVECs, flow cytometry, and mitochondrial superoxide staining revealed increased levels of ROS. Moreover, the mitochondrial tracker with dsDNA co-localization showed the release of mtDNA into the cytoplasm increased, which activated the DNA receptor cGAS-STING. Protein blotting and TUNEL staining revealed that IQGAP1 promoted NLRP3-mediated pyroptosis. Furthermore, cGAS or STING small-molecule inhibitors RU.521 or C-176 reverse IQGAP1-promoted HUVECs from undergoing NLRP3-mediated pyroptosis. These results suggest that IQGAP1 promotes oxidative stress and mtDNA release, activates the DNA sensor cGAS-STING, and leads to NLRP3-mediated pyroptosis. The present study provides new insights into the mechanisms underlying AS and identifies new pharmacological targets for treatment.
Collapse
Affiliation(s)
- Cheng An
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Fei Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Can Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Shaojun Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, China
| | - Chengxin Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China.
| | - Shenglin Ge
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China.
| |
Collapse
|
57
|
Sato A, Tsukiyama T, Komeno M, Iwatani C, Tsuchiya H, Kawamoto I, Murase M, Nakagawa T, Itagaki I, Seita Y, Matsumoto S, Nakaya M, Shimizu A, Yamada A, Ema M, Ogita H. Generation of a familial hypercholesterolemia model in non-human primate. Sci Rep 2023; 13:15649. [PMID: 37730951 PMCID: PMC10511719 DOI: 10.1038/s41598-023-42763-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 09/14/2023] [Indexed: 09/22/2023] Open
Abstract
Familial hypercholesterolemia (FH) is an inherited autosomal dominant disorder that is associated with a high plasma level of low-density lipoprotein (LDL) cholesterol, leading to an increased risk of cardiovascular diseases. To develop basic and translational research on FH, we here generated an FH model in a non-human primate (cynomolgus monkeys) by deleting the LDL receptor (LDLR) gene using the genome editing technique. Six LDLR knockout (KO) monkeys were produced, all of which were confirmed to have mutations in the LDLR gene by sequence analysis. The levels of plasma cholesterol and triglyceride were quite high in the monkeys, and were similar to those in FH patients with homozygous mutations in the LDLR gene. In addition, periocular xanthoma was observed only 1 year after birth. Lipoprotein profile analysis showed that the plasma very low-density lipoprotein and LDL were elevated, while the plasma high density lipoprotein was decreased in LDLR KO monkeys. The LDLR KO monkeys were also strongly resistant to medications for hypercholesterolemia. Taken together, we successfully generated a non-human primate model of hypercholesterolemia in which the phenotype is similar to that of homozygous FH patients.
Collapse
Affiliation(s)
- Akira Sato
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Tomoyuki Tsukiyama
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Masahiro Komeno
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Chizuru Iwatani
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Hideaki Tsuchiya
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Ikuo Kawamoto
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Mitsuru Murase
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Takahiro Nakagawa
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Iori Itagaki
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Yasunari Seita
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Shoma Matsumoto
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Masataka Nakaya
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Atsushi Yamada
- Medical Innovation Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Masatsugu Ema
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan.
| |
Collapse
|
58
|
Wakako A, Sadato A, Oeda M, Higashiguchi S, Hayakawa M, Oshima M, Hirose Y. Development of a Model for Plaque Induction in Rat Carotid Arteries. Asian J Neurosurg 2023; 18:499-507. [PMID: 38152536 PMCID: PMC10749859 DOI: 10.1055/s-0043-1763522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023] Open
Abstract
Objective Plaque induction through intimal injury using a balloon catheter in small animals and by artificial ligation of the carotid artery in large animals have been reported. However, these reports have not yet succeeded in inducing stable plaques nor creating a high degree of intimal thickening to be used as animal models. We have previously developed a plaque induction model in rats but have failed to obtain a plaque incidence frequency that can be used as a model. Thus, in the current study, we aimed to create a versatile disease model to examine the pharmacokinetics of drug administration, determine the efficacy of treatment, and examine the process of intimal thickening. We also attempted to create an improved model with shorter, more frequent, and more severe intimal thickening. Materials and Methods The common carotid artery of male Wistar rats was surgically exposed and completely ligated with a wire and 6-0 nylon thread. Then, the wire was removed to create a partial ligation. To create a high frequency and high degree of intimal thickening, 72 rats were divided into two groups: a single lesion group with a 0.25-mm wire and a single ligature point, and a tandem lesion group with a 0.3-mm wire and two ligature points. Each group was further divided into normal diet and high cholesterol diet groups. The presence and frequency of intimal thickening were examined for each group after 4, 8, and 16 weeks of growth. Results In the single lesion group, intimal thickening was observed in 42% of the 4-week group and 75% of the 8-week group. In the tandem lesion group, intimal thickening was observed in 75% of the 4-week group and 50% of the 8-week group. In addition, 50% of the individuals reared for 16 weeks developed intimal thickening. Conclusion We successfully induced intimal thickening in the carotid arteries of rats with high frequency in the single lesion and tandem lesion groups. The results also showed that the tandem lesion group tended to induce intimal thickening earlier than the single lesion group.
Collapse
Affiliation(s)
- Akira Wakako
- Department of Neurosurgery, Fujita Medical University Okazaki Medical Center, Okazaki, Aichi, Japan
| | - Akiyo Sadato
- Department of Neurosurgery, Fujita Medical University, Toyoake, Aichi, Japan
| | - Motoki Oeda
- Department of Neurosurgery, Toyota Memorial Hospital, Toyota, Aichi, Japan
| | - Saeko Higashiguchi
- Department of Neurosurgery, Fujita Medical University Okazaki Medical Center, Okazaki, Aichi, Japan
| | - Motoharu Hayakawa
- Department of Neurosurgery, Fujita Medical University Okazaki Medical Center, Okazaki, Aichi, Japan
| | - Marie Oshima
- Institute of Industrial Science/Graduate School of Interdisciplinary Information Studies, University of Tokyo, Tokyo, Japan
| | - Yuichi Hirose
- Department of Neurosurgery, Fujita Medical University, Toyoake, Aichi, Japan
| |
Collapse
|
59
|
Andreeva VD, Ehlers H, R C AK, Presselt M, J van den Broek L, Bonnet S. Combining nitric oxide and calcium sensing for the detection of endothelial dysfunction. Commun Chem 2023; 6:179. [PMID: 37644120 PMCID: PMC10465535 DOI: 10.1038/s42004-023-00973-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 08/01/2023] [Indexed: 08/31/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide and are not typically diagnosed until the disease has manifested. Endothelial dysfunction is an early, reversible precursor in the irreversible development of cardiovascular diseases and is characterized by a decrease in nitric oxide production. We believe that more reliable and reproducible methods are necessary for the detection of endothelial dysfunction. Both nitric oxide and calcium play important roles in the endothelial function. Here we review different types of molecular sensors used in biological settings. Next, we review the current nitric oxide and calcium sensors available. Finally, we review methods for using both sensors for the detection of endothelial dysfunction.
Collapse
Affiliation(s)
| | - Haley Ehlers
- Mimetas B.V., De limes 7, 2342 DH, Oegstgeest, The Netherlands
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Aswin Krishna R C
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Martin Presselt
- Leibniz Institute of Photonic Technology (Leibniz-IPHT), Albert-Einstein-Str. 9, 07745, Jena, Germany
- Sciclus GmbH & Co. KG, Moritz-von-Rohr-Str. 1a, 07745, Jena, Germany
| | | | - Sylvestre Bonnet
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
60
|
Szekeres R, Priksz D, Kiss R, Romanescu DD, Bombicz M, Varga B, Gesztelyi R, Szilagyi A, Takacs B, Tarjanyi V, Pelles-Tasko B, Forgacs I, Remenyik J, Szilvassy Z, Juhasz B. Therapeutic Aspects of Prunus cerasus Extract in a Rabbit Model of Atherosclerosis-Associated Diastolic Dysfunction. Int J Mol Sci 2023; 24:13253. [PMID: 37686067 PMCID: PMC10488229 DOI: 10.3390/ijms241713253] [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: 08/02/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
This study evaluates the potential therapeutic effects of anthocyanin-rich Prunus cerasus (sour cherry) extract (PCE) on atherosclerosis-associated cardiac dysfunction, described by the impairment of the NO-PKG (nitric oxide-protein kinase G) pathway and the antioxidant capacity. Initially, a rabbit model of atherosclerotic cardiovascular disease was established by administering a cholesterol-rich diet, enabling the examination of the impact of 9 g/kg PCE on the pre-existing compromised cardiovascular condition. After that, the animals were divided into four groups for 12 weeks: the (1) untreated control group; (2) PCE-administered healthy rabbits; (3) hypercholesterolemic (HC) group kept on an atherogenic diet; and (4) PCE-treated HC group. Dyslipidemia, impaired endothelial function, and signs of diastolic dysfunction were evident in hypercholesterolemic rabbits, accompanied by a reduced cardiac expression of eNOS (endothelial nitric oxide synthase), PKG, and SERCA2a (sarco/endoplasmic reticulum calcium ATPase 2a). Subsequent PCE treatment improved the lipid profile and the cardiac function. Additionally, PCE administration was associated with elevated myocardial levels of eNOS, PKG, and SERCA2a, while no significant changes in the vascular status were observed. Western blot analysis further revealed hypercholesterolemia-induced increase and PCE-associated reduction in heme oxygenase-1 expression. The observed effects of anthocyanins indicate their potential as a valuable addition to the treatment regimen for atherosclerosis-associated cardiac dysfunction.
Collapse
Affiliation(s)
- Reka Szekeres
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Daniel Priksz
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Rita Kiss
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Dana Diana Romanescu
- Department of Diabetology, Pelican Clinical Hospital, 410087 Oradea, Romania;
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Mariann Bombicz
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Balazs Varga
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Rudolf Gesztelyi
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Anna Szilagyi
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Barbara Takacs
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Vera Tarjanyi
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Beata Pelles-Tasko
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Ildiko Forgacs
- Center for Complex Systems and Microbiome Innovations, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, H-4032 Debrecen, Hungary; (I.F.); (J.R.)
| | - Judit Remenyik
- Center for Complex Systems and Microbiome Innovations, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, H-4032 Debrecen, Hungary; (I.F.); (J.R.)
| | - Zoltan Szilvassy
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Bela Juhasz
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| |
Collapse
|
61
|
Murphy JM, Jeong K, Tran DTK, Cioffi DL, Campbell PM, Kim JH, Jo H, Ahn EYE, Lim STS. Nuclear FAK in endothelium: An intrinsic inhibitor of NF-κB activation in atherosclerosis. Atherosclerosis 2023; 379:117189. [PMID: 37527611 PMCID: PMC10530536 DOI: 10.1016/j.atherosclerosis.2023.117189] [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: 02/25/2021] [Revised: 07/11/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023]
Abstract
BACKGROUND AND AIMS Hyperlipidemia leads to the accumulation of oxidized low-density lipoprotein (oxLDL) within the vessel wall where it causes chronic inflammation in endothelial cells (ECs) and drives atherosclerotic lesions. Although focal adhesion kinase (FAK) is critical in proinflammatory NF-κB activation in ECs, it is unknown if hyperlipidemia alters FAK-mediated NF-κB activity in vivo to affect atherosclerosis progression. METHODS We investigated changes in EC FAK and NF-κB activation using Apoe-/- mice fed a western diet (WD). Both pharmacological FAK inhibition and EC-specific FAK inhibited mouse models were utilized. FAK and NF-κB localization and activity were also analyzed in human atherosclerotic samples. RESULTS ECs of hyperlipidemic mice clearly showed much higher levels of FAK activation in the cytoplasm, which was associated with increased NF-κB activation compared to normal diet (ND) group. On the contrary, FAK is mostly localized in the nucleus and inactive in ECs under healthy conditions with a low NF-κB activity. Both pharmacological and EC-specific genetic FAK inhibition in WD fed Apoe-/- mice exhibited a significant decrease in FAK activity and cytoplasmic localization, NF-κB activation, macrophage recruitment, and atherosclerotic lesions compared to the vehicle or FAK wild-type groups. Analyses of human atherosclerotic specimens revealed a positive correlation between increased active cytoplasmic FAK within ECs and NF-κB activation in the lesions. CONCLUSIONS Hyperlipidemic conditions activate NF-κB pathway by increasing EC FAK activity and cytoplasmic localization in mice and human atherosclerotic samples. As FAK inhibition can efficiently reduce vascular inflammation and atherosclerotic lesions in mice by reversing EC FAK localization and NF-κB activation, these findings support a potential use for FAK inhibitors in treating atherosclerosis.
Collapse
Affiliation(s)
- James M Murphy
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Kyuho Jeong
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, Mobile, AL, 36688, USA
| | - Duyen Thi Kieu Tran
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Donna L Cioffi
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, Mobile, AL, 36688, USA
| | - Pamela Moore Campbell
- Department of Pathology, University of South Alabama College of Medicine, Mobile, AL, 36617, USA
| | - Jin H Kim
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL, 36688, USA
| | - Hanjoong Jo
- Department of Bioengineering, Emory University and Georgia Institute of Technology, Atlanta, GA, 30322, USA
| | - Eun-Young Erin Ahn
- Department of Pathology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Ssang-Taek Steve Lim
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
62
|
Jamuna S, Ashokkumar R, Raja IS, Devaraj SN. Anti-Atherogenic Protection by Oligomeric Proanthocyanidins via Regulating Collagen Crosslinking Against CC Diet-Induced Atherosclerosis in Rats. Appl Biochem Biotechnol 2023; 195:4881-4892. [PMID: 37097399 DOI: 10.1007/s12010-023-04487-w] [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] [Accepted: 04/11/2023] [Indexed: 04/26/2023]
Abstract
The synthesis of collagen and its turnover remained as critical determinants for the progression of atherosclerosis. During this condition, proteases secreted by SMCs and foam cells in the necrotic core degrade collagen. Growing evidences demonstrated that consumption of antioxidant rich diet is highly associated with a reduced risk of atherosclerosis. Oligomeric proanthocyanidins (OPC) have been proved to possess promising antioxidant, anti-inflammatory and cardioprotective activity, based on our previous studies. The present study aims to investigate the efficacy of OPC isolated from Crataegus oxyacantha berries as a natural collagen crosslinker and anti-atherogenic agent. Spectral studies like FTIR, ultraviolet and circular dichroism analysis confirmed the in vitro crosslinking ability of OPC with rat tail collagen when compared to the standard epigallocatechin gallate. The administration of cholesterol:cholic acid (CC) diet induces proteases-mediated collagen degradation that could result in plaque instability. Further, the CC diet fed rats showed significantly increased levels of total cholesterol and triacylglycerols which, in turn, increases the activities of collagen degrading proteases-MMPs (MMP 1, 2 and 9) and Cathepsin S and D. Upon OPC treatment, marked reduction in the lipid content, activation of proteases with concomitant increase in the mRNA levels of collagen Type I and Type III as similar to atorvastatin treatment were observed .Thus, OPC supplementation may contribute to the prevention of atherosclerotic plaque instability by acting as a natural crosslinker of collagen.
Collapse
Affiliation(s)
- Sankar Jamuna
- Affyclone Laboratories Pvt Ltd., 600044, Chrompet, Chennai, India
- Department of Biochemistry, University of Madras, Guindy campus, 600025, Chennai, India
| | - Rathinavel Ashokkumar
- Affyclone Laboratories Pvt Ltd., 600044, Chrompet, Chennai, India
- Department of Biochemistry, University of Madras, Guindy campus, 600025, Chennai, India
| | | | | |
Collapse
|
63
|
Liu B, Fang L, Mo P, Chen C, Ji Y, Pang L, Chen H, Deng Y, Ou W, Liu SM. Apoe-knockout induces strong vascular oxidative stress and significant changes in the gene expression profile related to the pathways implicated in redox, inflammation, and endothelial function. Cell Signal 2023; 108:110696. [PMID: 37409402 DOI: 10.1016/j.cellsig.2023.110696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/30/2023] [Accepted: 04/28/2023] [Indexed: 07/07/2023]
Abstract
Apolipoprotein E (APOE) was recognized as a key regulator of lipid metabolism, which prompted the Apoe-knockout (Apoe-/-) mouse to be the most widely used atherosclerotic model. However, with more and more important physiological roles of APOE being revealed, it is necessary to reacquaint its comprehensive function in the aorta. In this study, we aimed to reveal how Apoe-knockout impacts the gene pathways and phenotypes in the aorta of mice. We performed transcriptome sequencing to acquire the gene expression profile (GEP) for C57BL/6J and Apoe-/- mouse aorta, and used enrichment analysis to reveal the signal pathways enriched for differentially expressed genes (DEGs). In addition, we used immunofluorescence and ELISA to detect the phenotypic differences of vascular tissues and plasma in the two-group mice. Apoe-knockout resulted in significant changes in the expression of 538 genes, among which about 75% were up-regulated and 134 genes were altered more than twice. In addition to the lipid metabolism pathways, DEGs were also mainly enriched in the pathways implicated in endothelial cell proliferation, migration of epithelial cells, immune regulatory, and redox. GSEA shows that the up-regulated genes are mainly enriched in 'immune regulation pathways' and 'signal regulation' pathways, while the down-regulated genes are enriched in lipid metabolism pathways, 'regulation_of_nitric_oxide_synthase_activity' and the pathways involved in redox homeostasis, including 'monooxygenase regulation', 'peroxisomes' and 'oxygen binding'. A significant increase of reactive oxygen species and a remarkable reduction of GSH/GSSG ratio were respectively observed in the vascular tissues and plasma of Apoe-/- mice. In addition, endothelin-1 significantly increased in the vascular tissue and the plasma of Apoe-/- mice. Taken together, our results suggest that besides functioning in lipid metabolism, APOE may be an important signal regulator that mediates the expression of the genes related to the pathways involved in redox, inflammation, and endothelial function. Apoe-knockout-induced strong vascular oxidative stress is also the key factor contributing to atherosclerosis.
Collapse
Affiliation(s)
- Benrong Liu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China.
| | - Lei Fang
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Pei Mo
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Changnong Chen
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Yang Ji
- Department of Emergency, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Lihua Pang
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Huanzhen Chen
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Yichao Deng
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Wenchao Ou
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Shi-Ming Liu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China.
| |
Collapse
|
64
|
Wang B, Zhao P, Zhang P, Hu J, Liu Y, Xie M, He Y. 3D-printed tortuous vessels with Photodissociable and morphology-controllable ink. J Biomater Appl 2023:8853282231183984. [PMID: 37485893 DOI: 10.1177/08853282231183984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Acute ischemic stroke (AIS) is a high mortality cerebrovascular disease associated with vessel curvature. However, the relevant mechanism remains unclear due to a lack of appropriate tortuous vascular models to investigate and validate. This study explores the combination of projection-based 3D bioprinting (PBP) with photo-stimulus-responsive techniques to fabricate a sodium alginate (SA)/acrylamide (AAM) hydrogel vascular scaffold capable of bending deformation. The coordination of Fe3+ ions with carboxylate groups in the alginate chains of the vascular scaffold acts as a molecular switch, which can be dissociated through photoreduction to enable the deformation response. Fourier Transform Infrared (FTIR) and X-ray Photoelectron Spectroscopy (XPS) results verified the deformation principle. By subjecting the scaffold to UV light exposure, Fe3+ is reduced to Fe2+ in spatially selected regions, resulting in the release of strain and subsequent deformation. Furthermore, it also controlled the degree and direction of curvature of the vessels. The cell seeding experiment verified that the vascular scaffold showed excellent biocompatibility. Overall, our approach could be used to generate an in vitro model of curved vascular pathology to investigate the pathogenesis and provide new directions for the diagnosis and treatment of vascular diseases in the future.
Collapse
Affiliation(s)
- Biling Wang
- School of Mechatronics & Vehicle Engineering, East China Jiaotong University, Nanchang, China
- Engineering for Life Group (EFL), Suzhou, China
| | - Pengcheng Zhao
- School of Mechatronics & Vehicle Engineering, East China Jiaotong University, Nanchang, China
| | - Peng Zhang
- Engineering for Life Group (EFL), Suzhou, China
| | - Jun Hu
- School of Mechatronics & Vehicle Engineering, East China Jiaotong University, Nanchang, China
| | - Yande Liu
- School of Mechatronics & Vehicle Engineering, East China Jiaotong University, Nanchang, China
| | - Mingjun Xie
- Plastic and Reconstructive Surgery Center, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Yong He
- School of Mechatronics & Vehicle Engineering, East China Jiaotong University, Nanchang, China
- Engineering for Life Group (EFL), Suzhou, China
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
65
|
Bashore AC, Yan H, Xue C, Zhu LY, Kim E, Mawson T, Coronel J, Chung A, Ho S, Ross LS, Kissner M, Passegué E, Bauer RC, Maegdefessel L, Li M, Reilly MP. High-Dimensional Single-Cell Multimodal Landscape of Human Carotid Atherosclerosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.07.13.23292633. [PMID: 37502836 PMCID: PMC10370238 DOI: 10.1101/2023.07.13.23292633] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Background Atherosclerotic plaques are complex tissues composed of a heterogeneous mixture of cells. However, we have limited understanding of the comprehensive transcriptional and phenotypical landscape of the cells within these lesions. Methods To characterize the landscape of human carotid atherosclerosis in greater detail, we combined cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) and single-cell RNA sequencing (scRNA-seq) to classify all cell types within lesions (n=21; 13 symptomatic) to achieve a comprehensive multimodal understanding of the cellular identities of atherosclerosis and their association with clinical pathophysiology. Results We identified 25 distinct cell populations each having a unique multi-omic signature, including macrophages, T cells, NK cells, mast cells, B cells, plasma cells, neutrophils, dendritic cells, endothelial cells, fibroblasts, and smooth muscle cells (SMCs). Within the macrophage populations, we identified 2 proinflammatory subsets that were enriched in IL1B or C1Q expression, 2 distinct TREM2 positive foam cell subsets, one of which also expressed inflammatory genes, as well as subpopulations displaying a proliferative gene expression signature and one expressing SMC-specific genes and upregulation of fibrotic pathways. An in-depth characterization uncovered several subsets of SMCs and fibroblasts, including a SMC-derived foam cell. We localized this foamy SMC to the deep intima of coronary atherosclerotic lesions. Using CITE-seq data, we also developed the first flow cytometry panel, using cell surface proteins CD29, CD142, and CD90, to isolate SMC-derived cells from lesions. Last, we found that the proportion of efferocytotic macrophages, classically activated endothelial cells, contractile and modulated SMC-derived cell types were reduced, and inflammatory SMCs were enriched in plaques of clinically symptomatic vs. asymptomatic patients. Conclusions Our multimodal atlas of cell populations within atherosclerosis provides novel insights into the diversity, phenotype, location, isolation, and clinical relevance of the unique cellular composition of human carotid atherosclerosis. This facilitates both the mapping of cardiovascular disease susceptibility loci to specific cell types as well as the identification of novel molecular and cellular therapeutic targets for treatment of the disease.
Collapse
Affiliation(s)
- Alexander C Bashore
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York
| | - Hanying Yan
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Chenyi Xue
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York
| | - Lucie Y Zhu
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York
| | - Eunyoung Kim
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York
| | - Thomas Mawson
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York
| | - Johana Coronel
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York
| | - Allen Chung
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York
| | - Sebastian Ho
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York
| | - Leila S Ross
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York
| | - Michael Kissner
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University Irving Medical Center, New York
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University Irving Medical Center, New York
| | - Robert C Bauer
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York
| | - Lars Maegdefessel
- Department of Vascular and Endovascular Surgery, Technical University Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance
- Karolinksa Institute, Department of Medicine
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Muredach P Reilly
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York
- Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
66
|
Lee WE, Genetzakis E, Figtree GA. Novel Strategies in the Early Detection and Treatment of Endothelial Cell-Specific Mitochondrial Dysfunction in Coronary Artery Disease. Antioxidants (Basel) 2023; 12:1359. [PMID: 37507899 PMCID: PMC10376062 DOI: 10.3390/antiox12071359] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Although elevated cholesterol and other recognised cardiovascular risk factors are important in the development of coronary artery disease (CAD) and heart attack, the susceptibility of humans to this fatal process is distinct from other animals. Mitochondrial dysfunction of cells in the arterial wall, particularly the endothelium, has been strongly implicated in the pathogenesis of CAD. In this manuscript, we review the established evidence and mechanisms in detail and explore the potential opportunities arising from analysing mitochondrial function in patient-derived cells such as endothelial colony-forming cells easily cultured from venous blood. We discuss how emerging technology and knowledge may allow us to measure mitochondrial dysfunction as a potential biomarker for diagnosis and risk management. We also discuss the "pros and cons" of animal models of atherosclerosis, and how patient-derived cell models may provide opportunities to develop novel therapies relevant for humans. Finally, we review several targets that potentially alleviate mitochondrial dysfunction working both via direct and indirect mechanisms and evaluate the effect of several classes of compounds in the cardiovascular context.
Collapse
Affiliation(s)
- Weiqian E. Lee
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Elijah Genetzakis
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Gemma A. Figtree
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW 2065, Australia
| |
Collapse
|
67
|
Wang C, Meng XC, Huang C, Wang J, Liao YH, Huang Y, Liu R. Association between ambient air pollutants and lipid profile: A systematic review and meta-analysis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 262:115140. [PMID: 37348216 DOI: 10.1016/j.ecoenv.2023.115140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/29/2023] [Accepted: 06/11/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND Studies of the effects of atmospheric pollutants on lipid profiles remain inconsistent and controversial. AIM The study was aimed to investigate the relationship between the exposure to ambient air pollutants and variations in the blood lipid profiles in the population. METHODS A comprehensive search of three different databases (PubMed, Web of Science, and the Cochrane Library) until December 17, 2022, yielded 17 origional studies fulfilling the inclusion criteria for a meta-analysis. Aggregate effect measures and 95% confidence intervals (95% CI) for the relevant ambient air pollutants were deduced employing random effects models. RESULTS The collective meta-analysis indicated that long-term exposure to PM1, PM2.5, PM10 and CO showed a substantial correlation with TC (PM1: β = 2.04, 95%CI = 0.15-3.94; PM2.5: β = 1.11, 95%CI = 0.39-1.84; PM10: β = 1.70, 95%CI = 0.67-2.73; CO: β = 0.08, 95%CI = 0.06-0.10), PM10 exhibited a significant association with TG (β = 0. 537,95% CI = 0.09-0.97), whereas HDL-C demonstrated notable relationships with PM1, PM10, SO2 and CO (PM1: β = -2.38, 95%CI = -4.00 to -2.76; PM10: β = -0.77, 95%CI = -1.33 to -0.21; SO2: β = -0.91, 95%CI = -1.73 to -0.10; CO: β = -0.03, 95%CI = -0.05 to 0.00). PM2.5, PM10 also showed significant associations with LDL-C (PM2.5: β = 1.44 95%CI = 0.48-2.40; PM10: β = 1.62 95%CI = 0.90-2.34). Subgroup analysis revealed significant or stronger correlations predominantly in cohort study designs, with higher male comparisons, and in regions exhibiting elevated contaminant levels. CONCLUSION In summary, the analysis substantiates that ambient air pollutants can be recognized as potent contributors to alterations in lipid profiles, particularly particulate pollutants which exert more obvious effects on lipid profiles.
Collapse
Affiliation(s)
- Chun Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Xing-Chen Meng
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Chao Huang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Jia Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Ying-Hao Liao
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Yang Huang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Ran Liu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| |
Collapse
|
68
|
Srivastava RAK. A Review of Progress on Targeting LDL Receptor-Dependent and -Independent Pathways for the Treatment of Hypercholesterolemia, a Major Risk Factor of ASCVD. Cells 2023; 12:1648. [PMID: 37371118 DOI: 10.3390/cells12121648] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Since the discovery of the LDL receptor in 1973 by Brown and Goldstein as a causative protein in hypercholesterolemia, tremendous amounts of effort have gone into finding ways to manage high LDL cholesterol in familial hypercholesterolemic (HoFH and HeFH) individuals with loss-of-function mutations in the LDL receptor (LDLR) gene. Statins proved to be the first blockbuster drug, helping both HoFH and HeFH individuals by inhibiting the cholesterol synthesis pathway rate-limiting enzyme HMG-CoA reductase and inducing the LDL receptor. However, statins could not achieve the therapeutic goal of LDL. Other therapies targeting LDLR include PCSK9, which lowers LDLR by promoting LDLR degradation. Inducible degrader of LDLR (IDOL) also controls the LDLR protein, but an IDOL-based therapy is yet to be developed. Among the LDLR-independent pathways, such as angiopoietin-like 3 (ANGPTL3), apolipoprotein (apo) B, apoC-III and CETP, only ANGPTL3 offers the advantage of treating both HoFH and HeFH patients and showing relatively better preclinical and clinical efficacy in animal models and hypercholesterolemic individuals, respectively. While loss-of-LDLR-function mutations have been known for decades, gain-of-LDLR-function mutations have recently been identified in some individuals. The new information on gain of LDLR function, together with CRISPR-Cas9 genome/base editing technology to target LDLR and ANGPTL3, offers promise to HoFH and HeFH individuals who are at a higher risk of developing atherosclerotic cardiovascular disease (ASCVD).
Collapse
Affiliation(s)
- Rai Ajit K Srivastava
- Integrated Pharma Solutions LLC, Boston, MA 02101-02117, USA
- College of Professional Studies, Northeastern University, Boston, MA 02101-02117, USA
| |
Collapse
|
69
|
Gautam J, Kumari D, Aggarwal H, Gupta SK, Kasarla SS, Sarkar S, Priya MRK, Kamboj P, Kumar Y, Dikshit M. Characterization of lipid signatures in the plasma and insulin-sensitive tissues of the C57BL/6J mice fed on obesogenic diets. Biochim Biophys Acta Mol Cell Biol Lipids 2023:159348. [PMID: 37285928 DOI: 10.1016/j.bbalip.2023.159348] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 05/23/2023] [Accepted: 05/29/2023] [Indexed: 06/09/2023]
Abstract
Diet-induced obesity mouse models are widely utilized to investigate the underlying mechanisms of dyslipidemia, glucose intolerance, insulin resistance, hepatic steatosis, and type 2 diabetes mellitus (T2DM), as well as for screening potential drug compounds. However, there is limited knowledge regarding specific signature lipids that accurately reflect dietary disorders. In this study, we aimed to identify key lipid signatures using LC/MS-based untargeted lipidomics in the plasma, liver, adipose tissue (AT), and skeletal muscle tissues (SKM) of male C57BL/6J mice that were fed chow, LFD, or obesogenic diets (HFD, HFHF, and HFCD) for a duration of 20 weeks. Furthermore, we conducted a comprehensive lipid analysis to assess similarities and differences with human lipid profiles. The mice fed obesogenic diets exhibited weight gain, glucose intolerance, elevated BMI, glucose and insulin levels, and a fatty liver, resembling characteristics of T2DM and obesity in humans. In total, we identified approximately 368 lipids in plasma, 433 in the liver, 493 in AT, and 624 in SKM. Glycerolipids displayed distinct patterns across the tissues, differing from human findings. However, changes in sphingolipids, phospholipids, and the expression of inflammatory and fibrotic genes showed similarities to reported human findings. Significantly modulated pathways in the obesogenic diet-fed groups included ceramide de novo synthesis, sphingolipid remodeling, and the carboxylesterase pathway, while lipoprotein-mediated pathways were minimally affected.
Collapse
Affiliation(s)
- Jyoti Gautam
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Deepika Kumari
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Hobby Aggarwal
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Sonu Kumar Gupta
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Siva Swapna Kasarla
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Soumalya Sarkar
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - M R Kamla Priya
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Parul Kamboj
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Yashwant Kumar
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India.
| | - Madhu Dikshit
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India.
| |
Collapse
|
70
|
Nehme J, Altulea A, Gheorghe T, Demaria M. The effects of macronutrients metabolism on cellular and organismal aging. Biomed J 2023; 46:100585. [PMID: 36801257 PMCID: PMC10209809 DOI: 10.1016/j.bj.2023.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/08/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023] Open
Abstract
Evidence supports the notion that metabolic pathways are major regulators of organismal aging, and that metabolic perturbations can extend health- and lifespan. For this reason, dietary interventions and compounds perturbing metabolism are currently explored as anti-aging strategies. A common target for metabolic interventions delaying aging is cellular senescence, a state of stable growth arrest that is accompanied by various structural and functional changes including the activation of a pro-inflammatory secretome. Here, we summarize the current knowledge on the molecular and cellular events associated with carbohydrate, lipid and protein metabolism, and define how macronutrients can regulate induction or prevention of cellular senescence. We discuss how various dietary interventions can achieve prevention of disease and extension of healthy longevity by partially modulating senescence-associated phenotypes. We also emphasize the importance of developing personalized nutritional interventions that take into account the current health and age status of the individual.
Collapse
Affiliation(s)
- Jamil Nehme
- University of Groningen, European Research Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, Netherlands
| | - Abdullah Altulea
- University of Groningen, European Research Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, Netherlands
| | - Teodora Gheorghe
- University of Groningen, European Research Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, Netherlands
| | - Marco Demaria
- University of Groningen, European Research Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, Netherlands.
| |
Collapse
|
71
|
Wang J, Venugopal J, Silaghi P, Su EJ, Guo C, Lawrence DA, Eitzman DT. Beta1-receptor blockade attenuates atherosclerosis progression following traumatic brain injury in apolipoprotein E deficient mice. PLoS One 2023; 18:e0285499. [PMID: 37235594 PMCID: PMC10218730 DOI: 10.1371/journal.pone.0285499] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Traumatic brain injury (TBI) is associated with cardiovascular mortality in humans. Enhanced sympathetic activity following TBI may contribute to accelerated atherosclerosis. The effect of beta1-adrenergic receptor blockade on atherosclerosis progression induced by TBI was studied in apolipoprotein E deficient mice. Mice were treated with metoprolol or vehicle following TBI or sham operation. Mice treated with metoprolol experienced a reduced heart rate with no difference in blood pressure. Six weeks following TBI, mice were sacrificed for analysis of atherosclerosis. Total surface area and lesion thickness, analyzed at the level of the aortic valve, was found to be increased in mice receiving TBI with vehicle treatment but this effect was ameliorated in TBI mice receiving metoprolol. No effect of metoprolol on atherosclerosis was observed in mice receiving only sham operation. In conclusion, accelerated atherosclerosis following TBI is reduced with beta-adrenergic receptor antagonism. Beta blockers may be useful to reduce vascular risk associated with TBI.
Collapse
Affiliation(s)
- Jintao Wang
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jessica Venugopal
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Paul Silaghi
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Enming J. Su
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Chiao Guo
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Daniel A. Lawrence
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Daniel T. Eitzman
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
72
|
Zhang PP, Zhuo BY, Duan ZW, Li X, Huang SL, Cao Q, Zhao T, Wei SL, Hu XH, Zhang Y. Marein reduces lipid levels via modulating the PI3K/AKT/mTOR pathway to induce lipophagy. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116523. [PMID: 37080364 DOI: 10.1016/j.jep.2023.116523] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/04/2023] [Accepted: 04/18/2023] [Indexed: 05/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The capitulum of Coreopsis tinctoria Nutt. (CT, Xue-Ju in Chinese) is a precious medicine in Xinjiang Uygur Autonomous region of China. The Coreopsis tinctoria Nutt. is used to prevent and treat dyslipidemia, coronary heart disease, etc. Recent studies have shown that its extract has a pharmacological effect on hyperlipidemia and hyperglycemia. AIM OF THE STUDY The study aimed to systematically evaluate the lipid-lowering activity of CT through a mice model of hyperlipidemia and a human hepatoma G2 (HepG2) cells model of lipid accumulation, and to investigate its main active components and mechanism. MATERIALS AND METHODS Biochemical analysis of blood/liver lipids and liver histopathology were used to evaluate the effect of the aqueous extract of Coreopsis tinctoria Nutt. (AECT) on hyperlipidemia mice. High-performance liquid chromatography (HPLC) analysis was used to identify the main components in the AECT. Oil red O staining, immunofluorescence, western blotting, and determination of the total cholesterol (TC), total triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C) were used to further study the effect and potential mechanism of the AECT main components on sodium oleate-induced lipid accumulation in HepG2 cells. RESULTS We confirmed the lipid-lowering activity of the aqueous extract and further identified flavonoids as its main components. Among them, five Coreopsis tinctoria Nutt. flavonoids mixture (FM) significantly reduced lipid droplet area, lipid content, TC, TG, and LDL-C levels, and elevated HDL-C levels in HepG2 cells induced by sodium oleate. Furthermore, they increased lipophagy in HepG2 lipid-accumulating cells, while decreasing the ratio of p-PI3K/PI3K, p-AKT/AKT, and p-mTOR/mTOR. Most importantly, marein may be a key component. CONCLUSIONS Our study demonstrated that AECT, with flavonoids as the main component, can improve diet-induced hyperlipidemia in obese mice. Among the main five flavonoids, marein plays a key role in promoting lipophagy by regulating the PI3K/AKT/mTOR pathway, resulting in a lipid-lowering effect.
Collapse
Affiliation(s)
- Pei-Pei Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Bing-Yu Zhuo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Zi-Wei Duan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Xin Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Song-Li Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Qian Cao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Ting Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China; Engineering Research Center of Good Agricultural Practice for Chinese Crude Drugs, Ministry of Education, Beijing, 100102, China.
| | - Sheng-Li Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China; Engineering Research Center of Good Agricultural Practice for Chinese Crude Drugs, Ministry of Education, Beijing, 100102, China.
| | - Xiu-Hua Hu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China; Engineering Research Center of Good Agricultural Practice for Chinese Crude Drugs, Ministry of Education, Beijing, 100102, China.
| | - Yuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China; Engineering Research Center of Good Agricultural Practice for Chinese Crude Drugs, Ministry of Education, Beijing, 100102, China.
| |
Collapse
|
73
|
Mu X, Gerhard-Herman MD, Zhang YS. Building Blood Vessel Chips with Enhanced Physiological Relevance. ADVANCED MATERIALS TECHNOLOGIES 2023; 8:2201778. [PMID: 37693798 PMCID: PMC10489284 DOI: 10.1002/admt.202201778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Indexed: 09/12/2023]
Abstract
Blood vessel chips are bioengineered microdevices, consisting of biomaterials, human cells, and microstructures, which recapitulate essential vascular structure and physiology and allow a well-controlled microenvironment and spatial-temporal readouts. Blood vessel chips afford promising opportunities to understand molecular and cellular mechanisms underlying a range of vascular diseases. The physiological relevance is key to these blood vessel chips that rely on bioinspired strategies and bioengineering approaches to translate vascular physiology into artificial units. Here, we discuss several critical aspects of vascular physiology, including morphology, material composition, mechanical properties, flow dynamics, and mass transport, which provide essential guidelines and a valuable source of bioinspiration for the rational design of blood vessel chips. We also review state-of-art blood vessel chips that exhibit important physiological features of the vessel and reveal crucial insights into the biological processes and disease pathogenesis, including rare diseases, with notable implications for drug screening and clinical trials. We envision that the advances in biomaterials, biofabrication, and stem cells improve the physiological relevance of blood vessel chips, which, along with the close collaborations between clinicians and bioengineers, enable their widespread utility.
Collapse
Affiliation(s)
- Xuan Mu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Marie Denise Gerhard-Herman
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
74
|
Sridharan D, Pracha N, Rana SJ, Ahmed S, Dewani AJ, Alvi SB, Mergaye M, Ahmed U, Khan M. Preclinical Large Animal Porcine Models for Cardiac Regeneration and Its Clinical Translation: Role of hiPSC-Derived Cardiomyocytes. Cells 2023; 12:cells12071090. [PMID: 37048163 PMCID: PMC10093073 DOI: 10.3390/cells12071090] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/14/2023] Open
Abstract
Myocardial Infarction (MI) occurs due to a blockage in the coronary artery resulting in ischemia and necrosis of cardiomyocytes in the left ventricular heart muscle. The dying cardiac tissue is replaced with fibrous scar tissue, causing a decrease in myocardial contractility and thus affecting the functional capacity of the myocardium. Treatments, such as stent placements, cardiac bypasses, or transplants are beneficial but with many limitations, and may decrease the overall life expectancy due to related complications. In recent years, with the advent of human induced pluripotent stem cells (hiPSCs), newer avenues using cell-based approaches for the treatment of MI have emerged as a potential for cardiac regeneration. While hiPSCs and their derived differentiated cells are promising candidates, their translatability for clinical applications has been hindered due to poor preclinical reproducibility. Various preclinical animal models for MI, ranging from mice to non-human primates, have been adopted in cardiovascular research to mimic MI in humans. Therefore, a comprehensive literature review was essential to elucidate the factors affecting the reproducibility and translatability of large animal models. In this review article, we have discussed different animal models available for studying stem-cell transplantation in cardiovascular applications, mainly focusing on the highly translatable porcine MI model.
Collapse
Affiliation(s)
- Divya Sridharan
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Nooruddin Pracha
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Schaza Javed Rana
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, Northeast Georgia Medical Center, Gainesville, GA 30501, USA
| | - Salmman Ahmed
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
- Lake Erie College of Osteopathic Medicine (LECOM), Erie, PA 16509, USA
| | - Anam J Dewani
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Chemistry & Biochemistry, The University of Toledo, Toledo, OH 43606, USA
| | - Syed Baseeruddin Alvi
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Muhamad Mergaye
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Uzair Ahmed
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Mahmood Khan
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
75
|
Schwarz N, Fernando S, Chen YC, Salagaras T, Rao SR, Liyanage S, Williamson AE, Toledo-Flores D, Dimasi C, Sargeant TJ, Manavis J, Eddy E, Kanellakis P, Thompson PL, Tan JTM, Snel MF, Bursill CA, Nicholls SJ, Peter K, Psaltis PJ. Colchicine exerts anti-atherosclerotic and -plaque-stabilizing effects targeting foam cell formation. FASEB J 2023; 37:e22846. [PMID: 36856983 DOI: 10.1096/fj.202201469r] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 02/02/2023] [Accepted: 02/15/2023] [Indexed: 03/02/2023]
Abstract
Colchicine is a broad-acting anti-inflammatory agent that has attracted interest for repurposing in atherosclerotic cardiovascular disease. Here, we studied its ability at a human equivalent dose of 0.5 mg/day to modify plaque formation and composition in murine atherosclerosis and investigated its actions on macrophage responses to atherogenic stimuli in vitro. In atherosclerosis induced by high-cholesterol diet, Apoe-/- mice treated with colchicine had 50% reduction in aortic oil Red O+ plaque area compared to saline control (p = .001) and lower oil Red O+ staining of aortic sinus lesions (p = .03). In vitro, addition of 10 nM colchicine inhibited foam cell formation from murine and human macrophages after treatment with oxidized LDL (ox-LDL). Mechanistically, colchicine downregulated glycosylation and surface expression of the ox-LDL uptake receptor, CD36, and reduced CD36+ staining in aortic sinus plaques. It also decreased macrophage uptake of cholesterol crystals, resulting in lower intracellular lysosomal activity, inhibition of the NLRP3 inflammasome, and reduced secretion of IL-1β and IL-18. Colchicine's anti-atherosclerotic actions were accentuated in a mouse model of unstable plaque induced by carotid artery tandem stenosis surgery, where it decreased lesion size by 48% (p = .01), reduced lipid (p = .006) and necrotic core area (p = .007), increased collagen content and cap-to-necrotic core ratio (p = .05), and attenuated plaque neutrophil extracellular traps (p < .001). At low dose, colchicine's effects were not accompanied by the evidence of microtubule depolymerization. Together, these results show that colchicine exerts anti-atherosclerotic and plaque-stabilizing effects at low dose by inhibiting foam cell formation and cholesterol crystal-induced inflammation. This provides a new framework to support its repurposing for atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Nisha Schwarz
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Sanuja Fernando
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Yung-Chih Chen
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Thalia Salagaras
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Sushma R Rao
- Proteomics, Metabolomics and MS-Imaging Facility, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Sanuri Liyanage
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Anna E Williamson
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Deborah Toledo-Flores
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Catherine Dimasi
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Timothy J Sargeant
- Lifelong Health in Ageing, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Jim Manavis
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Eleanor Eddy
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter Kanellakis
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter L Thompson
- Faculty of Health and Medical Sciences, Internal Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Joanne T M Tan
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Marten F Snel
- Proteomics, Metabolomics and MS-Imaging Facility, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Christina A Bursill
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Stephen J Nicholls
- Monash Cardiovascular Research Centre, Monash University, Melbourne, Victoria, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter J Psaltis
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
76
|
Lohkamp KJ, van den Hoek AM, Solé-Guardia G, Lisovets M, Alves Hoffmann T, Velanaki K, Geenen B, Verweij V, Morrison MC, Kleemann R, Wiesmann M, Kiliaan AJ. The Preventive Effect of Exercise and Oral Branched-Chain Amino Acid Supplementation on Obesity-Induced Brain Changes in Ldlr−/−.Leiden Mice. Nutrients 2023; 15:nu15071716. [PMID: 37049556 PMCID: PMC10097391 DOI: 10.3390/nu15071716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Exercise and dietary interventions are promising approaches to tackle obesity and its obesogenic effects on the brain. We investigated the impact of exercise and possible synergistic effects of exercise and branched-chain amino acids (BCAA) supplementation on the brain and behavior in high-fat-diet (HFD)-induced obese Ldlr−/−.Leiden mice. Baseline measurements were performed in chow-fed Ldlr−/−.Leiden mice to assess metabolic risk factors, cognition, and brain structure using magnetic resonance imaging. Thereafter, a subgroup was sacrificed, serving as a healthy reference. The remaining mice were fed an HFD and divided into three groups: (i) no exercise, (ii) exercise, or (iii) exercise and dietary BCAA. Mice were followed for 6 months and aforementioned tests were repeated. We found that exercise alone changed cerebral blood flow, attenuated white matter loss, and reduced neuroinflammation compared to non-exercising HFD-fed mice. Contrarily, no favorable effects of exercise on the brain were found in combination with BCAA, and neuroinflammation was increased. However, cognition was slightly improved in exercising mice on BCAA. Moreover, BCAA and exercise increased the percentage of epididymal white adipose tissue and muscle weight, decreased body weight and fasting insulin levels, improved the circadian rhythm, and transiently improved grip strength. In conclusion, BCAA should be supplemented with caution, although beneficial effects on metabolism, behavior, and cognition were observed.
Collapse
Affiliation(s)
- Klara J. Lohkamp
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Anita M. van den Hoek
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands; (A.M.v.d.H.); (M.C.M.); (R.K.)
| | - Gemma Solé-Guardia
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Maria Lisovets
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Talissa Alves Hoffmann
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Konstantina Velanaki
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Bram Geenen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Vivienne Verweij
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Martine C. Morrison
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands; (A.M.v.d.H.); (M.C.M.); (R.K.)
| | - Robert Kleemann
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands; (A.M.v.d.H.); (M.C.M.); (R.K.)
| | - Maximilian Wiesmann
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Amanda J. Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
- Correspondence:
| |
Collapse
|
77
|
Karere GM, Glenn JP, Li G, Konar A, VandeBerg JL, Cox LA. Potential miRNA biomarkers and therapeutic targets for early atherosclerotic lesions. Sci Rep 2023; 13:3467. [PMID: 36859458 PMCID: PMC9977938 DOI: 10.1038/s41598-023-29074-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/30/2023] [Indexed: 03/03/2023] Open
Abstract
Identification of potential therapeutic targets and biomarkers indicative of burden of early atherosclerosis that occur prior to advancement to life-threatening unstable plaques is the key to eradication of CAD prevalence and incidences. We challenged 16 baboons with a high cholesterol, high fat diet for 2 years and evaluated early-stage atherosclerotic lesions (fatty streaks, FS, and fibrous plaques, FP) in formalin-fixed common iliac arteries (CIA). We used small RNA sequencing to identify expressed miRNAs in CIA and in baseline blood samples of the same animals. We found 412 expressed miRNAs in CIA and 356 in blood samples. Eight miRNAs (miR-7975, -486-5p, -451a, -191-5p, -148a-3p, -17-5p, -378c, and -144-3p) were differentially expressed between paired fatty streak lesion and no-lesion sites of the tissue, and 27 miRNAs (e.g., miR-92a-3p, -5001, -342-3p, miR-28-3p, -21-5p, -221-3p, 146a-5p, and -16-5p) in fibrous plaques. The expression of 14 blood miRNAs significantly correlated with extent of lesions and the number of plaques. We identified coordinately regulated miRNA-gene networks in which miR-17-5p and miR-146a-5p are central hubs and miR-5001 and miR-7975 are potentially novel miRNAs associated with early atherosclerosis. In summary, we have identified miRNAs expressed in lesions and in blood that correlate with lesion burden and are potential therapeutic targets and biomarkers. These findings are a first step in elucidating miRNA regulated molecular mechanisms that underlie early atherosclerosis in a baboon model, enabling translation of our findings to humans.
Collapse
Affiliation(s)
- Genesio M Karere
- Department of Internal Medicine, Section on Molecular Medicine, Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| | - Jeremy P Glenn
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Ge Li
- Department of Internal Medicine, Section on Molecular Medicine, Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Ayati Konar
- Department of Internal Medicine, Section on Molecular Medicine, Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - John L VandeBerg
- Department of Human Genetics, South Texas Diabetes and Obesity Institute, The University of Texas Rio Grande Valley, Brownville, Harlingen, Edinburg, TX, 78520, USA
| | - Laura A Cox
- Department of Internal Medicine, Section on Molecular Medicine, Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| |
Collapse
|
78
|
Huang HS, Lin YE, Panyod S, Chen RA, Lin YC, Chai LMX, Hsu CC, Wu WK, Lu KH, Huang YJ, Sheen LY. Anti-depressive-like and cognitive impairment alleviation effects of Gastrodia elata Blume water extract is related to gut microbiome remodeling in ApoE -/- mice exposed to unpredictable chronic mild stress. JOURNAL OF ETHNOPHARMACOLOGY 2023; 302:115872. [PMID: 36343797 DOI: 10.1016/j.jep.2022.115872] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGY RELEVANCE Gastrodia elata Blume (GE) is a traditional Chinese dietary therapy used to treat neurological disorders. Gastrodia elata Blume water extract (WGE) has been shown to ameliorate inflammation and improve social frustration in mice in a chronic social defeat model. However, studies on the anti-depressive-like effects and cognitive impairment alleviation related to the impact of WGE on the gut microbiome of ApoE-/- mice remain elusive. AIM OF THE STUDY The present study aimed to investigate the anti-depressive-like effect and cognitive impairment alleviation and mechanisms of WGE in ApoE-/- mice subjected to unpredictable chronic mild stress (UCMS), as well as its impact on the gut microbiome of the mice. MATERIALS AND METHODS Sixty ApoE-/- mice (6 months old) were randomly grouped into six groups: control, UCMS, WGE groups [5, 10, 20 mL WGE/kg body weight (bw) + UCMS], and a positive group (fluoxetine 20 mg/kg bw + UCMS). After four weeks of the UCMS paradigm, the sucrose preference, novel object recognition, and open field tests were conducted. The neurotransmitters serotonin (5-HT), dopamine (DA) and their metabolites were measured in the prefrontal cortex. Serum was collected to measure corticosterone and amyloid-42 (Aβ-42) levels. Feces were collected, and the gut microbiome was analyzed. RESULTS WGE restored sucrose preference, exploratory behavior, recognition ability, and decreased the levels of serum corticosterone and Aβ-42 in ApoE-/- mice to alleviate depressive-like behavior and cognitive impairment. Furthermore, WGE regulated the monoamine neurotransmitter via reduced the 5-HT and DA turnover rates in the prefrontal cortex. Moreover, WGE elevated the levels of potentially beneficial bacteria such as Bifidobacterium, Akkermansia, Alloprevotella, Defluviitaleaceae_UCG-011, and Bifidobacterium pseudolongum as well as balanced fecal short-chain fatty acids (SCFAs). CONCLUSION WGE demonstrates anti-depressive-like effects, cognitive impairment alleviation, and gut microbiome and metabolite regulation in ApoE-/- mice. Our results support the possibility of developing a functional and complementary medicine to prevent or alleviate depression and cognitive decline using WGE in CVDs patients.
Collapse
Affiliation(s)
- Huai-Syuan Huang
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan.
| | - Yu-En Lin
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan.
| | - Suraphan Panyod
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan.
| | - Rou-An Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan.
| | - Ying-Cheng Lin
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan.
| | | | - Cheng-Chih Hsu
- Department of Chemistry, National Taiwan University, Taipei, Taiwan.
| | - Wei-Kai Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan.
| | - Kuan-Hung Lu
- Institute of Food Safety and Health, National Taiwan University, Taipei, Taiwan; Institute of Environmental and Occupational Health Sciences, National Taiwan University, Taipei, Taiwan.
| | - Yun-Ju Huang
- Department of Biotechnology and Food Technology, Southern Taiwan University of Science and Technology, Tainan, Taiwan.
| | - Lee-Yan Sheen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan; Center for Food and Biomolecules, National Taiwan University, Taipei, Taiwan; National Center for Food Safety Education and Research, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
79
|
Windler E, Beil FU, Berthold HK, Gouni-Berthold I, Kassner U, Klose G, Lorkowski S, März W, Parhofer KG, Plat J, Silbernagel G, Steinhagen-Thiessen E, Weingärtner O, Zyriax BC, Lütjohann D. Phytosterols and Cardiovascular Risk Evaluated against the Background of Phytosterolemia Cases-A German Expert Panel Statement. Nutrients 2023; 15:nu15040828. [PMID: 36839186 PMCID: PMC9963617 DOI: 10.3390/nu15040828] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/08/2023] Open
Abstract
Phytosterols (PSs) have been proposed as dietary means to lower plasma LDL-C. However, concerns are raised that PSs may exert atherogenic effects, which would offset this benefit. Phytosterolemia was thought to mimic increased plasma PSs observed after the consumption of PS-enriched foods. This expert statement examines the possibility of specific atherogenicity of PSs based on sterol metabolism, experimental, animal, and human data. Observational studies show no evidence that plasma PS concentrations would be associated with an increased risk of atherosclerosis or cardiovascular (CV) events. Since variants of the ABCG5/8 transporter affect the absorption of cholesterol and non-cholesterol sterols, Mendelian randomization studies examining the effects of ABCG5/8 polymorphisms cannot support or refute the potential atherogenic effects of PSs due to pleiotropy. In homozygous patients with phytosterolemia, total PS concentrations are ~4000% higher than under physiological conditions. The prevalence of atherosclerosis in these individuals is variable and may mainly relate to concomitant elevated LDL-C. Consuming PS-enriched foods increases PS concentrations by ~35%. Hence, PSs, on a molar basis, would need to have 20-40 times higher atherogenicity than cholesterol to offset their cholesterol reduction benefit. Based on their LDL-C lowering and absence of adverse safety signals, PSs offer a dietary approach to cholesterol management. However, their clinical benefits have not been established in long-term CV endpoint studies.
Collapse
Affiliation(s)
- Eberhard Windler
- Preventive Medicine, University Heart Center, University Hospital Hamburg-Eppendorf, Hamburg-Eppendorf, Martinistr. 52-Bldg. N26, 20246 Hamburg, Germany
| | - Frank-Ulrich Beil
- Ambulanzzentrum, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Heiner K. Berthold
- Department of Internal Medicine and Geriatrics, Bethel Clinic, 33611 Bielefeld, Germany
| | - Ioanna Gouni-Berthold
- Center for Endocrinology, Diabetes and Preventive Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Ursula Kassner
- Lipid Clinic at the Interdisciplinary Metabolism Center, Charite-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Gerald Klose
- Praxen Dres. T. Beckenbauer & S. Maierhof, Am Markt 11, 28195 Bremen und Dres. I. van de Loo & K. Spieker, Gerold Janssen Straße 2 A, 28359 Bremen, Germany
| | - Stefan Lorkowski
- Institute of Nutritional Science and Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Friedrich Schiller University Jena, Dornburger Str. 25, 07743 Jena, Germany
| | - Winfried März
- SYNLAB Akademie für Ärztliche Fortbildung, SYNLAB Holding Deutschland GmbH, P5,7, 68161 Mannheim, Germany
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8010 Graz, Austria
- Correspondence:
| | - Klaus G. Parhofer
- Medizinische Klinik IV, Klinikum der Universität München, Grosshadern, Marchioninistr. 15, 81377 München, Germany
| | - Jogchum Plat
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Günter Silbernagel
- Division of Vascular Medicine, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Elisabeth Steinhagen-Thiessen
- Arbeitsbereich Lipidstoffwechsel der Medizinischen Klinik für Endokrinologie und Stoffwechselmedizin, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Oliver Weingärtner
- Klinik für Innere Medizin I, Universitätskliniken Jena, Friedrich-Schiller-Universität Jena, 07743 Jena, Germany
| | - Birgit-Christiane Zyriax
- Midwifery Science—Health Care Research and Prevention, Research Group, Preventive Medicine and Nutrition, Institute for Health Services Research in Dermatology and Nursing (IVDP), University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Clinics Bonn, 53127 Bonn, Germany
| |
Collapse
|
80
|
Wu Q, Liang X, Hou X, Song Z, Bouhamdan M, Qiu Y, Koike Y, Rajagopalan C, Wei HG, Jiang H, Hish G, Zhang J, Chen YE, Jin JP, Xu J, Zhang K, Sun F. Cystic fibrosis rabbits develop spontaneous hepatobiliary lesions and CF-associated liver disease (CFLD)-like phenotypes. PNAS NEXUS 2023; 2:pgac306. [PMID: 36712930 PMCID: PMC9832953 DOI: 10.1093/pnasnexus/pgac306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 12/22/2022] [Indexed: 12/25/2022]
Abstract
Cystic fibrosis (CF) is an autosomal recessive genetic disease affecting multiple organs. Approximately 30% CF patients develop CF-related liver disease (CFLD), which is the third most common cause of morbidity and mortality of CF. CFLD is progressive, and many of the severe forms eventually need liver transplantation. The mechanistic studies and therapeutic interventions to CFLD are unfortunately very limited. Utilizing the CRISPR/Cas9 technology, we recently generated CF rabbits by introducing mutations to the rabbit CF transmembrane conductance regulator (CFTR) gene. Here we report the liver phenotypes and mechanistic insights into the liver pathogenesis in these animals. CF rabbits develop spontaneous hepatobiliary lesions and abnormal biliary secretion accompanied with altered bile acid profiles. They exhibit nonalcoholic steatohepatitis (NASH)-like phenotypes, characterized by hepatic inflammation, steatosis, and fibrosis, as well as altered lipid profiles and diminished glycogen storage. Mechanistically, our data reveal that multiple stress-induced metabolic regulators involved in hepatic lipid homeostasis were up-regulated in the livers of CF-rabbits, and that endoplasmic reticulum (ER) stress response mediated through IRE1α-XBP1 axis as well as NF-κB- and JNK-mediated inflammatory responses prevail in CF rabbit livers. These findings show that CF rabbits manifest many CFLD-like phenotypes and suggest targeting hepatic ER stress and inflammatory pathways for potential CFLD treatment.
Collapse
Affiliation(s)
- Qingtian Wu
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xiubin Liang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Xia Hou
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Zhenfeng Song
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Mohamad Bouhamdan
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yining Qiu
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yui Koike
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Carthic Rajagopalan
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Hong-Guang Wei
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Hong Jiang
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Gerry Hish
- Laboratory Animal Resources, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jifeng Zhang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Y Eugene Chen
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jian-Ping Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jie Xu
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Fei Sun
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
81
|
Heriansyah T, Dimiati H, Hadi TF, Umara DA, Riandi LV, Fajri F, Santosa SF, Wihastuti TA, Kumboyono K. Ascorbic Acid vs Calcitriol in Influencing Monocyte Chemoattractant Protein-1, Nitric Oxide, Superoxide Dismutase, as Markers of Endothelial Dysfunction: In Vivo Study in Atherosclerosis Rat Model. Vasc Health Risk Manag 2023; 19:139-144. [PMID: 36936550 PMCID: PMC10019521 DOI: 10.2147/vhrm.s401521] [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: 12/19/2022] [Accepted: 02/21/2023] [Indexed: 03/13/2023] Open
Abstract
Introduction Ascorbic acid and calcitriol were frequently utilized in conjunction as therapy during the COVID-19 pandemic, and individuals with minor symptoms had notable improvements. There have been a few studies, often with conflicting findings, that examine the use of them for endothelium restoration and numerous clinical trial studies that failed to establish the efficacy. The aim of this study was to find the efficacy of ascorbic acid compared to calcitriol on the inflammatory markers monocyte chemoattractant protein-1 (MCP-1), nitric oxide (NO), and superoxide dismutase (SOD), as protective agents which play an important role in the early stages of atherosclerosis formation. This study was an experimental in vivo study. Methods The total of 24 male Rattus norvegicus strain Wistar rats were divided into 4 groups, namely: control/normal group (N), atherosclerosis group (DL) given atherogenic diet, atherosclerosis group given atherogenic diet and ascorbic acid (DLC), and atherosclerosis group given atherogenic diet and calcitriol (DLD) treatment for 30 days. Results Ascorbic acid and calcitriol treatment was significantly effective (P<0.05) in lowering expression of MCP-1 and increasing NO and SOD level. Calcitriol was superior to ascorbic acid in increasing SOD (P<0.05). There was no significant difference between ascorbic acid and calcitriol in decreasing MCP-1 and increasing NO (P>0.05). Discussion Both treatments could reduce MCP-1, and increase NO and SOD by increasing antioxidants. In this study calcitriol was superior to ascorbic acid in increasing SOD, but not NO and decreasing MCP-1. According to the theory, it was found that calcitriol through nuclear factor erythroid 2-related factor 2 (Nrf2) causes a direct increase in the amount of SOD. Nrf2 is an emerging regulator of cellular resistance to oxidants. Conclusion Ascorbic acid and calcitriol treatment was able to reduce MCP-1 and increase NO and SOD in atherosclerosis rat. Calcitriol was significantly superior in increasing SOD levels compared to ascorbic acid.
Collapse
Affiliation(s)
- Teuku Heriansyah
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| | - Herlina Dimiati
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| | - Tjut Farahiya Hadi
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
- Correspondence: Tjut Farahiya Hadi, Email
| | - Dimas Arya Umara
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| | - Lian Varis Riandi
- Department of Parasitology, Faculty of Veterinary Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| | - Fauzan Fajri
- Department of Animal Model, Faculty of Veterinary Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| | - Sukmawan Fajar Santosa
- Integrated Research Laboratory, Faculty of Veterinary Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| | - Titin Andri Wihastuti
- Department of Basic Nursing, Faculty of Health Sciences, Universitas Brawijaya, Malang, Jawa Timur, Indonesia
| | - Kumboyono Kumboyono
- Department of Nursing, Faculty of Health Sciences, Universitas Brawijaya, Malang, Indonesia
| |
Collapse
|
82
|
Busnelli M, Manzini S, Colombo A, Franchi E, Lääperi M, Laaksonen R, Chiesa G. Effect of Diets on Plasma and Aorta Lipidome: A Study in the apoE Knockout Mouse Model. Mol Nutr Food Res 2023; 67:e2200367. [PMID: 36419336 DOI: 10.1002/mnfr.202200367] [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: 06/06/2022] [Revised: 09/20/2022] [Indexed: 11/27/2022]
Abstract
SCOPE Specific lipid molecules circulating in plasma at low concentrations have emerged as biomarkers of atherosclerotic risk. The aim of the present study is that of evaluating, in an athero-prone mouse model, how different diets can affect plasma and aorta lipidome. METHODS AND RESULTS Thirty-six apoE knockout mice are divided in three groups and feed 12 weeks with diets differing for cholesterol and fatty acid content. Atherosclerosis is measured at the aortic sinus and aorta. Lipids are quantified in plasma and aorta with mass spectrometry. The cholesterol content of the diets is the main driver of lipid accumulation in plasma and aorta. The fatty acid composition of the diets affects plasma levels both of essential (linoleic acid) and nonessential (myristic and arachidonic acid) ones. Lipidomics show a comparable distribution, in plasma and aorta, of the main lipid components of oxidized LDL, including cholesteryl esters and lysophosphatidylcholines. Interestingly, lactosylceramide, glucosyl/galactosylceramide, and individual ceramide species are found to accumulate in diseased aortic segments. CONCLUSION Both the cholesterol and fatty acid content of the diets profoundly affect plasma lipidome. Aorta lipidome is likewise affected with the accumulation of specific lipids known as markers of atherosclerosis.
Collapse
Affiliation(s)
- Marco Busnelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | - Stefano Manzini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | - Alice Colombo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | - Elsa Franchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | | | - Reijo Laaksonen
- Zora Biosciences Oy, Espoo, 02150, Finland.,Finnish Cardiovascular Research Center, University of Tampere, Tampere, 33520, Finland
| | - Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| |
Collapse
|
83
|
Hindy G, Tyrrell DJ, Vasbinder A, Wei C, Presswalla F, Wang H, Blakely P, Ozel AB, Graham S, Holton GH, Dowsett J, Fahed AC, Amadi KM, Erne GK, Tekmulla A, Ismail A, Launius C, Sotoodehnia N, Pankow JS, Thørner LW, Erikstrup C, Pedersen OB, Banasik K, Brunak S, Ullum H, Eugen-Olsen J, Ostrowski SR, on behalf of the DBDS Consortium, Haas ME, Nielsen JB, Lotta LA, on behalf of the Regeneron Genetics Center, Engström G, Melander O, Orho-Melander M, Zhao L, Murthy VL, Pinsky DJ, Willer CJ, Heckbert SR, Reiser J, Goldstein DR, Desch KC, Hayek SS. Increased soluble urokinase plasminogen activator levels modulate monocyte function to promote atherosclerosis. J Clin Invest 2022; 132:e158788. [PMID: 36194491 PMCID: PMC9754000 DOI: 10.1172/jci158788] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 09/29/2022] [Indexed: 01/26/2023] Open
Abstract
People with kidney disease are disproportionately affected by atherosclerosis for unclear reasons. Soluble urokinase plasminogen activator receptor (suPAR) is an immune-derived mediator of kidney disease, levels of which are strongly associated with cardiovascular outcomes. We assessed suPAR's pathogenic involvement in atherosclerosis using epidemiologic, genetic, and experimental approaches. We found serum suPAR levels to be predictive of coronary artery calcification and cardiovascular events in 5,406 participants without known coronary disease. In a genome-wide association meta-analysis including over 25,000 individuals, we identified a missense variant in the plasminogen activator, urokinase receptor (PLAUR) gene (rs4760), confirmed experimentally to lead to higher suPAR levels. Mendelian randomization analysis in the UK Biobank using rs4760 indicated a causal association between genetically predicted suPAR levels and atherosclerotic phenotypes. In an experimental model of atherosclerosis, proprotein convertase subtilisin/kexin-9 (Pcsk9) transfection in mice overexpressing suPAR (suPARTg) led to substantially increased atherosclerotic plaques with necrotic cores and macrophage infiltration compared with those in WT mice, despite similar cholesterol levels. Prior to induction of atherosclerosis, aortas of suPARTg mice excreted higher levels of CCL2 and had higher monocyte counts compared with WT aortas. Aortic and circulating suPARTg monocytes exhibited a proinflammatory profile and enhanced chemotaxis. These findings characterize suPAR as a pathogenic factor for atherosclerosis acting at least partially through modulation of monocyte function.
Collapse
Affiliation(s)
- George Hindy
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Population Medicine, Qatar University College of Medicine, QU Health, Doha, Qatar
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Daniel J. Tyrrell
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Alexi Vasbinder
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Changli Wei
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Feriel Presswalla
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Hui Wang
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Pennelope Blakely
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ayse Bilge Ozel
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah Graham
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Grace H. Holton
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Joseph Dowsett
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Akl C. Fahed
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Kingsley-Michael Amadi
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Grace K. Erne
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Annika Tekmulla
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Anis Ismail
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher Launius
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - James S. Pankow
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lise Wegner Thørner
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Mary E. Haas
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, New York, USA
| | - Jonas B. Nielsen
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, New York, USA
| | - Luca A. Lotta
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, New York, USA
| | | | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | - Lili Zhao
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Venkatesh L. Murthy
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - David J. Pinsky
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Cristen J. Willer
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Susan R. Heckbert
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Daniel R. Goldstein
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Karl C. Desch
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Salim S. Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
84
|
Balamurugan K, Medishetti R, Rao P, K RV, Chatti K, Parsa KV. Protocol to evaluate hyperlipidemia in zebrafish larvae. STAR Protoc 2022; 3:101819. [DOI: 10.1016/j.xpro.2022.101819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
85
|
Sanchez MM, Bagdasarian IA, Darch W, Morgan JT. Organotypic cultures as aging associated disease models. Aging (Albany NY) 2022; 14:9338-9383. [PMID: 36435511 PMCID: PMC9740367 DOI: 10.18632/aging.204361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/21/2022] [Indexed: 11/24/2022]
Abstract
Aging remains a primary risk factor for a host of diseases, including leading causes of death. Aging and associated diseases are inherently multifactorial, with numerous contributing factors and phenotypes at the molecular, cellular, tissue, and organismal scales. Despite the complexity of aging phenomena, models currently used in aging research possess limitations. Frequently used in vivo models often have important physiological differences, age at different rates, or are genetically engineered to match late disease phenotypes rather than early causes. Conversely, routinely used in vitro models lack the complex tissue-scale and systemic cues that are disrupted in aging. To fill in gaps between in vivo and traditional in vitro models, researchers have increasingly been turning to organotypic models, which provide increased physiological relevance with the accessibility and control of in vitro context. While powerful tools, the development of these models is a field of its own, and many aging researchers may be unaware of recent progress in organotypic models, or hesitant to include these models in their own work. In this review, we describe recent progress in tissue engineering applied to organotypic models, highlighting examples explicitly linked to aging and associated disease, as well as examples of models that are relevant to aging. We specifically highlight progress made in skin, gut, and skeletal muscle, and describe how recently demonstrated models have been used for aging studies or similar phenotypes. Throughout, this review emphasizes the accessibility of these models and aims to provide a resource for researchers seeking to leverage these powerful tools.
Collapse
Affiliation(s)
- Martina M. Sanchez
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | | | - William Darch
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | - Joshua T. Morgan
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| |
Collapse
|
86
|
Vitamin C Deficiency Exacerbates Dysfunction of Atherosclerotic Coronary Arteries in Guinea Pigs Fed a High-Fat Diet. Antioxidants (Basel) 2022; 11:antiox11112226. [PMID: 36421412 PMCID: PMC9686655 DOI: 10.3390/antiox11112226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/31/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Vitamin C (vitC) deficiency has been associated with an increased risk of cardiovascular disease; while several putative mechanistic links have been proposed, functional evidence supporting a causal relationship is scarce. In this study, we investigated how vitC deficiency affects coronary artery vasomotor function and the development of coronary atherosclerotic plaques in guinea pigs subjected to chronic dyslipidemia by a high-fat diet regime. Female Hartley guinea pigs were fed either a control (low-fat diet and sufficient vitC) (N = 8) or a high-fat diet with either sufficient (N = 8) or deficient (N = 10) vitC for 32 weeks. Guinea pigs subjected to the high-fat diet developed significant atherosclerotic plaques in their coronary arteries, with no quantitative effect of vitC deficiency. In isolated coronary arteries, vasomotor responses to potassium, carbachol, nitric oxide, or bradykinin were studied in a wire myograph. Carbachol, bradykinin, and nitric oxide mediated relaxation in the coronary arteries of the control group. While vasorelaxation to carbachol and nitric oxide was preserved in the two high-fat diet groups, bradykinin-induced vasorelaxation was abolished. Interestingly, bradykinin induced a significant contraction in coronary arteries from vitC-deficient guinea pigs (p < 0.05). The bradykinin-induced contraction was unaffected by L-NAME but significantly inhibited by both indomethacin and vitC, suggesting that, during vitC deficiency, increased release of arachidonic acid metabolites and vascular oxidative stress are involved in the constrictor effects mediated by bradykinin. In conclusion, the present study shows supporting evidence that poor vitC status negatively affects coronary artery function.
Collapse
|
87
|
Huang J, El-Kersh K, Mann KK, James KA, Cai L. Overview of the cardiovascular effects of environmental metals: New preclinical and clinical insights. Toxicol Appl Pharmacol 2022; 454:116247. [PMID: 36122736 PMCID: PMC9941893 DOI: 10.1016/j.taap.2022.116247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 02/06/2023]
Abstract
Environmental causes of cardiovascular diseases (CVDs) are global health issues. In particular, an association between metal exposure and CVDs has become evident but causal evidence still lacks. Therefore, this symposium at the Society of Toxicology 2022 annual meeting addressed epidemiological, clinical, pre-clinical animal model-derived and mechanism-based evidence by five presentations: 1) An epidemiologic study on potential CVD risks of individuals exposed occupationally and environmentally to heavy metals; 2) Both presentations of the second and third were clinical studies focusing on the potential link between heavy metals and pulmonary arterial hypertension (PAH), by presenting altered blood metal concentrations of both non-essential and essential metals in the patients with PAH and potential therapeutic approaches; 3) Arsenic-induced atherosclerosis via inflammatory cells in mouse model; 4) Pathogenic effects on the heart by adult chronic exposure to very low-dose cadmium via epigenetic mechanisms and whole life exposure to low dose cadmium via exacerbating high-fat-diet-lipotoxicity. This symposium has brought epidemiologists, therapeutic industry, physicians, and translational scientists together to discuss the health risks of occupational and environmental exposure to heavy metals through direct cardiotoxicity and indirect disruption of homeostatic mechanisms regulating essential metals, as well as lipid levels. The data summarized by the presenters infers a potential causal link between multiple metals and CVDs and defines differences and commonalities. Therefore, summary of these presentations may accelerate the development of efficient preventive and therapeutic strategies by facilitating collaborations among multidisciplinary investigators.
Collapse
Affiliation(s)
- Jiapeng Huang
- Department of Anesthesiology and Perioperative Medicine, University of Louisville School of Medicine, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA; Cardiovascular Innovation Institute, Department of Cardiovascular and Thoracic Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | - Karim El-Kersh
- Department of Internal Medicine, Division of Pulmonary Critical Care and Sleep Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Koren K Mann
- Departments of Pharmacology & Therapeutics and Oncology and Medicine, McGill University, Canada; Segal Cancer Center, Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada
| | - Katherine A James
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA,.
| | - Lu Cai
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA; Pediatric Research Institute, Departments of Pediatrics and Radiation Oncology, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|
88
|
Hu YX, You HM, Zhu RF, Liang YL, Li FF, Qin YW, Zhao XX, Liang C, Jing Q. Establishment of a lipid metabolism disorder model in ApoEb mutant zebrafish. Atherosclerosis 2022; 361:18-29. [PMID: 36306655 DOI: 10.1016/j.atherosclerosis.2022.10.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/21/2022] [Accepted: 10/14/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS ApoEb is a zebrafish homologous to mammalian ApoE, whose deficiency would lead to lipid metabolism disorders (LMDs) like atherosclerosis. We attempted to knock out the zebrafish ApoEb, then establish a zebrafish model with LMD. METHODS ApoEb was knocked out using the CRISPR/Cas9 system, and the accumulation of lipids was confirmed by Oil Red O staining, confocal imaging, and lipid measurements. The lipid-lowering effects of simvastatin (SIM), ezetimibe (EZE) and Xuezhikang (XZK), an extract derived from red yeast rice, were evaluated through in vivo imaging in zebrafish larvae. RESULTS In the ApoEb mutant, significant vascular lipid deposition occurred, and lipid measurement performed in the whole-body homogenate of larvae and adult plasma showed significantly increased lipid levels. SIM, EZE and XZK apparently relieved hyperlipidemia in ApoEb mutants, and XZK had a significant inhibitory effect on the recruitment of neutrophils and macrophages. CONCLUSIONS In this study, an LMD model has been established in ApoEb mutant zebrafish. We suggest that this versatile model could be applied in studying hypercholesterolemia and related vascular pathology in the context of early atherosclerosis, as well as the physiological function of ApoE.
Collapse
Affiliation(s)
- Yang-Xi Hu
- Department of Cardiology, Changzheng Hospital, Shanghai, 200003, China
| | - Hong-Min You
- Department of Cardiology, Changhai Hospital, Shanghai, 200433, China
| | - Rong-Fang Zhu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu-Lai Liang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Fang-Fang Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yong-Wen Qin
- Department of Cardiology, Changhai Hospital, Shanghai, 200433, China
| | - Xian-Xian Zhao
- Department of Cardiology, Changhai Hospital, Shanghai, 200433, China
| | - Chun Liang
- Department of Cardiology, Changzheng Hospital, Shanghai, 200003, China.
| | - Qing Jing
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
89
|
Kamato D, Ilyas I, Xu S, Little PJ. Non-Mouse Models of Atherosclerosis: Approaches to Exploring the Translational Potential of New Therapies. Int J Mol Sci 2022; 23:12964. [PMID: 36361754 PMCID: PMC9656683 DOI: 10.3390/ijms232112964] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 09/26/2023] Open
Abstract
Cardiovascular disease is the largest single cause of disease-related mortality worldwide and the major underlying pathology is atherosclerosis. Atherosclerosis develops as a complex process of vascular lipid deposition and retention by modified proteoglycans, endothelial dysfunction and unresolved chronic inflammation. There are a multitude of current therapeutic agents, most based on lowering plasma lipid levels, but, overall, they have a lower than optimum level of efficacy and many deaths continue to arise from cardiovascular disease world-wide. To identify and evaluate potential novel cardiovascular drugs, suitable animal models that reproduce human atherosclerosis with a high degree of fidelity are required as essential pre-clinical research tools. Commonly used animal models of atherosclerosis include mice (ApoE-/-, LDLR-/- mice and others), rabbits (WHHL rabbits and others), rats, pigs, hamster, zebrafish and non-human primates. Models based on various wild-type and genetically modified mice have been extensively reviewed but mice may not always be appropriate. Thus, here, we provide an overview of the advantages and shortcomings of various non-mouse animal models of atherosclerotic plaque formation, and plaque rupture, as well as commonly used interventional strategies. Taken together, the combinatorial selection of suitable animal models readily facilitates reproducible and rigorous translational research in discovering and validating novel anti-atherosclerotic drugs.
Collapse
Affiliation(s)
- Danielle Kamato
- Discovery Biology, Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
- Pharmacy Australia Centre of Excellence, School of Pharmacy, University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Iqra Ilyas
- Laboratory of Metabolics and Cardiovascular Diseases, University of Science and Technology of China, Hefei 230027, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China
| | - Suowen Xu
- Laboratory of Metabolics and Cardiovascular Diseases, University of Science and Technology of China, Hefei 230027, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei 230001, China
| | - Peter J. Little
- Pharmacy Australia Centre of Excellence, School of Pharmacy, University of Queensland, Woolloongabba, QLD 4102, Australia
- Sunshine Coast Health Institute and School of Health and Behavioural Sciences, University of the Sunshine Coast, Birtinya, QLD 4575, Australia
| |
Collapse
|
90
|
Yamaguchi T, Morino K. Perivascular mechanical environment: A narrative review of the role of externally applied mechanical force in the pathogenesis of atherosclerosis. Front Cardiovasc Med 2022; 9:944356. [PMID: 36337892 PMCID: PMC9629355 DOI: 10.3389/fcvm.2022.944356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 09/26/2022] [Indexed: 12/04/2022] Open
Abstract
Atherosclerosis is promoted by systemic factors, such as dyslipidemia, hypertension, diabetes, and smoking, which cause atherosclerosis in blood vessels throughout the body. However, atherosclerotic lesions are characterized by their frequent occurrence in specific vessels and sites. Blood vessels are exposed to various mechanical forces related to blood pressure and flow. Although shear stress promotes the initiation and progression of atherosclerotic lesions, the pathogenesis of site specificity of atherosclerosis is not sufficiently explained by shear stress. We propose the concept of a perivascular mechanical environment (PVME). Compelling evidence suggests that site specificity in atherosclerotic lesions depends on a distinct local PVME. Atheroprone arteries, such as the coronary artery, are markedly affected by externally applied mechanical force (EMF), whereas atheroprotective arteries, such as the internal thoracic artery, are less affected. Recent studies have shown that the coronary artery is affected by cardiac muscle contraction, the carotid artery by the hyoid bone and the thyroid cartilage, and the abdominal aorta and lower extremity arteries by musculoskeletal motion. We speculate that the thoracic cage protects the internal thoracic artery from EMF owing to a favorable PVME. Furthermore, evidence suggests that plaque eccentricity is provided by EMF; plaques are frequently observed on an external force-applied side. In each vascular tree, site-specific characteristics of the PVME differ substantially, inducing individual atherogenicity. From the perspective of the mechanical environment, hemodynamic stress occurs in an inside-out manner, whereas EMF occurs in an outside-in manner. These inward and outward forces apply mechanical load individually, but interact synergistically. The concept of a PVME is a novel pathogenesis of atherosclerosis and also might be a pathogenesis of other arterial diseases.
Collapse
Affiliation(s)
| | - Katsutaro Morino
- Institutional Research Office, Shiga University of Medical Science, Otsu, Japan
- *Correspondence: Katsutaro Morino,
| |
Collapse
|
91
|
Wahl L, Raschke M, Wittmann J, Regler A, Heelemann S, Brandsch C, Stangl GI, Vervuert I. Effects of atherogenic diet supplemented with fermentable carbohydrates on metabolic responses and plaque formation in coronary arteries using a Saddleback pig model. PLoS One 2022; 17:e0275214. [PMID: 36206259 PMCID: PMC9543622 DOI: 10.1371/journal.pone.0275214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
Fermentable carbohydrates are gaining interest in the field of human nutrition because of their benefits in obesity-related comorbidities. The aim of this study was to investigate the influence of fermentable carbohydrates, such as pectin and inulin, in an atherogenic diet on metabolic responses and plaque formation in coronary arteries using a Saddleback pig model. Forty-eight healthy pigs aged five months were divided into four feeding groups (n = 10) and one baseline group (n = 8). Three feeding groups received an atherogenic diet (38% crisps, 10% palm fat, and 2% sugar with or without supplementation of 5% pectin or inulin), and one group received a conventional diet over 15 weeks. Feed intake, weight gain, body condition score, and back fat thickness were monitored regularly. Blood and fecal samples were collected monthly to assess the metabolites associated with high cardiovascular risk and fat content, respectively. At the end of 15 weeks, the coronary arteries of the pigs were analyzed for atherosclerotic plaque formation. Independent of supplementation, significant changes were observed in lipid metabolism, such as an increase in triglycerides, bile acids, and cholesterol in serum, in all groups fed atherogenic diets in comparison to the conventional group. Serum metabolome analysis showed differentiation of the feeding groups by diet (atherogenic versus conventional diet) but not by supplementation with pectin or inulin. Cardiovascular lesions were found in all feeding groups and in the baseline group. Supplementation of pectin or inulin in the atherogenic diet had no significant impact on cardiovascular lesion size. Saddleback pigs can develop naturally occurring plaques in coronary arteries. Therefore, this pig model offers potential for further research on the effects of dietary intervention on obesity-related comorbidities, such as cardiovascular lesions, in humans.
Collapse
Affiliation(s)
- Lisa Wahl
- Institute of Animal Nutrition, Nutrition Diseases and Dietetics, Leipzig University, Leipzig, Germany,Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany
| | - Melina Raschke
- Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany,Institute of Agricultural and Nutritional Science, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | | | | | | | - Corinna Brandsch
- Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany,Institute of Agricultural and Nutritional Science, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Gabriele I. Stangl
- Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany,Institute of Agricultural and Nutritional Science, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Ingrid Vervuert
- Institute of Animal Nutrition, Nutrition Diseases and Dietetics, Leipzig University, Leipzig, Germany,Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany,* E-mail:
| |
Collapse
|
92
|
Gómez-Pérez S, Ovando-Gómez V, Hernández-Contreras AC, Herón-Petri M, Díaz-López HO, García-Miranda R, Irecta-Nájera CA. Atherogenic indices in pediatric population in South-Southeast region of Mexico. J Trop Pediatr 2022; 68:6827489. [PMID: 36375036 DOI: 10.1093/tropej/fmac099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Atherosclerosis is a cardiovascular disease, highly predictable, and associated with different atherogenic indices (AI) in adults. However, such indexes in the pediatric population are far less explored. The objective of this study was to evaluate the AI and the cardiovascular factors in the pediatric population in the South-Southeast of México. METHODS A total of 481 children between 2 and 17 years old were recruited. Anthropometric evaluation, blood pressure (BP), lipid profile, apolipoprotein A-I (ApoA-I) and apolipoprotein B (ApoB) were measured, and AI were calculated. The population was grouped by age, binary logistic regression analysis was performed to analysis for associations of AI and cardiovascular risk factors. Sensibility and specificity of AI to detect metabolic alteration were evaluated for curve ROC. RESULTS The atherogenic risk presented a high prevalence in the pediatric population, such as LDL-c/ApoB (86.9%), AIP (78%) and AC (36.6%). Preschoolers showed a higher risk of ApoB/ApoA-I and ApoB/LDL-c, while adolescents have a high risk of AIP. CRI-I and AC were associated with elements of lipid profile and body mass index (BMI). ROC curves analysis shows that AIP is the best index evaluating metabolic syndrome (MS) (0.87) and dyslipidemia (0.91). CONCLUSION Such pediatric population showed a high risk of AI, mainly by LDL-c/ApoB and AIP. The BMI was the cardiovascular risk factors most frequently related to AI, AIP is the best index for detecting cases of MS and dyslipidemia. This is the first study carried out in the pediatric population from the South-Southeast of Mexico that evaluated the AI.
Collapse
Affiliation(s)
- Samuel Gómez-Pérez
- Health Department, El Colegio de la Frontera Sur, 86280 Villahermosa, Tabasco, Mexico
| | - Valeria Ovando-Gómez
- Health Department, El Colegio de la Frontera Sur, 86280 Villahermosa, Tabasco, Mexico
| | - Armando Camilo Hernández-Contreras
- Department of Pediatrics, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Comitán de Domínguez Chiapas 30090, Mexico
| | - Marcelo Herón-Petri
- Department of Cardiothoracic and Vascular Surgery, Örebro University Hospital, 70185 Örebro, Sweden
| | | | - Rosario García-Miranda
- Health Department, El Colegio de la Frontera Sur, 29290 San Cristóbal de las Casas, Mexico
- School of Languages-Campus San Cristóbal, Autonomous University of Chiapas, 29264 San Cristóbal de las Casas, Mexico
| | | |
Collapse
|
93
|
Targeting ABCC6 in Mesenchymal Stem Cells: Impairment of Mature Adipocyte Lipid Homeostasis. Int J Mol Sci 2022; 23:ijms23169218. [PMID: 36012482 PMCID: PMC9409192 DOI: 10.3390/ijms23169218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/12/2022] [Accepted: 08/14/2022] [Indexed: 11/16/2022] Open
Abstract
Mutations in ABCC6, an ATP-binding cassette transporter with a so far unknown substrate mainly expressed in the liver and kidney, cause pseudoxanthoma elasticum (PXE). Symptoms of PXE in patients originate from the calcification of elastic fibers in the skin, eye, and vessels. Previous studies suggested an involvement of ABCC6 in cholesterol and lipid homeostasis. The intention of this study was to examine the influence of ABCC6 deficiency during adipogenic differentiation of human bone marrow-derived stem cells (hMSCs). Induction of adipogenic differentiation goes along with significantly elevated ABCC6 gene expression in mature adipocytes. We generated an ABCC6-deficient cell culture model using clustered regulatory interspaced short palindromic repeat Cas9 (CRISPR–Cas9) system to clarify the role of ABCC6 in lipid homeostasis. The lack of ABCC6 in hMSCs does not influence gene expression of differentiation markers in adipogenesis but results in a decreased triglyceride content in cell culture medium. Protein and gene expression analysis of mature ABCC6-deficient adipocytes showed diminished intra- and extra-cellular lipolysis, release of lipids, and fatty acid neogenesis. Therefore, our results demonstrate impaired lipid trafficking in adipocytes due to ABCC6 deficiency, highlighting adipose tissue and peripheral lipid metabolism as a relevant target for uncovering systemic PXE pathogenesis.
Collapse
|
94
|
Zhang S, Cao Y, Ren R, Qi J, Chen Y, Li Y. Effects of Cervical Rotatory Manipulation on Hemodynamics and Plaque Stability of Atherosclerotic Internal Carotid Artery in Rabbits. J Manipulative Physiol Ther 2022; 45:261-272. [PMID: 35907659 DOI: 10.1016/j.jmpt.2022.03.022] [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: 05/18/2020] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 10/16/2022]
Abstract
OBJECTIVE The purpose of this study was to investigate the effects of cervical rotatory manipulation (CRM) on hemodynamics and plaque stability of atherosclerotic internal carotid artery (ICA) in rabbits. METHODS Forty rabbits were randomly divided into 4 groups: (1) internal carotid atherosclerosis (ICAS) rabbits treated with CRM (ICAS-CRM group); (2) ICAS rabbits treated without CRM (ICAS group), (3) Normal-CRM group (normal rabbits treated with CRM), and (4) blank control group. In the ICAS-CRM group and ICAS group, the ICAS model was induced by ICA balloon injury combined with a high-fat diet for 12 weeks. CRM was applied to rabbits in the ICAS-CRM and the Normal-CRM groups. During the study, an ultrasonography examination was performed for detecting plaque and hemodynamics on the ICAs. At the end of the study, all atherosclerotic ICAs were removed for histological and immunohistochemical detection. RESULTS The hemodynamics (especially end-diastolic velocity, resistance index, and pulsatility index) through the ICAs were adversely affected by atherosclerosis while not adversely affected by CRM. Compared with the ICAS group, the micro-vessel density and average integrated optical densities of macrophages in the ICAS-CRM group were significantly increased. Compared to the ICAS group, in the ICAS-CRM group, the atherosclerosis was more serious, and the tunica intima was more unstable. CONCLUSIONS Although CRM did not affect the hemodynamic index of ICA, it was observed to decrease the stability of severe ICAS plaques in rabbits, which may increase the plaque vulnerability.
Collapse
Affiliation(s)
- Shaoqun Zhang
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China, Shenzhen, Guangdong, China
| | - Yafei Cao
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China, Shenzhen, Guangdong, China
| | - Ruxia Ren
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Ji Qi
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yili Chen
- Wang Jing Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yikai Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
95
|
Vesnina A, Prosekov A, Atuchin V, Minina V, Ponasenko A. Tackling Atherosclerosis via Selected Nutrition. Int J Mol Sci 2022; 23:8233. [PMID: 35897799 PMCID: PMC9368664 DOI: 10.3390/ijms23158233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 12/02/2022] Open
Abstract
The development and pathogenesis of atherosclerosis are significantly influenced by lifestyle, particularly nutrition. The modern level of science and technology development promote personalized nutrition as an efficient preventive measure against atherosclerosis. In this survey, the factors were revealed that contribute to the formation of an individual approach to nutrition: genetic characteristics, the state of the microbiota of the gastrointestinal tract (GIT) and environmental factors (diets, bioactive components, cardioprotectors, etc.). In the course of the work, it was found that in order to analyze the predisposition to atherosclerosis associated with nutrition, genetic features affecting the metabolism of nutrients are significant. The genetic features include the presence of single nucleotide polymorphisms (SNP) of genes and epigenetic factors. The influence of telomere length on the pathogenesis of atherosclerosis and circadian rhythms was also considered. Relatively new is the study of the relationship between chrono-nutrition and the development of metabolic diseases. That is, to obtain the relationship between nutrition and atherosclerosis, a large number of genetic markers should be considered. In this relation, the question arises: "How many genetic features need to be analyzed in order to form a personalized diet for the consumer?" Basically, companies engaged in nutrigenetic research and choosing a diet for the prevention of a number of metabolic diseases use SNP analysis of genes that accounts for lipid metabolism, vitamins, the body's antioxidant defense system, taste characteristics, etc. There is no set number of genetic markers. The main diets effective against the development of atherosclerosis were considered, and the most popular were the ketogenic, Mediterranean, and DASH-diets. The advantage of these diets is the content of foods with a low amount of carbohydrates, a high amount of vegetables, fruits and berries, as well as foods rich in antioxidants. However, due to the restrictions associated with climatic, geographical, material features, these diets are not available for a number of consumers. The way out is the use of functional products, dietary supplements. In this approach, the promising biologically active substances (BAS) that exhibit anti-atherosclerotic potential are: baicalin, resveratrol, curcumin, quercetin and other plant metabolites. Among the substances, those of animal origin are popular: squalene, coenzyme Q10, omega-3. For the prevention of atherosclerosis through personalized nutrition, it is necessary to analyze the genetic characteristics (SNP) associated with the metabolism of nutrients, to assess the state of the microbiota of the GIT. Based on the data obtained and food preferences, as well as the individual capabilities of the consumer, the optimal diet can be selected. It is topical to exclude nutrients of which their excess consumption stimulates the occurrence and pathogenesis of atherosclerosis and to enrich the diet with functional foods (FF), BAS containing the necessary anti-atherosclerotic, and stimulating microbiota of the GIT nutrients. Personalized nutrition is a topical preventive measure and there are a number of problems hindering the active use of this approach among consumers. The key factors include weak evidence of the influence of a number of genetic features, the high cost of the approach, and difficulties in the interpretation of the results. Eliminating these deficiencies will contribute to the maintenance of a healthy state of the population through nutrition.
Collapse
Affiliation(s)
- Anna Vesnina
- Laboratory of Natural Nutraceuticals Biotesting, Research Department, Kemerovo State University, 650043 Kemerovo, Russia;
| | - Alexander Prosekov
- Laboratory of Biocatalysis, Kemerovo State University, 650043 Kemerovo, Russia;
| | - Victor Atuchin
- Laboratory of Optical Materials and Structures, Institute of Semiconductor Physics, 630090 Novosibirsk, Russia
- Research and Development Department, Kemerovo State University, 650000 Kemerovo, Russia
- Laboratory of Applied Physics, Novosibirsk State University, 630090 Novosibirsk, Russia
- Department of Industrial Machinery Design, Novosibirsk State Technical University, 630073 Novosibirsk, Russia
- R&D Center “Advanced Electronic Technologies”, Tomsk State University, 634034 Tomsk, Russia
| | - Varvara Minina
- Department of Genetic and Fundamental Medicine, Kemerovo State University, 650000 Kemerovo, Russia;
| | - Anastasia Ponasenko
- Laboratory of Genome Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 650002 Kemerovo, Russia;
| |
Collapse
|
96
|
Ilyas I, Little PJ, Liu Z, Xu Y, Kamato D, Berk BC, Weng J, Xu S. Mouse models of atherosclerosis in translational research. Trends Pharmacol Sci 2022; 43:920-939. [PMID: 35902281 DOI: 10.1016/j.tips.2022.06.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 06/12/2022] [Accepted: 06/17/2022] [Indexed: 12/21/2022]
Abstract
Atherosclerotic cardiovascular disease (CVD), the major cause of premature human mortality, is a chronic and progressive metabolic and inflammatory disease in large- and medium-sized arteries. Mouse models are widely used to gain mechanistic insights into the pathogenesis of atherosclerosis and have facilitated the discovery of anti-atherosclerotic drugs. Despite promising preclinical studies, many drug candidates have not translated to clinical use because of the complexity of disease patho-mechanisms including lipid metabolic traits and inflammatory, genetic, and hemodynamic factors. We review the current preclinical utility and translation potential of traditional [apolipoprotein E (APOE)- and low-density lipoprotein (LDL) receptor (LDLR)-deficient mice] and emerging mouse models that include partial carotid ligation and AAV8-Pcsk9-D377Y injection in atherosclerosis research and drug discovery. This article represents an important resource in atherosclerosis research.
Collapse
Affiliation(s)
- Iqra Ilyas
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia
| | - Zhiping Liu
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yanyong Xu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Pathology of School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Danielle Kamato
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia; Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, Australia
| | - Bradford C Berk
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Jianping Weng
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China; Laboratory of Metabolics and Cardiovascular Diseases, Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China.
| | - Suowen Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China; Laboratory of Metabolics and Cardiovascular Diseases, Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
97
|
Fernández-García V, González-Ramos S, Avendaño-Ortiz J, Martín-Sanz P, Gómez-Coronado D, Delgado C, Castrillo A, Boscá L. High-fat diet activates splenic NOD1 and enhances neutrophil recruitment and neutrophil extracellular traps release in the spleen of ApoE-deficient mice. Cell Mol Life Sci 2022; 79:396. [PMID: 35789437 PMCID: PMC9256580 DOI: 10.1007/s00018-022-04415-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/19/2022] [Accepted: 06/03/2022] [Indexed: 12/14/2022]
Abstract
In the course of atherogenesis, the spleen plays an important role in the regulation of extramedullary hematopoiesis, and in the control of circulating immune cells, which contributes to plaque progression. Here, we have investigated the role of splenic nucleotide-binding oligomerization domain 1 (NOD1) in the recruitment of circulating immune cells, as well as the involvement of this immune organ in extramedullary hematopoiesis in mice fed on a high-fat high-cholesterol diet (HFD). Under HFD conditions, the absence of NOD1 enhances the mobilization of immune cells, mainly neutrophils, from the bone marrow to the blood. To determine the effect of NOD1-dependent mobilization of immune cells under pro-atherogenic conditions, Apoe-/- and Apoe-/-Nod1-/- mice fed on HFD for 4 weeks were used. Splenic NOD1 from Apoe-/- mice was activated after feeding HFD as inferred by the phosphorylation of the NOD1 downstream targets RIPK2 and TAK1. Moreover, this activation was accompanied by the release of neutrophil extracellular traps (NETs), as determined by the increase in the expression of peptidyl arginine deiminase 4, and the identification of citrullinated histone H3 in this organ. This formation of NETs was significantly reduced in Apoe-/-Nod1-/- mice. Indeed, the presence of Ly6G+ cells and the lipidic content in the spleen of mice deficient in Apoe and Nod1 was reduced when compared to the Apoe-/- counterparts, which suggests that the mobilization and activation of circulating immune cells are altered in the absence of NOD1. Furthermore, confirming previous studies, Apoe-/-Nod1-/- mice showed a reduced atherogenic disease, and diminished recruitment of neutrophils in the spleen, compared to Apoe-/- mice. However, splenic artery ligation reduced the atherogenic burden in Apoe-/- mice an effect that, unexpectedly was lost in Apoe-/-Nod1-/- mice. Together, these results suggest that neutrophil accumulation and activity in the spleen are driven in part by NOD1 activation in mice fed on HFD, contributing in this way to regulating atherogenic progression.
Collapse
Affiliation(s)
- Victoria Fernández-García
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Monforte de Lemos 3-5, 28029, Madrid, Spain.
| | - Silvia González-Ramos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain
| | - José Avendaño-Ortiz
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz, IdiPAZ., C. de Pedro Rico, 6, 28029, Madrid, Spain
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Diego Gómez-Coronado
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Ctra. M-607 9,100, 28034, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Carmen Delgado
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain
- Unidad de Biomedicina (Unidad Asociada Al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Monforte de Lemos 3-5, 28029, Madrid, Spain.
- Unidad de Biomedicina (Unidad Asociada Al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain.
| |
Collapse
|
98
|
Suh J, Kim S, Lee S, Kim R, Park NH. Hyperlipidemia is necessary for the initiation and progression of atherosclerosis by severe periodontitis in mice. Mol Med Rep 2022; 26:273. [PMID: 35795972 PMCID: PMC9309540 DOI: 10.3892/mmr.2022.12789] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/07/2022] [Indexed: 11/08/2022] Open
Abstract
Hyperlipidemia is a major risk of atherosclerosis; however, systemic inflammatory diseases such as rheumatoid arthritis, psoriasis, systemic lupus erythematosus and systemic sclerosis are also known risks for the development of atherosclerosis. Periodontitis, a local and systemic inflammatory condition, has also been reported as a risk for atherosclerosis, but the specific link between periodontitis and atherosclerosis remains somewhat controversial. We previously reported that ligature-induced periodontitis exacerbates atherosclerosis in hyperlipidemic Apolipoprotein E-deficient (ApoE−/−) mice. To understand whether hyperlipidemia is necessary for the development and exacerbation of atherosclerosis associated with periodontitis, the present study created ligature-induced periodontitis in both wild-type (WT) and ApoE−/− mice. Subsequently, the status of local, systemic and vascular inflammation, serum lipid contents and arterial lipid deposition were examined with histological analysis, µCT, en face analysis, serum lipid and cytokine measurements, reverse transcription-quantitative PCR and immunohistochemical analysis. Ligature placement induced severe periodontitis in both WT and ApoE−/− mice at the local level as demonstrated by gingival inflammation, alveolar bone loss, increased osteoclastic activities and inflammation in alveolar bone. Systemic inflammation was also induced by ligature placement in both WT and ApoE−/− mice, albeit more so in ApoE−/− mice. The serum cholesterol levels were not altered by the ligature in both WT and ApoE−/− mice. However, the vascular inflammation and arterial lipid deposition were induced by ligature-induced periodontitis only in ApoE−/− mice, but not in WT mice. The present study indicated that the coupling of systemic inflammation and hyperlipidemia was necessary for the development and exacerbation of atherosclerosis induced by ligature-induced periodontitis in mice.
Collapse
Affiliation(s)
- Jin Suh
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, Los Angeles, CA 90095, USA
| | - Sharon Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, Los Angeles, CA 90095, USA
| | - Sung Lee
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, Los Angeles, CA 90095, USA
| | - Reuben Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, Los Angeles, CA 90095, USA
| | - No-Hee Park
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, Los Angeles, CA 90095, USA
| |
Collapse
|
99
|
Georgakis MK, Bernhagen J, Heitman LH, Weber C, Dichgans M. Targeting the CCL2-CCR2 axis for atheroprotection. Eur Heart J 2022; 43:1799-1808. [PMID: 35567558 DOI: 10.1093/eurheartj/ehac094] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 12/22/2021] [Accepted: 02/15/2022] [Indexed: 11/12/2022] Open
Abstract
Decades of research have established atherosclerosis as an inflammatory disease. Only recently though, clinical trials provided proof-of-concept evidence for the efficacy of anti-inflammatory strategies with respect to cardiovascular events, thus offering a new paradigm for lowering residual vascular risk. Efforts to target the inflammasome-interleukin-1β-interleukin-6 pathway have been highly successful, but inter-individual variations in drug response, a lack of reduction in all-cause mortality, and a higher rate of infections also highlight the need for a second generation of anti-inflammatory agents targeting atherosclerosis-specific immune mechanisms while minimizing systemic side effects. CC-motif chemokine ligand 2/monocyte-chemoattractant protein-1 (CCL2/MCP-1) orchestrates inflammatory monocyte trafficking between the bone marrow, circulation, and atherosclerotic plaques by binding to its cognate receptor CCR2. Adding to a strong body of data from experimental atherosclerosis models, a coherent series of recent large-scale genetic and observational epidemiological studies along with data from human atherosclerotic plaques highlight the relevance and therapeutic potential of the CCL2-CCR2 axis in human atherosclerosis. Here, we summarize experimental and human data pinpointing the CCL2-CCR2 pathway as an emerging drug target in cardiovascular disease. Furthermore, we contextualize previous efforts to interfere with this pathway, scrutinize approaches of ligand targeting vs. receptor targeting, and discuss possible pathway-intrinsic opportunities and challenges related to pharmacological targeting of the CCL2-CCR2 axis in human atherosclerotic disease.
Collapse
Affiliation(s)
- Marios K Georgakis
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, D-81377 Munich, Germany
- Center of Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jürgen Bernhagen
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, D-81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Christian Weber
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Germany
- Institute for Genetic and Biomedical Research, UoS of Milan, National Research Council, Milan, Italy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, D-81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
| |
Collapse
|
100
|
Chuang HL, Chiu CC, Lo C, Hsu CC, Liu JY, Hung SW, Tsai SC, Sung HH, Wang CKL, Huang YT. Circulating gut microbiota-related metabolites influence endothelium plaque lesion formation in ApoE knockout rats. PLoS One 2022; 17:e0264934. [PMID: 35522651 PMCID: PMC9075652 DOI: 10.1371/journal.pone.0264934] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 02/18/2022] [Indexed: 11/17/2022] Open
Abstract
Atherosclerosis is the main cause of cardiac and peripheral vessel infarction in developed countries. Recent studies have established that gut microbiota and their metabolites play important roles in the development and progression of cardiovascular disease; however, the underlying mechanisms remain unclear. The present study aimed to investigate endothelium plaque lesion formation in ApoE-deficient rats fed a normal chow diet under germ-free (GF) and specific-pathogen-free (SPF) conditions at various time points. There was no difference in serum cholesterol and triglyceride levels between SPF-rats and GF-rats. Histological studies revealed that the GF-rats developed endothelium plaques in the aorta from 26 to 52 weeks, but this was not observed in SPF-rats. GF-rat coronary arteries had moderate-to-severe endothelium lesions during this time period, but SPF-rat coronary arteries had only mild lesion formation. Immunohistochemical staining showed higher accumulation of CD68-positive and arginase-negative foamy-like macrophages on the arterial walls of GF-rats, and expression of TNF-α and IL-6 in foam cells was only observed in GF-rats. In addition, microbial metabolites, including equol derivatives, enterolactone derivatives, indole-3-propionate, indole-3-acrylic acid, cholic acid, hippuric acid, and isoquinolone, were significantly higher in the SPF group than in the GF group. In conclusion, our results indicate that gut microbiota may attenuate atherosclerosis development.
Collapse
Affiliation(s)
- Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Chien-Chao Chiu
- Division of Animal Industry, Animal Technology Laboratories, Agricultural Technology Research Institute, Miaoli, Taiwan
| | - Ching Lo
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Cheng-Chih Hsu
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Ju-Yun Liu
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Shao-Wen Hung
- Division of Animal Industry, Animal Technology Laboratories, Agricultural Technology Research Institute, Miaoli, Taiwan
| | - Shih-Chieh Tsai
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Hsiang-Hsuan Sung
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Chi-Kuang Leo Wang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Yen-Te Huang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
- * E-mail:
| |
Collapse
|