1
|
Kopytek M, Undas KW, Tarasiuk J, Wroński S, Ząbczyk M, Natorska J. Dysglycaemia is associated with the pattern of valvular calcification in micro-computed tomography analysis: an observational study in patients with severe aortic stenosis. Cardiovasc Diabetol 2025; 24:129. [PMID: 40114166 PMCID: PMC11927127 DOI: 10.1186/s12933-025-02691-y] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Diabetes mellitus (DM) has been shown to increase the rate of aortic stenosis (AS) progression. However, the impact of impaired plasma glucose on valvular calcification remains poorly understood. Using ex vivo micro-computed tomography (micro-CT), we aimed to determine whether plasma glucose, glycated haemoglobin (HbA1c), or concentrations of advanced glycation end products (AGEs) and their soluble receptor (sRAGE) are associated with a specific pattern of valvular calcification in severe AS. METHODS In this case-control study, 14 (48%) normoglycaemic patients with AS were compared to 15 individuals (52%) with elevated glucose levels (≥ 5.6 mmol/L), all with HbA1c ≤ 6.5%. Stenotic aortic valves obtained surgically were analysed using micro-CT to assess structure of tissue mineralization. Calcium volume (CV), surface volume (SV), CV/SV ratio, and trabecular thickness (TbTh) were evaluated. Plasma AGEs and sRAGE were assessed by ELISAs. DM patients or those using antidiabetic agents were excluded from the study. RESULTS Patients with impaired and high glucose, including 10 (67%) with glucose between 5.6 and 6.9 mmol/L and 5 (33%) ranging from 7 to 7.6 mmol/L, exhibited higher HbA1c (+ 17%) and AGEs levels (+ 44.6%), but not sRAGE compared to those with normal glucose. Patients with impaired and high glucose had also 19.2% higher maximal transvalvular pressure gradient (PGmax) and 9.3% higher peak transvalvular velocity (Vmax) compared to normoglycaemic individuals. Micro-CT indices correlated with fasting glucose, HbA1c, and AGEs levels (all p < 0.05), but not with sRAGE (p > 0.05). Valves extracted from patients with impaired and high glucose exhibited higher mineralization volume, folding, and structural integrity, as reflected by increased CV (+ 127.6%), CV/SV ratio (+ 59%) and calcium deposits microarchitecture as indicated by about 50% higher TbTh, compared to normoglycaemic patients. When patients with AS were divided into three groups based on their glucose levels (< 5.5 mmol/L, 5.6-6.9 mmol/L, and 7.0-7.6 mmol/L), micro-CT analysis showed more distinct structural differences among the groups. The valves in the highest glucose group were the most severely affected. Micro-CT parameters were also associated with both transvalvular pressure gradients (PGmean and PGmax), Vmax and aortic valve area (all p < 0.05). CONCLUSIONS Strict glycaemic control could potentially reduce the rate of valve mineralization and calcium deposit accumulation in patients with AS. RESEARCH INSIGHTS WHAT IS CURRENTLY KNOWN ABOUT THIS TOPIC?: Diabetes mellitus (DM) is a risk factor for the progression of aortic stenosis (AS). Accumulation of advanced glycation end products (AGEs) enhances glycation of valvular proteins. WHAT IS THE KEY RESEARCH QUESTION?: Is dysglycaemia associated with more severe aortic valve calcification in patients with severe AS? Is ex vivo micro-CT suitable for assessing differences in calcification pattern within stenoticvalves? WHAT IS NEW?: Pre-diabetic patients with AS show increased valvular calcium volume, surface corrugation, and calcium deposit integrity. Micro-CT parameters associate with glycaemic status and echocardiographic measures of AS severity. Micro-CT provides precise assessment of calcification, offering insights beyond traditional methods. HOW MIGHT THIS STUDY INFLUENCE CLINICAL PRACTICE?: Strict glycaemic control together with CT calcium scoring should be performed in patients with AS to monitor disease progression.
Collapse
Affiliation(s)
- Magdalena Kopytek
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- Krakow Centre for Medical Research and Technologies, St. John Paul II Hospital, Krakow, Poland
| | - Kamila W Undas
- Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Jacek Tarasiuk
- Department of Condensed Matter Physics, Faculty of Physics and Applied Computer Science, AGH University of Krakow, Krakow, Poland
| | - Sebastian Wroński
- Department of Condensed Matter Physics, Faculty of Physics and Applied Computer Science, AGH University of Krakow, Krakow, Poland
| | - Michał Ząbczyk
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- Krakow Centre for Medical Research and Technologies, St. John Paul II Hospital, Krakow, Poland
| | - Joanna Natorska
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland.
- Krakow Centre for Medical Research and Technologies, St. John Paul II Hospital, Krakow, Poland.
| |
Collapse
|
2
|
Koroleva EA, Romanov VV. Analysis of Associations Between Advanced Glycation end-Products and Vascular Complications of type 2 Diabetes. 2024 IEEE INTERNATIONAL MULTI-CONFERENCE ON ENGINEERING, COMPUTER AND INFORMATION SCIENCES (SIBIRCON) 2024:248-252. [DOI: 10.1109/sibircon63777.2024.10758526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Elena A. Koroleva
- Research Institute of Clinical and Experimental Lymphology - Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (RICEL - Branch of IC&G SB RAS),Laboratory of Endocrinology,Novosibirsk,Russia
| | - Vyacheslav V. Romanov
- Research Institute of Clinical and Experimental Lymphology - Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (RICEL - Branch of IC&G SB RAS),Laboratory of Endocrinology,Novosibirsk,Russia
| |
Collapse
|
3
|
Xu Z, Chen Y, Wang Y, Han W, Xu W, Liao X, Zhang T, Wang G. Matrix stiffness, endothelial dysfunction and atherosclerosis. Mol Biol Rep 2023; 50:7027-7041. [PMID: 37382775 DOI: 10.1007/s11033-023-08502-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 04/28/2023] [Indexed: 06/30/2023]
Abstract
Atherosclerosis (AS) is the leading cause of the human cardiovascular diseases (CVDs). Endothelial dysfunction promotes the monocytes infiltration and inflammation that participate fundamentally in atherogenesis. Endothelial cells (EC) have been recognized as mechanosensitive cells and have different responses to distinct mechanical stimuli. Emerging evidence shows matrix stiffness-mediated EC dysfunction plays a vital role in vascular disease, but the underlying mechanisms are not yet completely understood. This article aims to summarize the effect of matrix stiffness on the pro-atherosclerotic characteristics of EC including morphology, rigidity, biological behavior and function as well as the related mechanical signal. The review also discusses and compares the contribution of matrix stiffness-mediated phagocytosis of macrophages and EC to AS progression. These advances in our understanding of the relationship between matrix stiffness and EC dysfunction open the avenues to improve the prevention and treatment of now-ubiquitous atherosclerotic diseases.
Collapse
Affiliation(s)
- Zichen Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yi Chen
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection, Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Yi Wang
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Wenbo Han
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Wenfeng Xu
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection, Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Xiaoling Liao
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection, Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Tao Zhang
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection, Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
- Bioengineering College of Chongqing University, NO.174, Shazheng Street, Shapingba District, Chongqing, 400030, PR China.
| |
Collapse
|
4
|
Ma C, Du T, Niu X, Fan Y. Biomechanics and mechanobiology of the bone matrix. Bone Res 2022; 10:59. [PMID: 36042209 PMCID: PMC9427992 DOI: 10.1038/s41413-022-00223-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 05/13/2022] [Accepted: 05/27/2022] [Indexed: 11/23/2022] Open
Abstract
The bone matrix plays an indispensable role in the human body, and its unique biomechanical and mechanobiological properties have received much attention. The bone matrix has unique mechanical anisotropy and exhibits both strong toughness and high strength. These mechanical properties are closely associated with human life activities and correspond to the function of bone in the human body. None of the mechanical properties exhibited by the bone matrix is independent of its composition and structure. Studies on the biomechanics of the bone matrix can provide a reference for the preparation of more applicable bone substitute implants, bone biomimetic materials and scaffolds for bone tissue repair in humans, as well as for biomimetic applications in other fields. In providing mechanical support to the human body, bone is constantly exposed to mechanical stimuli. Through the study of the mechanobiology of the bone matrix, the response mechanism of the bone matrix to its surrounding mechanical environment can be elucidated and used for the health maintenance of bone tissue and defect regeneration. This paper summarizes the biomechanical properties of the bone matrix and their biological significance, discusses the compositional and structural basis by which the bone matrix is capable of exhibiting these mechanical properties, and studies the effects of mechanical stimuli, especially fluid shear stress, on the components of the bone matrix, cells and their interactions. The problems that occur with regard to the biomechanics and mechanobiology of the bone matrix and the corresponding challenges that may need to be faced in the future are also described.
Collapse
Affiliation(s)
- Chunyang Ma
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Tianming Du
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China
| | - Xufeng Niu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China. .,Research Institute of Beihang University in Shenzhen, Shenzhen, 518057, China.
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China. .,School of Engineering Medicine, Beihang University, Beijing, 100083, China.
| |
Collapse
|
5
|
Paapstel K, Kals J. Metabolomics of Arterial Stiffness. Metabolites 2022; 12:370. [PMID: 35629874 PMCID: PMC9146333 DOI: 10.3390/metabo12050370] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 12/18/2022] Open
Abstract
Arterial stiffness (AS) is one of the earliest detectable signs of structural and functional alterations of the vessel wall and an independent predictor of cardiovascular events and death. The emerging field of metabolomics can be utilized to detect a wide spectrum of intermediates and products of metabolism in body fluids that can be involved in the pathogenesis of AS. Research over the past decade has reinforced this idea by linking AS to circulating acylcarnitines, glycerophospholipids, sphingolipids, and amino acids, among other metabolite species. Some of these metabolites influence AS through traditional cardiovascular risk factors (e.g., high blood pressure, high blood cholesterol, diabetes, smoking), while others seem to act independently through both known and unknown pathophysiological mechanisms. We propose the term 'arteriometabolomics' to indicate the research that applies metabolomics methods to study AS. The 'arteriometabolomics' approach has the potential to allow more personalized cardiovascular risk stratification, disease monitoring, and treatment selection. One of its major goals is to uncover the causal metabolic pathways of AS. Such pathways could represent valuable treatment targets in vascular ageing.
Collapse
Affiliation(s)
- Kaido Paapstel
- Endothelial Research Centre, University of Tartu, 8 Puusepa Street, 51014 Tartu, Estonia;
- Department of Cardiology, Institute of Clinical Medicine, University of Tartu, 8 Puusepa Street, 51014 Tartu, Estonia
- Heart Clinic, Tartu University Hospital, 8 Puusepa Street, 51014 Tartu, Estonia
| | - Jaak Kals
- Endothelial Research Centre, University of Tartu, 8 Puusepa Street, 51014 Tartu, Estonia;
- Department of Surgery, Institute of Clinical Medicine, University of Tartu, 8 Puusepa Street, 51014 Tartu, Estonia
- Surgery Clinic, Tartu University Hospital, 8 Puusepa Street, 51014 Tartu, Estonia
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, Centre of Excellence for Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| |
Collapse
|
6
|
Vascular Pathobiology: Atherosclerosis and Large Vessel Disease. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00006-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
7
|
Basehore SE, Bohlman S, Weber C, Swaminathan S, Zhang Y, Jang C, Arany Z, Clyne AM. Laminar Flow on Endothelial Cells Suppresses eNOS O-GlcNAcylation to Promote eNOS Activity. Circ Res 2021; 129:1054-1066. [PMID: 34605247 DOI: 10.1161/circresaha.121.318982] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Sarah E Basehore
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA (S.E.B., S.S.).,Fischell Department of Biomedical Engineering, College of Engineering, University of Maryland, College Park (S.B., C.W., A.M.C.)
| | - Samantha Bohlman
- Fischell Department of Biomedical Engineering, College of Engineering, University of Maryland, College Park (S.B., C.W., A.M.C.)
| | - Callie Weber
- Fischell Department of Biomedical Engineering, College of Engineering, University of Maryland, College Park (S.B., C.W., A.M.C.)
| | - Swathi Swaminathan
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA (S.E.B., S.S.)
| | - Yuji Zhang
- Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore (Y.Z.)
| | - Cholsoon Jang
- Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine (C.J.)
| | - Zoltan Arany
- Perelman School of Medicine, University of Pennsylvania, Philadelphia (Z.A.)
| | - Alisa Morss Clyne
- Fischell Department of Biomedical Engineering, College of Engineering, University of Maryland, College Park (S.B., C.W., A.M.C.)
| |
Collapse
|
8
|
Vo J, Mastoor Y, Mathieu PS, Clyne AM. A simple method to align cells on 3D hydrogels using 3D printed molds. BIOMEDICAL ENGINEERING ADVANCES 2021; 1. [PMID: 35663509 PMCID: PMC9165732 DOI: 10.1016/j.bea.2021.100001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Vascular smooth muscle cells align circumferentially around the vessel lumen, which allows these cells to control vascular tone by contracting and relaxing. It is essential that this circumferential alignment is recapitulated in tissue engineered blood vessels. While many methods have been reported to align cells on 2D polymeric substrates, few techniques enable cell alignment on a 3D physiologically relevant hydrogel substrate. We hypothesized that the ridges inherent to the sides of fused deposition modeling 3D printed molds could be used to topographically pattern both stiff and soft substrates and thereby align cells on flat and curved surfaces. Flat and curved molds with 150, 250, and 350 μm ridges were 3D printed and used to topographically pattern polydimethylsiloxane and gelatin-methacryloyl. The ridges transferred to both substrates with less than 10% change in ridge size. Vascular smooth muscle cells were then seeded on each substrate, and nuclear and actin alignment were quantified. Cells were highly aligned with the molded ridges to a similar extent on both the stiffer polydimethylsiloxane and the softer gelatin-methacryloyl substrates. These data confirm that fused deposition modeling 3D printed molds are a rapid, cost-effective way to topographically pattern stiff and soft substrates in varied 3D shapes. This method will enable investigators to align cells on 3D polymeric and hydrogel structures for tissue engineering and other applications.
Collapse
|
9
|
Diabetes and Thrombosis: A Central Role for Vascular Oxidative Stress. Antioxidants (Basel) 2021; 10:antiox10050706. [PMID: 33946846 PMCID: PMC8146432 DOI: 10.3390/antiox10050706] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/19/2021] [Accepted: 04/27/2021] [Indexed: 11/17/2022] Open
Abstract
Diabetes mellitus is the fifth most common cause of death worldwide. Due to its chronic nature, diabetes is a debilitating disease for the patient and a relevant cost for the national health system. Type 2 diabetes mellitus is the most common form of diabetes mellitus (90% of cases) and is characteristically multifactorial, with both genetic and environmental causes. Diabetes patients display a significant increase in the risk of developing cardiovascular disease compared to the rest of the population. This is associated with increased blood clotting, which results in circulatory complications and vascular damage. Platelets are circulating cells within the vascular system that contribute to hemostasis. Their increased tendency to activate and form thrombi has been observed in diabetes mellitus patients (i.e., platelet hyperactivity). The oxidative damage of platelets and the function of pro-oxidant enzymes such as the NADPH oxidases appear central to diabetes-dependent platelet hyperactivity. In addition to platelet hyperactivity, endothelial cell damage and alterations of the coagulation response also participate in the vascular damage associated with diabetes. Here, we present an updated interpretation of the molecular mechanisms underlying vascular damage in diabetes, including current therapeutic options for its control.
Collapse
|
10
|
Endothelial response to glucose: dysfunction, metabolism, and transport. Biochem Soc Trans 2021; 49:313-325. [PMID: 33522573 DOI: 10.1042/bst20200611] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/23/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
The endothelial cell response to glucose plays an important role in both health and disease. Endothelial glucose-induced dysfunction was first studied in diabetic animal models and in cells cultured in hyperglycemia. Four classical dysfunction pathways were identified, which were later shown to result from the common mechanism of mitochondrial superoxide overproduction. More recently, non-coding RNA, extracellular vesicles, and sodium-glucose cotransporter-2 inhibitors were shown to affect glucose-induced endothelial dysfunction. Endothelial cells also metabolize glucose for their own energetic needs. Research over the past decade highlighted how manipulation of endothelial glycolysis can be used to control angiogenesis and microvascular permeability in diseases such as cancer. Finally, endothelial cells transport glucose to the cells of the blood vessel wall and to the parenchymal tissue. Increasing evidence from the blood-brain barrier and peripheral vasculature suggests that endothelial cells regulate glucose transport through glucose transporters that move glucose from the apical to the basolateral side of the cell. Future studies of endothelial glucose response should begin to integrate dysfunction, metabolism and transport into experimental and computational approaches that also consider endothelial heterogeneity, metabolic diversity, and parenchymal tissue interactions.
Collapse
|
11
|
Bai S, Chaurasiya AH, Banarjee R, Walke PB, Rashid F, Unnikrishnan AG, Kulkarni MJ. CD44, a Predominant Protein in Methylglyoxal-Induced Secretome of Muscle Cells, is Elevated in Diabetic Plasma. ACS OMEGA 2020; 5:25016-25028. [PMID: 33043179 PMCID: PMC7542587 DOI: 10.1021/acsomega.0c01318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/16/2020] [Indexed: 06/11/2023]
Abstract
Methylglyoxal (MG), a glycolytic intermediate and reactive dicarbonyl, is responsible for exacerbation of insulin resistance and diabetic complication. In this study, MG-induced secretome of rat muscle cells was identified and relatively quantified by SWATH-MS. A total of 643 proteins were identified in MG-induced secretome, of which 82 proteins were upregulated and 99 proteins were downregulated by more than 1.3-fold in SWATH analysis. Further, secretory proteins from the classical secretory pathway and nonclassical secretory pathway were identified using SignalP and SecretomeP, respectively. A total of 180 proteins were identified with SignalP, and 113 proteins were identified with SecretomeP. The differentially expressed proteins were functionally annotated by KEGG pathway analysis using Cytoscape software with plugin clusterMaker. The differentially expressed proteins were found to be involved in various pathways like extracellular matrix (ECM)-receptor interaction, leukocyte transendothelial migration, fluid shear stress and atherosclerosis, complement and coagulation cascades, and lysosomal pathway. Since the MG levels are high in diabetic conditions, the presence of MG-induced secreted proteins was inspected by profiling human plasma of healthy and diabetic subjects (n = 10 each). CD44, a predominant MG-induced secreted protein, was found to be elevated in the diabetic plasma and to have a role in the development of insulin resistance.
Collapse
Affiliation(s)
- Shakuntala Bai
- Proteomics
Facility, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Arvindkumar H. Chaurasiya
- Proteomics
Facility, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Reema Banarjee
- Proteomics
Facility, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India
| | - Prachi B. Walke
- Proteomics
Facility, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Faraz Rashid
- Sciex, 121 DHR, Udyog Vihar, Phase IV, Gurugram 122015, Haryana, India
| | | | - Mahesh J. Kulkarni
- Proteomics
Facility, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| |
Collapse
|
12
|
Haque E, Kamil M, Hasan A, Irfan S, Sheikh S, Khatoon A, Nazir A, Mir SS. Advanced glycation end products (AGEs), protein aggregation and their cross talk: new insight in tumorigenesis. Glycobiology 2020; 30:49-57. [PMID: 31508802 DOI: 10.1093/glycob/cwz073] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 08/21/2019] [Accepted: 08/28/2019] [Indexed: 12/15/2022] Open
Abstract
Protein glycation and protein aggregation are two distinct phenomena being observed in cancer cells as factors promoting cancer cell viability. Protein aggregation is an abnormal interaction between proteins caused as a result of structural changes in them after any mutation or environmental assault. Protein aggregation is usually associated with neurodegenerative diseases like Alzheimer's and Parkinson's, but of late, research findings have shown its association with the development of different cancers like lung, breast and ovarian cancer. On the contrary, protein glycation is a cascade of irreversible nonenzymatic reaction of reducing sugar with the amino group of the protein resulting in the modification of protein structure and formation of advanced glycation end products (AGEs). These AGEs are reported to obstruct the normal function of proteins. Lately, it has been reported that protein aggregation occurs as a result of AGEs. This aggregation of protein promotes the transformation of healthy cells to neoplasia leading to tumorigenesis. In this review, we underline the current knowledge of protein aggregation and glycation along with the cross talk between the two, which may eventually lead to the development of cancer.
Collapse
Affiliation(s)
- Ejazul Haque
- Department of Biosciences, Faculty of Science, Integral University, Kursi Road, Lucknow 226026, India.,Department of Immunology and Medical Genetics, School of Medicine, University of Split, Soltanskaul. 2, 21000, Split, Croatia
| | - Mohd Kamil
- Department of Biosciences, Faculty of Science, Integral University, Kursi Road, Lucknow 226026, India.,Department of Immunology and Medical Genetics, School of Medicine, University of Split, Soltanskaul. 2, 21000, Split, Croatia.,Department of Microbiology, Beykoz Life Sciences and Biotechnology Institute (BILSAB), Bezmialem Vakif University, Istanbul, Turkey
| | - Adria Hasan
- Department of Bioengineering, Faculty of Engineering, Integral University, Kursi Road, Lucknow 226026, India
| | - Safia Irfan
- Department of Bioengineering, Faculty of Engineering, Integral University, Kursi Road, Lucknow 226026, India
| | - Saba Sheikh
- Department of Biosciences, Faculty of Science, Integral University, Kursi Road, Lucknow 226026, India
| | - Aisha Khatoon
- Department of Bioengineering, Faculty of Engineering, Integral University, Kursi Road, Lucknow 226026, India
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram extension, Sitapur Road, Lucknow, 226031, India
| | - Snober S Mir
- Department of Bioengineering, Faculty of Engineering, Integral University, Kursi Road, Lucknow 226026, India
| |
Collapse
|
13
|
Segoviano-Ramirez JC, Lopez-Altamirano DF, Garcia-Juarez J, Aguirre-Garza JES, Cárdenas-Estrada E, Ancer-Rodriguez J. The Diethylcarbamazine Delays and Decreases the NETosis of Polymorphonuclear Cells of Humans with DM Type 2. J Diabetes Res 2020; 2020:4827641. [PMID: 32190698 PMCID: PMC7072105 DOI: 10.1155/2020/4827641] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/16/2020] [Indexed: 12/30/2022] Open
Abstract
Type 2 diabetes mellitus (DM2) is a disease that reports high morbidity and mortality rates worldwide. Between its complications, one of the most important is the development of plantar ulcers. The role of the polymorphonuclear cells (PMNs) is affected by metabolic diseases like DM2. Fifteen years ago, reports about a new mechanism of innate immune response where PMNs generate some kind of webs with their chromatin were published. This mechanism was called NETosis. Also, some researchers have demonstrated that NETosis is responsible for the delay of the ulcer healing both in patients with DM2 and in animal models of DM2. Purified PMNs from healthy and DM2 human volunteers were incubated with diethylcarbamazine (DEC) and then induced to NETosis using phorbol 12-myristate 13-acetate (PMA). In a randomized blind study model, the NETosis was documented by confocal microscopy. On microphotographs, the area of each extracellular neutrophil trap (NET) formed at different times after stimuli with PMA was bounded, and the intensity of fluorescence (IF) from the chromatin dyed with 4',6-diamidino-2-phenylindole dihydrochloride (DAPI) was quantified. PMNs from healthy volunteers showed the development of NETs at expected times according to the literature. The same phenomenon was seen in cultures of PMNs from metabolically controlled DM2 volunteers. The use of DEC one hour before of the challenge with PMA delayed the NETosis in both groups. The semiquantitative morphometric analysis of the IF from DAPI, as a measure of PMN's capacity to forming NETs, is consistent with these results. The ANOVA test demonstrated that NETosis was lower and appeared later than expected time, both in PMNs from healthy (p ≤ 0.000001) and from DM2 (p ≤ 0.000477) volunteers. In conclusion, the DEC delays and decreases the NETosis by PMNs from healthy as well as DM2 people.
Collapse
Affiliation(s)
- Juan C. Segoviano-Ramirez
- Departamento de Patología, Facultad de Medicina, Universidad Autónoma de Nuevo León, UANL, Madero y Dr. Aguirre Pequeño, Mitras Centro, C.P. 64460, Mexico
- Unidad de Bioimagen, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autónoma de Nuevo León, UANL, Gonzalitos y Dr. Carlos Canseco, Mitras Centro, C.P. 64460, Mexico
| | - Daniel F. Lopez-Altamirano
- Unidad de Bioimagen, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autónoma de Nuevo León, UANL, Gonzalitos y Dr. Carlos Canseco, Mitras Centro, C.P. 64460, Mexico
| | - Jaime Garcia-Juarez
- Unidad de Bioimagen, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autónoma de Nuevo León, UANL, Gonzalitos y Dr. Carlos Canseco, Mitras Centro, C.P. 64460, Mexico
| | - Juan E. S. Aguirre-Garza
- Unidad de Bioimagen, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autónoma de Nuevo León, UANL, Gonzalitos y Dr. Carlos Canseco, Mitras Centro, C.P. 64460, Mexico
| | - Eloy Cárdenas-Estrada
- Unidad de Ensayos Clínicos, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autónoma de Nuevo León, UANL, Gonzalitos y Dr. Carlos Canseco, Mitras Centro, C.P. 64460, Mexico
| | - Jesús Ancer-Rodriguez
- Departamento de Patología, Facultad de Medicina, Universidad Autónoma de Nuevo León, UANL, Madero y Dr. Aguirre Pequeño, Mitras Centro, C.P. 64460, Mexico
| |
Collapse
|
14
|
Kistenev YV, Vrazhnov DA, Nikolaev VV, Sandykova EA, Krivova NA. Analysis of Collagen Spatial Structure Using Multiphoton Microscopy and Machine Learning Methods. BIOCHEMISTRY (MOSCOW) 2019; 84:S108-S123. [PMID: 31213198 DOI: 10.1134/s0006297919140074] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pathogenesis of many diseases is associated with changes in the collagen spatial structure. Traditionally, the 3D structure of collagen in biological tissues is analyzed using histochemistry, immunohistochemistry, magnetic resonance imaging, and X-radiography. At present, multiphoton microscopy (MPM) is commonly used to study the structure of biological tissues. MPM has a high spatial resolution comparable to histological analysis and can be used for direct visualization of collagen spatial structure. Because of a large volume of data accumulated due to the high spatial resolution of MPM, special analytical methods should be used for identification of informative features in the images and quantitative evaluation of relationship between these features and pathological processes resulting in the destruction of collagen structure. Here, we describe current approaches and achievements in the identification of informative features in the MPM images of collagen in biological tissues, as well as the development on this basis of algorithms for computer-aided classification of collagen structures using machine learning as a type of artificial intelligence methods.
Collapse
Affiliation(s)
- Yu V Kistenev
- Tomsk State University, Tomsk, 634050, Russia. .,Siberian State Medical University, Tomsk, 634050, Russia.,Institute of Strength Physics and Materials Science, Siberian Branch of the Russian Academy of Sciences, Tomsk, 634055, Russia
| | - D A Vrazhnov
- Tomsk State University, Tomsk, 634050, Russia.,Siberian State Medical University, Tomsk, 634050, Russia
| | - V V Nikolaev
- Tomsk State University, Tomsk, 634050, Russia.,Siberian State Medical University, Tomsk, 634050, Russia
| | - E A Sandykova
- Tomsk State University, Tomsk, 634050, Russia.,Siberian State Medical University, Tomsk, 634050, Russia
| | - N A Krivova
- Tomsk State University, Tomsk, 634050, Russia
| |
Collapse
|
15
|
Giri B, Dey S, Das T, Sarkar M, Banerjee J, Dash SK. Chronic hyperglycemia mediated physiological alteration and metabolic distortion leads to organ dysfunction, infection, cancer progression and other pathophysiological consequences: An update on glucose toxicity. Biomed Pharmacother 2018; 107:306-328. [PMID: 30098549 DOI: 10.1016/j.biopha.2018.07.157] [Citation(s) in RCA: 264] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/15/2018] [Accepted: 07/31/2018] [Indexed: 02/09/2023] Open
Abstract
Chronic exposure of glucose rich environment creates several physiological and pathophysiological changes. There are several pathways by which hyperglycemia exacerbate its toxic effect on cells, tissues and organ systems. Hyperglycemia can induce oxidative stress, upsurge polyol pathway, activate protein kinase C (PKC), enhance hexosamine biosynthetic pathway (HBP), promote the formation of advanced glycation end-products (AGEs) and finally alters gene expressions. Prolonged hyperglycemic condition leads to severe diabetic condition by damaging the pancreatic β-cell and inducing insulin resistance. Numerous complications have been associated with diabetes, thus it has become a major health issue in the 21st century and has received serious attention. Dysregulation in the cardiovascular and reproductive systems along with nephropathy, retinopathy, neuropathy, diabetic foot ulcer may arise in the advanced stages of diabetes. High glucose level also encourages proliferation of cancer cells, development of osteoarthritis and potentiates a suitable environment for infections. This review culminates how elevated glucose level carries out its toxicity in cells, metabolic distortion along with organ dysfunction and elucidates the complications associated with chronic hyperglycemia.
Collapse
Affiliation(s)
- Biplab Giri
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India; Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India.
| | - Sananda Dey
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India; Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Tanaya Das
- Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Mrinmoy Sarkar
- Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Jhimli Banerjee
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India
| | - Sandeep Kumar Dash
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India.
| |
Collapse
|
16
|
Li J, Zhang K, Huang N. Engineering Cardiovascular Implant Surfaces to Create a Vascular Endothelial Growth Microenvironment. Biotechnol J 2017; 12. [PMID: 28941232 DOI: 10.1002/biot.201600401] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 09/14/2017] [Indexed: 12/16/2022]
Abstract
Cardiovascular disease (CVD) is generally accepted as the leading cause of morbidity and mortality worldwide, and an increasing number of patients suffer from atherosclerosis and thrombosis annually. To treat these disorders and prolong the sufferers' life, several cardiovascular implants have been developed and applied clinically. Nevertheless, thrombosis and hyperplasia at the site of cardiovascular implants are recognized as long-term problems in the practice of interventional cardiology. Here, we start this review from the clinical requirement of the implants, such as anti-hyperplasia, anti-thrombosis, and pro-endothelialization, wherein particularly focus on the natural factors which influence functional endothelialization in situ, including the healthy smooth muscle cells (SMCs) environment, blood flow shear stress (BFSS), and the extracellular matrix (ECM) microenvironment. Then, the currently available strategies on surface modification of cardiovascular biomaterials to create vascular endothelial growth microenvironment are introduced as the main topic, e.g., BFSS effect simulation by surface micro-patterning, ECM rational construction and SMCs phenotype maintain. Finally, the prospects for extending use of the in situ construction of endothelial cells growth microenvironment are discussed and summarized in designing the next generation of vascular implants.
Collapse
Affiliation(s)
- Jingan Li
- School of Materials Science and Engineering, Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China.,Key Lab. for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Kun Zhang
- Key Lab. for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China.,School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China
| | - Nan Huang
- Key Lab. for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| |
Collapse
|
17
|
Berger AJ, Linsmeier KM, Kreeger PK, Masters KS. Decoupling the effects of stiffness and fiber density on cellular behaviors via an interpenetrating network of gelatin-methacrylate and collagen. Biomaterials 2017; 141:125-135. [PMID: 28683337 PMCID: PMC5556948 DOI: 10.1016/j.biomaterials.2017.06.039] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 06/26/2017] [Accepted: 06/28/2017] [Indexed: 12/13/2022]
Abstract
The extracellular microenvironment provides critical cues that guide tissue development, homeostasis, and pathology. Deciphering the individual roles of these cues in tissue function necessitates the development of physically tunable culture platforms, but current approaches to create such materials have produced scaffolds that either exhibit a limited mechanical range or are unable to recapitulate the fibrous nature of in vivo tissues. Here we report a novel interpenetrating network (IPN) of gelatin-methacrylate (gelMA) and collagen I that enables independent tuning of fiber density and scaffold stiffness across a physiologically-relevant range of shear moduli (2-12 kPa), while maintaining constant extracellular matrix content. This biomaterial system was applied to examine how changes in the physical microenvironment affect cell types associated with the tumor microenvironment. By increasing fiber density while maintaining constant stiffness, we found that MDA-MB-231 breast tumor cells required the presence of fibers to invade the surrounding matrix, while endothelial cells (ECs) did not. Meanwhile, increasing IPN stiffness independently of fiber content yielded decreased invasion and sprouting for both MDA-MB-231 cells and ECs. These results highlight the importance of decoupling features of the microenvironment to uncover their individual effects on cell behavior, in addition to demonstrating that individual cell types within a tissue may be differentially affected by the same changes in physical features. The mechanical range and fibrous nature of this tunable biomaterial platform enable mimicry of a wide variety of tissues, and may yield more precise identification of targets which may be exploited to develop interventions to control tissue function.
Collapse
Affiliation(s)
- Anthony J Berger
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Kelsey M Linsmeier
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Pamela K Kreeger
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States; Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States; Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States.
| | - Kristyn S Masters
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States; Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States; Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, United States.
| |
Collapse
|
18
|
Wang Y, Qiu J, Luo S, Xie X, Zheng Y, Zhang K, Ye Z, Liu W, Gregersen H, Wang G. High shear stress induces atherosclerotic vulnerable plaque formation through angiogenesis. Regen Biomater 2016; 3:257-67. [PMID: 27482467 PMCID: PMC4966293 DOI: 10.1093/rb/rbw021] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 05/15/2016] [Accepted: 05/19/2016] [Indexed: 12/12/2022] Open
Abstract
Rupture of atherosclerotic plaques causing thrombosis is the main cause of acute coronary syndrome and ischemic strokes. Inhibition of thrombosis is one of the important tasks developing biomedical materials such as intravascular stents and vascular grafts. Shear stress (SS) influences the formation and development of atherosclerosis. The current review focuses on the vulnerable plaques observed in the high shear stress (HSS) regions, which localizes at the proximal region of the plaque intruding into the lumen. The vascular outward remodelling occurs in the HSS region for vascular compensation and that angiogenesis is a critical factor for HSS which induces atherosclerotic vulnerable plaque formation. These results greatly challenge the established belief that low shear stress is important for expansive remodelling, which provides a new perspective for preventing the transition of stable plaques to high-risk atherosclerotic lesions.
Collapse
Affiliation(s)
- Yi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Shisui Luo
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Xiang Xie
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Yiming Zheng
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Kang Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Zhiyi Ye
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Wanqian Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Hans Gregersen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| |
Collapse
|
19
|
Hou L, Coller J, Natu V, Hastie TJ, Huang NF. Combinatorial extracellular matrix microenvironments promote survival and phenotype of human induced pluripotent stem cell-derived endothelial cells in hypoxia. Acta Biomater 2016; 44:188-99. [PMID: 27498178 DOI: 10.1016/j.actbio.2016.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 07/14/2016] [Accepted: 08/03/2016] [Indexed: 10/21/2022]
Abstract
UNLABELLED Recent developments in cell therapy using human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) hold great promise for treating ischemic cardiovascular tissues. However, poor post-transplantation viability largely limits the potential of stem cell therapy. Although the extracellular matrix (ECM) has become increasingly recognized as an important cell survival factor, conventional approaches primarily rely on single ECMs for in vivo co-delivery with cells, even though the endothelial basement membrane is comprised of a milieu of different ECMs. To address this limitation, we developed a combinatorial ECM microarray platform to simultaneously interrogate hundreds of micro-scale multi-component chemical compositions of ECMs on iPSC-EC response. After seeding iPSC-ECs onto ECM microarrays, we performed high-throughput analysis of the effects of combinatorial ECMs on iPSC-EC survival, endothelial phenotype, and nitric oxide production under conditions of hypoxia (1% O2) and reduced nutrients (1% fetal bovine serum), as is present in ischemic injury sites. Using automated image acquisition and analysis, we identified combinatorial ECMs such as collagen IV+gelatin+heparan sulfate+laminin and collagen IV+fibronectin+gelatin+heparan sulfate+laminin that significantly improved cell survival, nitric oxide production, and CD31 phenotypic expression, in comparison to single-component ECMs. These results were further validated in conventional cell culture platforms and within three-dimensional scaffolds. Furthermore, this approach revealed complex ECM interactions and non-intuitive cell behavior that otherwise could not be easily determined using conventional cell culture platforms. Together these data suggested that iPSC-EC delivery within optimal combinatorial ECMs may improve their survival and function under the condition of hypoxia with reduced nutrients. STATEMENT OF SIGNIFICANCE Human endothelial cells (ECs) derived from induced pluripotent stem cells (iPSC-ECs) are promising for treating diseases associated with reduced nutrient and oxygen supply like heart failure. However, diminished iPSC-EC survival after implantation into diseased environments limits their therapeutic potential. Since native ECs interact with numerous extracellular matrix (ECM) proteins for functional maintenance, we hypothesized that combinatorial ECMs may improve cell survival and function under conditions of reduced oxygen and nutrients. We developed a high-throughput system for simultaneous screening of iPSC-ECs cultured on multi-component ECM combinations under the condition of hypoxia and reduced serum. Using automated image acquisition and analytical algorithms, we identified combinatorial ECMs that significantly improved cell survival and function, in comparison to single ECMs. Furthermore, this approach revealed complex ECM interactions and non-intuitive cell behavior that otherwise could not be easily determined.
Collapse
|
20
|
Yurdagul A, Orr AW. Blood Brothers: Hemodynamics and Cell-Matrix Interactions in Endothelial Function. Antioxid Redox Signal 2016; 25:415-34. [PMID: 26715135 PMCID: PMC5011636 DOI: 10.1089/ars.2015.6525] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 11/25/2015] [Accepted: 12/23/2015] [Indexed: 12/29/2022]
Abstract
SIGNIFICANCE Alterations in endothelial function contribute to a variety of vascular diseases. In pathological conditions, the endothelium shows a reduced ability to regulate vasodilation (endothelial dysfunction) and a conversion toward a proinflammatory and leaky phenotype (endothelial activation). At the interface between the vessel wall and blood, the endothelium exists in a complex microenvironment and must translate changes in these environmental signals to alterations in vessel function. Mechanical stimulation and endothelial cell interactions with the vascular matrix, as well as a host of soluble factors, coordinately contribute to this dynamic regulation. RECENT ADVANCES Blood hemodynamics play an established role in the regulation of endothelial function. However, a growing body of work suggests that subendothelial matrix composition similarly and coordinately regulates endothelial cell phenotype such that blood flow affects matrix remodeling, which affects the endothelial response to flow. CRITICAL ISSUES Hemodynamics and soluble factors likely affect endothelial matrix remodeling through multiple mechanisms, including transforming growth factor β signaling and alterations in cell-matrix receptors, such as the integrins. Likewise, differential integrin signaling following matrix remodeling appears to regulate several key flow-induced responses, including nitric oxide production, regulation of oxidant stress, and activation of proinflammatory signaling and gene expression. Microvascular remodeling responses, such as angiogenesis and arteriogenesis, may also show coordinated regulation by flow and matrix. FUTURE DIRECTIONS Identifying the mechanisms regulating the dynamic interplay between hemodynamics and matrix remodeling and their contribution to the pathogenesis of cardiovascular disease remains an important research area with therapeutic implications across a variety of conditions. Antioxid. Redox Signal. 25, 415-434.
Collapse
Affiliation(s)
- Arif Yurdagul
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center–Shreveport, Shreveport, Louisiana
| | - A. Wayne Orr
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center–Shreveport, Shreveport, Louisiana
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, Louisiana
| |
Collapse
|
21
|
Clyne AM, Billiar KL. Problem-Based Learning in Biomechanics: Advantages, Challenges, and Implementation Strategies. J Biomech Eng 2016; 138:2525709. [PMID: 27210616 DOI: 10.1115/1.4033671] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Indexed: 11/08/2022]
Abstract
Problem-based learning (PBL) has been shown to be effective in biomedical engineering education, particularly in motivating student learning, increasing knowledge retention, and developing problem solving, communication, and teamwork skills. However, PBL adoption remains limited by real challenges in effective implementation. In this paper, we review the literature on advantages and challenges of PBL and present our own experiences. We also provide practical guidelines for implementing PBL, including two examples of PBL modules from biomechanics courses at two different institutions. Overall, we conclude that the benefits for both professors and students support the use of PBL in biomedical engineering education.
Collapse
|
22
|
Mohanan Nair M, Zhao R, Xie X, Shen GX. Impact of glycated LDL on endothelial nitric oxide synthase in vascular endothelial cells: involvement of transmembrane signaling and endoplasmic reticulum stress. J Diabetes Complications 2016; 30:391-7. [PMID: 26853630 DOI: 10.1016/j.jdiacomp.2016.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 12/23/2015] [Accepted: 01/11/2016] [Indexed: 01/01/2023]
Abstract
Cardiovascular diseases are the major cause of mortality in diabetes patients. Increased levels of glycated low density lipoprotein (glyLDL) are detected in diabetic patients. Endothelial nitric oxide synthase (eNOS) generates nitric oxide, which is responsible to endothelium-dependent vasodilation. The impact of glyLDL on the expression and activity of eNOS in vascular endothelial cells (EC) remains unknown. The present study investigated the effect of glyLDL on the levels of protein, mRNA and activity of eNOS in cultured human umbilical vein EC. The results demonstrated that incubation of EC with physiological concentrations of glyLDL significantly reduced the abundances of eNOS protein in EC with the maximal inhibition at 100μg/ml for 24h. At the optimized condition, glyLDL decreased eNOS mRNA and reduced its activity in EC. Blocking antibody against the receptor for advanced glycation end products (RAGE) prevented glyLDL-induced downregulation of eNOS in EC. GlyLDL increased the translocation of H-Ras from cytoplasm to membrane in EC. Farnesyl-transferase inhibitor-276, an H-Ras antagonist, normalized glyLDL-induced downregulation of eNOS and prevented glyLDL-induced upregulation of H-Ras in EC membrane. Treatment with 4-phenylbutyric acid, an endoplasmic reticulum (ER) stress antagonist, prevented glyLDL-induced eNOS downregulation in EC. The results suggest that diabetes-associated metabolic stress inhibits the production and activity of eNOA in cultured human vascular EC through the activation of RAGE/H-Ras mediated upstream signaling pathway. ER stress induced by glyLDL is possibly involved in eNOS downregulation.
Collapse
Affiliation(s)
- Manoj Mohanan Nair
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Ruozhi Zhao
- Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - Xueping Xie
- Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - Garry X Shen
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, Canada; Department of Internal Medicine, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
23
|
|
24
|
Palombo C, Kozakova M. Arterial stiffness, atherosclerosis and cardiovascular risk: Pathophysiologic mechanisms and emerging clinical indications. Vascul Pharmacol 2015; 77:1-7. [PMID: 26643779 DOI: 10.1016/j.vph.2015.11.083] [Citation(s) in RCA: 325] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 08/25/2015] [Accepted: 11/26/2015] [Indexed: 12/14/2022]
Abstract
Arterial stiffness results from a degenerative process affecting mainly the extracellular matrix of elastic arteries under the effect of aging and risk factors. Changes in extracellular matrix proteins and in the mechanical properties of the vessel wall related to arterial stiffening may activate number of mechanisms involved also in the process of atherosclerosis. Several noninvasive methods are now available to estimate large artery stiffness in the clinical setting, including carotid-femoral pulse wave velocity, the reference for aortic stiffness estimate, and local distensibility measures of superficial arteries, namely carotid and femoral. An independent predictive value of arterial stiffness for cardiovascular events has been demonstrated in general as well as in selected populations, and reference values adjusted for age and blood pressure have been established. Thus, arterial stiffness is emerging as an interesting tissue biomarker for cardiovascular risk stratification and estimation of the individual "biological age". This paper overviews the mechanisms accounting for development and progression of arterial stiffness and for associations between arterial stiffness, atherosclerotic burden and incident cardiovascular events, summarizes the evidence and caveat for clinical use of stiffness as surrogate marker of cardiovascular risk, and briefly outlines some emerging methods for large artery stiffness characterization.
Collapse
Affiliation(s)
- Carlo Palombo
- Department of Surgical, Medical and Molecular Pathology and Critical Area Medicine, University of Pisa, Italy.
| | - Michaela Kozakova
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
| |
Collapse
|
25
|
Unterman S, Freiman A, Beckerman M, Abraham E, Stanley JR, Levy E, Artzi N, Edelman E. Tuning of collagen scaffold properties modulates embedded endothelial cell regulatory phenotype in repair of vascular injuries in vivo. Adv Healthc Mater 2015; 4:2220-8. [PMID: 26333178 PMCID: PMC4664078 DOI: 10.1002/adhm.201500457] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/31/2015] [Indexed: 01/08/2023]
Abstract
Perivascularly implanted matrix embedded endothelial cells (MEECs) are potent regulators of inflammation and intimal hyperplasia following vascular injuries. Endothelial cells (ECs) in collagen scaffolds adopt a reparative phenotype with significant therapeutic potential. Although the biology of MEECs is increasingly understood, tuning of scaffold properties to control cell-substrate interactions is less well-studied. It is hypothesized that modulating scaffold degradation would change EC phenotype. Scaffolds with differential degradation are prepared by cross-linking and predegradation. Vascular injury increases degradation and the presence of MEECs retards injury-mediated degradation. MEECs respond to differential scaffold properties with altered viability in vivo, suppressed smooth muscle cell (SMC) proliferation in vitro, and altered interleukin-6 and matrix metalloproteinase-9 expression. When implanted perivascularly to a murine carotid wire injury, tuned scaffolds change MEEC effects on vascular repair and inflammation. Live animal imaging enables real-time tracking of cell viability, inflammation, and scaffold degradation, affording an unprecedented understanding of interactions between cells, substrate, and tissue. MEEC-treated injuries improve endothelialization and reduce SMC hyperplasia over 14 d. These data demonstrate the potent role material design plays in tuning MEEC efficacy in vivo, with implications for the design of clinical therapies.
Collapse
Affiliation(s)
- Shimon Unterman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alina Freiman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Ort Braude College, Karmiel, Israel
| | - Margarita Beckerman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Ort Braude College, Karmiel, Israel
| | - Eytan Abraham
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - James R.L. Stanley
- CBSET, Inc., Concord Biomedical Sciences and Emerging Technologies, Lexington, MA 02421, USA
| | - Ela Levy
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Ort Braude College, Karmiel, Israel
| | - Natalie Artzi
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elazer Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
26
|
Prevention of protein glycation by natural compounds. Molecules 2015; 20:3309-34. [PMID: 25690291 PMCID: PMC6272653 DOI: 10.3390/molecules20023309] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 12/14/2022] Open
Abstract
Non-enzymatic protein glycosylation (glycation) contributes to many diseases and aging of organisms. It can be expected that inhibition of glycation may prolong the lifespan. The search for inhibitors of glycation, mainly using in vitro models, has identified natural compounds able to prevent glycation, especially polyphenols and other natural antioxidants. Extrapolation of results of in vitro studies on the in vivo situation is not straightforward due to differences in the conditions and mechanism of glycation, and bioavailability problems. Nevertheless, available data allow to postulate that enrichment of diet in natural anti-glycating agents may attenuate glycation and, in consequence, ageing.
Collapse
|
27
|
Figueroa DS, Kemeny SF, Clyne AM. Glycated Collagen Decreased Endothelial Cell Fibronectin Alignment in Response to Cyclic Stretch Via Interruption of Actin Alignment. J Biomech Eng 2014; 136:101010. [DOI: 10.1115/1.4028037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 07/18/2014] [Indexed: 11/08/2022]
Abstract
Hyperglycemia is a defining characteristic of diabetes, and uncontrolled blood glucose in diabetes is associated with accelerated cardiovascular disease. Chronic hyperglycemia glycates extracellular matrix (ECM) collagen, which can lead to endothelial cell dysfunction. In healthy conditions, endothelial cells respond to mechanical stimuli such as cyclic stretch (CS) by aligning their actin cytoskeleton. Other cell types, specifically fibroblasts, align their ECM in response to CS. We previously demonstrated that glycated collagen inhibits endothelial cell actin alignment in response to CS. The aim of this study was to determine the effect of glycated collagen on ECM remodeling and protein alignment in response to stretch. Porcine aortic endothelial cells (PAEC) seeded on native or glycated collagen coated elastic substrates were exposed to 10% CS. Cells on native collagen aligned subcellular fibronectin fibers in response to stretch, whereas cells on glycated collagen did not. The loss of fibronectin alignment was due to inhibited actin alignment in response to CS, since fibronectin alignment did not occur in cells on native collagen when actin alignment was inhibited with cytochalasin. Further, while ECM protein content did not change in cells on native or glycated collagen in response to CS, degradation activity decreased in cells on glycated collagen. Matrix metalloproteinase 2 (MMP-2) and membrane-associated type 1 matrix metalloproteinase (MT1-MMP) protein levels decreased, and therefore MMP-2 activity also decreased. These MMP changes may relate to c-Jun N-terminal kinase (Jnk) phosphorylation inhibition with CS, which has previously been linked to focal adhesion kinase (FAK). These data demonstrate the importance of endothelial cell actin tension in remodeling and aligning matrix proteins in response to mechanical stimuli, which is critical to vascular remodeling in health and disease.
Collapse
Affiliation(s)
- Dannielle S. Figueroa
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104
| | - Steven F. Kemeny
- Mechanical Engineering and Mechanics, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104
| | - Alisa Morss Clyne
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104
- Mechanical Engineering and Mechanics, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104 e-mail:
| |
Collapse
|
28
|
Andrews AM, Jaron D, Buerk DG, Barbee KA. Shear stress-induced NO production is dependent on ATP autocrine signaling and capacitative calcium entry. Cell Mol Bioeng 2014; 7:510-520. [PMID: 25386222 DOI: 10.1007/s12195-014-0351-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Flow-induced production of nitric oxide (NO) by endothelial cells plays a fundamental role in vascular homeostasis. However, the mechanisms by which shear stress activates NO production remain unclear due in part to limitations in measuring NO, especially under flow conditions. Shear stress elicits the release of ATP, but the relative contribution of autocrine stimulation by ATP to flow-induced NO production has not been established. Furthermore, the importance of calcium in shear stress-induced NO production remains controversial, and in particular the role of capacitive calcium entry (CCE) has yet to be determined. We have utilized our unique NO measurement device to investigate the role of ATP autocrine signaling and CCE in shear stress-induced NO production. We found that endogenously released ATP and downstream activation of purinergic receptors and CCE plays a significant role in shear stress-induced NO production. ATP-induced eNOS phophorylation under static conditions is also dependent on CCE. Inhibition of protein kinase C significantly inhibited eNOS phosphorylation and the calcium response. To our knowledge, we are the first to report on the role of CCE in the mechanism of acute shear stress-induced NO response. In addition, our work highlights the importance of ATP autocrine signaling in shear stress-induced NO production.
Collapse
Affiliation(s)
- Allison M Andrews
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St. Philadelphia, PA 19104, USA
| | - Dov Jaron
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St. Philadelphia, PA 19104, USA
| | - Donald G Buerk
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St. Philadelphia, PA 19104, USA
| | - Kenneth A Barbee
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St. Philadelphia, PA 19104, USA
| |
Collapse
|
29
|
Xu J, Shi GP. Vascular wall extracellular matrix proteins and vascular diseases. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2106-2119. [PMID: 25045854 DOI: 10.1016/j.bbadis.2014.07.008] [Citation(s) in RCA: 255] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 07/07/2014] [Accepted: 07/14/2014] [Indexed: 01/08/2023]
Abstract
Extracellular matrix proteins form the basic structure of blood vessels. Along with providing basic structural support to blood vessels, matrix proteins interact with different sets of vascular cells via cell surface integrin or non-integrin receptors. Such interactions induce vascular cell de novo synthesis of new matrix proteins during blood vessel development or remodeling. Under pathological conditions, vascular matrix proteins undergo proteolytic processing, yielding bioactive fragments to influence vascular wall matrix remodeling. Vascular cells also produce alternatively spliced variants that induce vascular cell production of different matrix proteins to interrupt matrix homeostasis, leading to increased blood vessel stiffness; vascular cell migration, proliferation, or death; or vascular wall leakage and rupture. Destruction of vascular matrix proteins leads to vascular cell or blood-borne leukocyte accumulation, proliferation, and neointima formation within the vascular wall; blood vessels prone to uncontrolled enlargement during blood flow diastole; tortuous vein development; and neovascularization from existing pathological tissue microvessels. Here we summarize discoveries related to blood vessel matrix proteins within the past decade from basic and clinical studies in humans and animals - from expression to cross-linking, assembly, and degradation under physiological and vascular pathological conditions, including atherosclerosis, aortic aneurysms, varicose veins, and hypertension.
Collapse
Affiliation(s)
- Junyan Xu
- Department of Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
30
|
Hyperglycemia, hypoglycemia, and glycemic complexity are associated with worse outcomes after surgery. J Crit Care 2014; 29:611-7. [PMID: 24768531 DOI: 10.1016/j.jcrc.2014.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 02/26/2014] [Accepted: 03/10/2014] [Indexed: 11/21/2022]
Abstract
PURPOSE The purpose of this study was to determine if glycemic complexity, along with hypoglycemia and hyperglycemia, was associated with worse outcomes after cardiac surgery. MATERIALS AND METHODS We conducted a retrospective analysis of 970 patients who had insulin infusions designed to keep blood glucose levels between 80 and 110 mg/dL. Glycemic complexity was calculated using jackknifed approximate entropy. Logistic regression was used to adjust for confounders. RESULTS A total of 495 patients (51%) developed complications, and 32 patients (3.3%) died. Along with older age, comorbidities, and complicated surgeries, any hypoglycemia (glucose<71 mg/dL) and the number of glucose values greater than 140 mg/dL were independent predictors of complications. Increased risk of mortality, after adjusting for other risk factors, was associated with older age, longer perfusion time, receiving intraoperative transfusions, and greater jackknifed approximate entropy of the glucose time series. CONCLUSION We found that hypoglycemia (glucose<71 mg/dL) and hyperglycemia (glucose>140 mg/dL) were associated with increased risk of complications, whereas greater complexity of the glucose time series was associated with mortality.
Collapse
|
31
|
Singh VP, Bali A, Singh N, Jaggi AS. Advanced glycation end products and diabetic complications. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2014; 18:1-14. [PMID: 24634591 PMCID: PMC3951818 DOI: 10.4196/kjpp.2014.18.1.1] [Citation(s) in RCA: 933] [Impact Index Per Article: 84.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 10/11/2013] [Accepted: 12/10/2013] [Indexed: 02/06/2023]
Abstract
During long standing hyperglycaemic state in diabetes mellitus, glucose forms covalent adducts with the plasma proteins through a non-enzymatic process known as glycation. Protein glycation and formation of advanced glycation end products (AGEs) play an important role in the pathogenesis of diabetic complications like retinopathy, nephropathy, neuropathy, cardiomyopathy along with some other diseases such as rheumatoid arthritis, osteoporosis and aging. Glycation of proteins interferes with their normal functions by disrupting molecular conformation, altering enzymatic activity, and interfering with receptor functioning. AGEs form intra- and extracellular cross linking not only with proteins, but with some other endogenous key molecules including lipids and nucleic acids to contribute in the development of diabetic complications. Recent studies suggest that AGEs interact with plasma membrane localized receptors for AGEs (RAGE) to alter intracellular signaling, gene expression, release of pro-inflammatory molecules and free radicals. The present review discusses the glycation of plasma proteins such as albumin, fibrinogen, globulins and collagen to form different types of AGEs. Furthermore, the role of AGEs in the pathogenesis of diabetic complications including retinopathy, cataract, neuropathy, nephropathy and cardiomyopathy is also discussed.
Collapse
Affiliation(s)
- Varun Parkash Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala-147002, India
| | - Anjana Bali
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala-147002, India
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala-147002, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala-147002, India
| |
Collapse
|
32
|
Singh VP, Bali A, Singh N, Jaggi AS. Advanced glycation end products and diabetic complications. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2014. [PMID: 24634591 DOI: 10.4196/kjpp] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During long standing hyperglycaemic state in diabetes mellitus, glucose forms covalent adducts with the plasma proteins through a non-enzymatic process known as glycation. Protein glycation and formation of advanced glycation end products (AGEs) play an important role in the pathogenesis of diabetic complications like retinopathy, nephropathy, neuropathy, cardiomyopathy along with some other diseases such as rheumatoid arthritis, osteoporosis and aging. Glycation of proteins interferes with their normal functions by disrupting molecular conformation, altering enzymatic activity, and interfering with receptor functioning. AGEs form intra- and extracellular cross linking not only with proteins, but with some other endogenous key molecules including lipids and nucleic acids to contribute in the development of diabetic complications. Recent studies suggest that AGEs interact with plasma membrane localized receptors for AGEs (RAGE) to alter intracellular signaling, gene expression, release of pro-inflammatory molecules and free radicals. The present review discusses the glycation of plasma proteins such as albumin, fibrinogen, globulins and collagen to form different types of AGEs. Furthermore, the role of AGEs in the pathogenesis of diabetic complications including retinopathy, cataract, neuropathy, nephropathy and cardiomyopathy is also discussed.
Collapse
Affiliation(s)
- Varun Parkash Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala-147002, India
| | - Anjana Bali
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala-147002, India
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala-147002, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala-147002, India
| |
Collapse
|
33
|
Boonkaew B, Tompkins K, Manokawinchoke J, Pavasant P, Supaphol P. Characterization and cytological effects of a novel glycated gelatine substrate. Biomed Mater 2014; 9:025001. [DOI: 10.1088/1748-6041/9/2/025001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
34
|
Kemeny SF, Cicalese S, Figueroa DS, Clyne AM. Glycated collagen and altered glucose increase endothelial cell adhesion strength. J Cell Physiol 2013; 228:1727-36. [PMID: 23280505 DOI: 10.1002/jcp.24313] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 12/10/2012] [Indexed: 01/08/2023]
Abstract
Cell adhesion strength is important to cell survival, proliferation, migration, and mechanotransduction, yet changes in endothelial cell adhesion strength have not yet been examined in diseases such as diabetes with high rates of cardiovascular complications. We therefore investigated porcine aortic endothelial cell adhesion strength on native and glycated collagen-coated substrates and in low, normal, and high glucose culture using a spinning disc apparatus. Adhesion strength increased by 30 dynes/cm(2) in cells on glycated collagen as compared to native collagen. Attachment studies revealed that cells use higher adhesion strength αv β3 integrins to bind to glycated collagen instead of the typical α2 β1 integrins used to bind to native collagen. Similarly, endothelial cells cultured in low and high glucose had 15 dynes/cm(2) higher adhesion strength than cells in normal glucose after 2 days. Increased adhesion strength was due to elevated VEGF release and intracellular PKC in low and high glucose cells, respectively. Thus glucose increased endothelial cell adhesion strength via different underlying mechanisms. These adhesion strength changes could contribute to diabetic vascular disease, including accelerated atherosclerosis and disordered angiogenesis.
Collapse
Affiliation(s)
- Steven Frank Kemeny
- Department of Mechanical Engineering and Mechanics, Drexel University, Philadelphia, Pennsylvania, USA
| | | | | | | |
Collapse
|
35
|
Kemeny SF, Figueroa DS, Clyne AM. Hypo- and hyperglycemia impair endothelial cell actin alignment and nitric oxide synthase activation in response to shear stress. PLoS One 2013; 8:e66176. [PMID: 23776627 PMCID: PMC3680428 DOI: 10.1371/journal.pone.0066176] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 05/07/2013] [Indexed: 12/25/2022] Open
Abstract
Uncontrolled blood glucose in people with diabetes correlates with endothelial cell dysfunction, which contributes to accelerated atherosclerosis and subsequent myocardial infarction, stroke, and peripheral vascular disease. In vitro, both low and high glucose induce endothelial cell dysfunction; however the effect of altered glucose on endothelial cell fluid flow response has not been studied. This is critical to understanding diabetic cardiovascular disease, since endothelial cell cytoskeletal alignment and nitric oxide release in response to shear stress from flowing blood are atheroprotective. In this study, porcine aortic endothelial cells were cultured in 1, 5.55, and 33 mM D-glucose medium (low, normal, and high glucose) and exposed to 20 dynes/cm2 shear stress for up to 24 hours in a parallel plate flow chamber. Actin alignment and endothelial nitric oxide synthase phosphorylation increased with shear stress for cells in normal glucose, but not cells in low and high glucose. Both low and high glucose elevated protein kinase C (PKC) levels; however PKC blockade only restored actin alignment in high glucose cells. Cells in low glucose instead released vascular endothelial growth factor (VEGF), which translocated β-catenin away from the cell membrane and disabled the mechanosensory complex. Blocking VEGF in low glucose restored cell actin alignment in response to shear stress. These data suggest that low and high glucose alter endothelial cell alignment and nitric oxide production in response to shear stress through different mechanisms.
Collapse
Affiliation(s)
- Steven Frank Kemeny
- Mechanical Engineering and Mechanics, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Dannielle Solomon Figueroa
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Alisa Morss Clyne
- Mechanical Engineering and Mechanics, Drexel University, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
36
|
Mason BN, Reinhart-King CA. Controlling the mechanical properties of three-dimensional matrices via non-enzymatic collagen glycation. Organogenesis 2013; 9:70-5. [PMID: 23811696 DOI: 10.4161/org.24942] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The mechanical properties of the extracellular matrix play an important role in maintaining cellular function and overall tissue homeostasis. Recently, a number of hydrogel systems have been developed to investigate the role of matrix mechanics in mediating cell behavior within three-dimensional environments. However, many of the techniques used to modify the stiffness of the matrix also alter properties that are important to cellular function including matrix density, porosity and binding site frequency, or rely on amorphous synthetic materials. In a recent publication, we described the fabrication, characterization and utilization of collagen gels that have been non-enzymatically glycated in their unpolymerized form to produce matrices of varying stiffness. Using these scaffolds, we showed that the mechanical properties of the resulting collagen gels could be increased 3-fold without significantly altering the collagen fiber architecture. Using these matrices, we found that endothelial cell spreading and outgrowth from multi-cellular spheroids changes as a function of the stiffness of the matrix. Our results demonstrate that non-enzymatic collagen glycation is a tractable technique that can be used to study the role of 3D stiffness in mediating cellular function. This commentary will review some of the current methods that are being used to modulate matrix mechanics and discuss how our recent work using non-enzymatic collagen glycation can contribute to this field.
Collapse
Affiliation(s)
- Brooke N Mason
- Department of Biomedical Engineering, Cornell University, Ithaca, NY USA
| | | |
Collapse
|
37
|
Tuning three-dimensional collagen matrix stiffness independently of collagen concentration modulates endothelial cell behavior. Acta Biomater 2013; 9:4635-44. [PMID: 22902816 DOI: 10.1016/j.actbio.2012.08.007] [Citation(s) in RCA: 269] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 08/08/2012] [Accepted: 08/08/2012] [Indexed: 12/11/2022]
Abstract
Numerous studies have described the effects of matrix stiffening on cell behavior using two-dimensional synthetic surfaces; however, less is known about the effects of matrix stiffening on cells embedded in three-dimensional in vivo-like matrices. A primary limitation in investigating the effects of matrix stiffness in three dimensions is the lack of materials that can be tuned to control stiffness independently of matrix density. Here, we use collagen-based scaffolds where the mechanical properties are tuned using non-enzymatic glycation of the collagen in solution, prior to polymerization. Collagen solutions glycated prior to polymerization result in collagen gels with a threefold increase in compressive modulus without significant changes to the collagen architecture. Using these scaffolds, we show that endothelial cell spreading increases with matrix stiffness, as does the number and length of angiogenic sprouts and the overall spheroid outgrowth. Differences in sprout length are maintained even when the receptor for advanced glycation end products is inhibited. Our results demonstrate the ability to de-couple matrix stiffness from matrix density and structure in collagen gels, and that increased matrix stiffness results in increased sprouting and outgrowth.
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Differences in local blood flow patterns along the endothelium may trigger abnormal vascular responses which can have profound pathophysiological consequences. While endothelial cells exposed to laminar blood flow (high shear stress) are protected from atherosclerosis formation, turbulent or disturbed blood flow, which occurs at bends and bifurcations of blood vessels, facilitates atherosclerosis formation. Here, we will highlight the endothelial cell mechanisms involved in detecting shear stress and their translation into downstream biochemical signals. RECENT FINDINGS Prior evidence supports a role for integrins as mechanotransducers in the endothelium by promoting phosphorylation of different targets through the activation of focal adhesion kinase. Our recent findings show that integrins contact integrin-linked kinase and regulate vasomotor responses by an endothelial nitric oxide synthase-dependent mechanism, which stabilizes the production of vasoactive factor nitric oxide. In addition, different structures of endothelial cells, mainly primary cilia, are investigated, as they can explain the differential responses to laminar versus disturbed flow. SUMMARY The discovery of a connection between endothelial cell structures such as cilia, integrin, extracellular matrix, and signaling events opens today a new chapter in our understanding of the molecular mechanisms regulating vascular responses to the changes in flow.
Collapse
Affiliation(s)
- Carlos Zaragoza
- National Center for Cardiovascular Research, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | | | | |
Collapse
|
39
|
The role of scaffold microarchitecture in engineering endothelial cell immunomodulation. Biomaterials 2012; 33:7019-27. [PMID: 22796162 DOI: 10.1016/j.biomaterials.2012.06.052] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Accepted: 06/22/2012] [Indexed: 01/13/2023]
Abstract
The implantation of matrix-embedded endothelial cells (MEECs) has been reported to have great therapeutic potential in controlling the vascular response to injury and maintaining patency in arteriovenous anastomoses. While there is an appreciation of their effectiveness in clinical and animal studies, the mechanisms through which they mediate these powerful effects remain relatively unknown. In this work, we examined the hypothesis that the 3-dimensional microarchitecture of the tissue engineering scaffold was a key regulator of endothelial behavior in MEEC constructs. Notably, we found that ECs in porous collagen scaffold had a markedly altered cytoskeletal structure with oriented actin fibers and rearrangement of the focal adhesion proteins in comparison to cells grown on 2D surfaces. We examined the immunomodulatory capabilities of MEECs and discovered that they were able to reduce the recruitment of monocytes to an inflamed endothelial monolayer by 5-fold compared to EC on 2D surfaces. An analysis of secreted factors from the cells revealed an 8-fold lower release of Monocyte Chemotactic Protein-1 (MCP-1) from MEECs. Differences between 3D and 2D cultured cells were abolished in the presence of inhibitors to the focal adhesion associated signaling molecule Src suggesting that adhesion-mediated signaling is essential in controlling the potent immunomodulatory effects of MEEC.
Collapse
|
40
|
DeVerse JS, Bailey KA, Jackson KN, Passerini AG. Shear stress modulates RAGE-mediated inflammation in a model of diabetes-induced metabolic stress. Am J Physiol Heart Circ Physiol 2012; 302:H2498-508. [PMID: 22467309 DOI: 10.1152/ajpheart.00869.2011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Atherosclerosis occurs preferentially at sites of disturbed blood flow despite the influence of risk factors contributing to systemic inflammation. The receptor for advanced glycation endproducts (RAGE) is a prominent mediator of inflammation in diabetes that is upregulated in atherosclerotic plaques. Our goal was to elucidate a role for arterial hemodynamics in the regulation of RAGE expression and activity. Endothelial RAGE expression was elevated at sites of flow disturbance in the aortas of healthy swine. To demonstrate a direct role for physiological shear stress (SS) in modulating RAGE expression, human aortic endothelial cells (HAEC) were exposed to high SS (HSS; 15 dyn/cm(2)), which downregulated RAGE by fourfold, or oscillatory SS (OSS; 0 ± 5 dyn/cm(2)), which upregulated RAGE by threefold, compared with static culture at 4 h. In a model of diabetes-induced metabolic stress, HAEC were chronically conditioned under high glucose (25 mM) and then simultaneously stimulated with TNF-α (0.5 ng/ml) and the RAGE ligand high mobility group box 1 (HMGB1). A 50% increase in VCAM-1 expression over TNF-α was associated with increased cytoplasmic and mitochondrial reactive oxygen species and NF-κB activity. This increase was RAGE-specific and NADPH oxidase dependent. In activated HAEC, OSS amplified HMGB1-induced VCAM-1 (3-fold) and RAGE (1.6-fold) expression and proportionally enhanced monocyte adhesion to HAEC in a RAGE-dependent manner, while HSS mitigated these increases to the level of TNF-α alone. We demonstrate that SS plays a fundamental role in regulating RAGE expression and inflammatory responses in the endothelium. These findings may provide mechanistic insight into how diabetes accelerates the nonrandom distribution of atherosclerosis in arteries.
Collapse
Affiliation(s)
- J Sherrod DeVerse
- Department of Biomedical Engineering, University of California, Davis, California 95616, USA
| | | | | | | |
Collapse
|