1
|
Malamud M, Brown GD. The Dectin-1 and Dectin-2 clusters: C-type lectin receptors with fundamental roles in immunity. EMBO Rep 2024:10.1038/s44319-024-00296-2. [PMID: 39482490 DOI: 10.1038/s44319-024-00296-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 11/03/2024] Open
Abstract
The ability of myeloid cells to recognize and differentiate endogenous or exogenous ligands rely on the presence of different transmembrane protein receptors. C-type lectin receptors (CLRs), defined by the presence of a conserved structural motif called C-type lectin-like domain (CTLD), are a crucial family of receptors involved in this process, being able to recognize a diverse range of ligands from glycans to proteins or lipids and capable of initiating an immune response. The Dectin-1 and Dectin-2 clusters involve two groups of CLRs, with genes genomically linked within the natural killer cluster of genes in both humans and mice, and all characterized by the presence of a single extracellular CTLD. Fundamental immune cell functions such as antimicrobial effector mechanisms as well as internalization and presentation of antigens are induced and/or regulated through activatory, or inhibitory signalling pathways triggered by these receptors after ligand binding. In this review, we will discuss the most recent concepts regarding expression, ligands, signaling pathways and functions of each member of the Dectin clusters of CLRs, highlighting the importance and diversity of their functions.
Collapse
Affiliation(s)
- Mariano Malamud
- Medical Research Council (MRC) Centre for Medical Mycology, University of Exeter, Exeter, UK.
| | - Gordon D Brown
- Medical Research Council (MRC) Centre for Medical Mycology, University of Exeter, Exeter, UK.
| |
Collapse
|
2
|
Kruglikov IL, Scherer PE. Is the endotoxin-complement cascade the major driver in lipedema? Trends Endocrinol Metab 2024; 35:769-780. [PMID: 38688780 PMCID: PMC11387139 DOI: 10.1016/j.tem.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024]
Abstract
Lipedema is a poorly understood disorder of adipose tissue characterized by abnormal but symmetrical deposition of subcutaneous white adipose tissue (WAT) in proximal extremities. Here, we propose that the underlying cause for lipedema could be triggered by a selective accumulation of bacterial lipopolysaccharides (LPS; also known as endotoxin) in gluteofemoral WAT. Together with a malfunctioning complement system, this induces low-grade inflammation in the depot and raises its uncontrollable expansion. Correspondingly, more attention should be paid in future research to the endotoxemia prevalent in patients with lipedema. We would like to propose that proper management of endotoxemia can reduce the progression and even improve the state of disease in patients with lipedema.
Collapse
Affiliation(s)
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA.
| |
Collapse
|
3
|
Wu Y, Liu W, Li J, Shi H, Ma S, Wang D, Pan B, Xiao R, Jiang H, Liu X. Decreased Tiam1-mediated Rac1 activation is responsible for impaired directional persistence of chondrocyte migration in microtia. J Cell Mol Med 2024; 28:e18443. [PMID: 38837873 PMCID: PMC11149491 DOI: 10.1111/jcmm.18443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/06/2024] [Accepted: 05/11/2024] [Indexed: 06/07/2024] Open
Abstract
The human auricle has a complex structure, and microtia is a congenital malformation characterized by decreased size and loss of elaborate structure in the affected ear with a high incidence. Our previous studies suggest that inadequate cell migration is the primary cytological basis for the pathogenesis of microtia, however, the underlying mechanism is unclear. Here, we further demonstrate that microtia chondrocytes show a decreased directional persistence during cell migration. Directional persistence can define a leading edge associated with oriented movement, and any mistakes would affect cell function and tissue morphology. By the screening of motility-related genes and subsequent confirmations, active Rac1 (Rac1-GTP) is identified to be critical for the impaired directional persistence of microtia chondrocytes migration. Moreover, Rho guanine nucleotide exchange factors (GEFs) and Rho GTPase-activating proteins (GAPs) are detected, and overexpression of Tiam1 significantly upregulates the level of Rac1-GTP and improves directional migration in microtia chondrocytes. Consistently, decreased expression patterns of Tiam1 and active Rac1 are found in microtia mouse models, Bmp5se/J and Prkralear-3J/GrsrJ. Collectively, our results provide new insights into microtia development and therapeutic strategies of tissue engineering for microtia patients.
Collapse
Affiliation(s)
- Yi Wu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Wei Liu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Jia Li
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Hang Shi
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Shize Ma
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Di Wang
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Bo Pan
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Ran Xiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
- Key Laboratory of External Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Haiyue Jiang
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Xia Liu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
- Key Laboratory of External Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Sánchez-León ME, Loaeza-Reyes KJ, Matias-Cervantes CA, Mayoral-Andrade G, Pérez-Campos EL, Pérez-Campos-Mayoral L, Hernández-Huerta MT, Zenteno E, Pérez-Cervera Y, Pina-Canseco S. LOX-1 in Cardiovascular Disease: A Comprehensive Molecular and Clinical Review. Int J Mol Sci 2024; 25:5276. [PMID: 38791315 PMCID: PMC11121106 DOI: 10.3390/ijms25105276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
LOX-1, ORL-1, or lectin-like oxidized low-density lipoprotein receptor 1 is a transmembrane glycoprotein that binds and internalizes ox-LDL in foam cells. LOX-1 is the main receptor for oxidized low-density lipoproteins (ox-LDL). The LDL comes from food intake and circulates through the bloodstream. LOX-1 belongs to scavenger receptors (SR), which are associated with various cardiovascular diseases. The most important and severe of these is the formation of atherosclerotic plaques in the intimal layer of the endothelium. These plaques can evolve into complicated thrombi with the participation of fibroblasts, activated platelets, apoptotic muscle cells, and macrophages transformed into foam cells. This process causes changes in vascular endothelial homeostasis, leading to partial or total obstruction in the lumen of blood vessels. This obstruction can result in oxygen deprivation to the heart. Recently, LOX-1 has been involved in other pathologies, such as obesity and diabetes mellitus. However, the development of atherosclerosis has been the most relevant due to its relationship with cerebrovascular accidents and heart attacks. In this review, we will summarize findings related to the physiologic and pathophysiological processes of LOX-1 to support the detection, diagnosis, and prevention of those diseases.
Collapse
Affiliation(s)
- Maria Eugenia Sánchez-León
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
| | - Karen Julissa Loaeza-Reyes
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
- Centro de Estudios en Ciencias de la Salud y la Enfermedad, Facultad de Odontología, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico
| | - Carlos Alberto Matias-Cervantes
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
| | - Gabriel Mayoral-Andrade
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
| | | | - Laura Pérez-Campos-Mayoral
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
| | - María Teresa Hernández-Huerta
- Consejo Nacional de Humanidades, Ciencias y Tecnologías, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Mexico;
| | - Edgar Zenteno
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Yobana Pérez-Cervera
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
- Centro de Estudios en Ciencias de la Salud y la Enfermedad, Facultad de Odontología, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico
| | - Socorro Pina-Canseco
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
| |
Collapse
|
5
|
Munno M, Mallia A, Greco A, Modafferi G, Banfi C, Eligini S. Radical Oxygen Species, Oxidized Low-Density Lipoproteins, and Lectin-like Oxidized Low-Density Lipoprotein Receptor 1: A Vicious Circle in Atherosclerotic Process. Antioxidants (Basel) 2024; 13:583. [PMID: 38790688 PMCID: PMC11118168 DOI: 10.3390/antiox13050583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Atherosclerosis is a complex condition that involves the accumulation of lipids and subsequent plaque formation in the arterial intima. There are various stimuli, cellular receptors, and pathways involved in this process, but oxidative modifications of low-density lipoprotein (ox-LDL) are particularly important in the onset and progression of atherosclerosis. Ox-LDLs promote foam-cell formation, activate proinflammatory pathways, and induce smooth-muscle-cell migration, apoptosis, and cell death. One of the major receptors for ox-LDL is LOX-1, which is upregulated in several cardiovascular diseases, including atherosclerosis. LOX-1 activation in endothelial cells promotes endothelial dysfunction and induces pro-atherogenic signaling, leading to plaque formation. The binding of ox-LDLs to LOX-1 increases the generation of reactive oxygen species (ROS), which can induce LOX-1 expression and oxidize LDLs, contributing to ox-LDL generation and further upregulating LOX-1 expression. This creates a vicious circle that is amplified in pathological conditions characterized by high plasma levels of LDLs. Although LOX-1 has harmful effects, the clinical significance of inhibiting this protein remains unclear. Further studies both in vitro and in vivo are needed to determine whether LOX-1 inhibition could be a potential therapeutic target to counteract the atherosclerotic process.
Collapse
Affiliation(s)
- Marco Munno
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
| | - Alice Mallia
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
- Dipartimento di Biologia e Biotecnologie “Lazzaro Spallanzani”, Università di Pavia, 27100 Pavia, Italy
| | - Arianna Greco
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
| | - Gloria Modafferi
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
| | - Cristina Banfi
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
| | - Sonia Eligini
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
| |
Collapse
|
6
|
Jiang W, Gan C, Zhou X, Yang Q, Chen D, Xiao H, Dai L, Chen Y, Wang M, Yang H, Li Q. Klotho inhibits renal ox-LDL deposition via IGF-1R/RAC1/OLR1 signaling to ameliorate podocyte injury in diabetic kidney disease. Cardiovasc Diabetol 2023; 22:293. [PMID: 37891556 PMCID: PMC10612302 DOI: 10.1186/s12933-023-02025-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
OBJECTIVE Diabetic kidney disease (DKD) is characterized by the abnormal deposition of oxidized low-density lipoprotein (ox-LDL), which contributes to podocyte damage. Klotho, an aging suppressor that plays a critical role in protecting podocytes in DKD, is mainly expressed in kidney tubular epithelium and secreted in the blood. However, it has not been established whether Klotho can alleviate podocyte injury by inhibiting renal ox-LDL deposition, and the potential molecular mechanisms require further investigation. METHODS We conducted a comprehensive analysis of serum and kidney biopsy samples obtained from patients diagnosed with DKD. Additionally, to explore the underlying mechanism of Klotho in the deposition of ox-LDL in the kidneys, we employed a mouse model of DKD with the Klotho genotype induced by streptozotocin (STZ). Furthermore, we conducted meticulous in vitro experiments on podocytes to gain further insights into the specific role of Klotho in the deposition of ox-LDL within the kidney. RESULTS Our groundbreaking study unveiled the remarkable ability of the soluble form of Klotho to effectively inhibit high glucose-induced ox-LDL deposition in podocytes affected by DKD. Subsequent investigations elucidated that Klotho achieved this inhibition by reducing the expression of the insulin/insulin-like growth factor 1 receptor (IGF-1R), consequently leading to a decrease in the expression of Ras-related C3 botulinum toxin substrate 1 (RAC1) and an enhancement of mitochondrial function. Ultimately, this series of events culminated in a significant reduction in the expression of the oxidized low-density lipoprotein receptor (OLR1), thereby resulting in a notable decrease in renal ox-LDL deposition in DKD. CONCLUSION Our findings suggested that Klotho had the potential to mitigate podocyte injury and reduced high glucose-induced ox-LDL deposition in glomerulus by modulating the IGF-1R/RAC1/OLR1 signaling. These results provided valuable insights that could inform the development of novel strategies for diagnosing and treating DKD.
Collapse
Affiliation(s)
- Wei Jiang
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Chun Gan
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Xindi Zhou
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Qing Yang
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Dan Chen
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Han Xiao
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Lujun Dai
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Yaxi Chen
- Department of Infectious Diseases, Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, People's Republic of China
| | - Mo Wang
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Haiping Yang
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Qiu Li
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
7
|
Villalobos-Labra R, Liu R, Spaans F, Sáez T, Semeria Maitret T, Quon A, Sawamura T, Cooke CLM, Davidge ST. Placenta-Derived Extracellular Vesicles From Preeclamptic Pregnancies Impair Vascular Endothelial Function via Lectin-Like Oxidized LDL Receptor-1. Hypertension 2023; 80:2226-2238. [PMID: 37615097 DOI: 10.1161/hypertensionaha.123.21205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND Preeclampsia is a complex syndrome that includes maternal vascular dysfunction. Syncytiotrophoblast-derived extracellular vesicles from preeclampsia placentas (preeclampsia-STBEVs) were shown to induce endothelial dysfunction, but an endothelial transmembrane mediator is still unexplored. The LOX-1 (lectin-like oxidized low-density lipoprotein receptor-1) is a transmembrane scavenger receptor that can cause endothelial dysfunction, and its expression is increased in the endothelium of preeclampsia women. In this study, we hypothesized that LOX-1 mediates the effects of preeclampsia-STBEVs on endothelial function. METHODS Preeclampsia-STBEVs were collected by perfusion of placentas from women with preeclampsia and in vitro and ex vivo endothelial cell function were assessed. RESULTS In human umbilical vein endothelial cells, inhibition of LOX-1 with LOX-1 blocking antibody (TS20) reduced the uptake of preeclampsia-STBEVs (61.3±8.8%). TS20 prevented the activation of ERK (extracellular signal-regulated kinase, a kinase downstream of LOX-1) and reduced the activation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells; 21.1±8.0%) and nitrative stress (23.2±10.3%) that was induced by preeclampsia-STBEVs. Vascular function was assessed by wire myography in isolated mesenteric arteries from pregnant rats that were incubated overnight with preeclampsia-STBEVs±TS20. TS20 prevented endothelium-dependent vasodilation impairment induced by preeclampsia-STBEVs. Nitric oxide contribution to the relaxation was reduced by preeclampsia-STBEVs, which was prevented by TS20. Superoxide dismutase or apocynin, an inhibitor of NOX (nicotinamide adenine dinucleotide phosphate oxidase), restored the impaired endothelium-dependent vasodilation in arteries exposed to preeclampsia-STBEVs. CONCLUSIONS Taken together, our findings demonstrate that LOX-1 mediates the endothelial dysfunction induced by preeclampsia-STBEVs. Our study further expands on the mechanisms that may lead to adverse outcomes in preeclampsia and proposes LOX-1 as a potential target for future interventions.
Collapse
Affiliation(s)
- Roberto Villalobos-Labra
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Escuela de Medicina sede San Felipe (R.V.-L.), Universidad de Valparaíso, Chile
| | - Ricky Liu
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Department of Physiology (R.L., S.T.D.), University of Alberta, Edmonton, Canada
| | - Floor Spaans
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Tamara Sáez
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Departamento de Medicina Interna (T. Sáez), Universidad de Valparaíso, Chile
- Centro de Investigaciones Biomédicas, Escuela de Medicina, Facultad de Medicina (T. Sáez), Universidad de Valparaíso, Chile
| | - Tamara Semeria Maitret
- Department of Laboratory Medicine and Pathology (T.S.M.), University of Alberta, Edmonton, Canada
| | - Anita Quon
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Tatsuya Sawamura
- Departments of Molecular Pathophysiology and Life Innovation, Shinshu University, Matsumoto, Japan (T. Sawamura)
| | - Christy-Lynn M Cooke
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Sandra T Davidge
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Department of Physiology (R.L., S.T.D.), University of Alberta, Edmonton, Canada
| |
Collapse
|
8
|
Buonfiglio F, Xia N, Yüksel C, Manicam C, Jiang S, Zadeh JK, Musayeva A, Elksne E, Pfeiffer N, Patzak A, Li H, Gericke A. Studies on the Effects of Hypercholesterolemia on Mouse Ophthalmic Artery Reactivity. Diseases 2023; 11:124. [PMID: 37873768 PMCID: PMC10594501 DOI: 10.3390/diseases11040124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/10/2023] [Accepted: 09/18/2023] [Indexed: 10/25/2023] Open
Abstract
Atherogenic lipoproteins may impair vascular reactivity, leading to tissue damage in various organs, including the eye. This study aimed to investigate whether ophthalmic artery reactivity is affected in mice lacking the apolipoprotein E gene (ApoE-/-), a model for hypercholesterolemia and atherosclerosis. Twelve-month-old male ApoE-/- mice and age-matched wild-type controls were used to assess vascular reactivity using videomicroscopy. Moreover, the vascular mechanics, lipid content, levels of reactive oxygen species (ROS), and expression of pro-oxidant redox enzymes and the lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) were determined in vascular tissue. Unlike the aorta, the ophthalmic artery of ApoE-/- mice developed no signs of endothelial dysfunction and no signs of excessive lipid deposition. Remarkably, the levels of ROS, nicotinamide adenine dinucleotide phosphate oxidase 1 (NOX1), NOX2, NOX4, and LOX-1 were increased in the aorta but not in the ophthalmic artery of ApoE-/- mice. Our findings suggest that ApoE-/- mice develop endothelial dysfunction in the aorta by increased oxidative stress via the involvement of LOX-1, NOX1, and NOX2, whereas NOX4 may participate in media remodeling. In contrast, the ophthalmic artery appears to be resistant to chronic apolipoprotein E deficiency. A lack of LOX-1 expression/overexpression in response to increased oxidized low-density lipoprotein levels may be a possible mechanism of action.
Collapse
Affiliation(s)
- Francesco Buonfiglio
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Can Yüksel
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Caroline Manicam
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Subao Jiang
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Jenia Kouchek Zadeh
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Aytan Musayeva
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Eva Elksne
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Andreas Patzak
- Institute of Translational Physiology, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Huige Li
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| |
Collapse
|
9
|
Chiu TH, Ku CW, Ho TJ, Tsai KL, Yang YD, Ou HC, Chen HI. Schisanhenol ameliorates oxLDL-caused endothelial dysfunction by inhibiting LOX-1 signaling. ENVIRONMENTAL TOXICOLOGY 2023. [PMID: 36999521 DOI: 10.1002/tox.23788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/06/2023] [Accepted: 03/12/2023] [Indexed: 06/19/2023]
Abstract
Atherosclerotic lesions play a critical role in leading cardiovascular diseases. Oxidized low-density lipoprotein (OxLDL) is a vital risk factor for atherosclerosis since it acts a crucial role in endothelial dysfunction and foam cell formation. Schisanhenol, a composition extracted from the fruit of Schisandra rubriflora, has been reported to have antioxidative effects on human LDL oxidation. This study investigates whether Schisanhenol protects against oxLDL-mediated endothelial damage by modulating the lectin-like oxLDL receptor-1 (LOX-1)-mediated inflammatory processes. Human umbilical vein endothelial cells (HUVECs) were pre-treated with 10 or 20 μM Schisanhenol for 2 h and then exposed to 150 μg/mL oxLDL. We revealed that Schisanhenol reduced oxLDL-enhanced LOX-1 expression. We also found that oxLDL down-regulated endothelial nitric oxide synthase (eNOS) as well as activated inducible NOS (iNOS), thereby enhancing the generation of nitric oxide (NO). Moreover, oxLDL elevated the expression levels of phosphorylated-p38MAPK, subsequently promoting NF-κB-modulated inflammatory responses. Pretreatment with Schisanhenol exerted significant cytoprotective function in all the above-mentioned detrimental events. Results from this present study reveal that Schisanhenol has a potential therapeutic effect on preventing oxLDL-induced endothelial injuries.
Collapse
Affiliation(s)
- Tsan-Hung Chiu
- Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Chang-Wen Ku
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Kun-Ling Tsai
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Dung Yang
- Department of Rehabilitation, Asia University Hospital, Taichung, Taiwan
| | - Hsiu-Chung Ou
- Department of Physical Therapy, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Hsiu-I Chen
- Department of Physical Therapy, College of Medical and Health Science, Asia University, Taichung, Taiwan
- Department of Physical Therapy, Hungkuang University, Taichung, Taiwan
| |
Collapse
|
10
|
Pratt J, Haidara K, Annabi B. MT1-MMP Expression Levels and Catalytic Functions Dictate LDL Receptor-Related Protein-1 Ligand Internalization Capacity in U87 Glioblastoma Cells. Int J Mol Sci 2022; 23:ijms232214214. [PMID: 36430705 PMCID: PMC9692856 DOI: 10.3390/ijms232214214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Modulations in cell surface receptor ectodomain proteolytic shedding impact on receptor function and cancer biomarker expression. As such, heavily pursued therapeutic avenues have exploited LDL receptor-related protein-1 (LRP-1)-mediated capacity in internalizing Angiopep-2 (An2), a brain-penetrating peptide that allows An2-drug conjugates to cross the blood-brain tumor barrier (BBTB). Given that LRP-1 is proteolytically shed from the cell surface through matrix metalloproteinase (MMP) activity, the balance between MMP expression/function and LRP-1-mediated An2 internalization is unknown. In this study, we found that membrane type-1 (MT1)-MMP expression increased from grade 1 to 4 brain tumors, while that of LRP-1 decreased inversely. MMP pharmacological inhibitors such as Ilomastat, Doxycycline and Actinonin increased in vitro An2 internalization by up to 2.5 fold within a human grade IV-derived U87 glioblastoma cell model. Transient siRNA-mediated MT1-MMP gene silencing resulted in increased basal An2 cell surface binding and intracellular uptake, while recombinant MT1-MMP overexpression reduced both cell surface LRP-1 expression as well as An2 internalization. The addition of Ilomastat to cells overexpressing recombinant MT1-MMP restored LRP-1 expression at the cell surface and An2 uptake to levels comparable to those observed in control cells. Collectively, our data suggest that MT1-MMP expression status dictates An2-mediated internalization processes in part by regulating cell surface LRP-1 functions. Such evidence prompts preclinical evaluations of combined MMP inhibitors/An2-drug conjugate administration to potentially increase the treatment of high-MT1-MMP-expressing brain tumors.
Collapse
|
11
|
Chen X, Pang X, Yeo AJ, Xie S, Xiang M, Shi B, Yu G, Li C. The Molecular Mechanisms of Ferroptosis and Its Role in Blood-Brain Barrier Dysfunction. Front Cell Neurosci 2022; 16:889765. [PMID: 35663422 PMCID: PMC9160190 DOI: 10.3389/fncel.2022.889765] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
The blood-brain barrier (BBB) is a selective, semi-permeable layer of endothelial cells that protects the central nervous system from harmful substances circulating in blood. It is one of the important barriers of the nervous system. BBB dysfunction is an early pathophysiological change observed in nervous system diseases. There are few treatments for BBB dysfunction, so this motivates the review. Ferroptosis is a novel cell death mode caused by iron-mediated lipid peroxidation accumulation, which has recently attracted more attention due to its possible role in nervous system disorders. Studies have shown that lipid peroxidation and iron accumulation are related to the barrier dysfunction, especially the expression of tight junction proteins. Therefore, examination of the relationship between ferroptosis and BBB dysfunction may reveal new targets for the treatment of brain diseases.
Collapse
Affiliation(s)
- Xiaoshu Chen
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xinru Pang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Abrey J. Yeo
- University of Queensland Centre for Clinical Research, Brisbane, QLD, Australia
| | - Siwen Xie
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Mengting Xiang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Bin Shi
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Gongchang Yu
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- *Correspondence: Gongchang Yu,
| | - Chao Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Chao Li,
| |
Collapse
|
12
|
Li X, Tang X, Liu B, Zhang J, Zhang Y, Lv H, Liu D, Mehta JL, Wang X. LOX-1 Deletion Attenuates Myocardial Fibrosis in the Aged Mice, Particularly Those With Hypertension. Front Cardiovasc Med 2021; 8:736215. [PMID: 34712709 PMCID: PMC8545876 DOI: 10.3389/fcvm.2021.736215] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 09/14/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is a transmembrane glycoprotein that mediates uptake of oxidized low-density lipoprotein (ox-LDL) into cells. Previous studies had shown that LOX-1 deletion had a potential to inhibit cardiac fibrosis in mouse models of hypertension and myocardial infarction. Whether LOX-1 deletion also affects cardiac fibrosis associated with aging still remains unknown. The aim of this study was to investigate the effect of LOX-1 deletion on myocardial fibrosis in the aged mice. Methods: C57BL/6 mice and LOX-1 knockout (KO) mice with C57BL/6 background were studied to the age of 60 weeks. Both genotypes of aged mice were exposed to angiotensin II (Ang II) or saline for additional 4 weeks. The mice were then sacrificed, and myocardial fibrosis, reactive oxygen species (ROS) and expression of LOX-1, fibronectin, collagens, p22phox, and gp91phox were measured. Results: LOX-1 deletion markedly reduced Ang II-mediated rise of blood pressure in the aged mice (vs. saline-treated mice). LOX-1 deletion also limited fibrosis and decreased fibronectin and collagen-3 expression in the hearts of aged mice, but not the expression of collagen-1 and collagen-4. LOX-1 deletion also inhibited ROS production and p22phox expression. As the aged mice were exposed to Ang II for 4 weeks (resulting in hypertension), LOX-1 deletion more pronounced inhibiting myocardial fibrosis and ROS production, and decreasing expression of fibronectin, collagen-1, collagen-2, collagen-3, p22phox, and gp91phox. Conclusion: LOX-1 deletion limited fibrosis and ROS production in the hearts of aged mice. This effect was more pronounced in the aged mice with hypertension induced by Ang II infusion.
Collapse
Affiliation(s)
- Xiao Li
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Xihe Tang
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Bo Liu
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Jinghang Zhang
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yongxi Zhang
- Department of Oncology, The Third Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Hefan Lv
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Dongling Liu
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Jawahar L Mehta
- Division of Cardiology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Xianwei Wang
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China.,Department of Human Anatomy & Histoembryology, Xinxiang Medical University, Xinxiang, China.,Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
13
|
Bodac A, Meylan E. Neutrophil metabolism in the cancer context. Semin Immunol 2021; 57:101583. [PMID: 34963565 DOI: 10.1016/j.smim.2021.101583] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 12/30/2022]
Abstract
Neutrophils are critical innate immune cells for the host anti-bacterial defense. Throughout their lifecycle, neutrophils are exposed to different microenvironments and modulate their metabolism to survive and sustain their functions. Although tumor cell metabolism has been intensively investigated, how neutrophil metabolism is affected in cancer remains largely to be discovered. Neutrophils are described as mainly glycolytic cells. However, distinct tumor-associated neutrophil (TAN) states may co-exist in tumors and adapt their metabolism to exert different or even opposing activities ranging from tumor cell killing to tumor support. In this review, we gather evidence about the metabolic mechanisms that underly TANs' pro- or anti-tumoral functions in cancer. We first discuss how tumor-secreted factors and the heterogenous tumor microenvironment can have a strong impact on TAN metabolism. We then describe alternative metabolic pathways used by TANs to exert their functions in cancer, from basic glycolysis to more recently-recognized but less understood metabolic shifts toward mitochondrial oxidative metabolism, lipid and amino acid metabolism and even autophagy. Last, we discuss promising strategies targeting neutrophil metabolism to combat cancer.
Collapse
Affiliation(s)
- Anita Bodac
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015, Lausanne, Switzerland
| | - Etienne Meylan
- Lung Cancer & Immuno-Oncology Laboratory, Bordet Cancer Research Laboratories, Institut Jules Bordet, Faculty of Medicine, Université Libre de Bruxelles, 1070, Anderlecht, Belgium; Laboratory of Immunobiology, Faculty of Sciences, Université Libre de Bruxelles, 6041, Gosselies, Belgium; ULB Cancer Research Center (U-CRC) and ULB Center for Research in Immunology (U-CRI), Belgium.
| |
Collapse
|
14
|
Oxidized LDL but not angiotensin II induces cardiomyocyte hypertrophic responses through the interaction between LOX-1 and AT 1 receptors. J Mol Cell Cardiol 2021; 162:110-118. [PMID: 34555408 DOI: 10.1016/j.yjmcc.2021.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/15/2021] [Accepted: 09/08/2021] [Indexed: 01/19/2023]
Abstract
It is well known that lectin-like oxidized low-density lipoprotein (ox-LDL) and its receptor LOX-1, angiotensin II (AngII) and its type 1 receptor (AT1-R) play an important role in the development of cardiac hypertrophy. However, the molecular mechanism is not clear. In this study, we found that ox-LDL-induced cardiac hypertrophy was suppressed by inhibition of LOX-1 or AT1-R but not by AngII inhibition. These results suggest that the receptors LOX-1 and AT1-R, rather than AngII, play a key role in the role of ox-LDL. The same results were obtained in mice lacking endogenous AngII and their isolated cardiomyocytes. Ox-LDL but not AngII could induce the binding of LOX-1 and AT1-R; inhibition of LOX-1 or AT1-R but not AngII could abolish the binding of these two receptors. Overexpression of wild type LOX-1 with AT1-R enhanced ox-LDL-induced binding of two receptors and phosphorylation of ERKs, however, transfection of LOX-1 dominant negative mutant (lys266ala / lys267ala) or an AT1-R mutant (glu257ala) not only reduced the binding of two receptors but also inhibited the ERKs phosphorylation. Phosphorylation of ERKs induced by ox-LDL in LOX-1 and AT1-R-overexpression cells was abrogated by an inhibitor of Gq protein rather than Jak2, Rac1 or RhoA. Genetically, an AT1-R mutant lacking Gq protein coupling ability inhibited ox-LDL induced ERKs phosphorylation. Furthermore, through bimolecular fluorescence complementation analysis, we confirmed that ox-LDL rather than AngII stimulation induced the direct binding of LOX-1 and AT1-R. We conclude that direct binding of LOX-1 and AT1-R and the activation of downstream Gq protein are important mechanisms of ox-LDL-induced cardiomyocyte hypertrophy.
Collapse
|
15
|
Tan ML, Ling L, Fischbach C. Engineering strategies to capture the biological and biophysical tumor microenvironment in vitro. Adv Drug Deliv Rev 2021; 176:113852. [PMID: 34197895 PMCID: PMC8440401 DOI: 10.1016/j.addr.2021.113852] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022]
Abstract
Despite decades of research and advancements in diagnostic and treatment modalities, cancer remains a major global healthcare challenge. This is due in part to a lack of model systems that allow investigating the mechanisms underlying tumor development, progression, and therapy resistance under relevant conditions in vitro. Tumor cell interactions with their surroundings influence all stages of tumorigenesis and are shaped by both biological and biophysical cues including cell-cell and cell-extracellular matrix (ECM) interactions, tissue architecture and mechanics, and mass transport. Engineered tumor models provide promising platforms to elucidate the individual and combined contributions of these cues to tumor malignancy under controlled and physiologically relevant conditions. This review will summarize current knowledge of the biological and biophysical microenvironmental cues that influence tumor development and progression, present examples of in vitro model systems that are presently used to study these interactions and highlight advancements in tumor engineering approaches to further improve these technologies.
Collapse
Affiliation(s)
- Matthew L Tan
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Lu Ling
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
16
|
Status of biomarkers for the identification of stable or vulnerable plaques in atherosclerosis. Clin Sci (Lond) 2021; 135:1981-1997. [PMID: 34414413 DOI: 10.1042/cs20210417] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/04/2021] [Accepted: 08/09/2021] [Indexed: 01/18/2023]
Abstract
Atherosclerosis is a systemic inflammation of the arteries characterized by atherosclerotic plaque due to the accumulation of lipids, inflammatory cells, apoptotic cells, calcium and extracellular matrix (ECM) proteins. Stable plaques present a chronic inflammatory infiltration, whereas vulnerable plaques present an 'active' inflammation involved in the thinning of the fibrous cap that predisposes to plaque rupture. Several complex biological cellular processes lead plaques to evolve from stable to vulnerable predisposing them to rupture and thrombosis. In this review, we analyze some emerging circulating biomarkers related to inflammation, ECM and lipid infiltration, angiogenesis, metalloproteinases and microRNA (miRNA), as possible diagnostic and prognostic indicators of plaque vulnerability.
Collapse
|
17
|
Sugimoto K, Yokokawa T, Misaka T, Kaneshiro T, Yoshihisa A, Nakazato K, Takeishi Y. High-fat diet attenuates the improvement of hypoxia-induced pulmonary hypertension in mice during reoxygenation. BMC Cardiovasc Disord 2021; 21:331. [PMID: 34229630 PMCID: PMC8258936 DOI: 10.1186/s12872-021-02143-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 06/14/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND It is widely recognized that metabolic disorder is associated with pulmonary hypertension (PH). It is known that hypoxia-induced elevated pulmonary artery pressure in mice returns to normal pressure during reoxygenation. However, it is still unclear how metabolic disorder affects the reverse remodeling of pulmonary arteries. In this study, we investigated the effects of high-fat diet (HFD) on the decrease in pulmonary artery pressure and reverse remodeling of pulmonary arteries in mice with hypoxia-induced PH. METHODS We used female C57BL/6 mice aged 8 weeks. After being exposed to hypoxia (10% oxygen for four weeks) to induce PH, the mice were returned to normoxic conditions and randomized into a normal diet (ND) group and HFD group. Both groups were fed with their respective diets for 12 weeks. RESULTS The Fulton index and right ventricular systolic pressure measured by a micro-manometer catheter were significantly higher in the HFD group than in the ND group at 12 weeks after reoxygenation. The medial smooth muscle area was larger in the HFD group. Caspase-3 activity in the lung tissue of the HFD group was decreased, and the apoptosis of pulmonary smooth muscle cells was suppressed after reoxygenation. Moreover, the expression levels of peroxisome proliferator-activated receptor-γ and apelin were lower in the HFD group than in the ND group. CONCLUSIONS The results suggest that metabolic disorder may suppress pulmonary artery reverse remodeling in mice with hypoxia-induced PH during reoxygenation.
Collapse
MESH Headings
- Animals
- Apelin/metabolism
- Apoptosis
- Arterial Pressure
- Caspase 3/metabolism
- Diet, High-Fat/adverse effects
- Disease Models, Animal
- Female
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/therapy
- Hypoxia/complications
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Obesity/etiology
- Obesity/metabolism
- Obesity/physiopathology
- Oxygen Inhalation Therapy
- PPAR gamma/metabolism
- Pulmonary Artery/metabolism
- Pulmonary Artery/physiopathology
- Vascular Remodeling
- Mice
Collapse
Affiliation(s)
- Koichi Sugimoto
- Department of Cardiovascular Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan.
- Department of Pulmonary Hypertension, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan.
| | - Tetsuro Yokokawa
- Department of Cardiovascular Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan
- Department of Pulmonary Hypertension, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan
| | - Tomofumi Misaka
- Department of Cardiovascular Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan
| | - Takashi Kaneshiro
- Department of Cardiovascular Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan
| | - Akiomi Yoshihisa
- Department of Cardiovascular Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan
| | - Kazuhiko Nakazato
- Department of Cardiovascular Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan
| | - Yasuchika Takeishi
- Department of Cardiovascular Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan
| |
Collapse
|
18
|
KAP1 silencing relieves OxLDL-induced vascular endothelial dysfunction by down-regulating LOX-1. Biosci Rep 2021; 40:225915. [PMID: 32725144 PMCID: PMC7414520 DOI: 10.1042/bsr20200821] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/18/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022] Open
Abstract
KRAB domain-associated protein 1 (KAP1) is highly expressed in atherosclerotic plaques. Here, we studied the role of KAP1 in atherosclerosis development using a cell model of endothelial dysfunction induced by oxidative low-density lipoprotein (OxLDL). The phosphorylation and protein levels of KAP1 were similar between OxLDL-treated and non-treated endothelial cells (ECs). KAP1 depletion significantly inhibited the production of OxLDL-enhanced reactive oxygen species and the expression of adhesion molecules in ECs. Treatment with OxLDL promoted the proliferation and migration of ECs, which was also confirmed by the elevated levels of the proliferative markers c-Myc and PCNA, as well as the migratory marker MMP-9. However, these effects were also abrogated by KAP1 depletion. Moreover, the depletion of KAP1 in OxLDL-treated ECs resulted in decreases in the LOX-1 level and increases in eNOS expression. Generally, the data suggest that strategies targeting KAP1 depletion might be particularly useful for the prevention or treatment of atherosclerosis.
Collapse
|
19
|
Sugimoto K, Yokokawa T, Misaka T, Kaneshiro T, Yamada S, Yoshihisa A, Nakazato K, Takeishi Y. Endothelin-1 Upregulates Activin Receptor-Like Kinase-1 Expression via G i/RhoA/Sp-1/Rho Kinase Pathways in Human Pulmonary Arterial Endothelial Cells. Front Cardiovasc Med 2021; 8:648981. [PMID: 33708809 PMCID: PMC7940194 DOI: 10.3389/fcvm.2021.648981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 01/25/2021] [Indexed: 12/21/2022] Open
Abstract
Background: Pulmonary arterial hypertension (PAH) is characterized by pulmonary vasoconstriction and organic stenosis. It has been demonstrated that endothelin-1 (ET-1) induces pulmonary vasoconstriction through the activation of RhoA. In addition, a gene mutation of activin receptor-like kinase (ACVRL)-1 is recognized in PAH patients. However, little is known about the association between ET-1 and ACVRL-1. Objective: In the present study, we aimed to investigate the effect of ET-1 on ACVRL-1 expression and delineate the involvement of the Gi/RhoA/Rho kinase pathway. Methods: ET-1 was added to culture medium of human pulmonary arterial endothelial cells (PAECs). Pre-treatment with pertussis toxin (PTX) or exoenzyme C3 transferase (C3T) was performed for inhibition of Gi or RhoA, respectively. Rho kinase was inhibited by Y27632. Mithramycin A was used for inhibition of Sp-1, which is a transcriptional factor of ACVRL-1. The active form of RhoA (GTP-RhoA) was assessed by pull-down assay. Results: ACVRL-1 expression was increased by ET-1 in the PAECs. Pull-down assay revealed that ET-1 induced GTP-loading of RhoA, which was suppressed by pre-treatment with PTX or C3T. Further, PTX, C3T, and Y27632 suppressed the ET-1-induced ACVRL-1 expression. ET-1 increased the activity of the ACVRL-1 promoter and stabilized the ACVRL-1 mRNA. Sp-1 peaked 15 min after adding ET-1 to the PAECs. PTX and C3T prevented the increase of Sp-1 induced by ET-1. Inhibition of Sp-1 by mithramycin A suppressed ET-1-induced ACVRL-1 upregulation. Conclusion: The present study demonstrated that ET-1 increases ACVRL-1 expression in human PAECs via the Gi/RhoA/Rho kinase pathway with the involvement of Sp-1.
Collapse
Affiliation(s)
- Koichi Sugimoto
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan.,Department of Pulmonary Hypertension, Fukushima Medical University, Fukushima, Japan
| | - Tetsuro Yokokawa
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan.,Department of Pulmonary Hypertension, Fukushima Medical University, Fukushima, Japan
| | - Tomofumi Misaka
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Takashi Kaneshiro
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Shinya Yamada
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Akiomi Yoshihisa
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Kazuhiko Nakazato
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Yasuchika Takeishi
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
20
|
Mentrup T, Cabrera-Cabrera F, Schröder B. Proteolytic Regulation of the Lectin-Like Oxidized Lipoprotein Receptor LOX-1. Front Cardiovasc Med 2021; 7:594441. [PMID: 33553253 PMCID: PMC7856673 DOI: 10.3389/fcvm.2020.594441] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/21/2020] [Indexed: 11/13/2022] Open
Abstract
The lectin-like oxidized-LDL (oxLDL) receptor LOX-1, which is broadly expressed in vascular cells, represents a key mediator of endothelial activation and dysfunction in atherosclerotic plaque development. Being a member of the C-type lectin receptor family, LOX-1 can bind different ligands, with oxLDL being the best characterized. LOX-1 mediates oxLDL uptake into vascular cells and by this means can promote foam cell formation. In addition, LOX-1 triggers multiple signaling pathways, which ultimately induce a pro-atherogenic and pro-fibrotic transcriptional program. However, the molecular mechanisms underlying this signal transduction remain incompletely understood. In this regard, proteolysis has recently emerged as a regulatory mechanism of LOX-1 function. Different proteolytic cleavages within the LOX-1 protein can initiate its turnover and control the cellular levels of this receptor. Thereby, cleavage products with individual biological functions and/or medical significance are produced. Ectodomain shedding leads to the release of a soluble form of the receptor (sLOX1) which has been suggested to have diagnostic potential as a biomarker. Removal of the ectodomain leaves behind a membrane-bound N-terminal fragment (NTF), which despite being devoid of the ligand-binding domain is actively involved in signal transduction. Degradation of this LOX-1 NTF, which represents an athero-protective mechanism, critically depends on the aspartyl intramembrane proteases Signal peptide peptidase-like 2a and b (SPPL2a/b). Here, we present an overview of the biology of LOX-1 focusing on how proteolytic cleavages directly modulate the function of this receptor and, what kind of pathophysiological implications this has in cardiovascular disease.
Collapse
Affiliation(s)
| | | | - Bernd Schröder
- Institute for Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
21
|
Ke LY, Law SH, Mishra VK, Parveen F, Chan HC, Lu YH, Chu CS. Molecular and Cellular Mechanisms of Electronegative Lipoproteins in Cardiovascular Diseases. Biomedicines 2020; 8:biomedicines8120550. [PMID: 33260304 PMCID: PMC7760527 DOI: 10.3390/biomedicines8120550] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023] Open
Abstract
Dysregulation of glucose and lipid metabolism increases plasma levels of lipoproteins and triglycerides, resulting in vascular endothelial damage. Remarkably, the oxidation of lipid and lipoprotein particles generates electronegative lipoproteins that mediate cellular deterioration of atherosclerosis. In this review, we examined the core of atherosclerotic plaque, which is enriched by byproducts of lipid metabolism and lipoproteins, such as oxidized low-density lipoproteins (oxLDL) and electronegative subfraction of LDL (LDL(−)). We also summarized the chemical properties, receptors, and molecular mechanisms of LDL(−). In combination with other well-known markers of inflammation, namely metabolic diseases, we concluded that LDL(−) can be used as a novel prognostic tool for these lipid disorders. In addition, through understanding the underlying pathophysiological molecular routes for endothelial dysfunction and inflammation, we may reassess current therapeutics and might gain a new direction to treat atherosclerotic cardiovascular diseases, mainly targeting LDL(−) clearance.
Collapse
Affiliation(s)
- Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (L.-Y.K.); (S.H.L.); (V.K.M.); (F.P.)
- Graduate Institute of Medicine, College of Medicine and Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan; (H.-C.C.); (Y.-H.L.)
| | - Shi Hui Law
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (L.-Y.K.); (S.H.L.); (V.K.M.); (F.P.)
| | - Vineet Kumar Mishra
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (L.-Y.K.); (S.H.L.); (V.K.M.); (F.P.)
| | - Farzana Parveen
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (L.-Y.K.); (S.H.L.); (V.K.M.); (F.P.)
| | - Hua-Chen Chan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan; (H.-C.C.); (Y.-H.L.)
| | - Ye-Hsu Lu
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan; (H.-C.C.); (Y.-H.L.)
- Division of Cardiology, Department of International Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
| | - Chih-Sheng Chu
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan; (H.-C.C.); (Y.-H.L.)
- Division of Cardiology, Department of International Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Correspondence: ; Tel.: +886-73121101 (ext. 2297); Fax: +886-73111996
| |
Collapse
|
22
|
Kakino A, Fujita Y, Ke LY, Chan HC, Tsai MH, Dai CY, Chen CH, Sawamura T. Adiponectin forms a complex with atherogenic LDL and inhibits its downstream effects. J Lipid Res 2020; 62:100001. [PMID: 33410750 PMCID: PMC7890179 DOI: 10.1194/jlr.ra120000767] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 12/31/2022] Open
Abstract
Adiponectin, an adipocyte-derived protein, has antiatherogenic and antidiabetic effects, but how it confers the atherogenic effects is not well known. To study the antiatherogenic mechanisms of adiponectin, we examined whether it interacts with atherogenic low density lipoprotein (LDL) to attenuate LDL's atherogenicity. L5, the most electronegative subfraction of LDL, induces atherogenic responses similarly to copper-oxidized LDL (oxLDL). Unlike the native LDL endocytosed via the LDL receptor, L5 and oxLDL are internalized by cells via the lectin-like oxidized LDL receptor-1 (LOX-1). Using enzyme-linked immunosorbent assays (ELISAs), we showed that adiponectin preferentially bound oxLDL but not native LDL. In Chinese hamster ovary (CHO) cells transfected with the LOX-1 or LDL receptor, adiponectin selectively inhibited the uptake of oxLDL but not of native LDL, respectively. Furthermore, adiponectin suppressed the internalization of oxLDL in human coronary artery endothelial cells (HCAECs) and THP-1-derived macrophages. Western blot analysis of human plasma showed that adiponectin was abundant in L5 but not in L1, the least electronegative subfraction of LDL. Sandwich ELISAs with anti-adiponectin and anti-apolipoprotein B antibodies confirmed the binding of adiponectin to L5 and oxLDL. In LOX-1-expressing CHO cells, adiponectin inhibited cellular responses to oxLDL and L5, including nuclear factor-κB activation and extracellular signal-regulated kinas phosphorylation. In HCAECs, adiponectin inhibited oxLDL-induced endothelin-1 secretion and extracellular signal-regulated kinase phosphorylation. Conversely, oxLDL suppressed the adiponectin-induced activation of adenosine monophosphate-activated protein kinase in COS-7 cells expressing adiponectin receptor AdipoR1. Our findings suggest that adiponectin binds and inactivates atherogenic LDL, providing novel insight into the antiatherogenic mechanisms of adiponectin.
Collapse
Affiliation(s)
- Akemi Kakino
- Department of Molecular Pathophysiology, School of Medicine, Shinshu University, Nagano, Japan; Institute for Biomedical Sciences, Shinshu University, Nagano, Japan; Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan; Department of Molecular Pathophysiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Yoshiko Fujita
- Department of Molecular Pathophysiology, School of Medicine, Shinshu University, Nagano, Japan
| | - Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hua-Chen Chan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Hsien Tsai
- Department of Child Care, College of Humanities and Social Sciences, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Chia-Yen Dai
- Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chu-Huang Chen
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Vascular and Medicinal Research, Texas Heart Institute, Houston, TX, USA
| | - Tatsuya Sawamura
- Department of Molecular Pathophysiology, School of Medicine, Shinshu University, Nagano, Japan; Institute for Biomedical Sciences, Shinshu University, Nagano, Japan; Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan; Department of Molecular Pathophysiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan; Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
23
|
Le Master E, Ahn SJ, Levitan I. Mechanisms of endothelial stiffening in dyslipidemia and aging: Oxidized lipids and shear stress. CURRENT TOPICS IN MEMBRANES 2020; 86:185-215. [PMID: 33837693 PMCID: PMC8168803 DOI: 10.1016/bs.ctm.2020.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vascular stiffening of the arterial walls is well-known as a key factor in aging and the development of cardiovascular disease; however, the role of endothelial stiffness in vascular dysfunction is still an emerging topic. In this review, the authors discuss the impact of dyslipidemia, oxidized lipids, substrate stiffness, age and pro-atherogenic disturbed flow have on endothelial stiffness. Furthermore, we investigate several mechanistic pathways that are key contributors in endothelial stiffness and discuss their physiological effects in the onset of atherogenesis in the disturbed flow regions of the aortic vasculature. The findings in this chapter describe a novel paradigm of synergistic interaction of plasma dyslipidemia/oxidized lipids and pro-atherogenic disturbed shear stress, as well as aging has on endothelial stiffness and vascular dysfunction.
Collapse
Affiliation(s)
- Elizabeth Le Master
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Sang Joon Ahn
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Irena Levitan
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
24
|
Hofmann A, Brunssen C, Wolk S, Reeps C, Morawietz H. Soluble LOX-1: A Novel Biomarker in Patients With Coronary Artery Disease, Stroke, and Acute Aortic Dissection? J Am Heart Assoc 2020; 9:e013803. [PMID: 31902328 PMCID: PMC6988168 DOI: 10.1161/jaha.119.013803] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Anja Hofmann
- Division of Vascular Endothelium and Microcirculation Department of Medicine III Medical Faculty Carl Gustav Carus and University Hospital Carl Gustav Carus Dresden Technische Universität Dresden Dresden Germany.,Division of Vascular and Endovascular Surgery Department for Visceral-, Thoracic and Vascular Surgery Medical Faculty Carl Gustav Carus and University Hospital Carl Gustav Carus Dresden Technische Universität Dresden Dresden Germany
| | - Coy Brunssen
- Division of Vascular Endothelium and Microcirculation Department of Medicine III Medical Faculty Carl Gustav Carus and University Hospital Carl Gustav Carus Dresden Technische Universität Dresden Dresden Germany
| | - Steffen Wolk
- Division of Vascular and Endovascular Surgery Department for Visceral-, Thoracic and Vascular Surgery Medical Faculty Carl Gustav Carus and University Hospital Carl Gustav Carus Dresden Technische Universität Dresden Dresden Germany
| | - Christian Reeps
- Division of Vascular and Endovascular Surgery Department for Visceral-, Thoracic and Vascular Surgery Medical Faculty Carl Gustav Carus and University Hospital Carl Gustav Carus Dresden Technische Universität Dresden Dresden Germany
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation Department of Medicine III Medical Faculty Carl Gustav Carus and University Hospital Carl Gustav Carus Dresden Technische Universität Dresden Dresden Germany
| |
Collapse
|
25
|
Couto NF, Rezende L, Fernandes-Braga W, Alves AP, Agero U, Alvarez-Leite J, Damasceno NRT, Castro-Gomes T, Andrade LO. OxLDL alterations in endothelial cell membrane dynamics leads to changes in vesicle trafficking and increases cell susceptibility to injury. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1862:183139. [PMID: 31812625 DOI: 10.1016/j.bbamem.2019.183139] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/14/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023]
Abstract
Plasma membrane repair (PMR) is an important process for cell homeostasis, especially for cells under constant physical stress. Repair involves a sequence of Ca2+-dependent events, including lysosomal exocytosis and subsequent compensatory endocytosis. Cholesterol sequestration from plasma membrane causes actin cytoskeleton reorganization and polymerization, increasing cell stiffness, which leads to exocytosis and reduction of a peripheral pool of lysosomes involved in PMR. These changes in mechanical properties are similar to those observed in cells exposed to oxidized Low Density Lipoprotein (oxLDL), a key molecule during atherosclerosis development. Using a human umbilical vein endothelial cell line (EAhY926) we evaluated the influence of mechanical modulation induced by oxLDL in PMR and its effect in endothelial fragility. Similar to MβCD (a drug capable of sequestering cholesterol) treatment, oxLDL exposure led to actin reorganization and de novo polymerization, as well as an increase in cell rigidity and lysosomal exocytosis. Additionally, for both MβCD and oxLDL treated cells, there was an initial increase in endocytic events, likely triggered by the peak of exocytosis induced by both treatments. However, no further endocytic events were observed, suggesting that constitutive endocytosis is blocked upon treatment and that the reorganized cytoskeleton function as a mechanical barrier to membrane traffic. Finally, the increase in cell rigidity renders cells more prone to mechanical injury. Together, these data show that mechanical modulation induced by oxLDL exposure not only alters membrane traffic in cells, but also makes them more susceptible to mechanical injury, which may likely contribute to the initial steps of atherosclerosis development.
Collapse
Affiliation(s)
- Natália Fernanda Couto
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luisa Rezende
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Weslley Fernandes-Braga
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana Paula Alves
- Department of Physics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ubirajara Agero
- Department of Physics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jacqueline Alvarez-Leite
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Thiago Castro-Gomes
- Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luciana O Andrade
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
26
|
Sugimoto K, Yokokawa T, Misaka T, Nakazato K, Ishida T, Takeishi Y. Senescence Marker Protein 30 Deficiency Exacerbates Pulmonary Hypertension in Hypoxia-Exposed Mice. Int Heart J 2019; 60:1430-1434. [PMID: 31735783 DOI: 10.1536/ihj.19-190] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Pulmonary arterial hypertension is a fatal disease caused by pulmonary arterial vasoconstriction and organic stenosis due to the proliferation of pulmonary smooth muscle cells and endothelial cells. Endothelial dysfunction, including impaired nitric oxide (NO) bioavailability, plays a crucial role in the pathogenesis of pulmonary hypertension, and endothelial nitric oxide synthase (eNOS) is an important modulator of pulmonary vasodilatation. Although senescence marker protein (SMP) 30 is known as an anti-aging protein, the role of SMP30 in pulmonary vessels is still unclear. In this study, we examined the role of SMP30 in pulmonary vasculature using SMP30-deficient mice.We used female SMP30-deficient mice and wild-type littermate (WT) mice at the age of 12 to 18 weeks. The WT and SMP30-deficient mice were exposed to normoxia or hypoxia (10% oxygen for 4 weeks). In normoxia, the right ventricular systolic pressure (RVSP) was not different between the WT and SMP30-deficient mice, but in hypoxia, the RVSP was significantly higher in the SMP30-deficient mice compared to the WT mice (P < 0.05). The hypoxia-induced increases in right ventricular hypertrophy and medial smooth muscle area of the pulmonary artery were comparable between the WT and the SMP30-deficient mice. Western blotting showed that eNOS phosphorylation in lung tissue was reduced in the SMP30-deficient mice compared to the WT mice in normoxia. However, in hypoxic conditions, eNOS phosphorylation was reduced in both the WT and SMP30-deficient mice with no differences in Akt phosphorylation.Our study demonstrated that SMP30 is involved in the development of hypoxia-induced pulmonary hypertension by impairment of eNOS activity.
Collapse
Affiliation(s)
- Koichi Sugimoto
- Department of Pulmonary Hypertension, Fukushima Medical University.,Department of Cardiovascular Medicine, Fukushima Medical University
| | - Tetsuro Yokokawa
- Department of Pulmonary Hypertension, Fukushima Medical University.,Department of Cardiovascular Medicine, Fukushima Medical University
| | - Tomofumi Misaka
- Department of Cardiovascular Medicine, Fukushima Medical University
| | | | - Takafumi Ishida
- Department of Cardiovascular Medicine, Fukushima Medical University
| | | |
Collapse
|
27
|
Li M, Li F, Wang T, Zhao L, Shi Y. Fabrication of carboxymethylcellulose hydrogel containing β-cyclodextrin–eugenol inclusion complexes for promoting diabetic wound healing. J Biomater Appl 2019; 34:851-863. [DOI: 10.1177/0885328219873254] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Mengdie Li
- School of Pharmacy, Jinzhou Medical University, Jinzhou, China
| | - Fang Li
- School of Pharmacy, Jinzhou Medical University, Jinzhou, China
| | - Tao Wang
- School of Pharmacy, Jinzhou Medical University, Jinzhou, China
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou, China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
28
|
LOX-1: Regulation, Signaling and Its Role in Atherosclerosis. Antioxidants (Basel) 2019; 8:antiox8070218. [PMID: 31336709 PMCID: PMC6680802 DOI: 10.3390/antiox8070218] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/05/2019] [Accepted: 07/08/2019] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis has long been known to be a chronic inflammatory disease. In addition, there is intense oxidative stress in atherosclerosis resulting from an imbalance between the excess reactive oxygen species (ROS) generation and inadequate anti-oxidant defense forces. The excess of the oxidative forces results in the conversion of low-density lipoproteins (LDL) to oxidized LDL (ox-LDL), which is highly atherogenic. The sub-endothelial deposition of ox-LDL, formation of foamy macrophages, vascular smooth muscle cell (VSMC) proliferation and migration, and deposition of collagen are central pathophysiologic steps in the formation of atherosclerotic plaque. Ox-LDL exerts its action through several different scavenger receptors, the most important of which is LOX-1 in atherogenesis. LOX-1 is a transmembrane glycoprotein that binds to and internalizes ox-LDL. This interaction results in variable downstream effects based on the cell type. In endothelial cells, there is an increased expression of cellular adhesion molecules, resulting in the increased attachment and migration of inflammatory cells to intima, followed by their differentiation into macrophages. There is also a worsening endothelial dysfunction due to the increased production of vasoconstrictors, increased ROS, and depletion of endothelial nitric oxide (NO). In the macrophages and VSMCs, ox-LDL causes further upregulation of the LOX-1 gene, modulation of calpains, macrophage migration, VSMC proliferation and foam cell formation. Soluble LOX-1 (sLOX-1), a fragment of the main LOX-1 molecule, is being investigated as a diagnostic marker because it has been shown to be present in increased quantities in patients with hypertension, diabetes, metabolic syndrome and coronary artery disease. LOX-1 gene deletion in mice and anti-LOX-1 therapy has been shown to decrease inflammation, oxidative stress and atherosclerosis. LOX-1 deletion also results in damage from ischemia, making LOX-1 a promising target of therapy for atherosclerosis and related disorders. In this article we focus on the different mechanisms for regulation, signaling and the various effects of LOX-1 in contributing to atherosclerosis.
Collapse
|
29
|
Kattoor AJ, Kanuri SH, Mehta JL. Role of Ox-LDL and LOX-1 in Atherogenesis. Curr Med Chem 2019; 26:1693-1700. [DOI: 10.2174/0929867325666180508100950] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/01/2017] [Accepted: 12/01/2017] [Indexed: 02/02/2023]
Abstract
Oxidized LDL (ox-LDL) plays a central role in atherosclerosis by acting on multiple
cells such as endothelial cells, macrophages, platelets, fibroblasts and smooth muscle cells
through LOX-1. LOX-1 is a 50 kDa transmembrane glycoprotein that serves as receptor for
ox-LDL, modified lipoproteins, activated platelets and advance glycation end-products. Ox-
LDL through LOX-1, in endothelial cells, causes increase in leukocyte adhesion molecules,
activates pathways of apoptosis, increases reactive oxygen species and cause endothelial dysfunction.
In vascular smooth muscle cells and fibroblasts, they stimulate proliferation, migration
and collagen synthesis. LOX-1 expressed on macrophages inhibit macrophage migration
and stimulate foam cell formation. They also stimulate generation of metalloproteinases and
contribute to plaque instability and thrombosis. Drugs that modulate LOX-1 are desirable targets
against atherosclerosis. Many naturally occurring compounds have been shown to modulate
LOX-1 expression and atherosclerosis. Currently, novel drug design techniques are used
to identify molecules that can bind to LOX-1 and inhibit its activation by ox-LDL. In addition,
techniques using RNA interference and monoclonal antibody against LOX-1 are currently
being investigated for clinical use.
Collapse
Affiliation(s)
- Ajoe John Kattoor
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Sri Harsha Kanuri
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Jawahar L. Mehta
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| |
Collapse
|
30
|
Severino P, D'Amato A, Netti L, Pucci M, Infusino F, Maestrini V, Mancone M, Fedele F. Myocardial Ischemia and Diabetes Mellitus: Role of Oxidative Stress in the Connection between Cardiac Metabolism and Coronary Blood Flow. J Diabetes Res 2019; 2019:9489826. [PMID: 31089475 PMCID: PMC6476021 DOI: 10.1155/2019/9489826] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/23/2019] [Accepted: 03/13/2019] [Indexed: 12/27/2022] Open
Abstract
Ischemic heart disease (IHD) has several risk factors, among which diabetes mellitus represents one of the most important. In diabetic patients, the pathophysiology of myocardial ischemia remains unclear yet: some have atherosclerotic plaque which obstructs coronary blood flow, others show myocardial ischemia due to coronary microvascular dysfunction in the absence of plaques in epicardial vessels. In the cross-talk between myocardial metabolism and coronary blood flow (CBF), ion channels have a main role, and, in diabetic patients, they are involved in the pathophysiology of IHD. The exposition to the different cardiovascular risk factors and the ischemic condition determine an imbalance of the redox state, defined as oxidative stress, which shows itself with oxidant accumulation and antioxidant deficiency. In particular, several products of myocardial metabolism, belonging to oxidative stress, may influence ion channel function, altering their capacity to modulate CBF, in response to myocardial metabolism, and predisposing to myocardial ischemia. For this reason, considering the role of oxidative and ion channels in the pathophysiology of myocardial ischemia, it is allowed to consider new therapeutic perspectives in the treatment of IHD.
Collapse
Affiliation(s)
- Paolo Severino
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Andrea D'Amato
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Lucrezia Netti
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Mariateresa Pucci
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Fabio Infusino
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Viviana Maestrini
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Massimo Mancone
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Francesco Fedele
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
31
|
Berg G, Barchuk M, Miksztowicz V. Behavior of Metalloproteinases in Adipose Tissue, Liver and Arterial Wall: An Update of Extracellular Matrix Remodeling. Cells 2019; 8:cells8020158. [PMID: 30769840 PMCID: PMC6406870 DOI: 10.3390/cells8020158] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/22/2019] [Accepted: 02/01/2019] [Indexed: 02/06/2023] Open
Abstract
Extracellular matrix (ECM) remodeling is required for many physiological and pathological processes. Metalloproteinases (MMPs) are endopeptidases which are able to degrade different components of the ECM and nucleus matrix and to cleave numerous non-ECM proteins. Among pathological processes, MMPs are involved in adipose tissue expansion, liver fibrosis, and atherosclerotic plaque development and vulnerability. The expression and the activity of these enzymes are regulated by different hormones and growth factors, such as insulin, leptin, and adiponectin. The controversial results reported up to this moment regarding MMPs behavior in ECM biology could be consequence of the different expression patterns among species and the stage of the studied pathology. The aim of the present review was to update the knowledge of the role of MMPs and its inhibitors in ECM remodeling in high incidence pathologies such as obesity, liver fibrosis, and cardiovascular disease.
Collapse
Affiliation(s)
- Gabriela Berg
- Laboratorio de Lípidos y Aterosclerosis, Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires 1113, Argentina.
- Facultad de Farmacia y Bioquímica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Buenos Aires 1113, Argentina.
- Facultad de Farmacia y Bioquímica, CONICET, Universidad de Buenos Aires, Buenos Aires C1425FQB, Argentina.
| | - Magalí Barchuk
- Laboratorio de Lípidos y Aterosclerosis, Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires 1113, Argentina.
- Facultad de Farmacia y Bioquímica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Buenos Aires 1113, Argentina.
| | - Verónica Miksztowicz
- Laboratorio de Lípidos y Aterosclerosis, Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires 1113, Argentina.
- Facultad de Farmacia y Bioquímica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Buenos Aires 1113, Argentina.
- Facultad de Farmacia y Bioquímica, CONICET, Universidad de Buenos Aires, Buenos Aires C1425FQB, Argentina.
| |
Collapse
|
32
|
Wang Y, Hong Y, Zhang C, Shen Y, Pan YS, Chen RZ, Zhang Q, Chen YH. Picroside II attenuates hyperhomocysteinemia-induced endothelial injury by reducing inflammation, oxidative stress and cell apoptosis. J Cell Mol Med 2018; 23:464-475. [PMID: 30394648 PMCID: PMC6307770 DOI: 10.1111/jcmm.13949] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 09/03/2018] [Indexed: 12/27/2022] Open
Abstract
Picroside II (P‐II), one of the main active components of scrophularia extract, which have anti‐oxidative, anti‐inflammatory effects, but its effect on hyperhomocysteinemia (HHcy) induced endothelial injury remains to be determined. Here, we test whether P‐II protects HHcy‐induced endothelial dysfunction against oxidative stress, inflammation and cell apoptosis. In vitro study using HUVECs, and in hyperhomocysteinemia mouse models, we found that HHcy decreased endothelial SIRT1 expression and increased LOX‐1 expression, subsequently causing reactive oxygen species generation, up‐regulation of NADPH oxidase activity and NF‐κB activation, thereby promoting pro‐inflammatory response and cell apoptosis. Blockade of Sirt1 with Ex527 or siRNASIRT1 increased LOX‐1 expression, whereas overexpression of SIRT1 decreased LOX‐1 expression markedly. P‐II treatment significantly increased SIRT1 expression and reduced LOX‐1 expression, and protected against endothelial cells from Hcy‐induced oxidative injury, inflammation and apoptosis. However, blockade of SIRT1 or overexpression of LOX‐1 attenuated the therapeutic effects of P‐II. In conclusion, our results suggest that P‐II prevents the Hcy induced endothelial damage probably through regulating the SIRT1/LOX‐1 signaling pathway.
Collapse
Affiliation(s)
- Yunkai Wang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yajun Hong
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chunyu Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunli Shen
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ye Shen Pan
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rui Zhen Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qi Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Han Chen
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Tian K, Ogura S, Little PJ, Xu SW, Sawamura T. Targeting LOX-1 in atherosclerosis and vasculopathy: current knowledge and future perspectives. Ann N Y Acad Sci 2018; 1443:34-53. [PMID: 30381837 DOI: 10.1111/nyas.13984] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/12/2018] [Accepted: 09/24/2018] [Indexed: 12/11/2022]
Abstract
LOX-1 (lectin-like oxidized low-density lipoprotein receptor-1; also known as OLR1) is the dominant receptor that recognizes and internalizes oxidized low-density lipoproteins (ox-LDLs) in endothelial cells. Several genetic variants of LOX-1 are associated with the risk and severity of coronary artery disease. The LOX-1-ox-LDL interaction induces endothelial dysfunction, leukocyte adhesion, macrophage-derived foam cell formation, smooth muscle cell proliferation and migration, and platelet activation. LOX-1 activation eventually leads to the rupture of atherosclerotic plaques and acute cardiovascular events. In addition, LOX-1 can be cleaved to generate soluble LOX-1 (sLOX-1), which is a useful diagnostic and prognostic marker for atherosclerosis-related diseases in human patients. Of therapeutic relevance, several natural products and clinically used drugs have emerged as LOX-1 inhibitors that have antiatherosclerotic actions. We hereby provide an updated overview of role of LOX-1 in atherosclerosis and associated vascular diseases, with an aim to highlighting the potential of LOX-1 as a novel theranostic tool for cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Kunming Tian
- Department of Preventive Medicine, School of Public Health, Zunyi Medical University, Zunyi, Guizhou, China
| | - Sayoko Ogura
- Division of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Wooloongabba, Queensland, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou, China
| | - Suo-Wen Xu
- Aab Cardiovascular Research Institute, University of Rochester, Rochester, New York
| | - Tatsuya Sawamura
- Department of Physiology, School of Medicine, Shinshu University, Nagano, Japan.,Research Center for Next Generation Medicine, Shinshu University, Nagano, Japan
| |
Collapse
|
34
|
A causal link between oxidative stress and inflammation in cardiovascular and renal complications of diabetes. Clin Sci (Lond) 2018; 132:1811-1836. [PMID: 30166499 DOI: 10.1042/cs20171459] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/22/2018] [Accepted: 07/26/2018] [Indexed: 12/14/2022]
Abstract
Chronic renal and vascular oxidative stress in association with an enhanced inflammatory burden are determinant processes in the development and progression of diabetic complications including cardiovascular disease (CVD), atherosclerosis and diabetic kidney disease (DKD). Persistent hyperglycaemia in diabetes mellitus increases the production of reactive oxygen species (ROS) and activates mediators of inflammation as well as suppresses antioxidant defence mechanisms ultimately contributing to oxidative stress which leads to vascular and renal injury in diabetes. Furthermore, there is increasing evidence that ROS, inflammation and fibrosis promote each other and are part of a vicious connection leading to development and progression of CVD and kidney disease in diabetes.
Collapse
|
35
|
Ayoub KF, Pothineni NVK, Rutland J, Ding Z, Mehta JL. Immunity, Inflammation, and Oxidative Stress in Heart Failure: Emerging Molecular Targets. Cardiovasc Drugs Ther 2018; 31:593-608. [PMID: 28956198 DOI: 10.1007/s10557-017-6752-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE Heart failure (HF) remains a major cause of morbidity and mortality worldwide. Although various therapies developed over the last two decades have shown improved long term outcomes in patients with established HF, there has been little progress in preventing the adverse cardiac remodeling that initiates HF. To fill the gap in treatment, current research efforts are focused on understanding novel mechanisms and signaling pathways. Immune activation, inflammation, oxidative stress, alterations in mitochondrial bioenergetics, and autophagy have been postulated as important pathophysiological events in this process. An improved understanding of these complex processes could facilitate a therapeutic shift toward molecular targets that can potentially alter the course of HF. METHODS In this review, we address the role of immunity, inflammation, and oxidative stress as well as other novel emerging concepts in the pathophysiology of HF that may have therapeutic implications. CONCLUSION Based on the experimental and clinical studies presented here, we anticipate that a better understanding of the pathophysiology of HF will open the door for new therapeutic targets. A one-size-fits-all approach may not be appropriate for all patients with HF, and further clinical trials utilizing molecular targeting in HF may result in improved outcomes.
Collapse
Affiliation(s)
- Karam F Ayoub
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Naga Venkata K Pothineni
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Joshua Rutland
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Zufeng Ding
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jawahar L Mehta
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA. .,Division of Cardiovascular Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, #532, Little Rock, AR, 72205, USA.
| |
Collapse
|
36
|
OLR1 scavenger receptor knockdown affects mitotic gene expression but is dispensable for oxidized phospholipid- mediated stress signaling in SZ 95 sebocytes. Mech Ageing Dev 2018; 172:35-44. [DOI: 10.1016/j.mad.2017.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 10/20/2017] [Accepted: 11/01/2017] [Indexed: 12/19/2022]
|
37
|
Liu J, Liu Y, Jia K, Huo Z, Huo Q, Liu Z, Li Y, Han X, Wang R. Clinical analysis of lectin-like oxidized low-density lipoprotein receptor-1 in patients with in-stent restenosis after percutaneous coronary intervention. Medicine (Baltimore) 2018; 97:e0366. [PMID: 29702981 PMCID: PMC5944531 DOI: 10.1097/md.0000000000010366] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In-stent restenosis (ISR) is the most common complication associated with percutaneous coronary intervention (PCI). Although some studies have reported an association between lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) and ISR, not enough clinical validation data are available to support this link. Here, we report our cross-sectional study aimed at exploring the feasibility of LOX-1 as a biomarker for the prognostic diagnosis of patients undergoing PCI.Three groups were included: ISR group, including 99 patients with ISR diagnosed with coronary arteriography (CAG) after PCI; lesion group, comprising 87 patients with coronary artery stenosis (<50%) diagnosed with CAG after PCI; and control group, consisting of 96 volunteers with no coronary artery disease. The levels of LOX-1 were measured in each patient by using an enzyme-linked immunosorbent assay, and their general information as well as laboratory parameters were recorded and followed up during a period of 2 years.LOX-1 levels gradually increased after PCI along with the progression of the lesion in the 3 groups. The levels of LOX-1 were significantly higher in the ISR group than in the other 2 groups (P < .001). LOX-1 levels were correlated with the levels of uric acid (UA) (r = 0.289, P = .007), creatinine (CREA) (r = .316, P = .003), and high-density lipoprotein cholesterol (HDL-C) (r = -0.271, P = .012), whereas no statistically significant correlation was detected with the Gensini score (r = 0.157, P = .141). The sensitivity and specificity of LOX-1 were 81.5% and 55.7%, respectively, with the most optimal threshold (5.04 μg/L). The area under curve (AUC) of the receiver operator characteristic (ROC) curve of LOX-1 was 0.720, and LOX-1 had the highest AUC compared with CREA, UA, and HDL-C, both individually and in combination.A high level of LOX-1 in the early period after PCI has a certain predictive power and diagnostic value for ISR. However, the level of LOX-1 is not related to the Gensini score of coronary artery after PCI, and CREA and UA, which are weakly related to LOX-1, have no obvious synergy in the diagnosis of ISR with LOX-1.
Collapse
Affiliation(s)
- Junfeng Liu
- Department of Clinical Laboratory, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Science and Beijing Union Medical College
| | - Yunde Liu
- School of Laboratory Medicine, Tianjin Medical University
| | - Kegang Jia
- Department of Clinical Laboratory, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Science and Beijing Union Medical College
| | - Zhixiao Huo
- The Second People's Hospital of Tianjin, Tianjin, China
| | - Qianyu Huo
- School of Laboratory Medicine, Tianjin Medical University
| | - Zhili Liu
- School of Laboratory Medicine, Tianjin Medical University
| | - Yongshu Li
- Department of Clinical Laboratory, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Science and Beijing Union Medical College
| | - Xuejing Han
- Department of Clinical Laboratory, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Science and Beijing Union Medical College
| | - Rong Wang
- School of Laboratory Medicine, Tianjin Medical University
| |
Collapse
|
38
|
Gao S, Zhao D, Qi Y, Wang W, Wang M, Sun J, Liu J, Li Y, Liu J. Circulating Oxidized Low-Density Lipoprotein Levels Independently Predict 10-Year Progression of Subclinical Carotid Atherosclerosis: A Community-Based Cohort Study. J Atheroscler Thromb 2018. [PMID: 29515051 PMCID: PMC6193183 DOI: 10.5551/jat.43299] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aims: To investigate the association between circulating oxidized low-density lipoprotein (ox-LDL) levels and progression of subclinical atherosclerosis and to examine whether this link is independent of other low-density lipoprotein (LDL)-related parameters. Methods: Totally, 804 subjects who were free of cardiovascular disease at baseline completed risk factor surveys and carotid ultrasound measurements in 2002 and 2012. Modified Poisson regression was performed to examine the association between baseline serum ox-LDL levels and the 10-year risk of progression of carotid atherosclerosis which was defined as the development of at least one new plaque in a previously plaque-free carotid segment at re-examination. Results: The mean age of the subjects was 58.6 ± 7.7 years at baseline and 43.3% were men. A total of 504 (62.7%) subjects had carotid plaque progression at re-examination. Subjects in the intermediate and highest tertiles of ox-LDL had a significantly higher adjusted risk of atherosclerosis progression than those in the lowest tertile [relative risk (95% confidence interval) 1.17 (1.01–1.34) for the intermediate tertile and 1.23 (1.07–1.42) for the highest tertile]. This association was independent of baseline levels of LDL-C, total LDL particle number, and small LDL particle number. Conclusion: This study demonstrates that serum ox-LDL levels predict 10-year progression of subclinical atherosclerosis. Moreover, this effect is independent of the cholesterol content, the number, and the size of LDL particles.
Collapse
Affiliation(s)
- Shen Gao
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Municipal Key Laboratory of Clinical Epidemiology
| | - Dong Zhao
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Municipal Key Laboratory of Clinical Epidemiology
| | - Yue Qi
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Municipal Key Laboratory of Clinical Epidemiology
| | - Wei Wang
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Municipal Key Laboratory of Clinical Epidemiology
| | - Miao Wang
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Municipal Key Laboratory of Clinical Epidemiology
| | - Jiayi Sun
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Municipal Key Laboratory of Clinical Epidemiology
| | - Jun Liu
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Municipal Key Laboratory of Clinical Epidemiology
| | - Yan Li
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Municipal Key Laboratory of Clinical Epidemiology
| | - Jing Liu
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Municipal Key Laboratory of Clinical Epidemiology
| |
Collapse
|
39
|
Balzan S, Lubrano V. LOX-1 receptor: A potential link in atherosclerosis and cancer. Life Sci 2018; 198:79-86. [PMID: 29462603 DOI: 10.1016/j.lfs.2018.02.024] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 02/07/2018] [Accepted: 02/16/2018] [Indexed: 12/19/2022]
Abstract
Altered production of reactive oxygen species (ROS), causing lipid peroxidation and DNA damage, contributes to the progression of atherosclerosis and cancer. Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is a lectin-like receptor for oxidized low-density lipoproteins (ox-LDL) primarily expressed in endothelial cells and vasculature-rich organs. LOX-1 receptors is a marker for atherosclerosis, and once activated by ox-LDL or other ligands, stimulates the expression of adhesion molecules, pro-inflammatory signaling pathways and proangiogenic proteins, including NF-kB and VEGF, in vascular endothelial cells and macrophages. Several different types of cancer reported LOX-1 gene upregulation, and numerous interplays exist concerning LOX-1 in atherosclerosis, metabolic diseases and cancer. One of them involves NF-kB, an oncogenic protein that regulates the transcription of several inflammatory genes response. In a model of cellular transformation, the MCF10A ER-Src, inhibition of LOX-1 gene reduces NF-kB activation and the inflammatory and hypoxia pathways, suggesting a mechanistic connection between cellular transformation and atherosclerosis. The remodeling proteins MMP-2 and MMP-9 have been found increased in angiogenesis in atherosclerotic plaque and also in human prostate cancer cells. In this review, we outlined the role of LOX-1 in atherogenesis and tumorigenesis as a potential link in these diseases, suggesting that LOX-1 inhibition could represent a promising strategy in the treatment of atherosclerosis and tumors.
Collapse
Affiliation(s)
- Silvana Balzan
- Institute of Clinical Physiology, CNR, Via Moruzzi 1, Pisa 56124, Italy.
| | - Valter Lubrano
- Fondazione CNR/Regione Toscana G. Monasterio, Via Moruzzi 1, Pisa 56124, Italy
| |
Collapse
|
40
|
Eelen G, de Zeeuw P, Treps L, Harjes U, Wong BW, Carmeliet P. Endothelial Cell Metabolism. Physiol Rev 2018; 98:3-58. [PMID: 29167330 PMCID: PMC5866357 DOI: 10.1152/physrev.00001.2017] [Citation(s) in RCA: 330] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 06/19/2017] [Accepted: 06/22/2017] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells (ECs) are more than inert blood vessel lining material. Instead, they are active players in the formation of new blood vessels (angiogenesis) both in health and (life-threatening) diseases. Recently, a new concept arose by which EC metabolism drives angiogenesis in parallel to well-established angiogenic growth factors (e.g., vascular endothelial growth factor). 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3-driven glycolysis generates energy to sustain competitive behavior of the ECs at the tip of a growing vessel sprout, whereas carnitine palmitoyltransferase 1a-controlled fatty acid oxidation regulates nucleotide synthesis and proliferation of ECs in the stalk of the sprout. To maintain vascular homeostasis, ECs rely on an intricate metabolic wiring characterized by intracellular compartmentalization, use metabolites for epigenetic regulation of EC subtype differentiation, crosstalk through metabolite release with other cell types, and exhibit EC subtype-specific metabolic traits. Importantly, maladaptation of EC metabolism contributes to vascular disorders, through EC dysfunction or excess angiogenesis, and presents new opportunities for anti-angiogenic strategies. Here we provide a comprehensive overview of established as well as newly uncovered aspects of EC metabolism.
Collapse
Affiliation(s)
- Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Pauline de Zeeuw
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Ulrike Harjes
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Brian W Wong
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| |
Collapse
|
41
|
Hung CH, Chan SH, Chu PM, Lin HC, Tsai KL. Metformin regulates oxLDL-facilitated endothelial dysfunction by modulation of SIRT1 through repressing LOX-1-modulated oxidative signaling. Oncotarget 2017; 7:10773-87. [PMID: 26885898 PMCID: PMC4905438 DOI: 10.18632/oncotarget.7387] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 01/31/2016] [Indexed: 12/20/2022] Open
Abstract
It is suggested that oxLDL is decisive in the initiation and development of atherosclerotic injuries. The up-regulation of oxidative stress and the generation of ROS act as key modulators in developing pro-atherosclerotic and anti-atherosclerotic processes in the human endothelial wall. In this present study, we confirmed that metformin enhanced SIRT1 and AMPK expression in human umbilical vein endothelial cells (HUVECs). Metformin also inhibited oxLDL-increased LOX-1 expression and oxLDL-collapsed AKT/eNOS levels. However, silencing SIRT1 and AMPK diminished the protective function of metformin against oxidative injuries. These results provide a new insight regarding the possible molecular mechanisms of metformin.
Collapse
Affiliation(s)
- Ching-Hsia Hung
- Department of Physical Therapy, College of Medicine, National Cheng Kung University,Tainan, Taiwan.,Institute of Allied Health Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shih-Hung Chan
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Ming Chu
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Huei-Chen Lin
- Institute of Allied Health Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Physical Therapy, Shu-Zen Junior College of Medicine and Management, Taiwan
| | - Kun-Ling Tsai
- Department of Physical Therapy, College of Medicine, National Cheng Kung University,Tainan, Taiwan
| |
Collapse
|
42
|
González-Chavarría I, Fernandez E, Gutierrez N, González-Horta EE, Sandoval F, Cifuentes P, Castillo C, Cerro R, Sanchez O, Toledo JR. LOX-1 activation by oxLDL triggers an epithelial mesenchymal transition and promotes tumorigenic potential in prostate cancer cells. Cancer Lett 2017; 414:34-43. [PMID: 29107109 DOI: 10.1016/j.canlet.2017.10.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/20/2017] [Accepted: 10/23/2017] [Indexed: 02/02/2023]
Abstract
Obesity is related to an increased risk of developing prostate cancer with high malignancy stages or metastasis. Recent results demonstrated that LOX-1, a receptor associated with obesity and atherosclerosis, is overexpressed in advanced and metastatic prostate cancer. Furthermore, high levels of oxLDL, the main ligand for LOX-1, have been found in patients with advanced prostate cancer. However, the role of LOX-1 in prostate cancer has not been unraveled completely yet. Here, we show that LOX-1 is overexpressed in prostate cancer cells and its activation by oxLDL promotes an epithelial to mesenchymal transition, through of lowered expression of epithelial markers (E-cadherin and plakoglobin) and an increased expression of mesenchymal markers (vimentin, N-cadherin, snail, slug, MMP-2 and MMP-9). Consequently, LOX-1 activation by oxLDL promotes actin cytoskeleton restructuration and MMP-2 and MMP-9 activity inducing prostate cancer cell invasion and migration. Additionally, LOX-1 increased the tumorigenic potential of prostate cancer cells and its expression was necessary for tumor growth in nude mice. In conclusion, our results suggest that oxLDL/LOX-1 could be ones of mechanisms that explain why obese patients with prostate cancer have an accelerated tumor progression and a greater probability of developing metastasis.
Collapse
Affiliation(s)
- I González-Chavarría
- Biotechnology and Biopharmaceuticals Laboratory, Department of Pathophysiology, School of Biological Science, Universidad de Concepión, Concepción, Chile
| | - E Fernandez
- Biotechnology and Biopharmaceuticals Laboratory, Department of Pathophysiology, School of Biological Science, Universidad de Concepión, Concepción, Chile
| | - N Gutierrez
- Biotechnology and Biopharmaceuticals Laboratory, Department of Pathophysiology, School of Biological Science, Universidad de Concepión, Concepción, Chile
| | - E E González-Horta
- Biotechnology and Biopharmaceuticals Laboratory, Department of Pathophysiology, School of Biological Science, Universidad de Concepión, Concepción, Chile
| | - F Sandoval
- Biotechnology and Biopharmaceuticals Laboratory, Department of Pathophysiology, School of Biological Science, Universidad de Concepión, Concepción, Chile
| | - P Cifuentes
- Biotechnology and Biopharmaceuticals Laboratory, Department of Pathophysiology, School of Biological Science, Universidad de Concepión, Concepción, Chile
| | - C Castillo
- Biotechnology and Biopharmaceuticals Laboratory, Department of Pathophysiology, School of Biological Science, Universidad de Concepión, Concepción, Chile
| | - R Cerro
- Biotechnology and Biopharmaceuticals Laboratory, Department of Pathophysiology, School of Biological Science, Universidad de Concepión, Concepción, Chile
| | - O Sanchez
- Department of Pharmacology, School of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - Jorge R Toledo
- Biotechnology and Biopharmaceuticals Laboratory, Department of Pathophysiology, School of Biological Science, Universidad de Concepión, Concepción, Chile.
| |
Collapse
|
43
|
Jiang X, Andjelkovic AV, Zhu L, Yang T, Bennett MVL, Chen J, Keep RF, Shi Y. Blood-brain barrier dysfunction and recovery after ischemic stroke. Prog Neurobiol 2017; 163-164:144-171. [PMID: 28987927 DOI: 10.1016/j.pneurobio.2017.10.001] [Citation(s) in RCA: 575] [Impact Index Per Article: 82.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 05/30/2017] [Accepted: 10/02/2017] [Indexed: 01/06/2023]
Abstract
The blood-brain barrier (BBB) plays a vital role in regulating the trafficking of fluid, solutes and cells at the blood-brain interface and maintaining the homeostatic microenvironment of the CNS. Under pathological conditions, such as ischemic stroke, the BBB can be disrupted, followed by the extravasation of blood components into the brain and compromise of normal neuronal function. This article reviews recent advances in our knowledge of the mechanisms underlying BBB dysfunction and recovery after ischemic stroke. CNS cells in the neurovascular unit, as well as blood-borne peripheral cells constantly modulate the BBB and influence its breakdown and repair after ischemic stroke. The involvement of stroke risk factors and comorbid conditions further complicate the pathogenesis of neurovascular injury by predisposing the BBB to anatomical and functional changes that can exacerbate BBB dysfunction. Emphasis is also given to the process of long-term structural and functional restoration of the BBB after ischemic injury. With the development of novel research tools, future research on the BBB is likely to reveal promising potential therapeutic targets for protecting the BBB and improving patient outcome after ischemic stroke.
Collapse
Affiliation(s)
- Xiaoyan Jiang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | | | - Ling Zhu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Michael V L Bennett
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Yejie Shi
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
44
|
Chan SH, Hung CH, Shih JY, Chu PM, Cheng YH, Lin HC, Tsai KL. SIRT1 inhibition causes oxidative stress and inflammation in patients with coronary artery disease. Redox Biol 2017; 13:301-309. [PMID: 28601780 PMCID: PMC5466584 DOI: 10.1016/j.redox.2017.05.027] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 05/31/2017] [Indexed: 01/22/2023] Open
Abstract
Coronary artery disease (CAD) is the primary critical cardiovascular event. Endothelial cell and monocyte dysfunction with subsequent extravagant inflammation are the main causes of vessel damage in CAD. Thus, strategies that repress cell death and manage unsuitable pro-inflammatory responses in CAD are potential therapeutic strategies for improving the clinical prognosis of patients with CAD. SIRT1 (Sirtuin 1) plays an important role in regulating cellular physiological processes. SIRT1 is also thought to protect the cardiovascular system by means of its antioxidant, anti-inflammation and anti-apoptosis activities. In the present study, we found that the SIRT1 expression levels were repressed and the acetylated p53 expression levels were enhanced in the monocytes of patients with CAD. LOX-1/oxidative stress was also up-regulated in the monocytes of patients with CAD, thereby increasing pro-apoptotic events and pro-inflammatory responses. We also demonstrated that monocytes from CAD patients caused endothelial adhesion molecule activation and the adherence of monocytes and endothelial cells. Our findings may explain why CAD patients remain at an increased risk of long-term recurrent ischemic events and provide new knowledge regarding the management of clinical CAD patients.
Collapse
Affiliation(s)
- Shih-Hung Chan
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Hsia Hung
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Allied Health Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jhih-Yuan Shih
- Department of Internal Medicine, Chi-Mei Hospital, Tainan, Taiwan
| | - Pei-Ming Chu
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Yung-Hsin Cheng
- Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| | - Huei-Chen Lin
- Institute of Allied Health Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Physical Therapy, Shu-Zen Junior College of Medicine and Management, Taiwan
| | - Kun-Ling Tsai
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
45
|
Sugimoto K, Nakazato K, Sato A, Suzuki S, Yoshihisa A, Machida T, Saitoh SI, Sekine H, Takeishi Y. Autoimmune disease mouse model exhibits pulmonary arterial hypertension. PLoS One 2017; 12:e0184990. [PMID: 28926602 PMCID: PMC5605000 DOI: 10.1371/journal.pone.0184990] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 09/05/2017] [Indexed: 11/30/2022] Open
Abstract
Background Pulmonary arterial hypertension is often associated with connective tissue disease. Although there are some animal models of pulmonary hypertension, an autoimmune disease-based model has not yet been reported. MRL/lpr mice, which have hypergammaglobulinemia, produce various autoimmune antibodies, and develop vasculitis and nephritis spontaneously. However, little is known about pulmonary circulation in these mice. In the present study, we examined the pulmonary arterial pressure in MRL/lpr mice. Methods and results We used female MRL/lpr mice aged between 12 and 14 weeks. Fluorescent immunostaining showed that there was no deposition of immunoglobulin or C3 in the lung tissue of the MRL/lpr mice. Elevation of interferon-γ and interleukin-6 was recognized in the lung tissue of the MRL/lpr mice. Right ventricular systolic pressure, Fulton index and the ratio of right ventricular weight to body weight in the MRL/lpr mice were significantly higher than those in wild type mice with same background (C57BL/6). The medial smooth muscle area and the proportion of muscularized vessels in the lung tissue of the MRL/lpr mice were larger than those of the C57BL/6 mice. Western blot analysis demonstrated markedly elevated levels of prepro-endothelin-1 and survivin as well as decreased endothelial nitric oxide synthase phosphorylation in the lung tissue of the MRL/lpr mice. Terminal deoxynucleotidyl-transferase-mediated dUTP nick end-labeling assay showed the resistance against apoptosis of pulmonary arterial smooth muscle cells in the MRL/lpr mice. Conclusion We showed that MRL/lpr mice were complicated with pulmonary hypertension. MRL/lpr mice appeared to be a useful model for studying the mechanism of pulmonary hypertension associated with connective tissue diseases.
Collapse
Affiliation(s)
- Koichi Sugimoto
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
- Department of Pulmonary Hypertension, Fukushima Medical University, Fukushima, Japan
- * E-mail:
| | - Kazuhiko Nakazato
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Akihiko Sato
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Satoshi Suzuki
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Akiomi Yoshihisa
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Takeshi Machida
- Department of Immunology, Fukushima Medical University, Fukushima, Japan
| | - Shu-ichi Saitoh
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Hideharu Sekine
- Department of Immunology, Fukushima Medical University, Fukushima, Japan
| | - Yasuchika Takeishi
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
- Department of Pulmonary Hypertension, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
46
|
Zhang C, Adamos C, Oh MJ, Baruah J, Ayee MAA, Mehta D, Wary KK, Levitan I. oxLDL induces endothelial cell proliferation via Rho/ROCK/Akt/p27 kip1 signaling: opposite effects of oxLDL and cholesterol loading. Am J Physiol Cell Physiol 2017; 313:C340-C351. [PMID: 28701359 DOI: 10.1152/ajpcell.00249.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 07/05/2017] [Accepted: 07/05/2017] [Indexed: 12/19/2022]
Abstract
Oxidized modifications of LDL (oxLDL) play a key role in the development of endothelial dysfunction and atherosclerosis. However, the underlying mechanisms of oxLDL-mediated cellular behavior are not completely understood. Here, we compared the effects of two major types of oxLDL, copper-oxidized LDL (Cu2+-oxLDL) and lipoxygenase-oxidized LDL (LPO-oxLDL), on proliferation of human aortic endothelial cells (HAECs). Cu2+-oxLDL enhanced HAECs' proliferation in a dose- and degree of oxidation-dependent manner. Similarly, LPO-oxLDL also enhanced HAEC proliferation. Mechanistically, both Cu2+-oxLDL and LPO-oxLDL enhance HAEC proliferation via activation of Rho, Akt phosphorylation, and a decrease in the expression of cyclin-dependent kinase inhibitor 1B (p27kip1). Both Cu2+-oxLDL or LPO-oxLDL significantly increased Akt phosphorylation, whereas an Akt inhibitor, MK2206, blocked oxLDL-induced increase in HAEC proliferation. Blocking Rho with C3 or its downstream target ROCK with Y27632 significantly inhibited oxLDL-induced Akt phosphorylation and proliferation mediated by both Cu2+- and LPO-oxLDL. Activation of RhoA was blocked by Rho-GDI-1, which also abrogated oxLDL-induced Akt phosphorylation and HAEC proliferation. In contrast, blocking Rac1 in these cells had no effect on oxLDL-induced Akt phosphorylation or cell proliferation. Moreover, oxLDL-induced Rho/Akt signaling downregulated cell cycle inhibitor p27kip1 Preloading these cells with cholesterol, however, prevented oxLDL-induced Akt phosphorylation and HAEC proliferation. These findings provide a new understanding of the effects of oxLDL on endothelial proliferation, which is essential for developing new treatments against neovascularization and progression of atherosclerosis.
Collapse
Affiliation(s)
- Chongxu Zhang
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Crystal Adamos
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Myung-Jin Oh
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Jugajyoti Baruah
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Manuela A A Ayee
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Dolly Mehta
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Kishore K Wary
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Irena Levitan
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| |
Collapse
|
47
|
LOX-1 and Its Splice Variants: A New Challenge for Atherosclerosis and Cancer-Targeted Therapies. Int J Mol Sci 2017; 18:ijms18020290. [PMID: 28146073 PMCID: PMC5343826 DOI: 10.3390/ijms18020290] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/15/2017] [Accepted: 01/23/2017] [Indexed: 12/13/2022] Open
Abstract
Alternative splicing (AS) is a process in which precursor messenger RNA (pre-mRNA) splicing sites are differentially selected to diversify the protein isoform population. Changes in AS patterns have an essential role in normal development, differentiation and response to physiological stimuli. It is documented that AS can generate both “risk” and “protective” splice variants that can contribute to the pathogenesis of several diseases including atherosclerosis. The main endothelial receptor for oxidized low-density lipoprotein (ox-LDLs) is LOX-1 receptor protein encoded by the OLR1 gene. When OLR1 undergoes AS events, it generates three variants: OLR1, OLR1D4 and LOXIN. The latter lacks exon 5 and two-thirds of the functional domain. Literature data demonstrate a protective role of LOXIN in pathologies correlated with LOX-1 overexpression such as atherosclerosis and tumors. In this review, we summarize recent developments in understanding of OLR1 AS while also highlighting data warranting further investigation of this process as a novel therapeutic target.
Collapse
|
48
|
He K, Yue LH, Zhao GQ, Li C, Lin J, Jiang N, Wang Q, Xu Q, Peng XD, Hu LT, Zhang J. The role of LOX-1 on innate immunity against Aspergillus keratitis in mice. Int J Ophthalmol 2016; 9:1245-50. [PMID: 27672585 DOI: 10.18240/ijo.2016.09.01] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/26/2016] [Indexed: 01/19/2023] Open
Abstract
AIM To explore the effects of lectin-like ox-LDL receptor (LOX-1) on innate immunity against Aspergillus fumigatus (A. fumigatus ) in mice cornea. METHODS The mRNA levels of LOX-1 were tested in normal and A. fumigatus infected corneas of C57BL/6 and BALB/c mice. The expression of LOX-1, pro-inflammatory cytokines TNF-α, CXCL1 and IL-6, anti-inflammatory cytokines IL-10, and matrix metalloproteinase 9 (MMP9) were tested with treatment with LOX-1 neutralizing antibody or control IgG in A. fumigatus infected corneas of C57BL/6. Macrophages and neutrophils were extracted from susceptible C57BL/6 mice, and pretreated with LOX-1 neutralizing antibody or IgG, then stimulated with A. fumigatus. The mRNA levels of LOX-1, TNF-α, CXCL1, IL-6, IL-10 and MMP9 were evaluated by polymerase chain reaction. RESULTS The expression of LOX-1 was significantly increased in C57BL/6 mice corneas after A. fumigatus infection compared with BABL/c mice. After treatment with LOX-1 neutralizing antibody, the expression of LOX-1, TNF-α, CXCL1, IL-6, MMP9 and IL-10 in C57BL/6 corneas were significantly decreased compared with treatment with control IgG; the expression of LOX-1, CXCL1, IL-6 and IL-10 were significantly decreased in macrophages, while TNF-α and MMP9 expressions had no change; LOX-1, TNF-α, CXCL1, IL-6, MMP9 and IL-10 expressions were significantly decreased in neutrophils. CONCLUSION The expression of LOX-1 can affect the expression of pro-inflammatory and anti-inflammatory cytokines in fungal infected corneas, macrophages and neutrophils of C57BL/6. LOX-1 inhibition rebalances the inflammatory response of fungal keratitis in mice.
Collapse
Affiliation(s)
- Kun He
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Li-Hui Yue
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Gui-Qiu Zhao
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Cui Li
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Jing Lin
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Nan Jiang
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Qian Wang
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Qiang Xu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Xu-Dong Peng
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Li-Ting Hu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Jie Zhang
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| |
Collapse
|
49
|
Pircher A, Treps L, Bodrug N, Carmeliet P. Endothelial cell metabolism: A novel player in atherosclerosis? Basic principles and therapeutic opportunities. Atherosclerosis 2016; 253:247-257. [PMID: 27594537 DOI: 10.1016/j.atherosclerosis.2016.08.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/09/2016] [Accepted: 08/18/2016] [Indexed: 12/28/2022]
Abstract
Atherosclerosis is a leading cause of morbidity and mortality in Western society. Despite improved insight into disease pathogenesis and therapeutic options, additional treatment strategies are required. Emerging evidence highlights the relevance of endothelial cell (EC) metabolism for angiogenesis, and indicates that EC metabolism is perturbed when ECs become dysfunctional to promote atherogenesis. In this review, we overview the latest insights on EC metabolism and discuss current knowledge on how atherosclerosis deregulates EC metabolism, and how maladaptation of deregulated EC metabolism can contribute to atherosclerosis progression. We will also highlight possible therapeutic avenues, based on targeting EC metabolism.
Collapse
Affiliation(s)
- Andreas Pircher
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven, B-3000, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven, B-3000, Belgium
| | - Natalia Bodrug
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven, B-3000, Belgium; Laboratory of Adhesion and Angiogenesis, Centre for Tumour Biology, Barts Cancer Institute - a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven, B-3000, Belgium.
| |
Collapse
|
50
|
Zeya B, Arjuman A, Chandra NC. Lectin-like Oxidized Low-Density Lipoprotein (LDL) Receptor (LOX-1): A Chameleon Receptor for Oxidized LDL. Biochemistry 2016; 55:4437-44. [DOI: 10.1021/acs.biochem.6b00469] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Bushra Zeya
- Department
of Biochemistry, All India Institute of Medical Sciences, Patna 801507, India
| | - Albina Arjuman
- Division of P&I, Indian Council of Medical Research, New Delhi 110 029, India
| | - Nimai Chand Chandra
- Department
of Biochemistry, All India Institute of Medical Sciences, Patna 801507, India
| |
Collapse
|