1
|
Kim S, Jung UJ, Kim SR. The Crucial Role of the Blood-Brain Barrier in Neurodegenerative Diseases: Mechanisms of Disruption and Therapeutic Implications. J Clin Med 2025; 14:386. [PMID: 39860392 PMCID: PMC11765772 DOI: 10.3390/jcm14020386] [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: 11/27/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The blood-brain barrier (BBB) is a crucial structure that maintains brain homeostasis by regulating the entry of molecules and cells from the bloodstream into the central nervous system (CNS). Neurodegenerative diseases such as Alzheimer's and Parkinson's disease, as well as ischemic stroke, compromise the integrity of the BBB. This leads to increased permeability and the infiltration of harmful substances, thereby accelerating neurodegeneration. In this review, we explore the mechanisms underlying BBB disruption, including oxidative stress, neuroinflammation, vascular dysfunction, and the loss of tight junction integrity, in patients with neurodegenerative diseases. We discuss how BBB breakdown contributes to neuroinflammation, neurotoxicity, and the abnormal accumulation of pathological proteins, all of which exacerbate neuronal damage and facilitate disease progression. Furthermore, we discuss potential therapeutic strategies aimed at preserving or restoring BBB function, such as anti-inflammatory treatments, antioxidant therapies, and approaches to enhance tight junction integrity. Given the central role of the BBB in neurodegeneration, maintaining its integrity represents a promising therapeutic approach to slow or prevent the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea;
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Republic of Korea
| |
Collapse
|
2
|
Schöl M, Schempp R, Hennig T, Wigger D, Schumacher F, Kleuser B, Stigloher C, van Ham M, Jänsch L, Schneider-Schaulies S, Dölken L, Avota E. Dynamic changes in the proximitome of neutral sphingomyelinase-2 (nSMase2) in TNFα stimulated Jurkat cells. Front Immunol 2024; 15:1435701. [PMID: 39044828 PMCID: PMC11263205 DOI: 10.3389/fimmu.2024.1435701] [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: 05/20/2024] [Accepted: 06/17/2024] [Indexed: 07/25/2024] Open
Abstract
Ceramides generated by the activity of the neutral sphingomyelinase 2 (nSMase2) play a pivotal role in stress responses in mammalian cells. Dysregulation of sphingolipid metabolism has been implicated in numerous inflammation-related pathologies. However, its influence on inflammatory cytokine-induced signaling is yet incompletely understood. Here, we used proximity labeling to explore the plasma membrane proximal protein network of nSMase2 and TNFα-induced changes thereof. We established Jurkat cells stably expressing nSMase2 C-terminally fused to the engineered ascorbate peroxidase 2 (APEX2). Removal of excess biotin phenol substantially improved streptavidin-based affinity purification of biotinylated proteins. Using our optimized protocol, we determined nSMase2-proximal biotinylated proteins and their changes within the first 5 min of TNFα stimulation by quantitative mass spectrometry. We observed significant dynamic changes in the nSMase2 microenvironment in response to TNFα stimulation consistent with rapid remodeling of protein networks. Our data confirmed known nSMase2 interactors and revealed that the recruitment of most proteins depended on nSMase2 enzymatic activity. We measured significant enrichment of proteins related to vesicle-mediated transport, including proteins of recycling endosomes, trans-Golgi network, and exocytic vesicles in the proximitome of enzymatically active nSMase2 within the first minutes of TNFα stimulation. Hence, the nSMase2 proximal network and its TNFα-induced changes provide a valuable resource for further investigations into the involvement of nSMase2 in the early signaling pathways triggered by TNFα.
Collapse
Affiliation(s)
- Marie Schöl
- Institute for Virology and Immunobiology, University of Wuerzburg, Würzburg, Germany
| | - Rebekka Schempp
- Institute for Virology and Immunobiology, University of Wuerzburg, Würzburg, Germany
| | - Thomas Hennig
- Institute for Virology and Immunobiology, University of Wuerzburg, Würzburg, Germany
| | - Dominik Wigger
- Institute of Pharmacy, Department of Pharmacology & Toxicology, Freie Universität Berlin, Berlin, Germany
| | - Fabian Schumacher
- Institute of Pharmacy, Department of Pharmacology & Toxicology, Freie Universität Berlin, Berlin, Germany
| | - Burkhard Kleuser
- Institute of Pharmacy, Department of Pharmacology & Toxicology, Freie Universität Berlin, Berlin, Germany
| | - Christian Stigloher
- Imaging Core Facility, Biocenter, University of Wuerzburg, Würzburg, Germany
| | - Marco van Ham
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lothar Jänsch
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Lars Dölken
- Institute of Virology, Medizinische Hochschule Hannover, Hannover, Germany
| | - Elita Avota
- Institute for Virology and Immunobiology, University of Wuerzburg, Würzburg, Germany
| |
Collapse
|
3
|
Wilkerson JL, Tatum SM, Holland WL, Summers SA. Ceramides are fuel gauges on the drive to cardiometabolic disease. Physiol Rev 2024; 104:1061-1119. [PMID: 38300524 PMCID: PMC11381030 DOI: 10.1152/physrev.00008.2023] [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: 02/14/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/02/2024] Open
Abstract
Ceramides are signals of fatty acid excess that accumulate when a cell's energetic needs have been met and its nutrient storage has reached capacity. As these sphingolipids accrue, they alter the metabolism and survival of cells throughout the body including in the heart, liver, blood vessels, skeletal muscle, brain, and kidney. These ceramide actions elicit the tissue dysfunction that underlies cardiometabolic diseases such as diabetes, coronary artery disease, metabolic-associated steatohepatitis, and heart failure. Here, we review the biosynthesis and degradation pathways that maintain ceramide levels in normal physiology and discuss how the loss of ceramide homeostasis drives cardiometabolic pathologies. We highlight signaling nodes that sense small changes in ceramides and in turn reprogram cellular metabolism and stimulate apoptosis. Finally, we evaluate the emerging therapeutic utility of these unique lipids as biomarkers that forecast disease risk and as targets of ceramide-lowering interventions that ameliorate disease.
Collapse
Affiliation(s)
- Joseph L Wilkerson
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - Sean M Tatum
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - William L Holland
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
4
|
Di Pietro P, Izzo C, Abate AC, Iesu P, Rusciano MR, Venturini E, Visco V, Sommella E, Ciccarelli M, Carrizzo A, Vecchione C. The Dark Side of Sphingolipids: Searching for Potential Cardiovascular Biomarkers. Biomolecules 2023; 13:168. [PMID: 36671552 PMCID: PMC9855992 DOI: 10.3390/biom13010168] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/31/2022] [Accepted: 01/11/2023] [Indexed: 01/14/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death and illness in Europe and worldwide, responsible for a staggering 47% of deaths in Europe. Over the past few years, there has been increasing evidence pointing to bioactive sphingolipids as drivers of CVDs. Among them, most studies place emphasis on the cardiovascular effect of ceramides and sphingosine-1-phosphate (S1P), reporting correlation between their aberrant expression and CVD risk factors. In experimental in vivo models, pharmacological inhibition of de novo ceramide synthesis averts the development of diabetes, atherosclerosis, hypertension and heart failure. In humans, levels of circulating sphingolipids have been suggested as prognostic indicators for a broad spectrum of diseases. This article provides a comprehensive review of sphingolipids' contribution to cardiovascular, cerebrovascular and metabolic diseases, focusing on the latest experimental and clinical findings. Cumulatively, these studies indicate that monitoring sphingolipid level alterations could allow for better assessment of cardiovascular disease progression and/or severity, and also suggest them as a potential target for future therapeutic intervention. Some approaches may include the down-regulation of specific sphingolipid species levels in the circulation, by inhibiting critical enzymes that catalyze ceramide metabolism, such as ceramidases, sphingomyelinases and sphingosine kinases. Therefore, manipulation of the sphingolipid pathway may be a promising strategy for the treatment of cardio- and cerebrovascular diseases.
Collapse
Affiliation(s)
- Paola Di Pietro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Carmine Izzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Angela Carmelita Abate
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Paola Iesu
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Maria Rosaria Rusciano
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | | | - Valeria Visco
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Eduardo Sommella
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy
| |
Collapse
|
5
|
Pal P, Atilla-Gokcumen GE, Frasor J. Emerging Roles of Ceramides in Breast Cancer Biology and Therapy. Int J Mol Sci 2022; 23:ijms231911178. [PMID: 36232480 PMCID: PMC9569866 DOI: 10.3390/ijms231911178] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
One of the classic hallmarks of cancer is the imbalance between elevated cell proliferation and reduced cell death. Ceramide, a bioactive sphingolipid that can regulate this balance, has long been implicated in cancer. While the effects of ceramide on cell death and therapeutic efficacy are well established, emerging evidence indicates that ceramide turnover to downstream sphingolipids, such as sphingomyelin, hexosylceramides, sphingosine-1-phosphate, and ceramide-1-phosphate, is equally important in driving pro-tumorigenic phenotypes, such as proliferation, survival, migration, stemness, and therapy resistance. The complex and dynamic sphingolipid network has been extensively studied in several cancers, including breast cancer, to find key sphingolipidomic alterations that can be exploited to develop new therapeutic strategies to improve patient outcomes. Here, we review how the current literature shapes our understanding of how ceramide synthesis and turnover are altered in breast cancer and how these changes offer potential strategies to improve breast cancer therapy.
Collapse
Affiliation(s)
- Purab Pal
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - G. Ekin Atilla-Gokcumen
- Department of Chemistry, University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
- Correspondence: (G.E.A.-G.); (J.F.)
| | - Jonna Frasor
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Correspondence: (G.E.A.-G.); (J.F.)
| |
Collapse
|
6
|
Choi RH, Tatum SM, Symons JD, Summers SA, Holland WL. Ceramides and other sphingolipids as drivers of cardiovascular disease. Nat Rev Cardiol 2021; 18:701-711. [PMID: 33772258 PMCID: PMC8978615 DOI: 10.1038/s41569-021-00536-1] [Citation(s) in RCA: 228] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2021] [Indexed: 02/03/2023]
Abstract
Increases in calorie consumption and sedentary lifestyles are fuelling a global pandemic of cardiometabolic diseases, including coronary artery disease, diabetes mellitus, cardiomyopathy and heart failure. These lifestyle factors, when combined with genetic predispositions, increase the levels of circulating lipids, which can accumulate in non-adipose tissues, including blood vessel walls and the heart. The metabolism of these lipids produces bioactive intermediates that disrupt cellular function and survival. A compelling body of evidence suggests that sphingolipids, such as ceramides, account for much of the tissue damage in these cardiometabolic diseases. In humans, serum ceramide levels are proving to be accurate biomarkers of adverse cardiovascular disease outcomes. In mice and rats, pharmacological inhibition or depletion of enzymes driving de novo ceramide synthesis prevents the development of diabetes, atherosclerosis, hypertension and heart failure. In cultured cells and isolated tissues, ceramides perturb mitochondrial function, block fuel usage, disrupt vasodilatation and promote apoptosis. In this Review, we discuss the body of literature suggesting that ceramides are drivers - and not merely passengers - on the road to cardiovascular disease. Moreover, we explore the feasibility of therapeutic strategies to lower ceramide levels to improve cardiovascular health.
Collapse
Affiliation(s)
- Ran Hee Choi
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Sean M Tatum
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - J David Symons
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA.
| | - William L Holland
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
7
|
Ermini L, Farrell A, Alahari S, Ausman J, Park C, Sallais J, Melland-Smith M, Porter T, Edson M, Nevo O, Litvack M, Post M, Caniggia I. Ceramide-Induced Lysosomal Biogenesis and Exocytosis in Early-Onset Preeclampsia Promotes Exosomal Release of SMPD1 Causing Endothelial Dysfunction. Front Cell Dev Biol 2021; 9:652651. [PMID: 34017832 PMCID: PMC8130675 DOI: 10.3389/fcell.2021.652651] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
Aberrant ceramide build-up in preeclampsia, a serious disorder of pregnancy, causes exuberant autophagy-mediated trophoblast cell death. The significance of ceramide accumulation for lysosomal biogenesis in preeclampsia is unknown. Here we report that lysosome formation is markedly increased in trophoblast cells of early-onset preeclamptic placentae, in particular in syncytiotrophoblasts. This is accompanied by augmented levels of transcription factor EB (TFEB). In vitro and in vivo experiments demonstrate that ceramide increases TFEB expression and nuclear translocation and induces lysosomal formation and exocytosis. Further, we show that TFEB directly regulates the expression of lysosomal sphingomyelin phosphodiesterase (L-SMPD1) that degrades sphingomyelin to ceramide. In early-onset preeclampsia, ceramide-induced lysosomal exocytosis carries L-SMPD1 to the apical membrane of the syncytial epithelium, resulting in ceramide accumulation in lipid rafts and release of active L-SMPD1 via ceramide-enriched exosomes into the maternal circulation. The SMPD1-containing exosomes promote endothelial activation and impair endothelial tubule formation in vitro. Both exosome-induced processes are attenuated by SMPD1 inhibitors. These findings suggest that ceramide-induced lysosomal biogenesis and exocytosis in preeclamptic placentae contributes to maternal endothelial dysfunction, characteristic of this pathology.
Collapse
Affiliation(s)
- Leonardo Ermini
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Abby Farrell
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Sruthi Alahari
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Jonathan Ausman
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Chanho Park
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Julien Sallais
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Megan Melland-Smith
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Tyler Porter
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Michael Edson
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Ori Nevo
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Michael Litvack
- Translational Medicine Program, Peter Gilgan Center, The Hospital for Sick Children, Toronto, ON, Canada
| | - Martin Post
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Translational Medicine Program, Peter Gilgan Center, The Hospital for Sick Children, Toronto, ON, Canada
| | - Isabella Caniggia
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Ferchaud-Roucher V, Zair Y, Aguesse A, Krempf M, Ouguerram K. Omega 3 Improves Both apoB100-containing Lipoprotein Turnover and their Sphingolipid Profile in Hypertriglyceridemia. J Clin Endocrinol Metab 2020; 105:5893579. [PMID: 32805740 DOI: 10.1210/clinem/dgaa459] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 08/08/2020] [Indexed: 01/17/2023]
Abstract
CONTEXT Evidence for an association between sphingolipids and metabolic disorders is increasingly reported. Omega-3 long-chain polyunsaturated fatty acids (n-3 LC-PUFAs) improve apolipoprotein B100 (apoB100)-containing lipoprotein metabolism, but their effects on the sphingolipid content in lipoproteins remain unknown. OBJECTIVES In subjects with hypertriglyceridemia, we analyzed the effect of n-3 LC-PUFAs on the turnover apoB100-containing lipoproteins and on their sphingolipid content and looked for the possible association between these lipid levels and apoB100-containing lipoprotein turnover parameters. METHODS Six subjects underwent a kinetic study before and after n-3 supplementation for 2 months with 1 g of fish oil 3 times day containing 360 mg of eicosapentaenoic acid (EPA) and 240 mg of docosahexaenoic acid (DHA) in the form of triglycerides. We examined apoB100-containing lipoprotein turnover by primed perfusion labeled [5,5,5-2H3]-leucine and determined kinetic parameters using a multicompartmental model. We quantified sphingolipid species content in lipoproteins using mass spectrometry. RESULTS Supplementation decreased very low-density lipoprotein (VLDL), triglyceride, and apoB100 concentrations. The VLDL neutral and polar lipids showed increased n-3 LC-PUFA and decreased n-6 LC-PUFA content. The conversion rate of VLDL1 to VLDL2 and of VLDL2 to LDL was increased. We measured a decrease in total apoB100 production and VLDL1 production. Supplementation reduced the total ceramide concentration in VLDL while the sphingomyelin content in LDL was increased. We found positive correlations between plasma palmitic acid and VLDL ceramide and between VLDL triglyceride and VLDL ceramide, and inverse correlations between VLDL n-3 LC-PUFA and VLDL production. CONCLUSION Based on these results, we hypothesize that the improvement in apoB100 metabolism during n-3 LC-PUFA supplementation is contributed to by changes in sphingolipids.
Collapse
Affiliation(s)
- Véronique Ferchaud-Roucher
- University of Nantes, CHU Nantes, INRAe, UMR 1280 Physiopathology of Nutritional Adaptations, Nantes, France
- CRNH, West Human Nutrition Research Center, Nantes, France
| | - Yassine Zair
- CRNH, West Human Nutrition Research Center, Nantes, France
| | - Audrey Aguesse
- University of Nantes, CHU Nantes, INRAe, UMR 1280 Physiopathology of Nutritional Adaptations, Nantes, France
- CRNH, West Human Nutrition Research Center, Nantes, France
| | - Michel Krempf
- University of Nantes, CHU Nantes, INRAe, UMR 1280 Physiopathology of Nutritional Adaptations, Nantes, France
- CRNH, West Human Nutrition Research Center, Nantes, France
| | - Khadija Ouguerram
- University of Nantes, CHU Nantes, INRAe, UMR 1280 Physiopathology of Nutritional Adaptations, Nantes, France
- CRNH, West Human Nutrition Research Center, Nantes, France
| |
Collapse
|
9
|
Najdaghi S, Razi S, Rezaei N. An overview of the role of interleukin-8 in colorectal cancer. Cytokine 2020; 135:155205. [PMID: 32721849 DOI: 10.1016/j.cyto.2020.155205] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023]
Abstract
Colorectal Cancer (CRC), a common malignancy, is developing globally among people. Mutagenic insults activate peripheral nucleated cells to secrete chemokines in order to cause an inflammatory state. Despite the presence of multi-retrieving factors, elevated production of minor cytokines may speed-up the sever stages of the baseline inflammation targeting normal compensatory mechanism. IL-8 is a pro-inflammatory cytokine that is believed to be up-regulated in CRC to proceed primary condition into tumor behavior via induction of proliferation, angiogenesis and metastasis. Here, we assess the role of IL-8 in every step of CRC from signaling pathway and formation to invasion and discuss around new perspective therapy that targets IL-8 to manage CRC worldwide incidence and survival rate, more precisely.
Collapse
Affiliation(s)
- Soroush Najdaghi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Sheffield, UK.
| |
Collapse
|
10
|
Hoffman M, Kyriazis ID, Dimitriou A, Mishra SK, Koch WJ, Drosatos K. B-type natriuretic peptide is upregulated by c-Jun N-terminal kinase and contributes to septic hypotension. JCI Insight 2020; 5:133675. [PMID: 32324169 PMCID: PMC7205432 DOI: 10.1172/jci.insight.133675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 03/18/2020] [Indexed: 02/06/2023] Open
Abstract
B-type natriuretic peptide (BNP) is secreted by ventricular cardiomyocytes in response to various types of cardiac stress and has been used as a heart failure marker. In septic patients, increased BNP suggests poor prognosis; however, no causal link has been established. Among various effects, BNP decreases systemic vascular resistance and increases natriuresis that leads to lower blood pressure. We previously observed that JNK inhibition corrects cardiac dysfunction and suppresses cardiac BNP mRNA in endotoxemia. In this study, we investigated the transcriptional mechanism that regulates BNP expression and the involvement of plasma BNP in causing septic hypotension. Our in vitro and in vivo findings confirmed that activation of JNK signaling increases BNP expression in sepsis via direct binding of c-Jun in activating protein–1 (AP-1) regulatory elements of the Nppb promoter. Accordingly, genetic ablation of BNP, as well as treatment with a potentially novel neutralizing anti-BNP monoclonal antibody (19B3) or suppression of its expression via administration of JNK inhibitor SP600125 improved cardiac output, stabilized blood pressure, and improved survival in mice with polymicrobial sepsis. Therefore, inhibition of JNK signaling or BNP in sepsis appears to stabilize blood pressure and improve survival.
Collapse
Affiliation(s)
- Matthew Hoffman
- Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ioannis D Kyriazis
- Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Alexandra Dimitriou
- Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Santosh K Mishra
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Walter J Koch
- Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Konstantinos Drosatos
- Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA.,Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
11
|
Dash BC, Setia O, Gorecka J, Peyvandi H, Duan K, Lopes L, Nie J, Berthiaume F, Dardik A, Hsia HC. A Dense Fibrillar Collagen Scaffold Differentially Modulates Secretory Function of iPSC-Derived Vascular Smooth Muscle Cells to Promote Wound Healing. Cells 2020; 9:E966. [PMID: 32295218 PMCID: PMC7226960 DOI: 10.3390/cells9040966] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/06/2020] [Accepted: 04/11/2020] [Indexed: 12/20/2022] Open
Abstract
The application of human-induced pluripotent stem cells (hiPSCs) to generate vascular smooth muscle cells (hiPSC-VSMCs) in abundance is a promising strategy for vascular regeneration. While hiPSC-VSMCs have already been utilized for tissue-engineered vascular grafts and disease modeling, there is a lack of investigations exploring their therapeutic secretory factors. The objective of this manuscript was to understand how the biophysical property of a collagen-based scaffold dictates changes in the secretory function of hiPSC-VSMCs while developing hiPSC-VSMC-based therapy for durable regenerative wound healing. We investigated the effect of collagen fibrillar density (CFD) on hiPSC-VSMC's paracrine secretion and cytokines via the construction of varying density of collagen scaffolds. Our study demonstrated that CFD is a key scaffold property that modulates the secretory function of hiPSC-VSMCs. This study lays the foundation for developing collagen-based scaffold materials for the delivery of hiPSC-VSMCs to promote regenerative healing through guiding paracrine signaling pathways.
Collapse
Affiliation(s)
- Biraja C. Dash
- Section of Plastic Surgery, Department of Surgery Yale School of Medicine, Yale University, New Haven, CT 06510, USA; (H.P.); (K.D.); (J.N.)
| | - Ocean Setia
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06510, USA; (O.S.); (J.G.); (L.L.); (A.D.)
| | - Jolanta Gorecka
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06510, USA; (O.S.); (J.G.); (L.L.); (A.D.)
| | - Hassan Peyvandi
- Section of Plastic Surgery, Department of Surgery Yale School of Medicine, Yale University, New Haven, CT 06510, USA; (H.P.); (K.D.); (J.N.)
| | - Kaiti Duan
- Section of Plastic Surgery, Department of Surgery Yale School of Medicine, Yale University, New Haven, CT 06510, USA; (H.P.); (K.D.); (J.N.)
| | - Lara Lopes
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06510, USA; (O.S.); (J.G.); (L.L.); (A.D.)
| | - James Nie
- Section of Plastic Surgery, Department of Surgery Yale School of Medicine, Yale University, New Haven, CT 06510, USA; (H.P.); (K.D.); (J.N.)
| | - Francois Berthiaume
- Department of Biomedical Engineering, Rutgers University, The State University New Jersey, Piscataway, NJ 08854, USA;
| | - Alan Dardik
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06510, USA; (O.S.); (J.G.); (L.L.); (A.D.)
| | - Henry C. Hsia
- Section of Plastic Surgery, Department of Surgery Yale School of Medicine, Yale University, New Haven, CT 06510, USA; (H.P.); (K.D.); (J.N.)
| |
Collapse
|
12
|
Zhang L, Chen W, Li Y, Hong W, Li H, Cui Z, Dong X, Han X, Bao G, Xiao L, Gao P, Wang Y. Effect of 650-nm low-level laser irradiation on c-Jun, c-Fos, ICAM-1, and CCL2 expression in experimental periodontitis. Lasers Med Sci 2020; 35:31-40. [PMID: 30341668 DOI: 10.1007/s10103-018-2662-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/09/2018] [Indexed: 02/05/2023]
Abstract
This study was designed to investigate the effect of 650-nm low-level laser irradiation (LLLI) as an adjunctive treatment of experimental periodontitis. To investigate possible LLLI-mediated anti-inflammatory effects, we utilized an experimental periodontitis (EP) rat model and analyzed c-Jun, c-Fos, ICAM-1, and CCL2 gene expressions on PB leukocytes and in the gingival tissue. Total RNA was isolated from the gingivae and peripheral blood (PB) leukocytes of normal, EP, scaling, and root planing (SRP)-treated EP and LLLI + SRP-treated EP rats, and gene expressions were analyzed by real-time PCR. The productions of c-Jun, c-Fos, ICAM-1, and CCL2 in gingivae were analyzed immunohistochemically. Tartrate-resistant acid phosphatase (TRAP) staining was used to determine osteoclast activity in alveolar bone. The c-Jun and ICAM-1 messenger RNA (mRNA) levels were significantly decreased in the EP rat gingival tissue treated by SRP + LLLI than by SRP, the c-Jun, ICAM-1, and c-Fos mRNA levels on PB leukocytes reduced after LLLI treatment but did not show any significant differences in both groups. There was no significant difference in CCL2 mRNA levels on PB leukocytes and in gingivae between the SRP + LLLI and the SRP groups. The c-Fos mRNA levels in gingivae did not show significant difference in both groups. Immunohistochemistry showed that the CCL2, ICAM-1, c-Jun, and c-Fos productions were significantly reduced in rats of the SRP + LLLI group compared with the only SRP group. LLLI significantly decreased the number of osteoclasts as demonstrated by TRAP staining. The 650-nm LLLI might be a useful treatment modality for periodontitis.
Collapse
Affiliation(s)
- Lin Zhang
- School of Dentistry, Hospital of Stomatology, Tianjin Medical University, 12 Qi Xiang Tai Street, Heping District, Tianjin, 300070, China
| | - Wenlei Chen
- School of Dentistry, Hospital of Stomatology, Tianjin Medical University, 12 Qi Xiang Tai Street, Heping District, Tianjin, 300070, China
| | - Yingxin Li
- Institute of Biomedical Engineering, Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Wei Hong
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Haidong Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhuang Cui
- School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xiaoxi Dong
- Institute of Biomedical Engineering, Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Xiaohui Han
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Gang Bao
- School of Dentistry, Hospital of Stomatology, Tianjin Medical University, 12 Qi Xiang Tai Street, Heping District, Tianjin, 300070, China
| | - Li Xiao
- School of Dentistry, Hospital of Stomatology, Tianjin Medical University, 12 Qi Xiang Tai Street, Heping District, Tianjin, 300070, China
| | - Pengfei Gao
- School of Dentistry, Hospital of Stomatology, Tianjin Medical University, 12 Qi Xiang Tai Street, Heping District, Tianjin, 300070, China
| | - Yonglan Wang
- School of Dentistry, Hospital of Stomatology, Tianjin Medical University, 12 Qi Xiang Tai Street, Heping District, Tianjin, 300070, China.
| |
Collapse
|
13
|
Mücke VT, Thomas D, Mücke MM, Waidmann O, Zeuzem S, Sarrazin C, Pfeilschifter J, Vermehren J, Finkelmeier F, Grammatikos G. Serum sphingolipids predict de novo hepatocellular carcinoma in hepatitis C cirrhotic patients with sustained virologic response. Liver Int 2019; 39:2174-2183. [PMID: 31207039 DOI: 10.1111/liv.14178] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/28/2019] [Accepted: 06/08/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Curing hepatitis C virus (HCV) infection reduces the risk of hepatocellular carcinoma (HCC) development, yet HCC occurs despite sustained virologic response (SVR) in 2%-8% of cirrhotic patients. Sphingolipids (SLs) have been identified as new biomarkers of chronic liver disease and HCC. The aim of this study was to evaluate serum SLs as diagnostic HCC biomarkers in patients with HCV-associated cirrhosis at SVR12. METHODS From 2014 to 2016, 166 patients with HCV-cirrhosis and SVR were recruited and SL profiles were measured at baseline and 12 weeks after completion of direct-acting antiviral (DAA) therapy. All patients received HCC surveillance in line with current guideline recommendations. Minimum follow-up period comprised 6 months. RESULTS Our study included 130 (78%) patients without history of HCC, 25 (15%) with history of HCC prior DAA therapy and 11 (7%) patients with de novo HCC after FU12. In those with upcoming de novo HCC serum C24DHC (P = 0.006), C24:1DHC (P = 0.048) and C16Cer (P = 0.011) were significantly upregulated at FU12, but not AFP (P = 0.138). Contemporaneous ultrasound did not visualize HCC, at this time. C16Cer stayed sole independent predictor with high diagnostic accuracy of AFP-positive (AUC = 0.741) and -negative (AUC = 0.766) HCC development. Serum SL parameters decreased from baseline to SVR12. CONCLUSIONS C24DHC, C24:1DHC and especially C16Cer were superior to AFP in early detection of AFP-positive and -negative de novo HCC development. We observed significant SL profile changes upon SVR. SLs may play a role in non-invasive HCC surveillance and hepatocarcinogenesis.
Collapse
Affiliation(s)
| | - Dominique Thomas
- Pharmazentrum Frankfurt, Institut für Klinische Pharmakologie, Frankfurt am Main, Germany
| | - Marcus M Mücke
- Universitätsklinikum Frankfurt, Frankfurt am Main, Germany
| | | | - Stefan Zeuzem
- Universitätsklinikum Frankfurt, Frankfurt am Main, Germany
| | - Christoph Sarrazin
- Universitätsklinikum Frankfurt, Frankfurt am Main, Germany.,St. Josefs-Hospital, Wiesbaden, Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Frankfurt am Main, Germany
| | | | | | - Georgios Grammatikos
- Universitätsklinikum Frankfurt, Frankfurt am Main, Germany.,St. Luke's Hospital Thessaloniki, Panorama, Greece
| |
Collapse
|
14
|
Mechanism of Action of the Tumor Vessel Targeting Agent NGR-hTNF: Role of Both NGR Peptide and hTNF in Cell Binding and Signaling. Int J Mol Sci 2019; 20:ijms20184511. [PMID: 31547231 PMCID: PMC6769691 DOI: 10.3390/ijms20184511] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/02/2019] [Accepted: 09/10/2019] [Indexed: 11/18/2022] Open
Abstract
NGR-hTNF is a therapeutic agent for a solid tumor that specifically targets angiogenic tumor blood vessels, through the NGR motif. Its activity has been assessed in several clinical studies encompassing tumors of different histological types. The drug’s activity is based on an improved permeabilization of newly formed tumor vasculature, which favors intratumor penetration of chemotherapeutic agents and leukocyte trafficking. This work investigated the binding and the signaling properties of the NGR-hTNF, to elucidate its mechanism of action. The crystal structure of NGR-hTNF and modeling of its interaction with TNFR suggested that the NGR region is available for binding to a specific receptor. Using 2D TR-NOESY experiments, this study confirmed that the NGR-peptides binds to a specific CD13 isoform, whose expression is restricted to tumor vasculature cells, and to some tumor cell lines. The interaction between hTNF or NGR-hTNF with immobilized TNFRs showed similar kinetic parameters, whereas the competition experiments performed on the cells expressing both TNFR and CD13 showed that NGR-hTNF had a higher binding affinity than hTNF. The analysis of the NGR-hTNF-triggered signal transduction events showed a specific impairment in the activation of pro-survival pathways (Ras, Erk and Akt), compared to hTNF. Since a signaling pattern identical to NGR-hTNF was obtained with hTNF and NGR-sequence given as distinct molecules, the inhibition observed on the survival pathways was presumably due to a direct effect of the NGR-CD13 engagement on the TNFR signaling pathway. The reduced activation of the pro survival pathways induced by NGR-hTNF correlated with the increased caspases activation and reduced cell survival. This study demonstrates that the binding of the NGR-motif to CD13 determines not only the homing of NGR-hTNF to tumor vessels, but also the increase in its antiangiogenic activity.
Collapse
|
15
|
Yao K, Wang Y, Xu D, Liu X, Shen C, Hu W, Wang Z, Wu R, Tang X, Sun A, Zou Y, Qian J, Wu G, Guo X, Cheng X, Ge J. Effect of combined testing of ceramides with high-sensitive troponin T on the detection of acute coronary syndrome in patients with chest pain in China: a prospective observational study. BMJ Open 2019; 9:e028211. [PMID: 31350245 PMCID: PMC6661622 DOI: 10.1136/bmjopen-2018-028211] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 05/24/2019] [Accepted: 05/30/2019] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVE Ceramides are associated with coronary plaque vulnerability. We aim to investigate the potential diagnostic value of ceramides for acute coronary syndrome (ACS) in Chinese patients with chest pain. DESIGN Prospective observational survey. SETTING Shanghai, China, 2016-2017. PARTICIPANTS A total of 2773 patients with chest pain from four hospitals in Shanghai, China, between August 2016 and October 2017. MAIN OUTCOME MEASURES Performance of metabolites model in detection of ACS cases including ST-elevation myocardial infarction (STEMI), non-STEMI (NSTEMI) and unstable angina. RESULTS Plasma levels of 12 ceramide molecules and corresponding ratios were compared between patients diagnosed with ACS and those without. Cer(d18:1/24:1(15Z))/Cer(d18:1/24:0) ratio, Cer(d18:1/14:0) and Cer(d18:1/22:0) were independent predictors of ACS after adjustment of traditional risk factors and high-sensitivecardiac troponin T. Receiver operating characteristic curve analysis showed a significant improvement in detecting ACS in the multivariable model with ceramides compared with that without (0.865 (0.840 to 0.889) vs 0.808 (0.776 to 0.841), p<0.001). CONCLUSION Distinct plasma ceramides are independent diagnostic predictors of ACS among patients with chest pain. Ceramides together with high-sensitive troponin and traditional factors showed great potential in identifying ACS among patients with chest pain.
Collapse
Affiliation(s)
- Kang Yao
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- Department of Cardiology, Xiamen Branch, Zhongshan Hospital Fudan University, China
| | - Yanzhong Wang
- School of Population Health and Environmental Sciences, Kings College London, London, UK
| | - Demin Xu
- Department of Cardiac Surgey, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cardiac Surgey, Xiamen Branch, Zhongshan HospitalFudan University, Shanghai, China
| | - Xuebo Liu
- Department of Cardiology, Tongji Hospital, Tongji University, Shanghai, China
| | - Chengxing Shen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Wei Hu
- Department of Cardiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Zhe Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Runda Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Xianglin Tang
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- Shanghai Institute of Biomedical Science, Fudan University, Shanghai, China
| | - Yunzeng Zou
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | | | - Xin Guo
- Qlife Lab Co., Ltd., Shanghai, China
| | | | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| |
Collapse
|
16
|
Govindpani K, McNamara LG, Smith NR, Vinnakota C, Waldvogel HJ, Faull RL, Kwakowsky A. Vascular Dysfunction in Alzheimer's Disease: A Prelude to the Pathological Process or a Consequence of It? J Clin Med 2019; 8:E651. [PMID: 31083442 PMCID: PMC6571853 DOI: 10.3390/jcm8050651] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia. Despite decades of research following several theoretical and clinical lines, all existing treatments for the disorder are purely symptomatic. AD research has traditionally been focused on neuronal and glial dysfunction. Although there is a wealth of evidence pointing to a significant vascular component in the disease, this angle has been relatively poorly explored. In this review, we consider the various aspects of vascular dysfunction in AD, which has a significant impact on brain metabolism and homeostasis and the clearance of β-amyloid and other toxic metabolites. This may potentially precede the onset of the hallmark pathophysiological and cognitive symptoms of the disease. Pathological changes in vessel haemodynamics, angiogenesis, vascular cell function, vascular coverage, blood-brain barrier permeability and immune cell migration may be related to amyloid toxicity, oxidative stress and apolipoprotein E (APOE) genotype. These vascular deficits may in turn contribute to parenchymal amyloid deposition, neurotoxicity, glial activation and metabolic dysfunction in multiple cell types. A vicious feedback cycle ensues, with progressively worsening neuronal and vascular pathology through the course of the disease. Thus, a better appreciation for the importance of vascular dysfunction in AD may open new avenues for research and therapy.
Collapse
Affiliation(s)
- Karan Govindpani
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Laura G McNamara
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Nicholas R Smith
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Chitra Vinnakota
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Richard Lm Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
17
|
Apostoli A, Bianchi V, Bono N, Dimasi A, Ammann KR, Moiia YR, Montisci A, Sheriff J, Bluestein D, Fiore GB, Pappalardo F, Candiani G, Redaelli A, Slepian MJ, Consolo F. Prothrombotic activity of cytokine-activated endothelial cells and shear-activated platelets in the setting of ventricular assist device support. J Heart Lung Transplant 2019; 38:658-667. [PMID: 30846234 DOI: 10.1016/j.healun.2019.02.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND We systematically analyzed the synergistic effect of: (i) cytokine-mediated inflammatory activation of endothelial cells (ECs) with and (ii) shear-mediated platelet activation (SMPA) as a potential contributory mechanism to intraventricular thrombus formation in the setting of left ventricular assist device (LVAD) support. METHODS Intact and shear-activated human platelets were exposed to non-activated and cytokine-activated ECs. To modulate the level of LVAD-related shear activation, platelets were exposed to shear stress patterns of varying magnitude (30, 50, and 70 dynes/cm2, 10 minutes) via a hemodynamic shearing device. ECs were activated via exposure to inflammatory tumor necrosis factor-α (TNF-α 10 and 100 ng/ml, 24 hours), consistent with inflammatory activation recorded in patients on LVAD circulatory support. RESULTS Adhesivity of shear-activated platelets to ECs was significantly higher than that of intact/unactivated platelets, regardless of the initial activation level (70 dynes/cm2 shear-activated platelets vs intact platelets: +80%, p < 0.001). Importantly, inflammatory activation of ECs amplified platelet prothrombinase activity progressively with increasing shear stress magnitude and TNF-α concentration: thrombin generation of 70 dynes/cm2 shear-activated platelets was 2.6-fold higher after exposure and adhesion to 100 ng/ml TNF-α‒activated ECs (p < 0.0001). CONCLUSIONS We demonstrated synergistic effect of SMPA and cytokine-mediated EC inflammatory activation to enhance EC‒platelet adhesion and platelet prothrombotic function. These mechanisms may contribute to intraventricular thrombosis in the setting of mechanical circulatory support.
Collapse
Affiliation(s)
- Alice Apostoli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Valentina Bianchi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Nina Bono
- Politecnico di Milano Research Unit, National Interuniversity Consortium of Materials Science and Technology, Milano, Italy
| | - Annalisa Dimasi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Kaitlyn R Ammann
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA
| | - Yana Roka Moiia
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA
| | - Andrea Montisci
- Anesthesia and Intensive Care, Sant'Ambrogio Cardiothoracic Center, Milano, Italy
| | - Jawaad Sheriff
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Danny Bluestein
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Gianfranco B Fiore
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Federico Pappalardo
- Advanced Heart Failure and Mechanical Circulatory Support Program, San Raffaele Scientific Institute, Milano, Italy; Università Vita Salute San Raffaele, Milano, Italy
| | - Gabriele Candiani
- Biocompatibility and Cell Culture Laboratory "BioCell," Department of Chemistry, Materials and Chemical Engineering "Giulio Natta," Politecnico di Milano, Milano, Italy
| | - Alberto Redaelli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Marvin J Slepian
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA
| | - Filippo Consolo
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy; Advanced Heart Failure and Mechanical Circulatory Support Program, San Raffaele Scientific Institute, Milano, Italy; Università Vita Salute San Raffaele, Milano, Italy.
| |
Collapse
|
18
|
Witzel II, Nasser R, Garcia-Sabaté A, Sapudom J, Ma C, Chen W, Teo JCM. Deconstructing Immune Microenvironments of Lymphoid Tissues for Reverse Engineering. Adv Healthc Mater 2019; 8:e1801126. [PMID: 30516005 DOI: 10.1002/adhm.201801126] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/25/2018] [Indexed: 01/01/2023]
Abstract
The immune microenvironment presents a diverse panel of cues that impacts immune cell migration, organization, differentiation, and the immune response. Uniquely, both the liquid and solid phases of every specific immune niche within the body play an important role in defining cellular functions in immunity at that particular location. The in vivo immune microenvironment consists of biomechanical and biochemical signals including their gradients, surface topography, dimensionality, modes of ligand presentation, and cell-cell interactions, and the ability to recreate these immune biointerfaces in vitro can provide valuable insights into the immune system. This manuscript reviews the critical roles played by different immune cells and surveys the current progress of model systems for reverse engineering of immune microenvironments with a focus on lymphoid tissues.
Collapse
Affiliation(s)
- Ini-Isabée Witzel
- Core Technology Platforms; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Rasha Nasser
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Anna Garcia-Sabaté
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Chao Ma
- Department of Mechanical and Aerospace Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
- Department of Biomedical Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
| | - Jeremy C. M. Teo
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
- Department of Mechanical and Aerospace Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
| |
Collapse
|
19
|
Chemotherapy and Inflammatory Cytokine Signalling in Cancer Cells and the Tumour Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1152:173-215. [PMID: 31456184 DOI: 10.1007/978-3-030-20301-6_9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer is the result of a cell's acquisition of a variety of biological capabilities or 'hallmarks' as outlined by Hanahan and Weinberg. These include sustained proliferative signalling, the ability to evade growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, and the ability to invade other tissue and metastasize. More recently, the ability to escape immune destruction has been recognized as another important hallmark of tumours. It is suggested that genome instability and inflammation accelerates the acquisition of a variety of the above hallmarks. Inflammation, is a product of the body's response to tissue damage or pathogen invasion. It is required for tissue repair and host defense, but prolonged inflammation can often be the cause for disease. In a cancer patient, it is often unclear whether inflammation plays a protective or deleterious role in disease progression. Chemotherapy drugs can suppress tumour growth but also induce pathways in tumour cells that have been shown experimentally to support tumour progression or, in other cases, encourage an anti-tumour immune response. Thus, with the goal of better understanding the context under which each of these possible outcomes occurs, recent progress exploring chemotherapy-induced inflammatory cytokine production and the effects of cytokines on drug efficacy in the tumour microenvironment will be reviewed. The implications of chemotherapy on host and tumour cytokine pathways and their effect on the treatment of cancer patients will also be discussed.
Collapse
|
20
|
Chandra S, Fulton DJR, Caldwell RB, Caldwell RW, Toque HA. Hyperglycemia-impaired aortic vasorelaxation mediated through arginase elevation: Role of stress kinase pathways. Eur J Pharmacol 2018; 844:26-37. [PMID: 30502342 DOI: 10.1016/j.ejphar.2018.11.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 11/15/2018] [Accepted: 11/20/2018] [Indexed: 11/15/2022]
Abstract
Diabetes-induced vascular endothelial dysfunction has been reported to involve hyperglycemia-induced increases in arginase activity. However, upstream mediators of this effect are not clear. Here, we have tested involvement of Rho kinase, ERK1/2 and p38 MAPK pathways in this process. Studies were performed with aortas isolated from wild type or hemizygous arginase 1 knockout (Arg1+/-) mice and bovine aortic endothelial cells exposed to high glucose (HG, 25 mmol/l) or normal glucose (NG, 5.5 mmol/l) conditions for different times. Effects of inhibitors of arginase, p38 MAPK, ERK1/2 or ROCK and ex vivo adenoviral delivery of active Arg1 and inactive (D128-Arg1) cDNA were also determined. Exposure in wild type aorta or endothelial cells to HG significantly increased arginase activity and Arg1 expression and impaired aortic relaxation. Transduction of wild type aorta with active Arg1 cDNA impaired vascular relaxation, whereas inactive Arg1 had no effect. The HG-induced vascular endothelial dysfunction was associated with increased phosphorylation (activation) of ERK1/2 and p38 MAPK. Pretreatment with inhibitors of ERK1/2, p38 MAPK, ROCK or arginase blocked HG-induced elevation of arginase activity and Arg1 expression and prevented the vascular dysfunction. Inhibition of ROCK blunted the HG-induced activation of ERK1/2 and p38 MAPK. In summary, activated ROCK and subsequent activation of ERK1/2 or p38 MAPK elevates arginase activity and Arg1 expression in hyperglycemic states. Targeting this pathway may provide an effective means for preventing diabetes/hyperglycemia-induced vascular endothelial dysfunction.
Collapse
Affiliation(s)
- Surabhi Chandra
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA; Department of Biology, University of Nebraska-Kearney, Kearney, NE, USA.
| | - David J R Fulton
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA; Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Cell Biology and Anatomy, Augusta University, Augusta, GA, USA; Veterans Administration Medical Center, Augusta, GA, USA
| | - R William Caldwell
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA; Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Haroldo A Toque
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA; Vascular Biology Center, Augusta University, Augusta, GA, USA
| |
Collapse
|
21
|
Jin J, Lu Z, Li Y, Ru JH, Lopes-Virella MF, Huang Y. LPS and palmitate synergistically stimulate sphingosine kinase 1 and increase sphingosine 1 phosphate in RAW264.7 macrophages. J Leukoc Biol 2018; 104:843-853. [PMID: 29882996 PMCID: PMC6162112 DOI: 10.1002/jlb.3a0517-188rrr] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 05/15/2018] [Accepted: 05/18/2018] [Indexed: 01/28/2023] Open
Abstract
It has been well established that patients with diabetes or metabolic syndrome (MetS) have increased prevalence and severity of periodontitis, an oral infection initiated by bacteria and characterized by tissue inflammation and destruction. To understand the underlying mechanisms, we have shown that saturated fatty acid (SFA), which is increased in patients with type 2 diabetes or MetS, and LPS, an important pathogenic factor for periodontitis, synergistically stimulate expression of proinflammatory cytokines in macrophages by increasing ceramide production. However, the mechanisms by which increased ceramide enhances proinflammatory cytokine expression have not been well understood. Since sphingosine 1 phosphate (S1P) is a metabolite of ceramide and a bioactive lipid, we tested our hypothesis that stimulation of ceramide production by LPS and SFA facilitates S1P production, which contributes to proinflammatory cytokine expression. Results showed that LPS and palmitate, a major SFA, synergistically increased not only ceramide, but also S1P, and stimulated sphingosine kinase (SK) expression and membrane translocation in RAW264.7 macrophages. Results also showed that SK inhibition attenuated the stimulatory effect of LPS and palmitate on IL-6 secretion. Moreover, results showed that S1P enhanced the stimulatory effect of LPS and palmitate on IL-6 secretion. Finally, results showed that targeting S1P receptors using either S1P receptor antagonists or small interfering RNA attenuated IL-6 upregulation by LPS and palmitate. Taken together, this study demonstrated that LPS and palmitate synergistically stimulated S1P production and S1P in turn contributed to the upregulation of proinflammatory cytokine expression in macrophages by LPS and palmitate.
Collapse
Affiliation(s)
- Junfei Jin
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Zhongyang Lu
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA
| | - Yanchun Li
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ji Hyun Ru
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Maria F Lopes-Virella
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA
| | - Yan Huang
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA
| |
Collapse
|
22
|
Ma W, Concha-Benavente F, Santegoets SJAM, Welters MJP, Ehsan I, Ferris RL, van der Burg SH. EGFR signaling suppresses type 1 cytokine-induced T-cell attracting chemokine secretion in head and neck cancer. PLoS One 2018; 13:e0203402. [PMID: 30192802 PMCID: PMC6128559 DOI: 10.1371/journal.pone.0203402] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 08/20/2018] [Indexed: 01/10/2023] Open
Abstract
Resistance to antitumor immunity can be promoted by the oncogenic pathways operational in human cancers, including the epidermal growth factor receptor (EGFR) pathway. Here we studied if and how EGFR downstream signaling in head and neck squamous cell carcinoma (HNSCC) can affect the attraction of immune cells. HPV-negative and HPV-positive HNSCC cell lines were analyzed in vitro for CCL2, CCL5, CXCL9, CXCL10, IL-6 and IL-1β expression and the attraction of T cells under different conditions, including cetuximab treatment and stimulation with IFNγ and TNFα using qPCR, ELISA and migration experiments. Biochemical analyses with chemical inhibitors and siRNA transfection were used to pinpoint the underlying mechanisms. Stimulation of HNSCC cells with IFNγ and TNFα triggered the production of T-cell attracting chemokines and required c-RAF activation. Blocking of the EGFR with cetuximab during this stimulation increased chemokine production in vitro, and augmented the attraction of T cells. Mechanistically, cetuximab decreased the phosphorylation of MEK1, ERK1/2, AKT, mTOR, JNK, p38 and ERK5. Chemical inhibition of EGFR signaling showed a consistent and pronounced chemokine production with MEK1/2 inhibitor PD98059 and JNK inhibitor SP600125, but not with inhibitors of p38, PI3K or mTOR. Combination treatment with cetuximab and a MEK1/2 or JNK inhibitor induced the highest chemokine expression. In conclusion, overexpression of EGFR results in the activation of multiple downstream signaling pathways that act simultaneously to suppress type 1 cytokine stimulated production of chemokines required to amplify the attraction of T cells.
Collapse
Affiliation(s)
- Wenbo Ma
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Fernando Concha-Benavente
- Department of Otolaryngology, University of Pittsburgh, and the University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States of America
| | | | - Marij J. P. Welters
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ilina Ehsan
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert L. Ferris
- Department of Otolaryngology, University of Pittsburgh, and the University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States of America
| | - Sjoerd H. van der Burg
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
23
|
Cho JG, Lee A, Chang W, Lee MS, Kim J. Endothelial to Mesenchymal Transition Represents a Key Link in the Interaction between Inflammation and Endothelial Dysfunction. Front Immunol 2018. [PMID: 29515588 PMCID: PMC5826197 DOI: 10.3389/fimmu.2018.00294] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Endothelial cells that line the inner walls of blood vessels are in direct contact with blood and display remarkable heterogeneity in their response to exogenous stimuli. These ECs have unique location-dependent properties determined by the corresponding vascular beds and play an important role in regulating the homeostasis of the vascular system. Evidence suggests that vascular endothelial cells exposed to various environments undergo dynamic phenotypic switching, a key biological program in the context of endothelial heterogeneity, but that might result in EC dysfunction and, in turn, cause a variety of human diseases. Emerging studies show the importance of endothelial to mesenchymal transition (EndMT) in endothelial dysfunction during inflammation. EndMT is a complex biological process in which ECs lose their endothelial characteristics, acquire mesenchymal phenotypes, and express mesenchymal cell markers, such as alpha smooth muscle actin and fibroblast-specific protein 1. EndMT is induced by inflammatory responses, leading to pathological states, including tissue fibrosis, pulmonary arterial hypertension, and atherosclerosis, via dysfunction of the vascular system. Although the mechanisms associated with inflammation-induced EndMT have been identified, unraveling the specific role of this phenotypic switching in vascular dysfunction remains a challenge. Here, we review the current understanding on the interactions between inflammatory processes, EndMT, and endothelial dysfunction, with a focus on the mechanisms that regulate essential signaling pathways. Identification of such mechanisms will guide future research and could provide novel therapeutic targets for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Jin Gu Cho
- Division of Biological Sciences, Sookmyung Women's University, Seoul, South Korea
| | - Aram Lee
- Division of Biological Sciences, Sookmyung Women's University, Seoul, South Korea
| | - Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, Busan, South Korea
| | - Myeong-Sok Lee
- Division of Biological Sciences, Sookmyung Women's University, Seoul, South Korea
| | - Jongmin Kim
- Division of Biological Sciences, Sookmyung Women's University, Seoul, South Korea
| |
Collapse
|
24
|
Scholl A, Ivanov I, Hinz B. Inhibition of interleukin-1β-induced endothelial tissue factor expression by the synthetic cannabinoid WIN 55,212-2. Oncotarget 2018; 7:61438-61457. [PMID: 27556861 PMCID: PMC5308663 DOI: 10.18632/oncotarget.11367] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 07/26/2016] [Indexed: 01/08/2023] Open
Abstract
The role of cannabinoids in thrombosis remains controversial. In view of the primary importance of tissue factor (TF) in blood coagulation and its involvement in the pathology of several cardiovascular, inflammatory and neoplastic diseases, a regulation of this initial procoagulant signal seems to be of particular interest. Using human umbilical vein endothelial cells (HUVEC) the present study investigated the impact of the synthetic cannabinoid WIN 55,212-2 on interleukin (IL)-1β-induced TF expression and activity. WIN 55,212-2 caused a time- and concentration-dependent suppression of IL-1β-induced TF protein accompanied by decreases in TF mRNA and activity. Inhibition of TF protein expression by WIN 55,212-2 was mimicked by its cannabinoid receptor-inactive enantiomer WIN 55,212-3 but not by structurally unrelated phyto-, endo- and synthetic cannabinoids. In addition, the inhibitory effect of WIN 55,212-2 was not reversed by antagonists to cannabinoid receptors (CB1, CB2) or transient receptor potential vanilloid 1. Mechanistic approaches revealed WIN 55,212-2 to suppress IL-1β-induced TF expression via inhibition of ceramide formation and via decreased phosphorylation of p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinases. Further inhibitor experiments demonstrated neutral sphingomyelinase (nSMase) to confer ceramide generation upon IL-1β treatment with the parallel IL-1β-mediated activation of MAPKs occurring via an nSMase-independent pathway. Finally, a receptor-independent inhibition of IL-1β-induced TF protein by WIN 55,212-2 was confirmed in human blood monocytes. Collectively, this data provide a hitherto unknown receptor-independent anticoagulatory action of the cannabinoid WIN 55,212-2.
Collapse
Affiliation(s)
- Antje Scholl
- Institute of Toxicology and Pharmacology, Rostock University Medical Center, D-18057 Rostock, Germany
| | - Igor Ivanov
- Institute of Toxicology and Pharmacology, Rostock University Medical Center, D-18057 Rostock, Germany
| | - Burkhard Hinz
- Institute of Toxicology and Pharmacology, Rostock University Medical Center, D-18057 Rostock, Germany
| |
Collapse
|
25
|
Kangsamaksin T, Chaithongyot S, Wootthichairangsan C, Hanchaina R, Tangshewinsirikul C, Svasti J. Lupeol and stigmasterol suppress tumor angiogenesis and inhibit cholangiocarcinoma growth in mice via downregulation of tumor necrosis factor-α. PLoS One 2017; 12:e0189628. [PMID: 29232409 PMCID: PMC5726636 DOI: 10.1371/journal.pone.0189628] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/29/2017] [Indexed: 02/01/2023] Open
Abstract
Lupeol and stigmasterol, major phytosterols in various herbal plants, possess anti-inflammatory activities and have been proposed as candidates for anti-cancer agents, but their molecular mechanisms are still unclear. Here, we investigated the effects of lupeol and stigmasterol on tumor and endothelial cells in vitro and their anti-cancer activities in vivo. Our results demonstrated that lupeol and stigmasterol suppressed cell viability, migration, and morphogenesis of human umbilical vein endothelial cells (HUVECs) but not cholangiocarcinoma (CCA) cells. Expression analyses showed that the treatment of both compounds significantly reduced the transcript level of tumor necrosis factor-α (TNF-α), and Western blot analyses further revealed a decrease in downstream effector levels of VEGFR-2 signaling, including phosphorylated forms of Src, Akt, PCL, and FAK, which were rescued by TNF-α treatment. In vivo, lupeol and stigmasterol disrupted tumor angiogenesis and reduced the growth of CCA tumor xenografts. Immunohistochemical analyses confirmed a decrease in CD31-positive vessel content and macrophage recruitment upon treatment. These findings indicate that lupeol and stigmasterol effectively target tumor endothelial cells and suppress CCA tumor growth by their anti-inflammatory activities and are attractive candidates for anti-cancer treatment of CCA tumors.
Collapse
Affiliation(s)
- Thaned Kangsamaksin
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
- * E-mail:
| | - Supattra Chaithongyot
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | | | - Chayada Tangshewinsirikul
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Jisnuson Svasti
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, Thailand
| |
Collapse
|
26
|
Jang SA, Park DW, Kwon JE, Song HS, Park B, Jeon H, Sohn EH, Koo HJ, Kang SC. Quinic acid inhibits vascular inflammation in TNF-α-stimulated vascular smooth muscle cells. Biomed Pharmacother 2017; 96:563-571. [PMID: 29032340 DOI: 10.1016/j.biopha.2017.10.021] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 09/11/2017] [Accepted: 10/02/2017] [Indexed: 02/04/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease, and the increased expression of adhesion molecules on vascular smooth muscle cells contributes to the progression of vascular disease. Quinic acid (QA) has been shown to possess radioprotection, anti-neuroinflammatory, and anti-oxidant activities; however, an anti-vascular inflammatory effect has not been reported. This study investigated the effect of QA on the expression of vascular cell adhesion molecule-1 (VCAM-1) stimulated by TNF-α in MOVAS cells. Pre-incubation of MOVAS cells, the mouse vascular smooth muscle cell line for 2h with QA (0.1, 1 and 10 μg/mL) dose-dependently inhibits TNF-α-induced mRNA and protein expression of VCAM-1 and monocyte adhesion. QA inhibits TNF-α-stimulated phosphorylation of MAP kinase and NK-κB activation. Our results indicate that QA inhibits the TNF-α-stimulated induction of VCAM-1 in VSMC by inhibiting the MAP kinase and NF-κB signaling pathways and the adhesion capacity of VSMC, which may explain the ability of QA to inhibit vascular inflammation such as atherosclerosis.
Collapse
Affiliation(s)
- Seon-A Jang
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Dae Won Park
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Jeong Eun Kwon
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Hae Seong Song
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Bongkyun Park
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Hyelin Jeon
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Eun-Hwa Sohn
- Department of Herbal Medicine Resources, Kangwon National University, Samcheok 25913, Republic of Korea
| | - Hyun Jung Koo
- Department of Medicinal and Industrial Crops, Korea National College of Agriculture and Fisheries, Jeonju 54874, Republic of Korea
| | - Se Chan Kang
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea.
| |
Collapse
|
27
|
Edwardson DW, Boudreau J, Mapletoft J, Lanner C, Kovala AT, Parissenti AM. Inflammatory cytokine production in tumor cells upon chemotherapy drug exposure or upon selection for drug resistance. PLoS One 2017; 12:e0183662. [PMID: 28915246 PMCID: PMC5600395 DOI: 10.1371/journal.pone.0183662] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 08/08/2017] [Indexed: 01/08/2023] Open
Abstract
Tumor Necrosis Factor alpha (TNF-α) has been shown to be released by tumor cells in response to docetaxel, and lipopolysaccharides (LPS), the latter through activation of toll-like receptor 4 (TLR4). However, it is unclear whether the former involves TLR4 receptor activation through direct binding of the drug to TLR4 at the cell surface. The current study was intended to better understand drug-induced TNF-α production in tumor cells, whether from short-term drug exposure or in cells selected for drug resistance. ELISAs were employed to measure cytokine release from breast and ovarian tumor cells in response to several structurally distinct chemotherapy agents and/or TLR4 agonists or antagonists. Drug uptake and drug sensitivity studies were also performed. We observed that several drugs induced TNF-αrelease from multiple tumor cell lines. Docetaxel-induced cytokine production was distinct from that of LPS in both MyD88-positive (MCF-7) and MyD88-deficient (A2780) cells. The acquisition of docetaxel resistance was accompanied by increased constitutive production of TNF-αand CXCL1, which waned at higher levels of resistance. In docetaxel-resistant MCF-7 and A2780 cell lines, the production of TNF-α could not be significantly augmented by docetaxel without the inhibition of P-gp, a transporter protein that promotes drug efflux from tumor cells. Pretreatment of tumor cells with LPS sensitized MyD88-positive cells (but not MyD88-deficient) to docetaxel cytotoxicity in both drug-naive and drug-resistant cells. Our findings suggest that taxane-induced inflammatory cytokine production from tumor cells depends on the duration of exposure, requires cellular drug-accumulation, and is distinct from the LPS response seen in breast tumor cells. Also, stimulation of the LPS-induced pathway may be an attractive target for treatment of drug-resistant disease.
Collapse
Affiliation(s)
- Derek W. Edwardson
- Ph.D. Program in Biomolecular Science, Laurentian University, Sudbury, Ontario, Canada
| | - Justin Boudreau
- Department of Biology, Laurentian University, Sudbury, Ontario, Canada
| | - Jonathan Mapletoft
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| | - Carita Lanner
- Ph.D. Program in Biomolecular Science, Laurentian University, Sudbury, Ontario, Canada
- Department of Biology, Laurentian University, Sudbury, Ontario, Canada
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
- Division of Medical Sciences, Northern Ontario School of Medicine, Sudbury, Ontario, Canada
| | - A. Thomas Kovala
- Ph.D. Program in Biomolecular Science, Laurentian University, Sudbury, Ontario, Canada
- Department of Biology, Laurentian University, Sudbury, Ontario, Canada
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
- Division of Medical Sciences, Northern Ontario School of Medicine, Sudbury, Ontario, Canada
| | - Amadeo M. Parissenti
- Ph.D. Program in Biomolecular Science, Laurentian University, Sudbury, Ontario, Canada
- Department of Biology, Laurentian University, Sudbury, Ontario, Canada
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
- Division of Medical Sciences, Northern Ontario School of Medicine, Sudbury, Ontario, Canada
- Health Sciences North Research Institute, Sudbury, Ontario, Canada
- Faculty of Medicine, Division of Oncology, University of Ottawa, Ottawa, Ontario, Canada
- * E-mail:
| |
Collapse
|
28
|
Yang L, Tang L, Dai F, Meng G, Yin R, Xu X, Yao W. Raf-1/CK2 and RhoA/ROCK signaling promote TNF-α-mediated endothelial apoptosis via regulating vimentin cytoskeleton. Toxicology 2017; 389:74-84. [PMID: 28743511 DOI: 10.1016/j.tox.2017.07.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/16/2017] [Accepted: 07/18/2017] [Indexed: 01/04/2023]
Abstract
Both RhoA/ROCK and Raf-1/CK2 pathway play essential roles in cell proliferation, apoptosis, differentiation, and multiple other common cellular functions. We previously reported that vimentin is responsible for TNF-α-induced cell apoptosis. Herein, we investigated the regulation of RhoA/ROCK and Raf-1/CK2 signaling on vimentin filaments and endothelial apoptosis mediated by TNF-α. Treatment with TNF-α significantly induced the activation of RhoA and ROCK, and the expression of ROCK1. RhoA deficiency could obviously inhibit ROCK activation and ROCK1 expression induced by TNF-α. Both RhoA deficiency and ROCK activity inhibition (Y-27632) greatly inhibited endothelial apoptosis and preserved cell viability in TNF-α-induced human umbilical vein endothelial cells (HUVECs). Also vimentin phosphorylation and the remodeling of vimentin or phospho-vimentin induced by TNF-α were obviously attenuated by RhoA suppression and ROCK inhibition. TNF-α-mediated vimentin cleavage was significantly inhibited by RhoA suppression and ROCK inhibition through decreasing the activation of caspase3 and 8. Furthermore, TNF-α treatment greatly enhanced the activation of Raf-1. Suppression of Raf-1 or CK2 by its inhibitor (GW5074 or TBB) blocked vimentin phosphorylation, remodeling and endothelial apoptosis, and preserved cell viability in TNF-α-induced HUVECs. However, Raf-1 inhibition showed no significant effect on TNF-α-induced ROCK expression and activation, suggesting that the regulation of Raf-1/CK2 signaling on vimentin was independent of ROCK. Taken together, these results indicate that both RhoA/ROCK and Raf-1/CK2 pathway are responsible for TNF-α-mediated endothelial cytotoxicity via regulating vimentin cytoskeleton.
Collapse
Affiliation(s)
- Lifeng Yang
- School of pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China
| | - Lian Tang
- School of pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China
| | - Fan Dai
- School of pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China
| | - Guoliang Meng
- School of pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China
| | - Runting Yin
- School of pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China
| | - Xiaole Xu
- School of pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China
| | - Wenjuan Yao
- School of pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China.
| |
Collapse
|
29
|
Farzan SF, Brickley EB, Li Z, Gilbert-Diamond D, Gossai A, Chen Y, Howe CG, Palys T, Karagas MR. Maternal and infant inflammatory markers in relation to prenatal arsenic exposure in a U.S. pregnancy cohort. ENVIRONMENTAL RESEARCH 2017; 156:426-433. [PMID: 28410520 PMCID: PMC5477637 DOI: 10.1016/j.envres.2017.03.056] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/27/2017] [Accepted: 03/30/2017] [Indexed: 05/21/2023]
Abstract
INTRODUCTION Accumulating evidence indicates that arsenic (As), a potent environmental toxicant, may increase cardiovascular disease risk and adversely affect endothelial function at high levels of exposure. Pregnancy is a vulnerable time for both mother and child; however, studies examining the association between prenatal As exposure and plasma biomarkers of inflammation and endothelial function in mothers and newborns are lacking. METHODS We examined maternal urinary As levels at gestational weeks 24-28 and levels of inflammatory biomarkers in plasma from 563 pregnant women and 500 infants' cord blood. We assessed a multiplexed panel of circulating inflammatory and endothelial function markers, including tumor necrosis factor alpha (TNFα), monocyte chemoattractant protein 1 (MCP1), intercellular adhesion molecule (ICAM1) and vascular cell adhesion molecule (VCAM1). RESULTS Compared with the bottom tertile, the highest tertile of maternal urinary As during pregnancy was associated with a 145.2ng/ml (95% CI 4.1, 286.3; p=0.04) increase in cord blood ICAM1 and 557.3ng/ml (95% CI -56.4, 1171.1; p=0.09) increase in cord blood VCAM1. Among mothers, the highest tertile of maternal urinary As during pregnancy was related to a 141.8ng/ml (95% CI 26.1, 257.5; p=0.02) increase maternal plasma VCAM1 levels. Urinary As was unrelated to MCP1 or TNFα in maternal plasma and cord blood. In structural equation models, the association between maternal urinary As and infant VCAM was mediated by maternal levels of VCAM (βmediation: 0.024, 95% CI: 0.002, 0.050). CONCLUSION Our observations indicate that As exposure during pregnancy may affect markers of vascular health and endothelial function in both pregnant women and children, and suggest further investigation of the potential impacts on cardiovascular health in these susceptible populations.
Collapse
Affiliation(s)
- Shohreh F Farzan
- Department of Preventive Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA.
| | - Elizabeth B Brickley
- Children's Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, NH, USA and Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Zhigang Li
- Children's Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, NH, USA and Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Diane Gilbert-Diamond
- Children's Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, NH, USA and Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Anala Gossai
- Children's Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, NH, USA and Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Yu Chen
- Department of Population Health, New York University School of Medicine, New York, NY, USA
| | - Caitlin G Howe
- Department of Preventive Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Thomas Palys
- Children's Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, NH, USA and Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Margaret R Karagas
- Children's Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, NH, USA and Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
30
|
Bulat K, Rygula A, Szafraniec E, Ozaki Y, Baranska M. Live endothelial cells imaged by Scanning Near-field Optical Microscopy (SNOM): capabilities and challenges. JOURNAL OF BIOPHOTONICS 2017; 10:928-938. [PMID: 27545579 DOI: 10.1002/jbio.201600081] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 07/23/2016] [Accepted: 08/05/2016] [Indexed: 06/06/2023]
Abstract
The scanning near-field optical microscopy (SNOM) shows a potential to study details of biological samples, since it provides the optical images of objects with nanometric spatial resolution (50-200 nm) and the topographic information at the same time. The goal of this work is to demonstrate the capabilities of SNOM in transmission configuration to study human endothelial cells and their morphological changes, sometimes very subtle, upon inflammation. Various sample preparations were tested for SNOM measurements and promising results are collected to show: 1) the influence of α tumor necrosis factor (TNF-α) on EA.hy 926 cells (measurements of the fixed cells); 2) high resolution images of various endothelial cell lines, i.e. EA.hy 926 and HLMVEC (investigations of the fixed cells in buffer environment); 3) imaging of live endothelial cells in physiological buffers. The study demonstrate complementarity of the SNOM measurements performed in air and in liquid environments, on fixed as well as on living cells. Furthermore, it is proved that the SNOM is a very useful method for analysis of cellular morphology and topography. Changes in the cell shape and nucleus size, which are the symptoms of inflammatory reaction, were noticed in TNF-α activated EA.hy 926 cells. The cellular structures of submicron size were observed in high resolution optical images of cells from EA.hy 926 and HLMVEC lines.
Collapse
Affiliation(s)
- Katarzyna Bulat
- Faculty of Chemistry, Jagiellonian University, Ingardena 3, Krakow, Poland
- Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, Kraków, Poland
| | - Anna Rygula
- Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, Kraków, Poland
| | - Ewelina Szafraniec
- Faculty of Chemistry, Jagiellonian University, Ingardena 3, Krakow, Poland
| | - Yukihiro Ozaki
- Kwasei Gakuin University, 2-1 Gakuen, Sanda, Hyougo, 669-1337, Japan
| | - Malgorzata Baranska
- Faculty of Chemistry, Jagiellonian University, Ingardena 3, Krakow, Poland
- Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, Kraków, Poland
| |
Collapse
|
31
|
Danoy M, Shinohara M, Rizki-Safitri A, Collard D, Senez V, Sakai Y. Alteration of pancreatic carcinoma and promyeloblastic cell adhesion in liver microvasculature by co-culture of hepatocytes, hepatic stellate cells and endothelial cells in a physiologically-relevant model. Integr Biol (Camb) 2017; 9:350-361. [DOI: 10.1039/c6ib00237d] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Mathieu Danoy
- LIMMS/CNRS UMI2820 Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
- Institut d'Electronique, de Microélectronique et de Nanotechnologies (IEMN), Université Lille, CNRS, ISEN, UMR 8520, F-59000 Lille, France
| | - Marie Shinohara
- Center for International Research on Integrative Biomedical Systems (CIBiS), Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Astia Rizki-Safitri
- Center for International Research on Integrative Biomedical Systems (CIBiS), Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Dominique Collard
- LIMMS/CNRS UMI2820 Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
- SMMIL-E: Institut pour la Recherche sur le Cancer de Lille, Boulevard du Pr Jules Leclercq, 59000 Lille, France
| | - Vincent Senez
- Institut d'Electronique, de Microélectronique et de Nanotechnologies (IEMN), Université Lille, CNRS, ISEN, UMR 8520, F-59000 Lille, France
| | - Yasuyuki Sakai
- Center for International Research on Integrative Biomedical Systems (CIBiS), Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
- Department of Chemical System Engineering, graduate school of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Max Planck - The University of Tokyo, Center for Integrative Inflammology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| |
Collapse
|
32
|
Ni Y, Teng T, Li R, Simonyi A, Sun GY, Lee JC. TNFα alters occludin and cerebral endothelial permeability: Role of p38MAPK. PLoS One 2017; 12:e0170346. [PMID: 28170408 PMCID: PMC5295672 DOI: 10.1371/journal.pone.0170346] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 01/03/2017] [Indexed: 11/22/2022] Open
Abstract
Occludin is a key tight junction (TJ) protein in cerebral endothelial cells (CECs) playing an important role in modulating blood-brain barrier (BBB) functions. This protein (65kDa) has been shown to engage in many signaling pathways and phosphorylation by both tyrosine and threonine kinases. Despite yet unknown mechanisms, pro-inflammatory cytokines and endotoxin (lipopolysaccharides, LPS) may alter TJ proteins in CECs and BBB functions. Here we demonstrate the responses of occludin in an immortalized human cerebral endothelial cell line (hCMEC/D3) to stimulation by TNFα (10 ng/mL), IL-1β (10 ng/mL) and LPS (100 ng/mL). Exposing cells to TNFα resulted in a rapid and transient upward band-shift of occludin, suggesting of an increase in phosphorylation. Exposure to IL-1β produced significantly smaller effects and LPS produced almost no effects on occludin band-shift. TNFα also caused transient stimulation of p38MAPK and ERK1/2 in hCMEC/D3 cells, and the occludin band-shift induced by TNFα was suppressed by SB202190, an inhibitor for p38MAPK, and partly by U0126, the MEK1/2-ERK1/2 inhibitor. Cells treated with TNFα and IL-1β but not LPS for 24 h resulted in a significant (p < 0.001) decrease in the expression of occludin, and the decrease could be partially blocked by SB202190, the inhibitor for p38MAPK. Treatment with TNFα also altered cell morphology and enhanced permeability of the CEC layer as measured by the FITC-dextran assay and the trans-endothelial electrical resistances (TEER). However, treatment with SB202190 alone could not effectively reverse the TNFα -induced morphology changes or the enhanced permeability changes. These results suggest that despite effects of TNFα on p38MAPK-mediated occludin phosphorylation and expression, these changes are not sufficient to avert the TNFα-induced alterations on cell morphology and permeability.
Collapse
Affiliation(s)
- Yawen Ni
- Department of Bioengineering, University of Missouri, Columbia, Missouri, United States of America
| | - Tao Teng
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Runting Li
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
| | - Agnes Simonyi
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
| | - Grace Y Sun
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
| | - James C Lee
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
33
|
Yan M, Leng T, Tang L, Zheng X, Lu B, Li Y, Sheng L, Lin S, Shi H, Yan G, Yin W. Neuroprotectant androst-3β, 5α, 6β-triol suppresses TNF-α-induced endothelial adhesion molecules expression and neutrophil adhesion to endothelial cells by attenuation of CYLD-NF-κB pathway. Biochem Biophys Res Commun 2017; 483:892-896. [PMID: 28082198 DOI: 10.1016/j.bbrc.2017.01.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 01/08/2017] [Indexed: 10/20/2022]
Abstract
Neuroinflammation is one of key pathologic element in neurological diseases including stroke, traumatic brain injury, Alzheimer' s Disease, Parkinson's Disease, and multiple sclerosis as well. Up-regulation of endothelial adhesion molecules, which facilitate leukocyte adhesion to the endothelium, is the vital process of endothelial cells mediated neuroinflammation. Androst-3β, 5α, 6β-triol (Triol) is a synthetic steroid which has been reported to have neuroprotective effects in hypoxia/re-oxygenation-induced neuronal injury model. In the present study, we firstly investigated whether Triol inhibited the TNF-α-induced inflammatory response in rat brain microvascular endothelial cells (RBMECs). Our data showed that Triol decreased TNF-α-induced expression of vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) and the adhesion of neutrophil to RBMECs. We also found that Triol inhibited TNF-α-induced degradation of IκBα and phosphorylation of NF-κBp65 that are required for NF-κB activation. Furthermore, Triol significantly reversed TNF-α-induced down-expression of CYLD, which is a deubiquitinase that negatively regulates activation of NF-κB. These results suggest that Triol displays an anti-inflammatory effect on TNF-α-induced RBMECs via downregulating of CYLD-NF-κB signaling pathways and might have a potential benefit in therapeutic neuroinflammation related diseases.
Collapse
Affiliation(s)
- Min Yan
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, GD 510080, PR China
| | - Tiandong Leng
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Lipeng Tang
- Department of Pharmacology of Traditional Chinese Medicine, Guangdong Province's Traditional Chinese Medical Hospital, Guangzhou, GD 510120, PR China
| | - Xiaoke Zheng
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, GD 510080, PR China
| | - Bingzheng Lu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, GD 510080, PR China
| | - Yuan Li
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, GD 510080, PR China
| | - Longxiang Sheng
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, GD 510080, PR China
| | - Suizhen Lin
- Guangzhou Cellprotek Pharmaceutical, G Building F/4, 3 Lanyue Road, Science City, Guangzhou, 510663, PR China
| | - Haitao Shi
- Guangzhou Cellprotek Pharmaceutical, G Building F/4, 3 Lanyue Road, Science City, Guangzhou, 510663, PR China
| | - Guangmei Yan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, GD 510080, PR China
| | - Wei Yin
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, GD 510080, PR China.
| |
Collapse
|
34
|
Bossi F, Bernardi S, De Nardo D, Bramante A, Candido R, Carretta R, Fischetti F, Fabris B. Angiotensin 1-7 significantly reduces diabetes-induced leukocyte recruitment both in vivo and in vitro. Atherosclerosis 2015; 244:121-30. [PMID: 26630181 DOI: 10.1016/j.atherosclerosis.2015.11.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 11/16/2015] [Accepted: 11/17/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Recent studies have demonstrated that Ang1-7 has anti-inflammatory effects. Since the formation of Ang1-7 is significantly altered in the setting of diabetes, here we aimed to evaluate whether Ang1-7 infusion could ameliorate diabetes-induced leukocyte recruitment. METHODS Wild-type male Wistar rats were randomly allocated to the following groups: control + saline, control + Ang1-7, diabetes + saline, diabetes + Ang1-7. Diabetes was induced by streptozotocin. Saline and Ang1-7 (576 μg/kg/day) were injected intraperitoneally daily. After 4 weeks leukocyte trafficking was studied in vivo by intravital microscopy in the mesenteric bed, where the expression of pro-oxidative, proinflammatory, and profibrotic molecules was also assessed. In parallel in vitro studies, HUVEC were grown in 5 mM, 22 mM, 30 mM, 40 mM, 50 mM, and 75 mM glucose media for 48 h, 72 h and 6 days and were treated either with placebo, or with Ang1-7, or with Ang1-7 and its inhibitor A779 in order to evaluate the expression of ICAM-1 and VCAM-1. We further studied leukocytes recruitment in vitro by evaluating PMN-HUVEC adhesion. RESULTS Ang1-7 prevented in vivo diabetes-induced leukocyte adhesion and extravasation, and it significantly reduced vascular hypertrophy and the other molecular changes due to diabetes. Ang 1-7 prevented also in vitro the hyperglycemia-induced increase of ICAM-1 and VCAM-1 as well as the hyperglycemia-induced PMN adhesion. A779 inhibited Ang 1-7 effects. CONCLUSIONS Ang1-7 significantly reduced diabetes-induced leukocyte recruitment both in vivo and in vitro. These findings emphasize the potential utility of ACE2/Ang1-7/Mas repletion as a strategy to reduce diabetes-induced atherosclerosis.
Collapse
Affiliation(s)
- Fleur Bossi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34100 Trieste, Italy.
| | - Stella Bernardi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34100 Trieste, Italy
| | - Daniele De Nardo
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34100 Trieste, Italy
| | - Alessandra Bramante
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34100 Trieste, Italy
| | - Riccardo Candido
- Centro Diabetologico ASS 1 Triestina, Via Puccini, 34100 Trieste, Italy
| | - Renzo Carretta
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34100 Trieste, Italy
| | - Fabio Fischetti
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34100 Trieste, Italy
| | - Bruno Fabris
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34100 Trieste, Italy
| |
Collapse
|
35
|
Tong YF, Liu Y, Hu ZX, Li ZC, A A. Protocatechuic aldehyde inhibits TNF-α-induced fibronectin expression in human umbilical vein endothelial cells via a c-Jun N-terminal kinase dependent pathway. Exp Ther Med 2015; 11:277-282. [PMID: 26889254 DOI: 10.3892/etm.2015.2896] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 08/11/2015] [Indexed: 12/13/2022] Open
Abstract
Fibronectin (FN) is one of the most important extracellular matrix proteins and plays an important role in the pathogenesis of atherosclerosis (AS). The aim of the present study was to evaluate the effect of a potent, water-soluble antioxidant, protocatechuic aldehyde (PA), which is derived from the Chinese herb Salvia miltiorrhiza, on the expression of FN in human umbilical vein endothelial cells (HUVECs) stimulated with tumor necrosis factor-α (TNF-α). The pharmacological effects of PA on the production of FN were investigated using ELISA and western blot analysis. In addition, ELISA and western blot analysis were used to examine the activation and suppression of the mitogen-activated protein kinase (MAPK) pathways and nuclear factor (NF)-κB in TNF-α-stimulated HUVECs, in order to explore the underlying pharmacological mechanism of PA. The inhibitory effect of PA on the total generation of reactive oxygen species (ROS) in TNF-α-stimulated HUVECs was assessed using 2',7'-dichlorofluorescein diacetate. Pretreatment of HUVECs with PA (0.15, 0.45 and 1.35 mM) for 18 h markedly attenuated the TNF-α-stimulated FN surface expression and secretion in a dose-dependent manner. Intracellular ROS generation and the expression of extracellular signal-regulated kinase 1 and 2 (ERK1/2), c-Jun N-terminal kinase (JNK) and p38 MAPK (p38) were significantly induced by TNF-α (2 ng/ml) in HUVECs. TNF-α-induced ROS generation and JNK activation were inhibited by PA in a concentration-dependent manner. By contrast, ERK1/2 and p38 activation was not significantly affected by PA. Pretreatment of HUVECs with PA for 18 h markedly attenuated TNF-α-stimulated NF-κB activation. In conclusion, the present findings suggest that PA inhibits TNF-α-induced FN expression in HUVECs through a mechanism that involves ROS/JNK and NF-κB.
Collapse
Affiliation(s)
- Yue-Feng Tong
- Division of Cardiology, The First Yongkang Municipal Hospital, Yongkang, Zhejiang 321300, P.R. China
| | - Yong Liu
- Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Zhi-Xing Hu
- Division of Cardiology, The First Yongkang Municipal Hospital, Yongkang, Zhejiang 321300, P.R. China
| | - Zhe-Cheng Li
- Division of Cardiology, The First Yongkang Municipal Hospital, Yongkang, Zhejiang 321300, P.R. China
| | - Agula A
- Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| |
Collapse
|
36
|
Yao W, Gu C, Shao H, Meng G, Wang H, Jing X, Zhang W. Tetrahydroxystilbene Glucoside Improves TNF-α-Induced Endothelial Dysfunction: Involvement of TGFβ/Smad Pathway and Inhibition of Vimentin Expression. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2015; 43:183-98. [DOI: 10.1142/s0192415x15500123] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Endothelial dysfunction plays an important role in the pathogenesis of atherogenesis. 2,3,5,4′-tetrahydroxystilbene-2-O-β-D-glucoside (TSG), an active component of the rhizome extract from Polygonum multiflorum (PM), exhibits significant anti-atherosclerotic activity. Here, we used human umbilical vein endothelial cells (HUVECs) induced by tumor necrosis factor-α (TNF-α) in vitro to investigate the cytoprotective effects of TSG on TNF-α-induced endothelial injury and the related mechanisms. Pretreatment with 50 and 100 μM TSG markedly attenuated TNF-α-induced loss of cell viability and release of lactate dehydrogenase (LDH) and inhibited TNF-α-induced cell apoptosis. The inhibition of vimentin expression was involved in the cytoprotection afforded by TSG. Using inhibitors for PI3K and TGFβ or siRNA for Akt and Smad2, we found that vimentin production in HUVECs is regulated by TGFβ/Smad signaling, but not by PI3K–Akt–mTOR signaling. Meanwhile, TSG inhibited both the expression of TGFβ1 and the phosphorylation of Smad2 and Smad3, and TSG suppressed the nuclear translocation of Smad4 induced by TNF-α. These results suggest that TSG protects HUVECs against TNF-α-induced cell damage by inhibiting vimentin expression via the interruption of the TGFβ/Smad signaling pathway.
Collapse
Affiliation(s)
- Wenjuan Yao
- Department of Pharmacology, Nantong University Medical College, Jiangsu, China
| | - Chengjing Gu
- Department of Pharmacology, Nantong University Medical College, Jiangsu, China
| | - Haoran Shao
- Department of Pharmacology, Nantong University Medical College, Jiangsu, China
| | - Guoliang Meng
- Department of Pharmacology, Nantong University Medical College, Jiangsu, China
| | - Huiming Wang
- Department of Pharmacology, Nantong University Medical College, Jiangsu, China
| | - Xiang Jing
- Department of Pharmacology, Nantong University Medical College, Jiangsu, China
| | - Wei Zhang
- Department of Pharmacology, Nantong University Medical College, Jiangsu, China
| |
Collapse
|
37
|
Perry DM, Newcomb B, Adada M, Wu BX, Roddy P, Kitatani K, Siskind L, Obeid LM, Hannun YA. Defining a role for acid sphingomyelinase in the p38/interleukin-6 pathway. J Biol Chem 2014; 289:22401-12. [PMID: 24951586 DOI: 10.1074/jbc.m114.589648] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Acid sphingomyelinase (ASM) is one of the key enzymes involved in regulating the metabolism of the bioactive sphingolipid ceramide in the sphingolipid salvage pathway, yet defining signaling pathways by which ASM exerts its effects has proven difficult. Previous literature has implicated sphingolipids in the regulation of cytokines such as interleukin-6 (IL-6), but the specific sphingolipid pathways and mechanisms involved in inflammatory signaling need to be further elucidated. In this work, we sought to define the role of ASM in IL-6 production because our previous work showed that a parallel pathway of ceramide metabolism, acid β-glucosidase 1, negatively regulates IL-6. First, silencing ASM with siRNA abrogated IL-6 production in response to the tumor promoter, 4β-phorbol 12-myristate 13-acetate (PMA), in MCF-7 cells, in distinction to acid β-glucosidase 1 and acid ceramidase, suggesting specialization of the pathways. Moreover, treating cells with siRNA to ASM or with the indirect pharmacologic inhibitor desipramine resulted in significant inhibition of TNFα- and PMA-induced IL-6 production in MDA-MB-231 and HeLa cells. Knockdown of ASM was found to significantly inhibit PMA-dependent IL-6 induction at the mRNA level, probably ruling out mechanisms of translation or secretion of IL-6. Further, ASM knockdown or desipramine blunted p38 MAPK activation in response to TNFα, revealing a key role for ASM in activating p38, a signaling pathway known to regulate IL-6 induction. Last, knockdown of ASM dramatically blunted invasion of HeLa and MDA-MB-231 cells through Matrigel. Taken together, these results demonstrate that ASM plays a critical role in p38 signaling and IL-6 synthesis with implications for tumor pathobiology.
Collapse
Affiliation(s)
- David M Perry
- From the Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425
| | | | | | - Bill X Wu
- From the Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Patrick Roddy
- From the Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Kazuyuki Kitatani
- the Tohoku Medical Megabank Organization and Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Leah Siskind
- the Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40202
| | - Lina M Obeid
- the Department of Medicine Stony Brook University, Stony Brook, New York 11794, and the Northport Veterans Affairs Hospital, Northport, New York 11768
| | | |
Collapse
|
38
|
Sanguis draconis, a dragon's blood resin, attenuates high glucose-induced oxidative stress and endothelial dysfunction in human umbilical vein endothelial cells. ScientificWorldJournal 2014; 2014:423259. [PMID: 24987732 PMCID: PMC4060585 DOI: 10.1155/2014/423259] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 05/10/2014] [Indexed: 01/01/2023] Open
Abstract
Hyperglycaemia, a characteristic feature of diabetes mellitus, induces endothelial dysfunction and vascular complications by limiting the proliferative potential of these cells. Here we aimed to investigate the effect of an ethanolic extract of Sanguis draconis (SD), a kind of dragon's blood resin that is obtained from Daemonorops draco (Palmae), on human umbilical vein endothelial cells (HUVEC) under high-glucose (HG) stimulation and its underlying mechanism. Concentration-dependent (0-50 μg/mL) assessment of cell viability showed that SD does not affect cell viability with a similar trend up to 48 h. Remarkably, SD (10-50 μg/mL) significantly attenuated the high-glucose (25 and 50 mM) induced cell toxicity in a concentration-dependent manner. SD inhibited high glucose-induced nitrite (NO) and lipid peroxidation (MDA) production and reactive oxygen species (ROS) formation in HUVEC. Western blot analysis revealed that SD treatments abolished HG-induced phosphorylation of extracellular signal-regulated kinase 1/2 (ERK 1/2), nuclear transcription factor, κB (NF-κB), VCAM-1, and E-selectin, and it also blocked the breakdown of PARP-116 kDa protein in a dose-dependent manner. Furthermore, we found that SD increased the expression of Bcl-2 and decreased Bax protein expression in HG-stimulated HUVEC. Thus, these results of this study demonstrate for the first time that SD inhibits glucose induced oxidative stress and vascular inflammation in HUVEC by inhibiting the ERK/NF-κB/PARP-1/Bax signaling cascade followed by suppressing the activation of VCAM-1 and E-selectin. These data suggest that SD may have a therapeutic potential in vascular inflammation due to the decreased levels of oxidative stress, apoptosis, and PARP-1 activation.
Collapse
|
39
|
Choi H, Nguyen HN, Lamb FS. Inhibition of endocytosis exacerbates TNF-α-induced endothelial dysfunction via enhanced JNK and p38 activation. Am J Physiol Heart Circ Physiol 2014; 306:H1154-63. [DOI: 10.1152/ajpheart.00885.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Tumor necrosis factor-α (TNF-α) is a pro-inflammatory cytokine that causes endothelial dysfunction. Endocytosis of TNF-α receptors (TNFR) precedes endosomal reactive oxygen species (ROS) production, which is required for NF-κB activation in vascular smooth muscle cells. It is unknown how endocytosis of TNFRs impacts signaling in endothelial cells. We hypothesized that TNF-α-induced endothelial dysfunction is induced by both endosomal and cell surface events, including NF-κB and mitogen-activated protein kinases (MAPKs) activation, and endocytosis of the TNFR modifies signaling. Mesenteric artery segments from C57BL/6 mice were treated with TNF-α (10 ng/ml) for 22 h in tissue culture, with or without signaling inhibitors (dynasore for endocytosis, SP600125 for JNK, SB203580 for p38, U0126 for ERK), and vascular function was assessed. Endothelium-dependent relaxation to acetylcholine (ACh) was impaired by TNF-α, and dynasore exacerbated this, whereas JNK or p38 inhibition prevented these effects. In cultured endothelial cells from murine mesenteric arteries, dynasore potentiated JNK and p38 but not ERK phosphorylation and promoted cell death. NF-κB activation by TNF-α was decreased by dynasore. JNK inhibition dramatically increased both the magnitude and duration of TNF-α-induced NF-κB activation and potentiated intercellular adhesion molecule-1 (ICAM-1) activation. Dynasore still inhibited NF-κB activation in the presence of SP600125. Thus TNF-α-induced endothelial dysfunction is both JNK and p38 dependent. Endocytosis modulates the balance of NF-κB and MAPK signaling, and inhibition of NF-κB activation by JNK limits this pro-proliferative signal, which may contribute to endothelial cell death in response to TNF-α.
Collapse
Affiliation(s)
- Hyehun Choi
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hong N. Nguyen
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Fred S. Lamb
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
40
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: enzymes. Br J Pharmacol 2013; 170:1797-867. [PMID: 24528243 PMCID: PMC3892293 DOI: 10.1111/bph.12451] [Citation(s) in RCA: 415] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. Enzymes are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, nuclear hormone receptors, catalytic receptors and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
41
|
Chakraborti S, Roy S, Mandal A, Chowdhury A, Chakraborti T. Role of PKC-ζ in NADPH oxidase-PKCα-Giα axis dependent inhibition of β-adrenergic response by U46619 in pulmonary artery smooth muscle cells. Arch Biochem Biophys 2013; 540:133-44. [PMID: 24184446 DOI: 10.1016/j.abb.2013.10.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 10/02/2013] [Accepted: 10/23/2013] [Indexed: 02/04/2023]
Abstract
Treatment of bovine pulmonary artery smooth muscle cells (BPASMCs) with U46619 attenuated isoproterenol caused stimulation of adenyl cyclase activity and cAMP production. Pretreatment with SQ29548 (Tp receptor antagonist), apocynin (NADPH oxidase inhibitor) and Go6976 (PKC-α inhibitor) eliminated U46619 caused attenuation of isoproterenol stimulated adenyl cyclase activity. Pretreatment with SQ29548 and apocynin prevented U46619 induced increase in NADPH oxidase activity, PKC-α activity and Giα phosphorylation. However, pretreatment with CZI, a PKC-ζ inhibitor, markedly, but not completely, inhibited U46619 induced increase in NADPH oxidase activity, PKC-α activity, Giα phosphorylation and also significantly eliminated U46619 caused attenuation of isoproterenol stimulated adenyl cyclase activity. Pretreatment with Go6976 inhibited U46619 induced increase in Giα phosphorylation, but not PKC-ζ activity and NADPH oxidase activity. Pretreatment with pertussis toxin eliminated U46619 caused attenuation of isoproterenol stimulated adenyl cyclase activity without any discernible change in PKC-ζ, NADPH oxidase and PKC-α activities. Transfection of the cells with Tp, PKC-ζ and PKC-α siRNA duplexes corroborate the findings observed with their respective pharmacological inhibitors on the responses produced by U46619. Taken together, we suggest involvement of PKC-ζ in U46619 caused attenuation of isoproterenol stimulated β-adrenergic response, which is regulated by NADPH oxidase-PKCα-Giα axis in pulmonary artery smooth muscle cells.
Collapse
Affiliation(s)
- Sajal Chakraborti
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia 741235, West Bengal, India.
| | | | | | | | | |
Collapse
|
42
|
Meiler S, Baumer Y, Huang Z, Hoffmann FW, Fredericks GJ, Rose AH, Norton RL, Hoffmann PR, Boisvert WA. Selenoprotein K is required for palmitoylation of CD36 in macrophages: implications in foam cell formation and atherogenesis. J Leukoc Biol 2013; 93:771-80. [PMID: 23444136 DOI: 10.1189/jlb.1212647] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Selk is an ER transmembrane protein important for calcium flux and macrophage activation, but its role in foam cell formation and atherosclerosis has not been evaluated. BMDMs from Selk(-/-) mice exhibited decreased uptake of modLDL and foam cell formation compared with WT controls, and the differences were eliminated with anti-CD36 blocking antibody. CD36 expression was decreased in TNF-α-stimulated Selk(-/-) BMDMs compared with WT controls. Fluorescence microscopy revealed TNF-α-induced clustering of CD36 in WT BMDMs indicative of lipid raft localization, which was absent in Selk(-/-) BMDMs. Fractionation revealed lower levels of CD36 reaching lipid rafts in TNF-α-stimulated Selk(-/-) BMDMs. Immunoprecipitation showed that Selk(-/-) BMDMs have decreased CD36 palmitoylation, which occurs at the ER membrane and is crucial for stabilizing CD36 expression and directing its localization to lipid rafts. To assess if this phenomenon had a role in atherogenesis, a HFD was fed to irradiated Ldlr(-/-) mice reconstituted with BM from Selk(-/-) or WT mice. Selk was detected in aortic plaques of controls, particularly in macrophages. Selk(-/-) in immune cells led to reduction in atherosclerotic lesion formation without affecting leukocyte migration into the arterial wall. These findings suggest that Selk is important for stable, localized expression of CD36 in macrophages during inflammation, thereby contributing to foam cell formation and atherogenesis.
Collapse
Affiliation(s)
- Svenja Meiler
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96813, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Tsuchiya K, Tanaka J, Shuiqing Y, Welch CL, DePinho RA, Tabas I, Tall AR, Goldberg IJ, Accili D. FoxOs integrate pleiotropic actions of insulin in vascular endothelium to protect mice from atherosclerosis. Cell Metab 2012; 15:372-81. [PMID: 22405072 PMCID: PMC3315846 DOI: 10.1016/j.cmet.2012.01.018] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 11/29/2011] [Accepted: 01/23/2012] [Indexed: 12/21/2022]
Abstract
Atherosclerotic cardiovascular disease is the leading cause of death in insulin-resistant (type 2) diabetes. Vascular endothelial dysfunction paves the way for atherosclerosis through impaired nitric oxide availability, inflammation, and generation of superoxide. Surprisingly, we show that ablation of the three genes encoding isoforms of transcription factor FoxO in endothelial cells prevents atherosclerosis in low-density lipoprotein receptor knockout mice by reversing these subphenotypes. Paradoxically, the atheroprotective effect of FoxO deletion is associated with a marked decrease of insulin-dependent Akt phosphorylation in endothelial cells, owing to reduced FoxO-dependent expression of the insulin receptor adaptor proteins Irs1 and Irs2. These findings support a model in which FoxO is the shared effector of multiple atherogenic pathways in endothelial cells. FoxO ablation lowers the threshold of Akt activity required for protection from atherosclerosis. The data demonstrate that FoxO inhibition in endothelial cells has the potential to mediate wide-ranging therapeutic benefits for diabetes-associated cardiovascular disease.
Collapse
Affiliation(s)
- Kyoichiro Tsuchiya
- Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Endogenous Mechanisms Underlying the Activation and Sensitization of Meningeal Nociceptors: The Role of Immuno-Vascular Interactions and Cortical Spreading Depression. Curr Pain Headache Rep 2012; 16:270-7. [PMID: 22328144 DOI: 10.1007/s11916-012-0255-1] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
45
|
Grammes F, Rørvik KA, Takle H. Tetradecylthioacetic acid modulates cardiac transcription in Atlantic salmon, Salmo salar L., suffering heart and skeletal muscle inflammation. JOURNAL OF FISH DISEASES 2012; 35:109-117. [PMID: 22233512 DOI: 10.1111/j.1365-2761.2011.01326.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Heart and skeletal muscle inflammation (HSMI) is a disease causing considerable mortality in farmed Atlantic salmon. We have previously reported that pre-feeding of tetradecylthioacetic acid (TTA) reduces the mortality during a natural outbreak of HSMI. In the present paper we show that in the cardiac ventricle, during HSMI infection, pre-feeding TTA increases the expression of the immune genes: TNFα, VCAM-1, IgM and CD8α. We also show that TTA increases the cardiosomatic index potentially by elevating cardiomyogenesis through activation of the cardiac transcription factors MEF2C and Nkx2.5. Using the recently published genomic sequence of a HSMI associated piscine reovirus (PRV), we could show that the PRV levels have no confounding effects on the mRNA expression of the investigated genes. The results suggest that TTA induced cardiac growth, together with an elevated cardiac recruitment of immune cells, which might lead to increased robustness during HSMI infection.
Collapse
|
46
|
Kim SK, Moon WK, Park JY, Jung H. Inflammatory mimetic microfluidic chip by immobilization of cell adhesion molecules for T cell adhesion. Analyst 2012; 137:4062-8. [DOI: 10.1039/c2an35424a] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
47
|
Lee JK, Kim JK, Park SH, Sim YB, Jung JS, Suh HW. Lactosylceramide Mediates the Expression of Adhesion Molecules in TNF-α and IFNγ-stimulated Primary Cultured Astrocytes. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2011; 15:251-8. [PMID: 22128256 DOI: 10.4196/kjpp.2011.15.5.251] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 09/19/2011] [Accepted: 09/24/2011] [Indexed: 01/08/2023]
Abstract
Here we have investigated how lactosylceramide (LacCer) modulates gene expression of adhesion molecules in TNF-α and IFNγ (CM)-stimulated astrocytes. We have observed that stimulation of astrocytes with CM increased the gene expression of ICAM-1 and VCAM-1. D-Threo-1-phenyl- 2-decanoylamino-3-morpholino-1-propanol (PDMP) and N-butyldeoxynojirimycin (NBDNJ), inhibitors of glucosylceramide synthase (GLS) and LacCer synthase (galactosyltransferase, GalT-2), inhibited the gene expression of ICAM-1 and VCAM-1 and activation of their gene promoter induced by CM, which were reversed by exogenously supplied LacCer. Silencing of GalT-2 gene using its antisense oligonucleotides also attenuated CM-induced ICAM-1 and VCAM-1 expression, which were reversed by LacCer. PDMP treatment and silencing of GalT-2 gene significantly reduced CM-induced luciferase activities in NF-KB, AP-1, GAS, and STAT-3 luciferase vectors-transfected cells. In addition, LacCer reversed the inhibition of NF-KB and STAT-1 luciferase activities by PDMP. Taken together, our results suggest that LacCer may play a crucial role in the expression of ICAM-1 and VCAM-1 via modulating transcription factors, such as NF-KB, AP-1, STAT-1, and STAT-3 in CM-stimulated astrocytes.
Collapse
Affiliation(s)
- Jin-Koo Lee
- Department of Pharmacology, College of Medicine, Dankook University, Cheonan 330-714, Korea
| | | | | | | | | | | |
Collapse
|
48
|
Fukuhara Y, Tsuchiya K, Horinouchi Y, Tajima S, Kihira Y, Hamano S, Kawazoe K, Ikeda Y, Ishizawa K, Tomita S, Tamaki T. Protective effect of photodegradation product of nifedipine against tumor necrosis factor alpha-induced oxidative stress in human glomerular endothelial cells. THE JOURNAL OF MEDICAL INVESTIGATION 2011; 58:118-26. [PMID: 21372496 DOI: 10.2152/jmi.58.118] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Recently, increasing evidence suggests that the antihypertensive drug nifedipine acts as a protective agent for endothelial cells, and that the activity is unrelated to its calcium channel blocking. Nitrosonifedipine (NO-NIF) is metabolically and photochemically produced from nifedipine, and NO-NIF has been recognized as a contaminant of nifedipine because it has no antihypertensive effect. Treatment of tumor necrosis factor-α (TNF-α) suppressed the cell viability and facilitated the expression of Inter-Cellular Adhesion Molecule 1(ICAM-1) in human glomerular endothelial cells (HGECs) though, pretreatment of NO-NIF significantly recovered the TNF-α-induced cell damage to the same extent as Trolox-C did, and suppressed the ICAM-1 expression in a concentration dependent manner. In addition, NO-NIF inhibited the cell toxicity induced by cumene hydroperoxide, which hampers the integrity of cell membrane through oxidative stress, as effective as Trolox-c. These data suggest that NO-NIF is a candidate for a new class of antioxidative drug that protect cells against oxidative stress in glomerular endothelial cells.
Collapse
Affiliation(s)
- Yayoi Fukuhara
- Department of Pharmacology, Institute of Health Bioscience, University of Tokushima Graduate School, Tokushima, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Hwa JS, Mun L, Kim HJ, Seo HG, Lee JH, Kwak JH, Lee DU, Chang KC. Genipin Selectively Inhibits TNF-α-activated VCAM-1 But Not ICAM-1 Expression by Upregulation of PPAR-γ in Human Endothelial Cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2011; 15:157-62. [PMID: 21860594 DOI: 10.4196/kjpp.2011.15.3.157] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 06/14/2011] [Accepted: 06/15/2011] [Indexed: 11/15/2022]
Abstract
Vascular inflammation process has been suggested to be an important risk factor in the development of atherosclerosis. Recently we reported that induction of peroxisome proliferator-activated receptor-γ (PPAR-γ) selectively inhibits vascular cell adhesion molecule-1 (VCAM-1) but not intercellular cell adhesion molecule-1 (ICAM-1) in tumor necrosis factor (TNF)-α-activated human umbilical vein endothelial cells (HUVEC). In this study, we investigated whether genipin inhibits expression of cellular adhesion molecules, which is relevant to inflammation. Pretreatment with genipin reduced reactive oxygen species (ROS) production and expression of VCAM-1, but not ICAM-1 in TNF-α-activated HUVEC. Genipin dose- and time-dependently increased PPAR-γ expression and inhibited TNF-α-induced phosphorylation of Akt and PKC with different degrees. Finally, genipin prevented TNF-α-induced adhesion of U937 monocytic cells to HUVEC. Taken together, these results indicate that upregualtion of PPAR-γ by genipin selectively inhibits TNF-α-induced expression of VCAM-1, in which regulation of Akt and/or PKC play a key role. We concluded that genipin can be used for the treatment of cardiovascular disorders such as atherosclerosis.
Collapse
Affiliation(s)
- Jung Seok Hwa
- Department of Urology, School of Medicine, and Institute of Health Sciences, Gyeongsang National University, Jinju 660-290, Korea
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Moon L, Ha YM, Jang HJ, Kim HS, Jun MS, Kim YM, Lee YS, Lee DH, Son KH, Kim HJ, Seo HG, Lee JH, Kim YS, Chang KC. Isoimperatorin, cimiside E and 23-O-acetylshengmanol-3-xyloside from Cimicifugae rhizome inhibit TNF-α-induced VCAM-1 expression in human endothelial cells: involvement of PPAR-γ upregulation and PI3K, ERK1/2, and PKC signal pathways. JOURNAL OF ETHNOPHARMACOLOGY 2011; 133:336-44. [PMID: 20937376 DOI: 10.1016/j.jep.2010.10.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2010] [Revised: 09/15/2010] [Accepted: 10/01/2010] [Indexed: 05/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The methanol extract of Cimicifugae Rhizome has been traditionally used in various disorders including inflammation. AIM OF THE STUDY The aim of the study is to explore whether anti-inflammatory action of 3 active compounds, two triterpenoid glycosides (cimiside E, 23-O-actylshengmanol-3-xyloside) and one furanocoumarin (isoimperatorin), isolated from Cimicifugae Rhizome is related with peroxisome proliferator-activated receptor-γ (PPAR-γ) expression in human umbilical endothelial cell line, EA.hy926 cells. MATERIALS AND METHODS Cell viability and production of reactive oxygen species were performed. In addition, adhesion of monocyte into endothelial cells and western blot for expression of adhesion molecules and signal proteins were investigated in tumor necrosis factor-α (TNF-α)-activated cells. RESULTS Pretreatment of test compounds significantly reduced reactive oxygen species (ROS) production and expression of vascular cell adhesion molecule-1 (VCAM-1), but not intercellular cell adhesion molecule-1 (ICAM-1). Three compounds all dose-dependently increased not only PPAR-γ expression in EA.hy926 cells but inhibited TNF-α-induced phosphorylation of Akt, extracellular-signal-regulated kinase (ERK) and protein kinase C (PKC) with different specificity. Finally, they prevented TNF-α-induced adhesion of U937 monocytic cells to EA.hy926 cells. CONCLUSIONS The present results show that cimiside E, 23-O-actylshengmanol-3-xyloside, isoimperatorin isolated from Cimicifugae Rhizome selectively inhibits TNF-α-induced expression of VCAM-1 at least by upregulation of PPAR-γ, and signals for ERK1/2, PI3K, and PKC are involved in this effect.
Collapse
Affiliation(s)
- Lidiya Moon
- Department of Pharmacology School of Medicine, and Institute of Health Sciences, Gyeongsang National University, 92 Chilam-dong, Jinju 660-751, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|