1
|
Gounder SK, Chuturgoon AA, Ghazi T. Exploring the cardiotoxic potential of fumonisin B1 through inflammatory pathways and epigenetic modifications: A mini review. Toxicon 2025; 261:108374. [PMID: 40286825 DOI: 10.1016/j.toxicon.2025.108374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
This review is centered around the cardiotoxic effects of fumonisin B1 (FB1), particularly its impact on sphingolipid metabolism, inflammation, and epigenetics. FB1 is a mycotoxin produced by Fusarium fungi, which mainly contaminates cereal grains and poses an adverse health risk to both humans and animals; however, its disease-causing capabilities remain to be uncovered, specifically its ability to exacerbate and cause cardiovascular disease. It disrupts sphingolipid metabolism by inhibiting ceramide synthase, leading to cellular dysfunction and contributes to conditions such as hypertension and eventual heart failure. FB1 is responsible for an altered inflammatory response, whereby it increases pro-inflammatory cytokines such as IL-6 and IL-1β, which contribute to cardiovascular diseases. Moreover, FB1 induces significant epigenetic changes, including DNA hypermethylation, histone modifications such as increased H3K9me2 and H3K9me3, inhibition of histone acetyltransferase activity, and changes in microRNA expression profiles. These epigenetic alterations can silence or activate inflammatory genes, exacerbating disease progression. This review thus highlights the need for further research to elucidate the connections between FB1, inflammation, epigenetic modifications, and cardiotoxicity, which could lead to better strategies for managing FB1-related adverse health risks.
Collapse
Affiliation(s)
- Selwyn Kyle Gounder
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4041, South Africa
| | - Anil Amichund Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4041, South Africa.
| | - Terisha Ghazi
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4041, South Africa.
| |
Collapse
|
2
|
Liang C, Liu L, Yu W, Shi Q, Zheng J, Lyu J, Zhong J. Construction and validation of risk prediction models for different subtypes of retinal vein occlusion. ADVANCES IN OPHTHALMOLOGY PRACTICE AND RESEARCH 2025; 5:107-116. [PMID: 40230508 PMCID: PMC11995075 DOI: 10.1016/j.aopr.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 03/02/2025] [Accepted: 03/13/2025] [Indexed: 04/16/2025]
Abstract
Purpose While prognostic models for retinal vein occlusion (RVO) exist, subtype-specific risk prediction tools for central retinal vein occlusion (CRVO) and branch retinal vein occlusion (BRVO) remain limited. This study aimed to construct and validate distinct CRVO and BRVO risk stratification nomograms. Methods We retrospectively analyzed electronic medical records from a tertiary hospital in Guangzhou (January 2010-November 2024). Non-RVO controls were matched 1:4 (CRVO) and 1:2 (BRVO) by sex and year of admission. The final cohorts included 630 patients (126 CRVO cases and 504 controls) and 813 patients (271 BRVO cases and 542 controls). Predictors encompassed clinical histories and laboratory indices. Multivariate regression identified independent risk factors, and model performance was evaluated using the area under the receiver operating characteristic curve (AUC), calibration plots, and decision curve analysis (DCA). Results The CRVO-nom and BRVO-nom highlighted significant predictors, including the neutrophil-to-lymphocyte ratio (NLR). Additional risk factors for CRVO included high-density lipoprotein cholesterol (HDL-C), platelet distribution width (PDW), history of diabetes, cerebral infarction, and coronary artery disease (CAD). For BRVO, significant predictors included a history of hypertension, age, and body mass index (BMI). The AUC for CRVO-nom was 0.80 (95% CI: 0.73-0.87) in the training set and 0.77 (95% CI: 0.65-0.86) in the validation set, while BRVO-nom yielded an AUC of 0.95 (95 %CI: 0.91-0.97) in the training set and 0.95 (95% CI: 0.89-0.98) in the validation set. Conclusions CRVO and BRVO exhibit distinct risk profiles. The developed nomograms-CRVO-nom and BRVO-nom-provide subtype-specific risk stratification with robust discrimination and clinical applicability. An online Shiny calculator facilitates real-time risk estimation, enabling targeted prevention for high-risk populations.
Collapse
Affiliation(s)
- Chunlan Liang
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Lian Liu
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Wenjuan Yu
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Qi Shi
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jiang Zheng
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jingxiang Zhong
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Department of Ophthalmology, The Sixth Affiliated Hospital of Jinan University, Dongguan, China
| |
Collapse
|
3
|
Mohammed S, Kalogeropoulos AP, Alvarado V, Weisfelner-Bloom M, Clarke CJ. Serum and plasma sphingolipids as biomarkers of chemotherapy-induced cardiotoxicity in female patients with breast cancer. J Lipid Res 2025; 66:100798. [PMID: 40189207 PMCID: PMC12127548 DOI: 10.1016/j.jlr.2025.100798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/25/2025] Open
Abstract
Although effective as a chemotherapeutic, the utility of Doxorubicin (Dox) is hampered by cardiotoxicity. Despite this, the ability to predict and guide monitoring of patients receiving Dox is hampered by a lack of effective biomarkers to identify susceptible patients and detect early signs of subclinical cardiotoxicity. Based on their well-established roles in the response to Dox and other chemotherapies, we performed a retrospective analysis of serum and plasma sphingolipids (SLs) from female patients with breast cancer (BC) undergoing anthracycline-containing therapy, correlating with cardiac parameters assessed by echocardiography. Results showed substantial changes in both plasma and serum SL species during therapy including ceramide (Cer), deoxydihydroCer, and dihydrosphingosine with reversion toward baseline after treatment. Linear mixed-effects model analysis revealed that baseline levels of a number of SLs correlated with adverse cardiac outcomes. Here, serum sphingosine-1-phosphate (S1P), dihydroS1P, and plasma Cer performed comparably to the prognostic value of pro-NT-BNP, an established biomarker of cardiotoxicity. Intriguingly, while pro-NT-BNP had no predictive value at mid- and post-therapy timepoints, serum S1P and dihydroS1P, and plasma Cer levels showed a correlation with adverse outcomes, particularly at the post-therapy timepoint. Finally, analysis of plasma and serum C16:C24-Cer ratios-previously linked with adverse cardiac outcomes-showed no correlation in the context of chemotherapy treatment. Overall, this pilot study provides initial evidence that plasma and serum SLs may have benefits as both prognostic and diagnostic biomarkers for female BC patients undergoing anthracycline-containing chemotherapy. Consequently, diagnostic SL measurements-recently implemented for metabolic-associated cardiac disorders-could have wider utility.
Collapse
Affiliation(s)
- Samia Mohammed
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | | | - Victoria Alvarado
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | | | - Christopher J Clarke
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Cancer Center, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
4
|
Du ZW, Li YS, Jiang XC, Gao JQ. Nanoparticles Designed Based on the Blood-Brain Barrier for the Treatment of Cerebral Ischemia-Reperfusion Injury. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410404. [PMID: 40042407 DOI: 10.1002/smll.202410404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/11/2025] [Indexed: 05/13/2025]
Abstract
Cerebral ischemia-reperfusion injury (CI/RI) is currently considered a significant factor affecting the prognosis of ischemic stroke. The blood-brain barrier (BBB) plays multiple roles in the treatment ofCI/RI. BBB leakage allows bloodborne toxins to exacerbate the stroke pathology. Yet as the physiological barrier that separates the blood from the brain, BBB also poses a significant obstacle to therapeutic drug delivery. Therefore, it is essential to consider both crossing and repairing the BBB in the process of the treatment of CI/RI. Leveraging the exceptional benefits of nanoparticles (NPs) for BBB penetration and targeted repair, numerous NPs are developed as promising drug delivery platforms. Considering the complex role of the BBB in CI/RI, this review delves into the strategies for designing NPs to cross the BBB, focusing on peptide-modified NPs, cell-mediated NPs, cell membrane-derived NPs, and BBB-modulating NPs. Additionally, it summarizes design strategies of NPs targeting endothelial cells (ECs), astrocytes, and those aimed at regulating the microenvironment to repair the BBB. On this basis, it reveals the prospects and challenges of NPs designed around the BBB in CI/RI treatment. And it highlights the need to combine BBB permeability promotion and BBB repair in nanoparticle strategies designed based on the BBB to achieve more effective treatment.
Collapse
Affiliation(s)
- Zhi-Wei Du
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Yao-Sheng Li
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Xin-Chi Jiang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Jian-Qing Gao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| |
Collapse
|
5
|
Mitra R, Pentland K, Kolev S, Eden M, Levine E, Oakes JM, Ebong EE. Co-therapy with S1P and heparan sulfate derivatives to restore endothelial glycocalyx and combat pro-atherosclerotic endothelial dysfunction. Life Sci 2025; 377:123662. [PMID: 40280298 DOI: 10.1016/j.lfs.2025.123662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
AIMS Endothelial cell (EC) glycocalyx (GCX) shedding from disturbed blood flow and chemical factors leads to low-density lipoprotein infiltration, reduced nitric oxide synthesis, vascular dysfunction and atherosclerosis. This study evaluates a therapy combining sphingosine-1-phosphate (S1P) and heparin (heparan sulfate derivative). We hypothesized that heparin/S1P co-treatment repairs mechanically damaged EC GCX in disturbed flow (DF) regions and restores anti-atherosclerotic mechanotransduction to treat cardiovascular disease. MATERIALS AND METHODS We used a parallel-plate flow chamber to simulate flow conditions in vitro and a partial carotid ligation mouse model to mimic DF in vivo. Heparin and albumin-bound S1P were administered to assess their reparative effects on the endothelial GCX. Fluorescent staining, confocal microscopy, and ultrasound evaluated endothelial cell function and endothelial-dependent vascular function. Barrier functionality was assessed via macrophage uptake. Heparin/S1P mechanism-of-action insights were gained through fluid dynamics simulations and staining of GCX synthesis enzyme and S1P receptor. Statistical analyses validated the results. KEY FINDINGS The in vitro data showed that heparin/S1P therapy improves DF-conditioned ECs by restoring GCX and elevating vasodilator eNOS (endothelial-type nitric oxide synthase) expression. In vivo studies confirmed GCX degradation, vessel inflammation, hyperpermeability, and wall thickening in the mouse model's partially ligated left carotid artery. Heparin/S1P treatment restored GCX thickness and coverage, reduced inflammation and hyperpermeability, and inhibited vessel wall thickening. SIGNIFICANCE This work introduces a new approach to regenerating the EC GCX and restoring its function in ECs under DF conditions, offering a groundbreaking solution for preventing cardiovascular diseases like atherosclerosis.
Collapse
Affiliation(s)
- Ronodeep Mitra
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Kaleigh Pentland
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Svilen Kolev
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Matthew Eden
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Erel Levine
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Jessica M Oakes
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Eno E Ebong
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States; Department of Bioengineering, Northeastern University, Boston, MA, United States; Department of Neuroscience, Albert Einstein College of Medicine, NY, New York, United States.
| |
Collapse
|
6
|
Jung B, Yagi H, Kuo A, Dorweiler TF, Aikawa M, Kasai T, Singh SA, Dannenberg AJ, Fu Z, Niaudet C, Smith LEH, Hla T. ApoM-bound S1P acts via endothelial S1PR1 to suppress choroidal neovascularization and vascular leakage. Angiogenesis 2025; 28:24. [PMID: 40266369 PMCID: PMC12018641 DOI: 10.1007/s10456-025-09975-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/13/2025] [Indexed: 04/24/2025]
Abstract
Neovascular age-related macular degeneration (nAMD) is a major cause of vision loss worldwide. Current standard of care is repetitive intraocular injections of vascular endothelial growth factor (VEGF) inhibitors, although responses may be partial and non-durable. We report that circulating sphingosine 1-phosphate (S1P) carried by apolipoprotein M (ApoM) acts through the endothelial S1P receptor 1 (S1PR1) to suppress choroidal neovascularization (CNV) in mouse laser-induced CNV, modeling nAMD. In humans, low plasma ApoM levels were associated with increased choroidal and retinal pathology. Additionally, endothelial S1pr1 knockout and overexpressing transgenic mice showed increased and reduced CNV lesion size, respectively. Systemic administration of ApoM-Fc, an engineered S1P chaperone protein, not only attenuated CNV to an equivalent degree as anti-VEGF antibody treatment but also suppressed pathological vascular leakage. We suggest that modulating circulating ApoM-bound S1P action on endothelial S1PR1 provides a novel therapeutic strategy to treat nAMD.
Collapse
Affiliation(s)
- Bongnam Jung
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Hitomi Yagi
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Andrew Kuo
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Tim F Dorweiler
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Taku Kasai
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Colin Niaudet
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA.
- CRCI2NA, INSERM, CNRS, Nantes University, 44000, Nantes, France.
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
7
|
Alidjan FM, Hoedt SD, Rashid M, Zee-van Vark LCVD, Voortman GJ, Dorst-Lagerwerf KY, Christoffersen C, Lafeber M, Roeters van Lennep JE, Verhoeven AJM, Friesema ECH, Mulder MT. Triglyceride-rich lipoprotein sphingolipids are altered in primary hypertension: A pilot case-control study. J Clin Lipidol 2025:S1933-2874(25)00067-4. [PMID: 40263067 DOI: 10.1016/j.jacl.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Sphingolipids modulate vascular function and alterations in plasma sphingolipid profiles have been associated with hypertension. Plasma sphingolipids, such as ceramides (Cer) and sphingosine-1-phosphate (S1P), are predominantly carried by lipoproteins. OBJECTIVE We compared sphingolipid profiles in plasma and isolated lipoproteins of patients with primary hypertension with those of normotensive controls. METHODS Blood was obtained from 19 patients with hypertension and 19 age- and sex-matched normotensive controls. S1P and the 7 most abundant Cer were quantified by liquid chromatography-tandem mass spectrometry in plasma and in lipoproteins. RESULTS Total plasma Cer were significantly higher in patients with hypertension compared to controls (14.3 ± 1.0 vs 11.9 ± 0.7 µM; P = .047), while there were no differences in plasma S1P levels (1.8 ± 0.1 vs 2.1 ± 0.1 µM; P = .128). Total Cer carried by patient triglyceride-rich lipoproteins (TRL; ie, predominantly very low-density lipoproteins) were also significantly higher (1.33 ± 0.15 vs 0.58 ± 0.10 µM; P = .001), which held for all Cer tested. Systolic blood pressure positively correlated with plasma levels of Cer(d18:1/20:0) and Cer(d18:1/24:1), and diastolic blood pressure positively correlated with total Cer, Cer(d18:1/18:0), Cer(d18:1/20:0) and Cer(d18:1/24:0). Relative to plasma Cer(d18:1/24:0), levels of Cer(d18:1/18:0), Cer(d18:1/20:0) and Cer(d18:1/24:1) were significantly higher in patients with hypertension than in controls. CONCLUSION Patients with hypertension display higher plasma Cer levels than normotensive controls, which is mainly explained by elevated concentrations in TRLs. Cer levels positively correlate with systolic and diastolic blood pressure, and ratios of Cer relative to Cer(d18:1/24:0) suggest an increased cardiovascular risk.
Collapse
Affiliation(s)
- Fazil M Alidjan
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder); Department of Internal Vascular Medicine, Haga Hospital, the Hague, the Netherlands (Dr Alidjan)
| | - Sandra den Hoedt
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| | - Mardin Rashid
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| | - Leonie C van der Zee-van Vark
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| | - Gardi J Voortman
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| | - Kristien Y Dorst-Lagerwerf
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet and Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (Dr Christoffersen)
| | - Melvin Lafeber
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder); Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands (Dr Lafeber)
| | - Jeanine E Roeters van Lennep
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| | - Adrie J M Verhoeven
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| | - Edith C H Friesema
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder).
| | - Monique T Mulder
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands (Drs Alidjan, Hoedt, Rashid, Zee-van Vark, Voortman. Dorst-Lagerwerf, Lafeber, Roeters van Lennep, Verhoeven, Friesema and Mulder)
| |
Collapse
|
8
|
Vincent V, Thakkar H, Sen A, Bansal A, Das US, Gunasekaran A, Bhatla N, Velpandian T, Singh A. Adiponectin mediated metabolic and sphingolipid alterations in preventing endothelial dysfunction. Mol Cell Biochem 2025:10.1007/s11010-025-05268-1. [PMID: 40140228 DOI: 10.1007/s11010-025-05268-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 03/22/2025] [Indexed: 03/28/2025]
Abstract
Endothelial dysfunction is an early indicator of atherosclerosis. Adiponectin, a hormone secreted by adipose tissue with insulin-sensitizing and anti-inflammatory properties, offers protection against atherosclerosis. This study investigated the metabolic and sphingolipid alterations in endothelial cells linked to the protective effects of adiponectin against endothelial dysfunction. Human Umbilical Endothelial Cells (HUVECs) were treated with Tumor Necrosis Factor-alpha (TNF-α) to induce endothelial dysfunction. AdipoRon and SKI-I were used to study the effects of adiponectin and sphingosine kinase inhibition in HUVECs. Metabolic changes and sphingolipid alterations were assessed to understand changes in lipid metabolism, and RNA sequencing was used to quantify the transcriptomics changes. TNF-α treatment significantly upregulated glycolysis and downregulated long-chain fatty acid oxidation and mitochondrial ATP production, while AdipoRon co-treatment partially reversed these metabolic effects. In HUVECs, TNF-α treatment increased intracellular C16 and C18 ceramides and Sphingosine 1-Phosphate (S1P) while decreasing extracellular S1P. AdipoRon Co-treatment reversed these effects; AdipoRon also reversed the transcriptional changes induced by TNF-α. Sphingosine kinase inhibition in HUVECs led to mitochondrial dysfunction at the metabolic and transcriptional levels. This study provides insights into potential therapeutic strategies targeting endothelial metabolism while unraveling a novel mitochondrial modulation mediated by sphingosine kinases in endothelial cells.
Collapse
Affiliation(s)
- Vinnyfred Vincent
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Himani Thakkar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Atanu Sen
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Ashutosh Bansal
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Ujjalkumar Subhash Das
- Ocular Pharmacology and Pharmacy Division, Dr. RP Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Abishek Gunasekaran
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Neerja Bhatla
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Thirumurthy Velpandian
- Ocular Pharmacology and Pharmacy Division, Dr. RP Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Archna Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
9
|
Bielawiec P, Harasim-Symbor E, Gołaszewska K, Chabowski A, Hodun K, Sztolsztener K. Apolipoprotein and sphingolipid measurements: Can be used in the clinical practice of atrial fibrillation diagnosing and evaluating the cryoablation effectiveness? PLoS One 2025; 20:e0315905. [PMID: 40036186 PMCID: PMC11878926 DOI: 10.1371/journal.pone.0315905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/29/2024] [Indexed: 03/06/2025] Open
Abstract
Atrial fibrillation (AF) has become the most common arrhythmia of clinical importance. A well-established and recommended therapeutic option for AF is the balloon-based cryoablation (CBA) method. There are still no sensitive biomarkers for AF prediction and cryoablation effectiveness assessment, therefore in our prospective study, we examined the plasma content of apolipoproteins (Apo) and sphingolipids, as well as the distribution of selected sphingolipids among lipoprotein fractions. The study included 33 patients with AF on admission and 24 h after cryoablation therapy, while 20 healthy volunteers were recruited to the control group. Plasma Apo concentrations were determined using a multiplex assay kit measuring fluorescence signal, whereas the high-performance liquid chromatography (HPLC) method was applied to assess the total plasma sphingolipid levels as well as their content in isolated lipoprotein fractions. Our results showed that cryoballoon ablation in AF patients markedly reduced the level of almost all Apo compared to the individuals from the control and Pre-CBA groups (Apo-A1: -25.9% and -20.0%, Apo-A2: -19.9% and -17.3%, Apo-B: -26.8% and -14.4%, Apo-C1: -20.3% and -13.4%, Apo-D: -15.9% and -22.2%, Apo-E: -18.3% and -14.3%, and Apo-J: -36.4% and -21.5%, p < 0.05, respectively). Importantly, the area under the curve of Apo-J (AUC 0.81; 95% CI, 0.71-0.92) indicates that it might be a useful biomarker of cryotherapy success in AF patients. Moreover, we also observed a pronounced increase in sphinganine (Sa; +33.5%), sphingosine (So; +24.6%), sphinganine-1-phosphate (Sa1P; +34.3%), and sphingosine-1-phosphate (So1P; +22.3%) concentrations in the Pre-CBA group in comparison with controls. This is the first study that evaluates such a broad panel of Apo and sphingolipids in patients with AF undergoing the CBA procedure, however, to confirm whether any of these parameters could be a clinically useful biomarker for predicting AF or assessing the effectiveness of treatment, further research will be necessary due to limitations of the study.
Collapse
Affiliation(s)
- Patrycja Bielawiec
- Department of Physiology, Medical University of Bialystok, Białystok, Poland
| | - Ewa Harasim-Symbor
- Department of Physiology, Medical University of Bialystok, Białystok, Poland
| | - Karolina Gołaszewska
- Department of Cardiology, Ministry of Interior and Administration Hospital in Bialystok, Białystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Białystok, Poland
| | - Katarzyna Hodun
- Department of Physiology, Medical University of Bialystok, Białystok, Poland
| | | |
Collapse
|
10
|
Kim D, Lee JW, Kim YT, Choe J, Kim G, Ha CM, Kim JG, Song KH, Yang S. Minimally Invasive Syringe-Injectable Hydrogel with Angiogenic Factors for Ischemic Stroke Treatment. Adv Healthc Mater 2025; 14:e2403119. [PMID: 39520382 PMCID: PMC11874675 DOI: 10.1002/adhm.202403119] [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: 09/13/2024] [Revised: 10/06/2024] [Indexed: 11/16/2024]
Abstract
Ischemic stroke (IS) accounts for most stroke incidents and causes intractable damage to brain tissue. This condition manifests as diverse aftereffects, such as motor impairment, emotional disturbances, and dementia. However, a fundamental approach to curing IS remains unclear. This study proposes a novel approach for treating IS by employing minimally invasive and injectable jammed gelatin-norbornene nanofibrous hydrogels (GNF) infused with growth factors (GFs). The developed GNF/GF hydrogels are administered to the motor cortex of a rat IS model to evaluate their therapeutic effects on IS-induced motor dysfunction. GNFs mimic a natural fibrous extracellular matrix architecture and can be precisely injected into a targeted brain area. The syringe-injectable jammed nanofibrous hydrogel system increased angiogenesis, inflammation, and sensorimotor function in the IS-affected brain. For clinical applications, the biocompatible GNF hydrogel has the potential to efficiently load disease-specific drugs, enabling targeted therapy for treating a wide range of neurological diseases.
Collapse
Affiliation(s)
- Donggue Kim
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
| | - Ji Woo Lee
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
| | - Yang Tae Kim
- Division of Life SciencesCollege of Life Sciences and BioengineeringIncheon National UniversityIncheon22012Republic of Korea
| | - Junhyeok Choe
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
| | - Gaeun Kim
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
| | - Chang Man Ha
- Research Division and Brain Research Core Facilities of Korea Brain Research InstituteDaegu41068Republic of Korea
| | - Jae Geun Kim
- Division of Life SciencesCollege of Life Sciences and BioengineeringIncheon National UniversityIncheon22012Republic of Korea
- Research Center of Brain‐Machine InterfaceIncheon National UniversityIncheon22012Republic of Korea
| | - Kwang Hoon Song
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
- Research Center of Brain‐Machine InterfaceIncheon National UniversityIncheon22012Republic of Korea
| | - Sunggu Yang
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
- Research Center of Brain‐Machine InterfaceIncheon National UniversityIncheon22012Republic of Korea
- gBrain Inc.Incheon21984Republic of Korea
| |
Collapse
|
11
|
Rubinelli L, Manzo OL, Sungho J, Del Gaudio I, Bareja R, Marino A, Palikhe S, Di Mauro V, Bucci M, Falcone DJ, Elemento O, Ersoy B, Diano S, Sasset L, Di Lorenzo A. Suppression of endothelial ceramide de novo biosynthesis by Nogo-B contributes to cardiometabolic diseases. Nat Commun 2025; 16:1968. [PMID: 40000621 PMCID: PMC11862206 DOI: 10.1038/s41467-025-56869-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Accrual of ceramides, membrane and bioactive sphingolipids, has been implicated in endothelial dysfunction preceding cardiometabolic diseases. Yet, direct in vivo evidence, underlying mechanisms, and pathological implications are lacking. Here we show that suppression of ceramides and sphingosine-1-phosphate (S1P), a product of ceramide degradation, are causally linked to endothelial dysfunction and activation, contributing to vascular and metabolic disorders in high fat diet fed (HFD) male mice. Mechanistically, the upregulation of Nogo-B and ORMDL proteins suppress ceramide de novo biosynthesis in endothelial cells (EC) of HFD mice, resulting in vascular and metabolic dysfunctions. Systemic and endothelial specific deletion of Nogo-B restore sphingolipid signaling and functions, lowers hypertension, and hepatic glucose production in HFD. Our results demonstrate in vivo that ceramide and S1P suppression rather than accrual contributes to endothelial dysfunction and cardiometabolic diseases in HFD mice. Our study also sets a framework for the development of therapeutic strategies to treat these conditions.
Collapse
Affiliation(s)
- Luisa Rubinelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Onorina Laura Manzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Jin Sungho
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, USA
| | - Ilaria Del Gaudio
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Rohan Bareja
- The Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, NY, USA
| | - Alice Marino
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Sailesh Palikhe
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Vittoria Di Mauro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Domenick J Falcone
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Olivier Elemento
- The Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, NY, USA
| | - Baran Ersoy
- Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sabrina Diano
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, USA
| | - Linda Sasset
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
12
|
Zhao F, Shao M, Li M, Li T, Zheng Y, Sun W, Ni C, Li L. Sphingolipid metabolites involved in the pathogenesis of atherosclerosis: perspectives on sphingolipids in atherosclerosis. Cell Mol Biol Lett 2025; 30:18. [PMID: 39920588 PMCID: PMC11804087 DOI: 10.1186/s11658-024-00679-2] [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: 07/29/2024] [Accepted: 12/17/2024] [Indexed: 02/09/2025] Open
Abstract
Atherosclerosis, with its complex pathogenesis, is a leading underlying cause of many cardiovascular diseases, which are increasingly prevalent in the population. Sphingolipids play an important role in the development of atherosclerosis. Key metabolites and enzymes in sphingolipid metabolism influence the pathogenesis of atherosclerosis in a variety of ways, including inflammatory responses and oxidative stress. Thus, an investigation of sphingolipid metabolism-related metabolites and key enzymes may provide novel insights and treatment targets for atherosclerosis. This review discusses various mechanisms and research progress on the relationship between various sphingolipid metabolites, related enzymes, and atherosclerosis. Finally, we look into the future research direction of phytosphingolipids.
Collapse
Affiliation(s)
- Fufangyu Zhao
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Mingyan Shao
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Mingrui Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tianxing Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yanfei Zheng
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Wenlong Sun
- Institute of Biomedical Research, School of Life Sciences, Shandong University of Technology, Zibo, 255000, Shandong, China.
| | - Cheng Ni
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Lingru Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
13
|
Xiong Y, Ye Q, Liu L, Lin W, Liao Y, Gao R, Xu J, Zhang X, Chen R, Chen S, Chen Q, Wei L. The compensatory enrichment of sphingosine-1-phosphate on HDL in FSGS enhances the protective function of glomerular endothelial cells compared to MCD. Sci Rep 2025; 15:1530. [PMID: 39789110 PMCID: PMC11718056 DOI: 10.1038/s41598-025-85865-8] [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: 07/15/2024] [Accepted: 01/07/2025] [Indexed: 01/12/2025] Open
Abstract
Glomerular endothelial cells (GECs) are pivotal in developing glomerular sclerosis disorders. The advancement of focal segmental glomerulosclerosis (FSGS) is intimately tied to disruptions in lipid metabolism. Sphingosine-1-phosphate (S1P), a molecule transported by high-density lipoproteins (HDL), exhibits protective effects on vascular endothelial cells by upregulating phosphorylated endothelial nitric oxide synthase (p-eNOS) and enhancing nitric oxide (NO) production. Nevertheless, the abundance of S1P within HDL in individuals with FSGS and minimal change disease (MCD) is yet to be elucidated, and its defensive role in GECs necessitates empirical confirmation. A total of 14 FSGS patients, 16 MCD patients, and 16 healthy controls (NC) were included in the study, with FSGS and MCD confirmed by renal biopsy. After blood sample collection, HDL was isolated and categorized into intact HDL, phospholipid-depleted HDL(apo-HDL), phospholipid-remained HDL(phoHDL), and recombinant HDL (rHDL). Various HDL samples, comprising intact, apo-HDL, pho-HDL and rHDL, were co-cultivated with human renal glomerular endothelial cells (HRGECs). Western blotting was utilized to quantify p-eNOS levels and assess PI3K-AKT pathway activation. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) analyzed S1P concentrations, while real-time quantitative PCR evaluated the expression of enzymes involved in S1P metabolism. Fluorescence labeling methods measured NO levels, and an immunofluorescence colocalization assay investigated Sphingosine-1-phosphate receptor 1 (S1PR1) expression in GECs across distinct kidney tissue groups. The HDL from FSGS patients demonstrated a significantly enhanced ability to promote p-eNOS expression and NO release in HRGECs compared to MCD patients and healthy controls. Additionally, the synthesis activity of S1P in renal tissues of FSGS patients was markedly higher than that observed in MCD patients and healthy controls, suggesting that S1P may play a crucial protective role in the progression of FSGS. Immunofluorescence staining showed that compared with MCD and NC, the expression of S1PR1 in GECs of FSGS patients was significantly decreased. Recombinant HDL with added S1P promoted the increase of p-eNOS in HRGECs. Knockdown of S1PR1 using siRNA reduced the expression of p-eNOS and NO release. The mechanism underlying the regulation of p-eNOS expression by rHDL was associated with the PI3K-AKT signaling pathway. The enhanced presence of S1P on HDL could serve as a diagnostic marker to differentiate FSGS from MCD. Incorporating S1P into HDL enhances glomerular endothelial cell function, suggesting that the S1P/S1PR pathway might offer a promising therapeutic avenue for FSGS.
Collapse
Affiliation(s)
- Yunfeng Xiong
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Qiuping Ye
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
- Fujian Institute of Clinical Immunology, Fuzhou, 350001, China
| | - Lifang Liu
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
- Fujian Institute of Clinical Immunology, Fuzhou, 350001, China
| | - Wanjun Lin
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
- Fujian Institute of Clinical Immunology, Fuzhou, 350001, China
| | - Yonggen Liao
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
- Fujian Institute of Clinical Immunology, Fuzhou, 350001, China
| | - Ruiyu Gao
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Jiaming Xu
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Xinyu Zhang
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Ruoyan Chen
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Sihui Chen
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Qiaoling Chen
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
- Fujian Institute of Clinical Immunology, Fuzhou, 350001, China.
| | - Lixin Wei
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
- Fujian Institute of Clinical Immunology, Fuzhou, 350001, China.
| |
Collapse
|
14
|
Potì F, Scalera E, Feuerborn R, Fischer J, Arndt L, Varga G, Pardali E, Seidl MD, Fobker M, Liebisch G, Hesse B, Lukasz AH, Rossaint J, Kehrel BE, Rosenbauer F, Renné T, Christoffersen C, Simoni M, Burkhardt R, Nofer JR. Sphingosine 1-phosphate receptor 1signaling in macrophages reduces atherosclerosis in LDL receptor-deficient mice. JCI Insight 2024; 9:e158127. [PMID: 39531328 PMCID: PMC11665566 DOI: 10.1172/jci.insight.158127] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Sphingosine 1-phosphate (S1P) is a lysosphingolipid with antiatherogenic properties, but mechanisms underlying its effects remain unclear. We here investigated atherosclerosis development in cholesterol-rich diet-fed LDL receptor-deficient mice with high or low overexpression levels of S1P receptor 1 (S1P1) in macrophages. S1P1-overexpressing macrophages showed increased activity of transcription factors PU.1, interferon regulatory factor 8 (IRF8), and liver X receptor (LXR) and were skewed toward an M2-distinct phenotype characterized by enhanced production of IL-10, IL-1RA, and IL-5; increased ATP-binding cassette transporter A1- and G1-dependent cholesterol efflux; increased expression of MerTK and efferocytosis; and reduced apoptosis due to elevated B cell lymphoma 6 and Maf bZIP B. A similar macrophage phenotype was observed in mice administered S1P1-selective agonist KRP203. Mechanistically, the enhanced PU.1, IRF8, and LXR activity in S1P1-overexpressing macrophages led to downregulation of the cAMP-dependent PKA and activation of the signaling cascade encompassing protein kinases AKT and mTOR complex 1 as well as the late endosomal/lysosomal adaptor MAPK and mTOR activator 1. Atherosclerotic lesions in aortic roots and brachiocephalic arteries were profoundly or moderately reduced in mice with high and low S1P1 overexpression in macrophages, respectively. We conclude that S1P1 signaling polarizes macrophages toward an antiatherogenic functional phenotype and countervails the development of atherosclerosis in mice.
Collapse
Affiliation(s)
- Francesco Potì
- Unit of Neuroscience, Department of Medicine and Surgery, University of Parma, Parma, Italy
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy
| | - Enrica Scalera
- Department of Food and Drug, University of Parma, Parma, Italy
- Central Laboratory Facility, University Hospital Münster, Münster, Germany
| | - Renata Feuerborn
- Central Laboratory Facility, University Hospital Münster, Münster, Germany
| | - Josephine Fischer
- Institute of Molecular Tumor Biology, University of Münster, Münster, Germany
| | - Lilli Arndt
- Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Germany
| | - Georg Varga
- Department of Pediatric Rheumatology and Immunology, University Children’s Hospital Münster, Münster, Germany
| | - Evangelia Pardali
- Department of Cardiology, University Hospital Münster, Münster, Germany
- Pharvaris GmbH, Zug, Switzerland
| | - Matthias D. Seidl
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Manfred Fobker
- Central Laboratory Facility, University Hospital Münster, Münster, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Bettina Hesse
- Division of General Internal Medicine, Nephrology, and Rheumatology, Department of Medicine D, and
| | - Alexander H. Lukasz
- Division of General Internal Medicine, Nephrology, and Rheumatology, Department of Medicine D, and
| | - Jan Rossaint
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Beate E. Kehrel
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Frank Rosenbauer
- Institute of Molecular Tumor Biology, University of Münster, Münster, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, and Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Jerzy-Roch Nofer
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy
- Central Laboratory Facility, University Hospital Münster, Münster, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute for Laboratory Medicine, Marien-Hospital, Niels-Stensen-Kliniken, Osnabrück, Germany
| |
Collapse
|
15
|
Del Gaudio I, Nitzsche A, Boyé K, Bonnin P, Poulet M, Nguyen TQ, Couty L, Ha HTT, Nguyen DT, Cazenave-Gassiot A, Ben Alaya K, Thérond P, Chun J, Wenk MR, Proia RL, Henrion D, Nguyen LN, Eichmann A, Camerer E. Zonation and ligand and dose dependence of sphingosine 1-phosphate receptor-1 signalling in blood and lymphatic vasculature. Cardiovasc Res 2024; 120:1794-1810. [PMID: 39086170 PMCID: PMC11587562 DOI: 10.1093/cvr/cvae168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/25/2024] [Accepted: 06/12/2024] [Indexed: 08/02/2024] Open
Abstract
AIMS Circulating levels of sphingosine 1-phosphate (S1P), an HDL-associated ligand for the endothelial cell (EC) protective S1P receptor-1 (S1PR1), are reduced in disease states associated with endothelial dysfunction. Yet, as S1PR1 has high affinity for S1P and can be activated by ligand-independent mechanisms and EC autonomous S1P production, it is unclear if relative reductions in circulating S1P can cause endothelial dysfunction. It is also unclear how EC S1PR1 insufficiency, whether induced by deficiency in circulating ligand or by S1PR1-directed immunosuppressive therapy, affects different vascular subsets. METHODS AND RESULTS We here fine map the zonation of S1PR1 signalling in the murine blood and lymphatic vasculature, superimpose cell-type-specific and relative deficiencies in S1P production to define ligand source and dose dependence, and correlate receptor engagement to essential functions. In naïve blood vessels, despite broad expression, EC S1PR1 engagement was restricted to resistance-size arteries, lung capillaries, and a subset of high-endothelial venules (HEVs). Similar zonation was observed for albumin extravasation in EC S1PR1-deficient mice, and brain extravasation was reproduced with arterial EC-selective S1pr1 deletion. In lymphatic ECs, S1PR1 engagement was high in collecting vessels and lymph nodes and low in blind-ended capillaries that drain tissue fluids. While EC S1P production sustained S1PR1 signalling in lymphatics and HEV, haematopoietic cells provided ∼90% of plasma S1P and sustained signalling in resistance arteries and lung capillaries. S1PR1 signalling and endothelial function were both surprisingly sensitive to reductions in plasma S1P with apparent saturation around 50% of normal levels. S1PR1 engagement did not depend on sex or age but modestly increased in arteries in hypertension and diabetes. Sphingosine kinase (Sphk)-2 deficiency also increased S1PR1 engagement selectively in arteries, which could be attributed to Sphk1-dependent S1P release from perivascular macrophages. CONCLUSION This study highlights vessel subtype-specific S1PR1 functions and mechanisms of engagement and supports the relevance of S1P as circulating biomarker for endothelial function.
Collapse
Affiliation(s)
- Ilaria Del Gaudio
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Anja Nitzsche
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Kevin Boyé
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Philippe Bonnin
- Physiologie Clinique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Lariboisière, Paris, France
- Université Paris Cité, INSERM U1144, UFR de Pharmacie, Paris, France
| | - Mathilde Poulet
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Toan Q Nguyen
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Ludovic Couty
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Hoa T T Ha
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Dat T Nguyen
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Amaury Cazenave-Gassiot
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Khaoula Ben Alaya
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Patrice Thérond
- Service de Biochimie, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital de Bicêtre, Le Kremlin Bicêtre, France
- UFR de Pharmacie, EA 4529, Châtenay-Malabry, France
| | - Jerold Chun
- Neuroscience Drug Discovery, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Markus R Wenk
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Richard L Proia
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Institutes of Health, Bethesda, MD, USA
| | - Daniel Henrion
- MitoVasc Department, Angers University, Team 2 (CarMe), Angers University Hospital (CHU of Angers), CNRS, INSERM U1083, Angers, France
| | - Long N Nguyen
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Anne Eichmann
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
- Department of Internal Medicine and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, USA
| | - Eric Camerer
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| |
Collapse
|
16
|
Seidita I, Ziegler A, Qalaj A, Winkler MS, Nierhaus A, Kluge S, Levkau B, Gräler MH. Rapid determination of sphingosine 1-phosphate association with carrier molecules by flow-induced dispersion analysis to predict sepsis outcome. iScience 2024; 27:111168. [PMID: 39524325 PMCID: PMC11544382 DOI: 10.1016/j.isci.2024.111168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/26/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Flow-induced dispersion analysis (FIDA) was used to investigate the association of fluorescein isothiocyanate-labeled signaling lipid sphingosine 1-phosphate (S1P) with its carrier molecules human serum albumin (HSA) and high-density lipoprotein (HDL). Associations were measured in plasma samples of patients after surgery, with sepsis or septic shock. All patients demonstrated a significant shift between the carrier binding: decrease of S1P bound to HSA with a concomitant increase of S1P bound to HDL. The molecular sizes of binding complexes correlated well with the relative amounts of S1P bound to HSA and HDL detected by liquid chromatography-tandem mass spectrometry. Very low complex formation of S1P with HDL was observed in several septic shock patients and correlated with the need for mechanical ventilation and intensive care unit (ICU) mortality. Determination of S1P binding to HSA and HDL by FIDA could therefore be useful in the clinical setting to predict disease progression, severity, and outcome.
Collapse
Affiliation(s)
- Isabelle Seidita
- Department of Anesthesiology and Intensive Care Medicine, Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knöll-Straße 2, D-07745 Jena, Germany
| | - Anke Ziegler
- Department of Anesthesiology and Intensive Care Medicine, Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knöll-Straße 2, D-07745 Jena, Germany
| | - Auron Qalaj
- Department of Anesthesiology and Intensive Care Medicine, Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knöll-Straße 2, D-07745 Jena, Germany
| | - Martin Sebastian Winkler
- Department of Anesthesiology, Emergency and Intensive Care Medicine, University of Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Axel Nierhaus
- Department of Intensive Care, Universitätsklinikum Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Stefan Kluge
- Department of Intensive Care, Universitätsklinikum Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Bodo Levkau
- Institute of Molecular Medicine III, Heinrich Heine University, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Markus H. Gräler
- Department of Anesthesiology and Intensive Care Medicine, Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knöll-Straße 2, D-07745 Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07740 Jena, Germany
| |
Collapse
|
17
|
Mitra R, Pentland K, Kolev S, Eden M, Levine E, Oakes JM, Ebong EE. Co-Therapy with S1P and Heparan Sulfate Derivatives to Restore Endothelial Glycocalyx and Combat Pro-Atherosclerotic Endothelial Dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.06.622347. [PMID: 39574692 PMCID: PMC11581019 DOI: 10.1101/2024.11.06.622347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Endothelial cell (EC) glycocalyx (GCX) shedding due to disturbed blood flow and chemical factors leads to low-density lipoprotein infiltration and reduced nitric oxide synthesis, causing vascular dysfunction and atherosclerosis. This study evaluates a novel therapy combining sphingosine-1-phosphate (S1P) and heparin (heparan sulfate derivative). We hypothesized that heparin/S1P would repair mechanically damaged EC GCX in disturbed flow (DF) regions and restore anti-atherosclerotic mechanotransduction function, addressing cardiovascular disease. We used a parallel-plate flow chamber to simulate flow conditions in vitro and a partial carotid ligation mouse model to mimic DF in vivo. Heparin and albumin-bound S1P were administered to assess their reparative effects on the endothelial GCX. Immunocytochemistry, fluorescent staining, confocal microscopy, cellular alignment studies, and ultrasound were performed to evaluate EC function and endothelial-dependent vascular function. Barrier functionality was assessed via macrophage uptake. Heparin/S1P mechanism-of-action insights were gained through fluid dynamics simulations and staining of GCX synthesis enzyme as well as S1P receptor. Statistical analyses validated results. In vitro data showed that heparin/S1P therapy improves the function of DF-conditioned ECs by restoring EC GCX and promoting EC alignment and elevated vasodilator eNOS (endothelial-type nitric oxide synthase) expression. The in vivo studies confirmed GCX degradation, increased vessel inflammation and hyperpermeability, and vessel wall thickening in the partially ligated left carotid artery. Heparin/S1P treatment restored GCX in the left carotid artery, enhancing GCX thickness and coverage of the blood vessel wall. This work advances a new approach to regenerating the EC GCX and restoring its function in ECs under DF conditions.
Collapse
|
18
|
Silvain J, Materne C, Zeitouni M, Procopi N, Guedeney P, Brugier D, Galier S, Lhomme M, Ponnaiah M, Guillas I, Kc P, Dahik VD, Frisdal E, Vicaut E, Lesnik P, Rahoual G, Le Goff W, Montalescot G, Kerneis M, Guerin M. Defective biological activities of high-density lipoprotein identify patients at highest risk of recurrent cardiovascular event. Eur J Prev Cardiol 2024:zwae356. [PMID: 39506545 DOI: 10.1093/eurjpc/zwae356] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/07/2024] [Accepted: 11/05/2024] [Indexed: 11/08/2024]
Abstract
AIMS Low cholesterol efflux capacity and elevated levels of Interleukin-1ß (IL-1ß) are both associated with residual cardiovascular risk in patients with acute myocardial infarction (MI) and may be used as new biomarkers to identify patients at higher cardiovascular risk. METHODS We evaluated potential synergetic effect of cholesterol efflux capacity and IL-1ß on recurrent major adverse cardiovascular events (MACE) at one-year in 2012 patients with acute ST- segment elevation MI who underwent primary percutaneous coronary intervention. In addition, we evaluated the contribution to residual risk of HDL biological functions from 20 patients of the two extreme subgroups, focusing on cholesterol efflux capacity and anti-inflammatory properties. RESULTS Patients with MACE during the first year after the MI had significantly lower serum cholesterol efflux capacity as compared to those without recurrent events and higher level of IL-1ß, both associations were confirmed after multivariate analysis. We found an inverse relationship between CEC and circulating levels of the inflammatory markers IL-1ß, defining a very high risk (Low CEC/High IL-1ß) and a low risk (High CEC/Low IL-1ß) group of patients. Patients combining Low CEC/High IL-1ß exhibited the highest risk of recurrent MACE at one year showing an additive prognostic value of these biomarkers, regardless of all the other clinical or biological factors. In this very high-risk subgroup, patients exhibited reduced HDL-efflux capacity and defective ABCA1 and SR-BI with enhanced pro-inflammatory activity as a potential explanation for our clinical findings. CONCLUSION Impaired cholesterol efflux capacity and elevated IL-1β synergistically increase the residual cardiovascular risk in MI patients, which could be explained by reduced HDL-efflux capacity and enhanced HDL pro-inflammatory activity.
Collapse
Affiliation(s)
- Johanne Silvain
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Clément Materne
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Michel Zeitouni
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Niki Procopi
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Paul Guedeney
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Delphine Brugier
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Sophie Galier
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Marie Lhomme
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN, ICAN OMICS and ICAN I/O), F-75013 Paris, France
| | - Maharajah Ponnaiah
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN, ICAN OMICS and ICAN I/O), F-75013 Paris, France
| | - Isabelle Guillas
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Pukar Kc
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Veronica D Dahik
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Eric Frisdal
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Eric Vicaut
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Unité de Recherche Clinique, Hôpital Fernand Widal (AP-HP), Paris, France. SAMM - Statistique, Analyse et Modélisation Multidisciplinaire EA 4543, Université Paris 1 Panthéon Sorbonne, France
| | - Philippe Lesnik
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Ghilas Rahoual
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
| | - Wilfried Le Goff
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Gilles Montalescot
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Mathieu Kerneis
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Maryse Guerin
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| |
Collapse
|
19
|
Abstract
Cardiovascular disease is the leading cause of death worldwide, and it commonly results from atherosclerotic plaque progression. One of the increasingly recognized drivers of atherosclerosis is dysfunctional efferocytosis, a homeostatic mechanism responsible for the clearance of dead cells and the resolution of inflammation. In atherosclerosis, the capacity of phagocytes to participate in efferocytosis is hampered, leading to the accumulation of apoptotic and necrotic tissue within the plaque, which results in enlargement of the necrotic core, increased luminal stenosis and plaque inflammation, and predisposition to plaque rupture or erosion. In this Review, we describe the different forms of programmed cell death that can occur in the atherosclerotic plaque and highlight the efferocytic machinery that is normally implicated in cardiovascular physiology. We then discuss the mechanisms by which efferocytosis fails in atherosclerosis and other cardiovascular and cardiometabolic diseases, including myocardial infarction and diabetes mellitus, and discuss therapeutic approaches that might reverse this pathological process.
Collapse
Affiliation(s)
- Shaunak S Adkar
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Nicholas J Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford, CA, USA.
| |
Collapse
|
20
|
Jones Q, Zheng J, Li Z, He M, Li X, Dai K, Worgall TS, Yu Y, Jiang XC. Effect of phospholipid transfer protein on plasma sphingosine-1-phosphate. J Biol Chem 2024; 300:107837. [PMID: 39343001 PMCID: PMC11532956 DOI: 10.1016/j.jbc.2024.107837] [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: 07/10/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Plasma phospholipid transfer protein (PLTP) is a risk factor for cardiovascular diseases. Sphingosine-1-phosphate (S1P), carried by high-density lipoprotein (HDL), is a potent lipid mediator and is also associated with cardiovascular diseases. We found that germline Pltp gene knockout (KO) mice have decreased circulating S1P without influencing apoM, a major S1P carrier on HDL. We then hypothesized that, like apoM, PLTP is another S1P carrier. We established inducible Pltp-KO, Apom-KO, and Pltp/Apom double KO mice and measured plasma lipoprotein and S1P levels under different diets. We found that PLTP deficiency, and the double deficiency have a similar effect on HDL reduction. Importantly, we found that all mice have about 50% reduction in plasma S1P levels, compared to WT mice, and PLTP deficiency significantly reduces apoM levels (about 40%), while apoM deficiency has no effect on PLTP activity, indicating that PLTP depletion reduces S1P through HDL reduction. To further evaluate this HDL reduction-mediated effect, we overexpressed PLTP which also caused a reduction of HDL. We found that the overexpression reduces S1P and apoM as well as apoA-I, a major apolipoprotein on HDL. Furthermore, we found that albumin (another reported S1P carrier) deficiency in mice has no effect on plasma S1P. We also found that the influence of PLTP on HDL may not require its direct binding to the particle. In conclusion, PLTP is not a direct S1P carrier. PLTP depletion or overexpression in adulthood dramatically reduces plasma S1P through HDL reduction. ApoM, but not albumin, deficiency reduces plasma S1P levels.
Collapse
Affiliation(s)
- Quiana Jones
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Jiao Zheng
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Zhiqiang Li
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Mulin He
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Xiang Li
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Kezhi Dai
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Tilla S Worgall
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA
| | - Yang Yu
- School of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, New York, USA.
| |
Collapse
|
21
|
Phan F, Bourron O, Foufelle F, Le Stunff H, Hajduch E. Sphingosine-1-phosphate signalling in the heart: exploring emerging perspectives in cardiopathology. FEBS Lett 2024; 598:2641-2655. [PMID: 38965662 DOI: 10.1002/1873-3468.14973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/23/2024] [Accepted: 06/12/2024] [Indexed: 07/06/2024]
Abstract
Cardiometabolic disorders contribute to the global burden of cardiovascular diseases. Emerging sphingolipid metabolites like sphingosine-1-phosphate (S1P) and its receptors, S1PRs, present a dynamic signalling axis significantly impacting cardiac homeostasis. S1P's intricate mechanisms extend to its transportation in the bloodstream by two specific carriers: high-density lipoprotein particles and albumin. This intricate transport system ensures the accessibility of S1P to distant target tissues, influencing several physiological processes critical for cardiovascular health. This review delves into the diverse functions of S1P and S1PRs in both physiological and pathophysiological conditions of the heart. Emphasis is placed on their diverse roles in modulating cardiac health, spanning from cardiac contractility, angiogenesis, inflammation, atherosclerosis and myocardial infarction. The intricate interplays involving S1P and its receptors are analysed concerning different cardiac cell types, shedding light on their respective roles in different heart diseases. We also review the therapeutic applications of targeting S1P/S1PRs in cardiac diseases, considering existing drugs like Fingolimod, as well as the prospects and challenges in developing novel therapies that selectively modulate S1PRs.
Collapse
Affiliation(s)
- Franck Phan
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Paris, France
- Diabetology Department, Assistance Publique-Hôpitaux de Paris (APHP), La Pitié-Salpêtrière-Charles Foix University Hospital, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Olivier Bourron
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Paris, France
- Diabetology Department, Assistance Publique-Hôpitaux de Paris (APHP), La Pitié-Salpêtrière-Charles Foix University Hospital, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Fabienne Foufelle
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Hervé Le Stunff
- Institut des Neurosciences Paris-Saclay, CNRS UMR 9197, Université Paris-Saclay, France
| | - Eric Hajduch
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| |
Collapse
|
22
|
Abstract
One hundred and fifty years ago, Johann Thudichum described sphingolipids as unusual "Sphinx-like" lipids from the brain. Today, we know that thousands of sphingolipid molecules mediate many essential functions in embryonic development and normal physiology. In addition, sphingolipid metabolism and signalling pathways are dysregulated in a wide range of pathologies, and therapeutic agents that target sphingolipids are now used to treat several human diseases. However, our understanding of sphingolipid regulation at cellular and organismal levels and their functions in developmental, physiological and pathological settings is rudimentary. In this Review, we discuss recent advances in sphingolipid pathways in different organelles, how secreted sphingolipid mediators modulate physiology and disease, progress in sphingolipid-targeted therapeutic and diagnostic research, and the trans-cellular sphingolipid metabolic networks between microbiota and mammals. Advances in sphingolipid biology have led to a deeper understanding of mammalian physiology and may lead to progress in the management of many diseases.
Collapse
Affiliation(s)
- Andrew Kuo
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
23
|
Foran D, Antoniades C, Akoumianakis I. Emerging Roles for Sphingolipids in Cardiometabolic Disease: A Rational Therapeutic Target? Nutrients 2024; 16:3296. [PMID: 39408263 PMCID: PMC11478599 DOI: 10.3390/nu16193296] [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: 08/08/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Cardiovascular disease is a leading cause of morbidity and mortality. New research elucidates increasingly complex relationships between cardiac and metabolic health, giving rise to new possible therapeutic targets. Sphingolipids are a heterogeneous class of bioactive lipids with critical roles in normal human physiology. They have also been shown to play both protective and deleterious roles in the pathogenesis of cardiovascular disease. Ceramides are implicated in dysregulating insulin signalling, vascular endothelial function, inflammation, oxidative stress, and lipoprotein aggregation, thereby promoting atherosclerosis and vascular disease. Ceramides also advance myocardial disease by enhancing pathological cardiac remodelling and cardiomyocyte death. Glucosylceramides similarly contribute to insulin resistance and vascular inflammation, thus playing a role in atherogenesis and cardiometabolic dysfunction. Sphingosing-1-phosphate, on the other hand, may ameliorate some of the pathological functions of ceramide by protecting endothelial barrier integrity and promoting cell survival. Sphingosine-1-phosphate is, however, implicated in the development of cardiac fibrosis. This review will explore the roles of sphingolipids in vascular, cardiac, and metabolic pathologies and will evaluate the therapeutic potential in targeting sphingolipids with the aim of prevention and reversal of cardiovascular disease in order to improve long-term cardiovascular outcomes.
Collapse
Affiliation(s)
| | | | - Ioannis Akoumianakis
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (D.F.); (C.A.)
| |
Collapse
|
24
|
Endo Y, Sasaki K, Ikewaki K. Bridging the Gap Between the Bench and Bedside: Clinical Applications of High-density Lipoprotein Function. J Atheroscler Thromb 2024; 31:1239-1248. [PMID: 38925924 PMCID: PMC11374562 DOI: 10.5551/jat.rv22020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Decades of research have reshaped our understanding of high-density lipoprotein (HDL) , shifting our focus from cholesterol (C) levels to multifaceted functionalities. Epidemiological studies initially suggested an association between HDL-C levels and cardiovascular disease (CVD) risk; however, such a simple association has not been indicated by recent studies. Notably, genome-wide studies have highlighted discrepancies between HDL-C levels and CVD outcomes, urging a deeper exploration of the role of HDL. The key to this shift lies in elucidating the role of HDL in reverse cholesterol transport (RCT), which is a fundamental anti-atherosclerotic mechanism. Understanding RCT has led to the identification of therapeutic targets and novel interventions for atherosclerosis. However, clinical trials have underscored the limitations of HDL-C as a therapeutic target, prompting the re-evaluation of the role of HDL in disease prevention. Further investigations have revealed the involvement of HDL composition in various diseases other than CVD, including chronic kidney disease, Alzheimer's disease, and autoimmune diseases. The anti-inflammatory, antioxidative, and anti-infectious properties of HDL have emerged as crucial aspects of its protective function, opening new avenues for novel biomarkers and therapeutic targets. Omics technologies have provided insights into the diverse composition of HDL, revealing disease-specific alterations in the HDL proteome and lipidome. In addition, combining cell-based and cell-free assays has facilitated the evaluation of the HDL functionality across diverse populations, offering the potential for personalized medicine. Overall, a comprehensive understanding of HDL multifunctionality leads to promising prospects for future clinical applications and therapeutic developments, extending beyond cardiovascular health.
Collapse
Affiliation(s)
- Yasuhiro Endo
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan
- Division of Environmental Medicine, National Defense Medical College Research Institute, Saitama, Japan
| | - Kei Sasaki
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Katsunori Ikewaki
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| |
Collapse
|
25
|
Poteryaeva ON, Usynin IF. Molecular mechanisms of the regulatory action of high-density lipoproteins on the endothelial function. BIOMEDITSINSKAIA KHIMIIA 2024; 70:206-217. [PMID: 39239895 DOI: 10.18097/pbmc20247004206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Endothelial dysfunction underlies the pathogenesis of many diseases, primarily cardiovascular diseases. Epidemiological studies have shown an inverse dependence between the plasma level of high-density lipoproteins (HDL) and cardiovascular diseases. The results of experimental studies indicate that the antiatherogenic effect of HDL is associated not only with their participation in the reverse transport of excess cholesterol, but also with their regulatory effect on the functions of cells of various organs and tissues, including endothelial cells. The purpose of this review is to consider recent data on the participation of plasma receptors and related intracellular signaling pathways in the mechanism of protective effect of HDL on endothelial cell functions. Understanding the mechanisms of cell function regulation under the influence of HDL is an important step for the development of new ways of pharmacological correction of impaired endothelial functions and creation of effective endothelial protection drugs.
Collapse
Affiliation(s)
- O N Poteryaeva
- Institute of Biochemistry, Federal Research Center of Fundamental and Translation Medicine, Novosibirsk, Russia
| | - I F Usynin
- Institute of Biochemistry, Federal Research Center of Fundamental and Translation Medicine, Novosibirsk, Russia
| |
Collapse
|
26
|
SenthilKumar G, Zirgibel Z, Cohen KE, Katunaric B, Jobe AM, Shult CG, Limpert RH, Freed JK. Ying and Yang of Ceramide in the Vascular Endothelium. Arterioscler Thromb Vasc Biol 2024; 44:1725-1736. [PMID: 38899471 PMCID: PMC11269027 DOI: 10.1161/atvbaha.124.321158] [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] [Indexed: 06/21/2024]
Abstract
Ceramides, a group of biologically active sphingolipids, have been described as the new cholesterol given strong evidence linking high plasma ceramide with endothelial damage, risk for early adverse cardiovascular events, and development of cardiometabolic disease. This relationship has sparked great interest in investigating therapeutic targets with the goal of suppressing ceramide formation. However, the growing data challenge this paradigm of ceramide as solely eliciting detrimental effects to the cardiovascular system. Studies show that ceramides are necessary for maintaining proper endothelial redox states, mechanosensation, and membrane integrity. Recent work in preclinical models and isolated human microvessels highlights that the loss of ceramide formation can in fact propagate vascular endothelial dysfunction. Here, we delve into these conflicting findings to evaluate how ceramide may be capable of exerting both beneficial and damaging effects within the vascular endothelium. We propose a unifying theory that while basal levels of ceramide in response to physiological stimuli are required for the production of vasoprotective metabolites such as S1P (sphingosine-1-phosphate), the chronic accumulation of ceramide can promote activation of pro-oxidative stress pathways in endothelial cells. Clinically, the evidence discussed here highlights the potential challenges associated with therapeutic suppression of ceramide formation as a means of reducing cardiovascular disease risk.
Collapse
Affiliation(s)
- Gopika SenthilKumar
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee WI
- Department of Physiology, Medical College of Wisconsin, Milwaukee WI
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee WI
| | - Zachary Zirgibel
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee WI
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee WI
| | - Katie E. Cohen
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee WI
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee WI
| | - Boran Katunaric
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee WI
- Department of Physiology, Medical College of Wisconsin, Milwaukee WI
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee WI
| | - Alyssa M. Jobe
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee WI
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee WI
| | - Carolyn G. Shult
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee WI
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee WI
| | - Rachel H. Limpert
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee WI
- Department of Physiology, Medical College of Wisconsin, Milwaukee WI
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee WI
| | - Julie K. Freed
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee WI
- Department of Physiology, Medical College of Wisconsin, Milwaukee WI
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee WI
| |
Collapse
|
27
|
Kravitz MS, Kattouf N, Stewart IJ, Ginde AA, Schmidt EP, Shapiro NI. Plasma for prevention and treatment of glycocalyx degradation in trauma and sepsis. Crit Care 2024; 28:254. [PMID: 39033135 PMCID: PMC11265047 DOI: 10.1186/s13054-024-05026-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 07/06/2024] [Indexed: 07/23/2024] Open
Abstract
The endothelial glycocalyx, a gel-like layer that lines the luminal surface of blood vessels, is composed of proteoglycans, glycoproteins, and glycosaminoglycans. The endothelial glycocalyx plays an essential role in vascular homeostasis, and its degradation in trauma and sepsis can lead to microvascular dysfunction and organ injury. While there are no proven therapies for preventing or treating endothelial glycocalyx degradation, some initial literature suggests that plasma may have a therapeutic role in trauma and sepsis patients. Overall, the literature suggesting the use of plasma as a therapy for endothelial glycocalyx degradation is non-clinical basic science or exploratory. Plasma is an established therapy in the resuscitation of patients with hemorrhage for restoration of coagulation factors. However, plasma also contains other bioactive components, including sphingosine-1 phosphate, antithrombin, and adiponectin, which may protect and restore the endothelial glycocalyx, thereby helping to maintain or restore vascular homeostasis. This narrative review begins by describing the endothelial glycocalyx in health and disease: we discuss the overlapping disease mechanisms in trauma and sepsis that lead to its damage and introduce plasma transfusion as a potential therapy for prevention and treatment of endothelial glycocalyx degradation. Second, we review the literature on plasma as an exploratory therapy for endothelial glycocalyx degradation in trauma and sepsis. Third, we discuss the safety of plasma transfusion by reviewing the adverse events associated with plasma and other blood product transfusions, and we examine modern transfusion precautions that have enhanced the safety of plasma transfusion. We conclude that the literature proposes that plasma may have the potential to prevent and treat endothelial glycocalyx degradation in trauma and sepsis, indicating the need for further research.
Collapse
Affiliation(s)
- M S Kravitz
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - N Kattouf
- Department of Emergency Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | - I J Stewart
- Department of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - A A Ginde
- Department of Emergency Medicine, University of Colorado School of Medicines, Aurora, CO, USA
| | - E P Schmidt
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - N I Shapiro
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
28
|
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
|
29
|
Bhosale SD, Moulder R, Suomi T, Ruohtula T, Honkanen J, Virtanen SM, Ilonen J, Elo LL, Knip M, Lahesmaa R. Serum proteomics of mother-infant dyads carrying HLA-conferred type 1 diabetes risk. iScience 2024; 27:110048. [PMID: 38883825 PMCID: PMC11176638 DOI: 10.1016/j.isci.2024.110048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 03/22/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
In-utero and dietary factors make important contributions toward health and development in early childhood. In this respect, serum proteomics of maturing infants can provide insights into studies of childhood diseases, which together with perinatal proteomes could reveal further biological perspectives. Accordingly, to determine differences between feeding groups and changes in infancy, serum proteomics analyses of mother-infant dyads with HLA-conferred susceptibility to type 1 diabetes (n = 22), weaned to either an extensively hydrolyzed or regular cow's milk formula, were made. The LC-MS/MS analyses included samples from the beginning of third trimester, the time of delivery, 3 months postpartum, cord blood, and samples from the infants at 3, 6, 9, and 12 months. Correlations between ranked protein intensities were detected within the dyads, together with perinatal and age-related changes. Comparison with intestinal permeability data revealed a number of significant correlations, which could merit further consideration in this context.
Collapse
Affiliation(s)
- Santosh D Bhosale
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Robert Moulder
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Tomi Suomi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Terhi Ruohtula
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jarno Honkanen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Suvi M Virtanen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
- Center for Child Health Research and Research, Development and Innovation Center, Tampere University Hospital, Tampere, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Laura L Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Mikael Knip
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Center for Child Health Research and Research, Development and Innovation Center, Tampere University Hospital, Tampere, Finland
| | - Riitta Lahesmaa
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
30
|
Burg N, Malpass R, Alex L, Tran M, Englebrecht E, Kuo A, Pannelini T, Minett M, Athukorala K, Worgall T, Faust HJ, Goodman S, Mehta B, Brenner M, Vestweber D, Wei K, Blobel C, Hla T, Salmon JE. Endothelial cell sphingosine 1-phosphate receptor 1 restrains VE-cadherin cleavage and attenuates experimental inflammatory arthritis. JCI Insight 2024; 9:e171467. [PMID: 38855867 PMCID: PMC11382883 DOI: 10.1172/jci.insight.171467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 05/01/2024] [Indexed: 06/11/2024] Open
Abstract
In rheumatoid arthritis, inflammatory mediators extravasate from blood into joints via gaps between endothelial cells (ECs), but the contribution of ECs is not known. Sphingosine 1-phosphate receptor 1 (S1PR1), widely expressed on ECs, maintains the vascular barrier. Here, we assessed the contribution of vascular integrity and EC S1PR1 signaling to joint damage in mice exposed to serum-induced arthritis (SIA). EC-specific deletion of S1PR1 or pharmacological blockade of S1PR1 promoted vascular leak and amplified SIA, whereas overexpression of EC S1PR1 or treatment with an S1PR1 agonist delayed SIA. Blockade of EC S1PR1 induced membrane metalloproteinase-dependent cleavage of vascular endothelial cadherin (VE-cadherin), a principal adhesion molecule that maintains EC junctional integrity. We identified a disintegrin and a metalloproteinase domain 10 (ADAM10) as the principal VE-cadherin "sheddase." Mice expressing a stabilized VE-cadherin construct had decreased extravascular VE-cadherin and vascular leakage in response to S1PR1 blockade, and they were protected from SIA. Importantly, patients with active rheumatoid arthritis had decreased circulating S1P and microvascular expression of S1PR1, suggesting a dysregulated S1P/S1PR1 axis favoring vascular permeability and vulnerability. We present a model in which EC S1PR1 signaling maintains homeostatic vascular barrier function by limiting VE-cadherin shedding mediated by ADAM10 and suggest this signaling axis as a therapeutic target in inflammatory arthritis.
Collapse
Affiliation(s)
- Nathalie Burg
- Hospital for Special Surgery, New York, New York, USA
| | - Ryan Malpass
- Hospital for Special Surgery, New York, New York, USA
| | - Linda Alex
- Hospital for Special Surgery, New York, New York, USA
| | - Miles Tran
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eric Englebrecht
- School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Andrew Kuo
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | - Tilla Worgall
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Heather J. Faust
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Susan Goodman
- Hospital for Special Surgery, New York, New York, USA
| | - Bella Mehta
- Hospital for Special Surgery, New York, New York, USA
| | - Michael Brenner
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Kevin Wei
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Carl Blobel
- Hospital for Special Surgery, New York, New York, USA
| | - Timothy Hla
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
31
|
Bassila C, Kluck GEG, Thyagarajan N, Chathely KM, Gonzalez L, Trigatti BL. Ligand-dependent interactions between SR-B1 and S1PR1 in macrophages and atherosclerotic plaques. J Lipid Res 2024; 65:100541. [PMID: 38583587 PMCID: PMC11087725 DOI: 10.1016/j.jlr.2024.100541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 03/16/2024] [Accepted: 04/01/2024] [Indexed: 04/09/2024] Open
Abstract
HDLs carry sphingosine-1-phosphate (S1P) and stimulate signaling pathways in different cells including macrophages and endothelial cells, involved in atherosclerotic plaque development. HDL signaling via S1P relies on the HDL receptor scavenger receptor class B, type I (SR-B1) and the sphingosine-1-phosphate receptor 1 (S1PR1), which interact when both are heterologously overexpressed in the HEK293 cell line. In this study, we set out to test if SR-B1 and S1PR1 interacted in primary murine macrophages in culture and atherosclerotic plaques. We used knock-in mice that endogenously expressed S1PR1 tagged with eGFP-(S1pr1eGFP/eGFP mice), combined with proximity ligation analysis to demonstrate that HDL stimulates the physical interaction between SR-B1 and S1PR1 in primary macrophages, that this is dependent on HDL-associated S1P and can be blocked by an inhibitor of SR-B1's lipid transfer activity or an antagonist of S1PR1. We also demonstrate that a synthetic S1PR1-selective agonist, SEW2871, stimulates the interaction between SR-B1 and S1PR1 and that this was also blocked by an inhibitor of SR-B1's lipid transport activity. Furthermore, we detected abundant SR-B1/S1PR1 complexes in atherosclerotic plaques of S1pr1eGFP/eGFP mice that also lacked apolipoprotein E. Treatment of mice with the S1PR1 antagonist, Ex26, for 12 h disrupted the SR-B1-S1PR1 interaction in atherosclerotic plaques. These findings demonstrate that SR-B1 and S1PR1 form ligand-dependent complexes both in cultured primary macrophages and within atherosclerotic plaques in mice and provide mechanistic insight into how SR-B1 and S1PR1 participate in mediating HDL signaling to activate atheroprotective responses in macrophages.
Collapse
Affiliation(s)
- Christine Bassila
- Department of Biochemistry and Biomedical Sciences, Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON, Canada
| | - George E G Kluck
- Department of Biochemistry and Biomedical Sciences, Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Narmadaa Thyagarajan
- Department of Biochemistry and Biomedical Sciences, Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Kevin M Chathely
- Department of Biochemistry and Biomedical Sciences, Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Leticia Gonzalez
- Department of Biochemistry and Biomedical Sciences, Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Bernardo L Trigatti
- Department of Biochemistry and Biomedical Sciences, Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
32
|
Velagapudi S, Wang D, Poti F, Feuerborn R, Robert J, Schlumpf E, Yalcinkaya M, Panteloglou G, Potapenko A, Simoni M, Rohrer L, Nofer JR, von Eckardstein A. Sphingosine-1-phosphate receptor 3 regulates the transendothelial transport of high-density lipoproteins and low-density lipoproteins in opposite ways. Cardiovasc Res 2024; 120:476-489. [PMID: 38109696 PMCID: PMC11060483 DOI: 10.1093/cvr/cvad183] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 10/08/2023] [Accepted: 10/20/2023] [Indexed: 12/20/2023] Open
Abstract
AIMS The entry of lipoproteins from blood into the arterial wall is a rate-limiting step in atherosclerosis. It is controversial whether this happens by filtration or regulated transendothelial transport.Because sphingosine-1-phosphate (S1P) preserves the endothelial barrier, we investigated in vivo and in vitro, whether S1P and its cognate S1P-receptor 3 (S1P3) regulate the transendothelial transport of lipoproteins. METHODS AND RESULTS Compared to apoE-haploinsufficient mice (CTRL), apoE-haploinsufficient mice with additional endothelium-specific knock-in of S1P3 (S1P3-iECKI) showed decreased transport of LDL and Evan's Blue but increased transport of HDL from blood into the peritoneal cave. After 30 weeks of high-fat diet feeding, S1P3-iECKI mice had lower levels of non-HDL-cholesterol and less atherosclerosis than CTRL mice. In vitro stimulation with an S1P3 agonist increased the transport of 125I-HDL but decreased the transport of 125I-LDL through human aortic endothelial cells (HAECs). Conversely, inhibition or knock-down of S1P3 decreased the transport of 125I-HDL but increased the transport of 125I-LDL. Silencing of SCARB1 encoding scavenger receptor B1 (SR-BI) abrogated the stimulation of 125I-HDL transport by the S1P3 agonist. The transendothelial transport of 125I-LDL was decreased by silencing of SCARB1 or ACVLR1 encoding activin-like kinase 1 but not by interference with LDLR. None of the three knock-downs prevented the stimulatory effect of S1P3 inhibition on transendothelial 125I-LDL transport. CONCLUSION S1P3 regulates the transendothelial transport of HDL and LDL oppositely by SR-BI-dependent and SR-BI-independent mechanisms, respectively. This divergence supports a contention that lipoproteins pass the endothelial barrier by specifically regulated mechanisms rather than passive filtration.
Collapse
Affiliation(s)
- Srividya Velagapudi
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Dongdong Wang
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Francesco Poti
- Department of Medicine and Surgery—Unit of Neurosciences, University of Parma, Parma, Italy
- Department of Biomedical, Metabolic and Neural Sciences—Unit of Endocrinology, University of Modena and Reggio Emilia, Modena, Italy
| | - Renata Feuerborn
- Central Laboratory Facility, University Hospital of Münster, Münster, Germany
| | - Jerome Robert
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Eveline Schlumpf
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Mustafa Yalcinkaya
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Grigorios Panteloglou
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Anton Potapenko
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences—Unit of Endocrinology, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucia Rohrer
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Jerzy-Roch Nofer
- Central Laboratory Facility, University Hospital of Münster, Münster, Germany
- Institute of Laboratory Medicine, Marien-Hospital Osnabrück, Niels-Stensen-Kliniken, Osnabrück, Germany
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| |
Collapse
|
33
|
Manzo OL, Nour J, Sasset L, Marino A, Rubinelli L, Palikhe S, Smimmo M, Hu Y, Bucci MR, Borczuk A, Elemento O, Freed JK, Norata GD, Di Lorenzo A. Rewiring Endothelial Sphingolipid Metabolism to Favor S1P Over Ceramide Protects From Coronary Atherosclerosis. Circ Res 2024; 134:990-1005. [PMID: 38456287 PMCID: PMC11009055 DOI: 10.1161/circresaha.123.323826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Growing evidence correlated changes in bioactive sphingolipids, particularly S1P (sphingosine-1-phosphate) and ceramides, with coronary artery diseases. Furthermore, specific plasma ceramide species can predict major cardiovascular events. Dysfunction of the endothelium lining lesion-prone areas plays a pivotal role in atherosclerosis. Yet, how sphingolipid metabolism and signaling change and contribute to endothelial dysfunction and atherosclerosis remain poorly understood. METHODS We used an established model of coronary atherosclerosis in mice, combined with sphingolipidomics, RNA-sequencing, flow cytometry, and immunostaining to investigate the contribution of sphingolipid metabolism and signaling to endothelial cell (EC) activation and dysfunction. RESULTS We demonstrated that hemodynamic stress induced an early metabolic rewiring towards endothelial sphingolipid de novo biosynthesis, favoring S1P signaling over ceramides as a protective response. This finding is a paradigm shift from the current belief that ceramide accrual contributes to endothelial dysfunction. The enzyme SPT (serine palmitoyltransferase) commences de novo biosynthesis of sphingolipids and is inhibited by NOGO-B (reticulon-4B), an ER membrane protein. Here, we showed that NOGO-B is upregulated by hemodynamic stress in myocardial EC of ApoE-/- mice and is expressed in the endothelium lining coronary lesions in mice and humans. We demonstrated that mice lacking NOGO-B specifically in EC (Nogo-A/BECKOApoE-/-) were resistant to coronary atherosclerosis development and progression, and mortality. Fibrous cap thickness was significantly increased in Nogo-A/BECKOApoE-/- mice and correlated with reduced necrotic core and macrophage infiltration. Mechanistically, the deletion of NOGO-B in EC sustained the rewiring of sphingolipid metabolism towards S1P, imparting an atheroprotective endothelial transcriptional signature. CONCLUSIONS These data demonstrated that hemodynamic stress induced a protective rewiring of sphingolipid metabolism, favoring S1P over ceramide. NOGO-B deletion sustained the rewiring of sphingolipid metabolism toward S1P protecting EC from activation under hemodynamic stress and refraining coronary atherosclerosis. These findings also set forth the foundation for sphingolipid-based therapeutics to limit atheroprogression.
Collapse
Affiliation(s)
- Onorina Laura Manzo
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Jasmine Nour
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Department of Excellence of Pharmacological and Biomolecular Sciences, University of Milan, Via G. Balzaretti, 9 – 20133, Milano, Italy
| | - Linda Sasset
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Alice Marino
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Luisa Rubinelli
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Sailesh Palikhe
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Martina Smimmo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano 49, Naples 80131, Italy
| | - Yang Hu
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA
| | - Maria Rosaria Bucci
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano 49, Naples 80131, Italy
| | - Alain Borczuk
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Olivier Elemento
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA
| | - Julie K. Freed
- Department of Anesthesiology, Medical College of Wisconsin Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Rd. Milwaukee, WI 53226, USA
| | - Giuseppe Danilo Norata
- Department of Excellence of Pharmacological and Biomolecular Sciences, University of Milan, Via G. Balzaretti, 9 – 20133, Milano, Italy
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| |
Collapse
|
34
|
Polenz CK, Scipione CA, Hyduk SJ, Althagafi MG, Ibrahim HM, Cybulsky MI. Plasma S1P Orchestrates the Reverse Transendothelial Migration of Aortic Intimal Myeloid Cells in Mice. Arterioscler Thromb Vasc Biol 2024; 44:883-897. [PMID: 38328936 DOI: 10.1161/atvbaha.123.320227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/24/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND Myeloid cells (MCs) reside in the aortic intima at regions predisposed to atherosclerosis. Systemic inflammation triggers reverse transendothelial migration (RTM) of intimal MCs into the arterial blood, which orchestrates a protective immune response that clears intracellular pathogens from the arterial intima. Molecular pathways that regulate RTM remain poorly understood. S1P (sphingosine-1-phosphate) is a lipid mediator that regulates immune cell trafficking by signaling via 5 G-protein-coupled receptors (S1PRs [S1P receptors]). We investigated the role of S1P in the RTM of aortic intimal MCs. METHODS Intravenous injection of lipopolysaccharide was used to model a systemic inflammatory stimulus that triggers RTM. CD11c+ intimal MCs in the lesser curvature of the ascending aortic arch were enumerated by en face confocal microscopy. Local gene expression was evaluated by transcriptomic analysis of microdissected intimal cells. RESULTS In wild-type C57BL/6 mice, lipopolysaccharide induced intimal cell expression of S1pr1, S1pr3, and Sphk1 (a kinase responsible for S1P production). Pharmacological modulation of multiple S1PRs blocked lipopolysaccharide-induced RTM and modulation of S1PR1 and S1PR3 reduced RTM in an additive manner. Cre-mediated deletion of S1pr1 in MCs blocked lipopolysaccharide-induced RTM, confirming a role for myeloid-specific S1PR1 signaling. Global or hematopoietic deficiency of Sphk1 reduced plasma S1P levels, the abundance of CD11c+ MCs in the aortic intima, and blunted lipopolysaccharide-induced RTM. In contrast, plasma S1P levels, the abundance of intimal MCs, and lipopolysaccharide-induced RTM were rescued in Sphk1-/- mice transplanted with Sphk1+/+ or mixed Sphk1+/+ and Sphk1-/- bone marrow. Stimulation with lipopolysaccharide increased endothelial permeability and intimal MC exposure to circulating factors such as S1P. CONCLUSIONS Functional and expression studies support a novel role for S1P signaling in the regulation of lipopolysaccharide-induced RTM and the homeostatic maintenance of aortic intimal MCs. Our data provide insight into how circulating plasma mediators help orchestrate intimal MC dynamics.
Collapse
Affiliation(s)
- Chanele K Polenz
- Departments of Laboratory Medicine and Pathobiology (C.K.P., C.A.S., M.G.A., H.M.I., M.I.C.), University of Toronto, Canada
- Toronto General Hospital Research Institute, University Health Network, Canada (C.K.P., C.A.S., S.J.H., M.G.A., H.M.I., M.I.C.)
| | - Corey A Scipione
- Departments of Laboratory Medicine and Pathobiology (C.K.P., C.A.S., M.G.A., H.M.I., M.I.C.), University of Toronto, Canada
- Immunology (C.A.S., M.I.C.), University of Toronto, Canada
- Toronto General Hospital Research Institute, University Health Network, Canada (C.K.P., C.A.S., S.J.H., M.G.A., H.M.I., M.I.C.)
| | - Sharon J Hyduk
- Toronto General Hospital Research Institute, University Health Network, Canada (C.K.P., C.A.S., S.J.H., M.G.A., H.M.I., M.I.C.)
| | - Marwan G Althagafi
- Departments of Laboratory Medicine and Pathobiology (C.K.P., C.A.S., M.G.A., H.M.I., M.I.C.), University of Toronto, Canada
- Toronto General Hospital Research Institute, University Health Network, Canada (C.K.P., C.A.S., S.J.H., M.G.A., H.M.I., M.I.C.)
| | - Hisham M Ibrahim
- Departments of Laboratory Medicine and Pathobiology (C.K.P., C.A.S., M.G.A., H.M.I., M.I.C.), University of Toronto, Canada
- Toronto General Hospital Research Institute, University Health Network, Canada (C.K.P., C.A.S., S.J.H., M.G.A., H.M.I., M.I.C.)
| | - Myron I Cybulsky
- Departments of Laboratory Medicine and Pathobiology (C.K.P., C.A.S., M.G.A., H.M.I., M.I.C.), University of Toronto, Canada
- Immunology (C.A.S., M.I.C.), University of Toronto, Canada
- Toronto General Hospital Research Institute, University Health Network, Canada (C.K.P., C.A.S., S.J.H., M.G.A., H.M.I., M.I.C.)
- Peter Munk Cardiac Centre, University Health Network, Toronto, Canada (M.I.C.)
| |
Collapse
|
35
|
Alkafaas SS, Elsalahaty MI, Ismail DF, Radwan MA, Elkafas SS, Loutfy SA, Elshazli RM, Baazaoui N, Ahmed AE, Hafez W, Diab M, Sakran M, El-Saadony MT, El-Tarabily KA, Kamal HK, Hessien M. The emerging roles of sphingosine 1-phosphate and SphK1 in cancer resistance: a promising therapeutic target. Cancer Cell Int 2024; 24:89. [PMID: 38419070 PMCID: PMC10903003 DOI: 10.1186/s12935-024-03221-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 01/09/2024] [Indexed: 03/02/2024] Open
Abstract
Cancer chemoresistance is a problematic dilemma that significantly restrains numerous cancer management protocols. It can promote cancer recurrence, spreading of cancer, and finally, mortality. Accordingly, enhancing the responsiveness of cancer cells towards chemotherapies could be a vital approach to overcoming cancer chemoresistance. Tumour cells express a high level of sphingosine kinase-1 (SphK1), which acts as a protooncogenic factor and is responsible for the synthesis of sphingosine-1 phosphate (S1P). S1P is released through a Human ATP-binding cassette (ABC) transporter to interact with other phosphosphingolipids components in the interstitial fluid in the tumor microenvironment (TME), provoking communication, progression, invasion, and tumor metastasis. Also, S1P is associated with several impacts, including anti-apoptotic behavior, metastasis, mesenchymal transition (EMT), angiogenesis, and chemotherapy resistance. Recent reports addressed high levels of S1P in several carcinomas, including ovarian, prostate, colorectal, breast, and HCC. Therefore, targeting the S1P/SphK signaling pathway is an emerging therapeutic approach to efficiently attenuate chemoresistance. In this review, we comprehensively discussed S1P functions, metabolism, transport, and signaling. Also, through a bioinformatic framework, we pointed out the alterations of SphK1 gene expression within different cancers with their impact on patient survival, and we demonstrated the protein-protein network of SphK1, elaborating its sparse roles. Furthermore, we made emphasis on different machineries of cancer resistance and the tight link with S1P. We evaluated all publicly available SphK1 inhibitors and their inhibition activity using molecular docking and how SphK1 inhibitors reduce the production of S1P and might reduce chemoresistance, an approach that might be vital in the course of cancer treatment and prognosis.
Collapse
Affiliation(s)
- Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Mohamed I Elsalahaty
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Doha F Ismail
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mustafa Ali Radwan
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Sara Samy Elkafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Menofia, Egypt
- Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, 197101, Russia
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
- Nanotechnology Research Center, British University, Cairo, Egypt
| | - Rami M Elshazli
- Biochemistry and Molecular Genetics Unit, Department of Basic Sciences, Faculty of Physical Therapy, Horus University-Egypt, New Damietta, 34517, Egypt
| | - Narjes Baazaoui
- Biology Department, College of Sciences and Arts Muhayil Assir, King Khalid University, Abha 61421, Saudi Arabia
| | - Ahmed Ezzat Ahmed
- Biology Department, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Wael Hafez
- NMC Royal Hospital, 16th Street, 35233, Khalifa, Abu Dhabi, United Arab Emirates
- Medical Research Division, Department of Internal Medicine, The National Research Centre, Cairo 11511, Egypt
| | - Mohanad Diab
- Burjeel Hospital Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Mohamed Sakran
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
- Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk 47512, Saudi Arabia
| | - Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Hani K Kamal
- Anatomy and Histology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohamed Hessien
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
36
|
Lin YC, Swendeman S, Moreira IS, Ghosh A, Kuo A, Rosário-Ferreira N, Guo S, Culbertson A, Levesque MV, Cartier A, Seno T, Schmaier A, Galvani S, Inoue A, Parikh SM, FitzGerald GA, Zurakowski D, Liao M, Flaumenhaft R, Gümüş ZH, Hla T. Designer high-density lipoprotein particles enhance endothelial barrier function and suppress inflammation. Sci Signal 2024; 17:eadg9256. [PMID: 38377179 PMCID: PMC10954247 DOI: 10.1126/scisignal.adg9256] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 01/31/2024] [Indexed: 02/22/2024]
Abstract
High-density lipoprotein (HDL) nanoparticles promote endothelial cell (EC) function and suppress inflammation, but their utility in treating EC dysfunction has not been fully explored. Here, we describe a fusion protein named ApoA1-ApoM (A1M) consisting of apolipoprotein A1 (ApoA1), the principal structural protein of HDL that forms lipid nanoparticles, and ApoM, a chaperone for the bioactive lipid sphingosine 1-phosphate (S1P). A1M forms HDL-like particles, binds to S1P, and is signaling competent. Molecular dynamics simulations showed that the S1P-bound ApoM moiety in A1M efficiently activated EC surface receptors. Treatment of human umbilical vein ECs with A1M-S1P stimulated barrier function either alone or cooperatively with other barrier-enhancing molecules, including the stable prostacyclin analog iloprost, and suppressed cytokine-induced inflammation. A1M-S1P injection into mice during sterile inflammation suppressed neutrophil influx and inflammatory mediator secretion. Moreover, systemic A1M administration led to a sustained increase in circulating HDL-bound S1P and suppressed inflammation in a murine model of LPS-induced endotoxemia. We propose that A1M administration may enhance vascular endothelial barrier function, suppress cytokine storm, and promote resilience of the vascular endothelium.
Collapse
Affiliation(s)
- Yueh-Chien Lin
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Steven Swendeman
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Irina S. Moreira
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456, Coimbra, Portugal
- CNC - Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-456, Coimbra, Portugal
| | - Avishek Ghosh
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Andrew Kuo
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Nícia Rosário-Ferreira
- CNC - Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-456, Coimbra, Portugal
| | | | - Alan Culbertson
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Michel V. Levesque
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Andreane Cartier
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Takahiro Seno
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Alec Schmaier
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02115, USA
| | - Sylvain Galvani
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Samir M. Parikh
- Division of Nephrology and Department of Medicine, Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, 75235, USA
| | - Garret A. FitzGerald
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - David Zurakowski
- Department of Anesthesia and Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Maofu Liao
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, 518055, China
| | | | - Zeynep H. Gümüş
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Timothy Hla
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
37
|
Das K, Keshava S, Kolesnick R, Pendurthi UR, Rao LVM. MicroRNA-10a enrichment in factor VIIa-released endothelial extracellular vesicles: potential mechanisms. J Thromb Haemost 2024; 22:441-454. [PMID: 37926194 PMCID: PMC10872460 DOI: 10.1016/j.jtha.2023.10.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/09/2023] [Accepted: 10/24/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Factor VIIa induces the release of extracellular vesicles (EVs) from endothelial cells (EEVs). Factor VIIa-released EEVs are enriched with microRNA-10a (miR10a) and elicit miR10a-dependent cytoprotective responses. OBJECTIVES To investigate mechanisms by which FVIIa induces miR10a expression in endothelial cells and sorts miR10a into the EVs. METHODS Activation of Elk-1 and TWIST1 expression was analyzed by immunofluorescence microscopy and immunoblot analysis. Small interfering RNA silencing approach was used to knock down the expression of specific genes in endothelial cells. EVs secreted from endothelial cells or released into circulation in mice were isolated by centrifugation and quantified by nanoparticle tracking analysis. Factor VIIa or EVs were injected into mice; mice were challenged with lipopolysaccharides to assess the cytoprotective effects of FVIIa or EVs. RESULTS FVIIa activation of ERK1/2 triggered the activation of Elk-1, which led to the induction of TWIST1, a key transcription factor involved in miR10a expression. Factor VIIa also induced the expression of La, a small RNA-binding protein. Factor VIIa-driven acid sphingomyelinase (ASM) activation and the subsequent activation of the S1P receptor pathway were responsible for the induction of La. Silencing of ASM or La significantly reduced miR10a levels in FVIIa-released EEVs without affecting the cellular expression of miR10a. Factor VIIa-EEVs from ASM knocked-down cells failed to provide cytoprotective responses in cell and murine model systems. Administration of FVIIa protected wild-type but not ASM-/- mice against lipopolysaccharide-induced inflammation and vascular leakage. CONCLUSION Our data suggest that enhanced cellular expression of miR10a coupled with La-dependent sorting of miR10a is responsible for enriching FVIIa-released EVs with miR10a.
Collapse
Affiliation(s)
- Kaushik Das
- Department of Cellular and Molecular Biology, UT Tyler School of Medicine, the University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Shiva Keshava
- Department of Cellular and Molecular Biology, UT Tyler School of Medicine, the University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | | | - Usha R Pendurthi
- Department of Cellular and Molecular Biology, UT Tyler School of Medicine, the University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - L Vijaya Mohan Rao
- Department of Cellular and Molecular Biology, UT Tyler School of Medicine, the University of Texas Health Science Center at Tyler, Tyler, Texas, USA.
| |
Collapse
|
38
|
Duan J, Zhao Q, He Z, Tang S, Duan J, Xing W. Current understanding of macrophages in intracranial aneurysm: relevant etiological manifestations, signaling modulation and therapeutic strategies. Front Immunol 2024; 14:1320098. [PMID: 38259443 PMCID: PMC10800944 DOI: 10.3389/fimmu.2023.1320098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Macrophages activation and inflammatory response play crucial roles in intracranial aneurysm (IA) formation and progression. The outcome of ruptured IA is considerably poor, and the mechanisms that trigger IA progression and rupture remain to be clarified, thereby developing effective therapy to prevent subarachnoid hemorrhage (SAH) become difficult. Recently, climbing evidences have been expanding our understanding of the macrophages relevant IA pathogenesis, such as immune cells population, inflammatory activation, intra-/inter-cellular signaling transductions and drug administration responses. Crosstalk between macrophages disorder, inflammation and cellular signaling transduction aggravates the devastating consequences of IA. Illustrating the pros and cons mechanisms of macrophages in IA progression are expected to achieve more efficient treatment interventions. In this review, we summarized the current advanced knowledge of macrophages activation, infiltration, polarization and inflammatory responses in IA occurrence and development, as well as the most relevant NF-κB, signal transducer and activator of transcription 1 (STAT1) and Toll-Like Receptor 4 (TLR4) regulatory signaling modulation. The understanding of macrophages regulatory mechanisms is important for IA patients' clinical outcomes. Gaining insight into the macrophages regulation potentially contributes to more precise IA interventions and will also greatly facilitate the development of novel medical therapy.
Collapse
Affiliation(s)
- Jian Duan
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Qijie Zhao
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zeyuan He
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Shuang Tang
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Jia Duan
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Wenli Xing
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| |
Collapse
|
39
|
Schoch L, Alcover S, Padró T, Ben-Aicha S, Mendieta G, Badimon L, Vilahur G. Update of HDL in atherosclerotic cardiovascular disease. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2023; 35:297-314. [PMID: 37940388 DOI: 10.1016/j.arteri.2023.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Epidemiologic evidence supported an inverse association between HDL (high-density lipoprotein) cholesterol (HDL-C) levels and atherosclerotic cardiovascular disease (ASCVD), identifying HDL-C as a major cardiovascular risk factor and postulating diverse HDL vascular- and cardioprotective functions beyond their ability to drive reverse cholesterol transport. However, the failure of several clinical trials aimed at increasing HDL-C in patients with overt cardiovascular disease brought into question whether increasing the cholesterol cargo of HDL was an effective strategy to enhance their protective properties. In parallel, substantial evidence supports that HDLs are complex and heterogeneous particles whose composition is essential for maintaining their protective functions, subsequently strengthening the "HDL quality over quantity" hypothesis. The following state-of-the-art review covers the latest understanding as per the roles of HDL in ASCVD, delves into recent advances in understanding the complexity of HDL particle composition, including proteins, lipids and other HDL-transported components and discusses on the clinical outcomes after the administration of HDL-C raising drugs with particular attention to CETP (cholesteryl ester transfer protein) inhibitors.
Collapse
Affiliation(s)
- Leonie Schoch
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain; Faculty of Medicine, University of Barcelona (UB), 08036 Barcelona, Spain
| | - Sebastián Alcover
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain
| | | | - Guiomar Mendieta
- Cardiology Unit, Cardiovascular Clinical Institute, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain; Cardiovascular Research Chair, UAB, 08025 Barcelona, Spain; CiberCV, Institute of Health Carlos III, Madrid, Spain
| | - Gemma Vilahur
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain; CiberCV, Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
40
|
Kurano M, Uranbileg B, Yatomi Y. Apolipoprotein M bound sphingosine 1-phosphate suppresses NETosis through activating S1P1 and S1P4. Biomed Pharmacother 2023; 166:115400. [PMID: 37657263 DOI: 10.1016/j.biopha.2023.115400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/23/2023] [Accepted: 08/26/2023] [Indexed: 09/03/2023] Open
Abstract
The pleiotropic effects of high-density lipoprotein (HDL), including its protective properties against sepsis, are attributed to the sphingosine 1-phosphate and apolipoprotein M (ApoM) that are carried on the lipoproteins. In this study, we attempted to elucidate the possible mechanisms underlying the sepsis coagulopathic state by considering the modulation of NETosis. Our results revealed that in a lipopolysaccharide-induced sepsis mouse model, the levels of NETosis markers, such as plasma DNA and histone, were elevated in ApoM-knockout (KO) mice and attenuated in ApoM-overexpressing mice. In ApoM-KO mice, the survival rate decreased and the occurrence rates of coagulopathy and organ injury increased following the administration of histone. Treatment with a conditioned medium of ApoM-overexpressing cells attenuated the observed NETosis in HL-60S cells that differentiated into neutrophils and were inhibited through the suppression of S1P1 or S1P4. The attenuation of PKCδ and PKCα/β by S1P1 and S1P4 activation may also be involved. In ApoM-overexpressing mice, coagulopathy and organ injuries were attenuated following an injection of histone; these effects were partially inhibited by S1P1, 3, S1P4, or S1P1 antagonists. Furthermore, the exogenous administration of ApoM protected ApoM-KO mice that were challenged with histone from developing NETosis. In conclusion, the ApoM/S1P axis protects against NETosis through the attenuation of PKC activation by S1P1 and S1P4. The development of drugs targeting the ApoM/S1P axis may be beneficial for the treatment of pathological conditions involving uncontrolled NETosis, such as sepsis.
Collapse
Affiliation(s)
- Makoto Kurano
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan.
| | - Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
41
|
Mannion AJ, Holmgren L. Nuclear mechanosensing of the aortic endothelium in health and disease. Dis Model Mech 2023; 16:dmm050361. [PMID: 37909406 PMCID: PMC10629673 DOI: 10.1242/dmm.050361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Abstract
The endothelium, the monolayer of endothelial cells that line blood vessels, is exposed to a number of mechanical forces, including frictional shear flow, pulsatile stretching and changes in stiffness influenced by extracellular matrix composition. These forces are sensed by mechanosensors that facilitate their transduction to drive appropriate adaptation of the endothelium to maintain vascular homeostasis. In the aorta, the unique architecture of the vessel gives rise to changes in the fluid dynamics, which, in turn, shape cellular morphology, nuclear architecture, chromatin dynamics and gene regulation. In this Review, we discuss recent work focusing on how differential mechanical forces exerted on endothelial cells are sensed and transduced to influence their form and function in giving rise to spatial variation to the endothelium of the aorta. We will also discuss recent developments in understanding how nuclear mechanosensing is implicated in diseases of the aorta.
Collapse
Affiliation(s)
- Aarren J. Mannion
- Department of Oncology-Pathology, Karolinska Institute, Stockholm 171 64, Sweden
| | - Lars Holmgren
- Department of Oncology-Pathology, Karolinska Institute, Stockholm 171 64, Sweden
| |
Collapse
|
42
|
Ghaderi S, Levkau B. An erythrocyte-centric view on the MFSD2B sphingosine-1-phosphate transporter. Pharmacol Ther 2023; 249:108483. [PMID: 37390971 DOI: 10.1016/j.pharmthera.2023.108483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/12/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
MFSD2B has been identified as the exclusive sphingosine-1-phosphate (S1P) transporter in red blood cells (RBC) and platelets. MFSD2B-mediated S1P export from platelets is required for aggregation and thrombus formation, whereas RBC MFSD2B maintains plasma S1P levels in concert with SPNS2, the vascular and lymphatic endothelial cell S1P exporter, to control endothelial permeability and ensure normal vascular development. However, the physiological function of MFSD2B in RBC remains rather elusive despite mounting evidence that the intracellular S1P pool plays important roles in RBC glycolysis, adaptation to hypoxia and the regulation of cell shape, hydration, and cytoskeletal organisation. The large accumulation of S1P and sphingosine in MFSD2B-deficient RBC coincides with stomatocytosis and membrane abnormalities, the reasons for which have remained obscure. MFS family members transport substrates in a cation-dependent manner along electrochemical gradients, and disturbances in cation permeability are known to alter cell hydration and shape in RBC. Furthermore, the mfsd2 gene is a transcriptional target of GATA together with mylk3, the gene encoding myosin light chain kinase (MYLK). S1P is known to activate MYLK and thereby impact on myosin phosphorylation and cytoskeletal architecture. This suggests that metabolic, transcriptional and functional interactions may exist between MFSD2B-mediated S1P transport and RBC deformability. Here, we review the evidence for such interactions and the implications for RBC homeostasis.
Collapse
Affiliation(s)
- Shahrooz Ghaderi
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany
| | - Bodo Levkau
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany.
| |
Collapse
|
43
|
Frey K, Rohrer L, Frommelt F, Ringwald M, Potapenko A, Goetze S, von Eckardstein A, Wollscheid B. Mapping the dynamic high-density lipoprotein synapse. Atherosclerosis 2023; 380:117200. [PMID: 37619408 DOI: 10.1016/j.atherosclerosis.2023.117200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND AND AIMS Heterogeneous high-density lipoprotein (HDL) particles, which can contain hundreds of proteins, affect human health and disease through dynamic molecular interactions with cell surface proteins. How HDL mediates its long-range signaling functions and interactions with various cell types is largely unknown. Due to the complexity of HDL, we hypothesize that multiple receptors engage with HDL particles resulting in condition-dependent receptor-HDL interaction clusters at the cell surface. METHODS Here we used the mass spectrometry-based and light-controlled proximity labeling strategy LUX-MS in a discovery-driven manner to decode HDL-receptor interactions. RESULTS Surfaceome nanoscale organization analysis of hepatocytes and endothelial cells using LUX-MS revealed that the previously known HDL-binding protein scavenger receptor B1 (SCRB1) is embedded in a cell surface protein community, which we term HDL synapse. Modulating the endothelial HDL synapse, composed of 60 proteins, by silencing individual members, showed that the HDL synapse can be assembled in the absence of SCRB1 and that the members are interlinked. The aminopeptidase N (AMPN) (also known as CD13) was identified as an HDL synapse member that directly influences HDL uptake into the primary human aortic endothelial cells (HAECs). CONCLUSIONS Our data indicate that preformed cell surface residing protein complexes modulate HDL function and suggest new theragnostic opportunities.
Collapse
Affiliation(s)
- Kathrin Frey
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland; Institute for Clinical Chemistry University Hospital Zurich, Zurich, Switzerland.
| | - Lucia Rohrer
- Institute for Clinical Chemistry University Hospital Zurich, Zurich, Switzerland
| | - Fabian Frommelt
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Meret Ringwald
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Anton Potapenko
- Institute for Clinical Chemistry University Hospital Zurich, Zurich, Switzerland
| | - Sandra Goetze
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland; ETH PHRT Swiss Multi-Omics Center (SMOC), Switzerland
| | | | - Bernd Wollscheid
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland; ETH PHRT Swiss Multi-Omics Center (SMOC), Switzerland.
| |
Collapse
|
44
|
Birch CA, Wedegaertner H, Orduña-Castillo LB, Gonzalez Ramirez ML, Qin H, Trejo J. Endothelial APC/PAR1 distinctly regulates cytokine-induced pro-inflammatory VCAM-1 expression. Front Mol Biosci 2023; 10:1211597. [PMID: 37692066 PMCID: PMC10483999 DOI: 10.3389/fmolb.2023.1211597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/04/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction: Dysfunction of the endothelium impairs its' protective role and promotes inflammation and progression of vascular diseases. Activated Protein C (APC) elicits endothelial cytoprotective responses including barrier stabilization, anti-inflammatory and anti-apoptotic responses through the activation of the G protein-coupled receptor (GPCR) protease-activated receptor-1 (PAR1) and is a promising therapeutic. Despite recent advancements in developing new Activated protein C variants with clinical potential, the mechanism by which APC/PAR1 promotes different cytoprotective responses remains unclear and is important to understand to advance Activated protein C and new targets as future therapeutics. Here we examined the mechanisms by which APC/PAR1 attenuates cytokine-induced pro-inflammatory vascular cell adhesion molecule (VCAM-1) expression, a key mediator of endothelial inflammatory responses. Methods: Quantitative multiplexed mass spectrometry analysis of Activated protein C treated endothelial cells, endothelial cell transcriptomics database (EndoDB) online repository queries, biochemical measurements of protein expression, quantitative real-time polymerase chain reaction (RT-qPCR) measurement of mRNA transcript abundance, pharmacological inhibitors and siRNA transfections of human cultured endothelial cells. Results: Here we report that Activated Protein C modulates phosphorylation of tumor necrosis factor (TNF)-α signaling pathway components and attenuates of TNF-α induced VCAM-1 expression independent of mRNA stability. Unexpectedly, we found a critical role for the G protein-coupled receptor co-receptor sphingosine-1 phosphate receptor-1 (S1PR1) and the G protein receptor kinase-2 (GRK2) in mediating APC/PAR1 anti-inflammatory responses in endothelial cells. Discussion: This study provides new knowledge of the mechanisms by which different APC/PAR1 cytoprotective responses are mediated through discrete β-arrestin-2-driven signaling pathways modulated by specific G protein-coupled receptor co-receptors and GRKs.
Collapse
Affiliation(s)
- Cierra A. Birch
- Department of Pharmacology, School of Medicine, University of California, San Diego, CA, United States
| | - Helen Wedegaertner
- Department of Pharmacology, School of Medicine, University of California, San Diego, CA, United States
- Biomedical Sciences Graduate Program, University of California, San Diego, CA, United States
| | - Lennis B. Orduña-Castillo
- Department of Pharmacology, School of Medicine, University of California, San Diego, CA, United States
| | | | - Huaping Qin
- Department of Pharmacology, School of Medicine, University of California, San Diego, CA, United States
| | - JoAnn Trejo
- Department of Pharmacology, School of Medicine, University of California, San Diego, CA, United States
| |
Collapse
|
45
|
Li Y, Luo X, Hua Z, Xue X, Wang X, Pang M, Wang T, Lyu A, Liu Y. Apolipoproteins as potential communicators play an essential role in the pathogenesis and treatment of early atherosclerosis. Int J Biol Sci 2023; 19:4493-4510. [PMID: 37781031 PMCID: PMC10535700 DOI: 10.7150/ijbs.86475] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/15/2023] [Indexed: 10/03/2023] Open
Abstract
Atherosclerosis as the leading cause of the cardiovascular disease is closely related to cholesterol deposition within subendothelial areas of the arteries. Significantly, early atherosclerosis intervention is the critical phase for its reversal. As atherosclerosis progresses, early foam cells formation may evolve into fibrous plaques and atheromatous plaque, ulteriorly rupture of atheromatous plaque increases risks of myocardial infarction and ischemic stroke, resulting in high morbidity and mortality worldwide. Notably, amphiphilic apolipoproteins (Apos) can concomitantly combine with lipids to form soluble lipoproteins that have been demonstrated to associate with atherosclerosis. Apos act as crucial communicators of lipoproteins, which not only can mediate lipids metabolism, but also can involve in pro-atherogenic and anti-atherogenic processes of atherosclerosis via affecting subendothelial retention and aggregation of low-density lipoprotein (LDL), oxidative modification of LDL, foam cells formation and reverse cholesterol transport (RCT) in macrophage cells. Correspondingly, Apos can be used as endogenous and/or exogenous targeting agents to effectively attenuate the development of atherosclerosis. The article reviews the classification, structure, and relationship between Apos and lipids, how Apos serve as communicators of lipoproteins to participate in the pathogenesis progression of early atherosclerosis, as well as how Apos as the meaningful targeting mass is used in early atherosclerosis treatment.
Collapse
Affiliation(s)
- Yang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xinyi Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhenglai Hua
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaoxia Xue
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiangpeng Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mingshi Pang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Tieshan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Aiping Lyu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong 999077, China
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
46
|
Ya'ar Bar S, Pintel N, Abd Alghne H, Khattib H, Avni D. The therapeutic potential of sphingolipids for cardiovascular diseases. Front Cardiovasc Med 2023; 10:1224743. [PMID: 37608809 PMCID: PMC10440740 DOI: 10.3389/fcvm.2023.1224743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/17/2023] [Indexed: 08/24/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide and Inflammation plays a critical role in the development of CVD. Despite considerable progress in understanding the underlying mechanisms and various treatment options available, significant gaps in therapy necessitate the identification of novel therapeutic targets. Sphingolipids are a family of lipids that have gained attention in recent years as important players in CVDs and the inflammatory processes that underlie their development. As preclinical studies have shown that targeting sphingolipids can modulate inflammation and ameliorate CVDs, targeting sphingolipids has emerged as a promising therapeutic strategy. This review discusses the current understanding of sphingolipids' involvement in inflammation and cardiovascular diseases, the existing therapeutic approaches and gaps in therapy, and explores the potential of sphingolipids-based drugs as a future avenue for CVD treatment.
Collapse
Affiliation(s)
- Sapir Ya'ar Bar
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
| | - Noam Pintel
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
| | - Hesen Abd Alghne
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Tel-Hai College Department of Biotechnology, Kiryat Shmona, Israel
| | - Hamdan Khattib
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Department of Gastroenterology and Hepatology, Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Dorit Avni
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Tel-Hai College Department of Biotechnology, Kiryat Shmona, Israel
| |
Collapse
|
47
|
Kiyozuka K, Zhao X, Konishi A, Minamishima YA, Obinata H. Apolipoprotein M supports S1P production and conservation and mediates prolonged Akt activation via S1PR1 and S1PR3. J Biochem 2023; 174:253-266. [PMID: 37098187 DOI: 10.1093/jb/mvad037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 04/11/2023] [Accepted: 04/19/2023] [Indexed: 04/27/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) is one of the lipid mediators involved in diverse physiological functions. S1P circulates in blood and lymph bound to carrier proteins. Three S1P carrier proteins have been reported, albumin, apolipoprotein M (ApoM) and apolipoprotein A4 (ApoA4). The carrier-bound S1P exerts its functions via specific S1P receptors (S1PR1-5) on target cells. Previous studies showed several differences in physiological functions between albumin-bound S1P and ApoM-bound S1P. However, molecular mechanisms underlying the carrier-dependent differences have not been clarified. In addition, ApoA4 is a recently identified S1P carrier protein, and its functional differences from albumin and ApoM have not been addressed. Here, we compared the three carrier proteins in the processes of S1P degradation, release from S1P-producing cells and receptor activation. ApoM retained S1P more stable than albumin and ApoA4 in the cell culture medium when compared in the equimolar amounts. ApoM facilitated theS1P release from endothelial cells most efficiently. Furthermore, ApoM-bound S1P showed a tendency to induce prolonged activation of Akt via S1PR1 and S1PR3. These results suggest that the carrier-dependent functional differences of S1P are partly ascribed to the differences in the S1P stability, S1P-releasing efficiency and signaling duration.
Collapse
Key Words
- Apolipoprotein A4
- Apolipoprotein M
- LC–MS/MS
- Sphingosine 1-phosphate.Abbreviations: ApoA4, Apolipoprotein A4; ApoM, Apolipoprotein M; CHO, Chinese hamster ovary; ERK, Extracellular signal-regulated kinase; LC–MS/MS, Liquid chromatography–tandem mass spectrometry; LPP, Lipid phosphate phosphatase; Mfsd2b, Multiple facilitator superfamily domain containing 2B; PBS, Phosphate-buffered saline; S1P, Sphingosine 1-phosphate; S1PR1, Sphingosine 1-phosphate receptor 1; S1PR3, Sphingosine 1-phosphate receptor 3; SphK, Sphingosine kinase; Spns2, Spinster homolog 2; TBS-T, Tris-buffed saline containing 0.1% Tween20
Collapse
Affiliation(s)
- Keisuke Kiyozuka
- Department of Biochemistry, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Xian Zhao
- Department of Biochemistry, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Akimitsu Konishi
- Department of Biochemistry, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Yoji Andrew Minamishima
- Department of Biochemistry, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Hideru Obinata
- Education and Research Support Center, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
48
|
Wilkins GC, Gilmour J, Giannoudaki E, Kirby JA, Sheerin NS, Ali S. Dissecting the Therapeutic Mechanisms of Sphingosine-1-Phosphate Receptor Agonism during Ischaemia and Reperfusion. Int J Mol Sci 2023; 24:11192. [PMID: 37446370 PMCID: PMC10342646 DOI: 10.3390/ijms241311192] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/21/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) and S1P receptors (S1PR) regulate many cellular processes, including lymphocyte migration and endothelial barrier function. As neutrophils are major mediators of inflammation, their transendothelial migration may be the target of therapeutic approaches to inflammatory conditions such as ischaemia-reperfusion injury (IRI). The aim of this project was to assess whether these therapeutic effects are mediated by S1P acting on neutrophils directly or indirectly through the endothelial cells. First, our murine model of peritoneum cell recruitment demonstrated the ability of S1P to reduce CXCL8-mediated neutrophil recruitment. Mechanistic in vitro studies revealed that S1P signals in neutrophils mainly through the S1PR1 and 4 receptors and induces phosphorylation of ERK1/2; however, this had no effect on neutrophil transmigration and adhesion. S1P treatment of endothelial cells significantly reduced TNF-α-induced neutrophil adhesion under flow (p < 0.01) and transendothelial migration towards CXCL8 during in vitro chemotaxis assays (p < 0.05). S1PR1 agonist CYM5442 treatment of endothelial cells also reduced neutrophil transmigration (p < 0.01) and endothelial permeability (p < 0.005), as shown using in vitro permeability assays. S1PR3 agonist had no effects on chemotaxis or permeability. In an in vivo mouse model of renal IRI, S1PR agonism with CYM5442 reduced endothelial permeability as shown by reduced Evan's Blue dye extravasation. Western blot was used to assess phosphorylation at different sites on vascular endothelial (VE)-cadherin and showed that CYM5442 reduced VEGF-mediated phosphorylation. Taken together, the results of this study suggest that reductions in neutrophil infiltration during IRI in response to S1P are mediated primarily by S1PR1 signalling on endothelial cells, possibly by altering phosphorylation of VE-cadherin. The results also demonstrate the therapeutic potential of S1PR1 agonist during IRI.
Collapse
Affiliation(s)
| | | | | | | | - Neil S. Sheerin
- Immunity and Inflammation, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (G.C.W.); (J.G.); (E.G.); (J.A.K.)
| | - Simi Ali
- Immunity and Inflammation, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (G.C.W.); (J.G.); (E.G.); (J.A.K.)
| |
Collapse
|
49
|
Cerutis DR, Weston MD, Miyamoto T. Entering, Linked with the Sphinx: Lysophosphatidic Acids Everywhere, All at Once, in the Oral System and Cancer. Int J Mol Sci 2023; 24:10278. [PMID: 37373424 PMCID: PMC10299546 DOI: 10.3390/ijms241210278] [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: 05/04/2023] [Revised: 06/08/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Oral health is crucial to overall health, and periodontal disease (PDD) is a chronic inflammatory disease. Over the past decade, PDD has been recognized as a significant contributor to systemic inflammation. Here, we relate our seminal work defining the role of lysophosphatidic acid (LPA) and its receptors (LPARs) in the oral system with findings and parallels relevant to cancer. We discuss the largely unexplored fine-tuning potential of LPA species for biological control of complex immune responses and suggest approaches for the areas where we believe more research should be undertaken to advance our understanding of signaling at the level of the cellular microenvironment in biological processes where LPA is a key player so we can better treat diseases such as PDD, cancer, and emerging diseases.
Collapse
Affiliation(s)
- D. Roselyn Cerutis
- Department of Oral Biology, Creighton University School of Dentistry, Omaha, NE 68178, USA;
| | - Michael D. Weston
- Department of Oral Biology, Creighton University School of Dentistry, Omaha, NE 68178, USA;
| | - Takanari Miyamoto
- Department of Periodontics, Creighton University School of Dentistry, Omaha, NE 68178, USA;
| |
Collapse
|
50
|
Weigel C, Bellaci J, Spiegel S. Sphingosine-1-phosphate and its receptors in vascular endothelial and lymphatic barrier function. J Biol Chem 2023; 299:104775. [PMID: 37142226 PMCID: PMC10220486 DOI: 10.1016/j.jbc.2023.104775] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023] Open
Abstract
The vascular and lymphatic systems both comprise a series of structurally distinct vessels lined with an inner layer of endothelial cells that function to provide a semipermeable barrier to blood and lymph. Regulation of the endothelial barrier is critical for maintaining vascular and lymphatic barrier homeostasis. One of the regulators of endothelial barrier function and integrity is sphingosine-1-phosphate (S1P), a bioactive sphingolipid metabolite secreted into the blood by erythrocytes, platelets, and endothelial cells and into the lymph by lymph endothelial cells. Binding of S1P to its G protein-coupled receptors, known as S1PR1-5, regulates its pleiotropic functions. This review outlines the structural and functional differences between vascular and lymphatic endothelium and describes current understanding of the importance of S1P/S1PR signaling in regulation of barrier functions. Most studies thus far have been primarily focused on the role of the S1P/S1PR1 axis in vasculature and have been summarized in several excellent reviews, and thus, we will only discuss new perspectives on the molecular mechanisms of action of S1P and its receptors. Much less is known about the responses of the lymphatic endothelium to S1P and the functions of S1PRs in lymph endothelial cells, and this is the major focus of this review. We also discuss current knowledge related to signaling pathways and factors regulated by the S1P/S1PR axis that control lymphatic endothelial cell junctional integrity. Gaps and limitations in current knowledge are highlighted together with the need to further understand the role of S1P receptors in the lymphatic system.
Collapse
Affiliation(s)
- Cynthia Weigel
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Jacqueline Bellaci
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.
| |
Collapse
|