1
|
Kharel Y, Huang T, Dunnavant K, Foster D, Souza GMPR, Nimchuk KE, Merchak AR, Pavelec CM, Juskiewicz ZJ, Alexander SS, Gaultier A, Abbott SBG, Shin JB, Isakson BE, Xu W, Leitinger N, Santos WL, Lynch KR. Assessment of Spinster homologue 2 (Spns2)-dependent transport of sphingosine-1-phosphate as a therapeutic target. Br J Pharmacol 2025; 182:2014-2030. [PMID: 39894457 PMCID: PMC12034028 DOI: 10.1111/bph.17456] [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: 08/31/2024] [Revised: 11/21/2024] [Accepted: 12/17/2024] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND AND PURPOSE Sphingosine-1-phosphate (S1P) receptor modulator (SRM) drugs suppress immune system function by disrupting lymphocyte trafficking, but SRMs are broadly immunosuppressive with on-target liabilities. Another strategy to modulate the immune system is to block S1P transport. This study tests the hypothesis that blockers of S1P transport (STBs) mediated by Spinster homologue 2 (Spns2) approximate the efficacy of SRMs without their adverse events. EXPERIMENTAL APPROACH We have discovered and optimized STBs to enable investigations of S1P biology and to determine whether S1P transport is a valid drug target. The STB SLF80821178 was administered to rodents to assess its efficacy in a multiple sclerosis model and to test for toxicities associated with SRMs or Spns2-deficient mice. Further, potential biomarkers of STBs, absolute lymphocyte counts (ALCs) in blood and S1P concentrations in plasma and lymph, were measured. KEY RESULTS SLF80821178 resembles SRMs in that it is efficacious in a standard multiple sclerosis model but does not evoke bradycardia or lung leakage, common to the SRM drug class. Also, chronic SLF80821178 administration does not affect auditory responses in adult mice despite the neurosensorial hearing defect observed in Spns2-null mice. While both SRM and STB administration decrease ALCs, the maximal effect is less with an STB (45% vs. 90%). STBs have minimal effects on S1P concentration in plasma or thoracic duct lymph. CONCLUSION AND IMPLICATIONS We found nothing to invalidate Spns2-dependent S1P transport as a drug target. Indeed, STBs could be superior to SRMs as a therapy to modulate immune system function.
Collapse
Affiliation(s)
- Yugesh Kharel
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Tao Huang
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Kyle Dunnavant
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia, USA
| | - Daniel Foster
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia, USA
| | - George M P R Souza
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Katherine E Nimchuk
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| | - Andrea R Merchak
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| | - Caitlin M Pavelec
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Zuzanna J Juskiewicz
- Robert M. Berne Cardiovascular Research Center and Department of Molecular Physiology and Biophysics, University of Virginia, Charlottesville, Virginia, USA
| | - Simon S Alexander
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Alban Gaultier
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| | - Stephen B G Abbott
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Jung-Bum Shin
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center and Department of Molecular Physiology and Biophysics, University of Virginia, Charlottesville, Virginia, USA
| | - Wehao Xu
- Department of Microbiology, Immunology and Cancer Biology and Genetically Engineered Murine Model Core, University of Virginia, Charlottesville, Virginia, USA
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Webster L Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia, USA
| | - Kevin R Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
2
|
Du B, Geng J, Wu B, Wang H, Luo R, Liu H, Zhang R, Shan F, Liu L, Zhang S. Pipelines for lymphocyte homeostasis maintenance during cancer immunotherapy. Front Immunol 2025; 16:1522417. [PMID: 40196122 PMCID: PMC11973195 DOI: 10.3389/fimmu.2025.1522417] [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: 11/04/2024] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
In general, increasing lymphocyte entry into tumor microenvironment (TME) and limiting their efflux will have a positive effect on the efficacy of immunotherapy. Current studies suggest maintenance lymphocyte homeostasis during cancer immunotherapy through the two pipelines tumor-associated high endothelial venules and lymphatic vessels. Tumor-associated high endothelial venules (TA-HEVs) play a key role in cancer immunotherapy through facilitating lymphocyte trafficking to the tumor. While tumor-associated lymphatic vessels, in contrast, may promote the egress of lymphocytes and restrict their function. Therefore, the two traffic control points might be potential to maintain lymphocyte homeostasis in cancer during immunotherapy. Herein, we highlight the unexpected roles of lymphocyte circulation regulated by the two gateways for through reviewing the biological characters and functions of TA-HEVs and tumor-associated lymphatic vessels in the entry, positioning and exit of lymphocyte cells in TME during anti-tumor immunity.
Collapse
Affiliation(s)
- Bensu Du
- China Medical University, Shenyang, China
| | - Jin Geng
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Bin Wu
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Houru Wang
- Northeast Yucai Foreign Language School, Shenyang, China
| | - Ru Luo
- Jinzhou Medical University, Jinzhou, China
| | | | - Rui Zhang
- China Medical University, Shenyang, China
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Lei Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuling Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
3
|
Wang Y, Dowling SD, Rodriguez V, Maciuch J, Mayer M, Therron T, Shaw TN, Gurra MG, Shah CL, Makinde HKM, Ginhoux F, Voehringer D, Harrington CA, Lawrence T, Grainger JR, Cuda CM, Winter DR, Perlman HR. Comprehensive analysis of myeloid reporter mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639159. [PMID: 40060446 PMCID: PMC11888320 DOI: 10.1101/2025.02.24.639159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
Macrophages are a pivotal cell type within the synovial lining and sub-lining of the joint, playing a crucial role in maintaining homeostasis of synovium. Although fate-mapping techniques have been employed to differentiate synovial macrophages from other synovial myeloid cells, no comprehensive study has yet been conducted within the mouse synovial macrophage compartment. In this study, we present, for the first time, lineage tracing results from 18 myeloid-specific fate-mapping models in mouse peripheral blood (PB) and synovial tissue. The identification of synovial macrophages and monocyte-lineage cells through flow cytometry was further validated using cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) datasets. These findings provide a valuable methodological tool for researchers to select appropriate models for studying the function of synovial myeloid cells and serve as a reference for investigations in other tissue types.
Collapse
Affiliation(s)
- Yidan Wang
- Northwestern University, Feinberg School of Medicine. Department of Medicine, Division of Rheumatology. Chicago, IL 60611, USA
| | - Samuel D Dowling
- Northwestern University, Feinberg School of Medicine. Department of Medicine, Division of Rheumatology. Chicago, IL 60611, USA
- Northwestern University, Feinberg School of Medicine. Department of Pediatrics, Division of Rheumatology. Chicago, IL 60611, USA
| | - Vanessa Rodriguez
- Northwestern University, Feinberg School of Medicine. Department of Medicine, Division of Rheumatology. Chicago, IL 60611, USA
| | - Jessica Maciuch
- Northwestern University, Feinberg School of Medicine. Department of Medicine, Division of Rheumatology. Chicago, IL 60611, USA
| | - Meghan Mayer
- Northwestern University, Feinberg School of Medicine. Department of Medicine, Division of Rheumatology. Chicago, IL 60611, USA
| | - Tyler Therron
- Northwestern University, Feinberg School of Medicine. Department of Medicine, Division of Rheumatology. Chicago, IL 60611, USA
| | - Tovah N Shaw
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Miranda G Gurra
- Northwestern University, Feinberg School of Medicine. Department of Medicine, Division of Rheumatology. Chicago, IL 60611, USA
| | - Caroline L Shah
- Northwestern University, Feinberg School of Medicine. Department of Medicine, Division of Rheumatology. Chicago, IL 60611, USA
| | - Hadijat-Kubura M Makinde
- Northwestern University, Feinberg School of Medicine. Department of Medicine, Division of Rheumatology. Chicago, IL 60611, USA
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR). 8A Biomedical Grove IMMUNOS Bldg, Level 3, SINGAPORE 138648
| | - David Voehringer
- University Hospital Erlangen, Department of Infection Biology and Friedrich-Alexander University Erlangen-Nuremberg (FAU). Wasserturmstrasse 3-5, 91054 Erlangen, Germany
| | - Cole A Harrington
- The Ohio State University Wexner Medical Center, Department of Neurology, The Neuroscience Research Institute, College of Medicine, Columbus, OH, USA
| | - Toby Lawrence
- King's College London, Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, London, UK
| | - John R Grainger
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester; Manchester, UK
| | - Carla M Cuda
- Northwestern University, Feinberg School of Medicine. Department of Medicine, Division of Rheumatology. Chicago, IL 60611, USA
| | - Deborah R Winter
- Northwestern University, Feinberg School of Medicine. Department of Medicine, Division of Rheumatology. Chicago, IL 60611, USA
- Center for Human Immunobiology (CHI), Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Harris R Perlman
- Northwestern University, Feinberg School of Medicine. Department of Medicine, Division of Rheumatology. Chicago, IL 60611, USA
| |
Collapse
|
4
|
Askanase AD, D'Cruz D, Kalunian K, Merrill JT, Navarra SV, Cahuzac C, Cornelisse P, Murphy MJ, Strasser DS, Trokan L, Berkani O. Cenerimod, a sphingosine-1-phosphate receptor modulator, versus placebo in patients with moderate-to-severe systemic lupus erythematosus (CARE): an international, double-blind, randomised, placebo-controlled, phase 2 trial. THE LANCET. RHEUMATOLOGY 2025; 7:e21-e32. [PMID: 39586304 DOI: 10.1016/s2665-9913(24)00246-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Sphingosine-1-phosphate (S1P) is a signalling molecule that has an inhibitory role in atherosclerosis, inflammation, cell proliferation, and immunity. Cenerimod is a selective S1P1 receptor modulator under investigation for the treatment of systemic lupus erythematosus (SLE). We aimed to determine the efficacy, safety, and tolerability of four doses of cenerimod in adults with moderate-to-severe SLE receiving standard of care background therapy. METHODS CARE was a double-blind, randomised, placebo-controlled, phase 2 trial, in adults (aged 18-75 years) with moderate-to-severe SLE (a score of at least 6 out of 105 on the SLE disease activity index-2000, modified to exclude leukopenia [mSLEDAI-2K] score). Participants were recruited from 189 hospitals, specialist centres, and outpatient clinics in 22 countries in Asia Pacific, Latin America, Europe, and the USA. Participants were randomly assigned (1:1:1:1:1), using an interactive response technology via balanced block randomisation (block size of 5) and stratified by oral glucocorticoid dose at randomisation and disease activity at screening, to once-daily oral cenerimod at 0·5 mg, 1·0 mg, 2·0 mg, or 4·0 mg or placebo, in addition to stable background SLE therapy, and followed up for 12 months. After 6 months, participants assigned to cenerimod 4·0 mg were randomly assigned again (1:1) to either cenerimod 2.0 mg or placebo for a further 6 months. The primary endpoint was change from baseline to month 6 in mSLEDAI-2K score, assessed in all participants randomly assigned to treatment (full analysis set). To meet the primary endpoint, the doses had to show a significant improvement over placebo, when adjusting for multiplicity, considering the hierarchical testing strategy, per a prespecified plan. Safety analyses included all participants who received at least one dose of study treatment. This study is registered with ClinicalTrials.gov, NCT03742037, and is complete. FINDINGS Between Dec 21, 2018, and Aug 25, 2022, 810 patients were screened and 427 were randomly assigned to 0·5 mg (n=85), 1·0 mg (n=85), 2·0 mg (n=86), and 4·0 mg (n=85) cenerimod or placebo (n=86). Median age was 42 years (IQR 33-51), 406 (95%) of 427 participants were women, 21 (5%) were men, and 337 (79%) were White. At month 6, the least squares mean change from baseline in mSLEDAI-2K score was -2·85 (95% CI -3·60 to -2·10) for the placebo group and -3·24 (-3·98 to -2·49; difference vs placebo -0·39 [95% CI -1·45 to 0·68]; p=0·47) for the cenerimod 0·5 mg group, -3·41 (-4·16 to -2·67; difference vs placebo -0·57 [-1·62 to 0·49]; p=0·29) for the 1·0 mg group, -2·84 (-3·58 to -2·09; difference vs placebo 0·01 [-1·05 to 1·08]; p=0·98) for the 2·0 mg group, and -4·04 (-4·79 to -3·28; difference vs placebo -1·19 [-2·25 to -0·12]; p=0·029) for the 4·0 mg group; hence, the primary endpoint was not met. Treatment-emergent adverse events up to 12 months had no clear treatment-related or dose-related pattern across the groups. At month 6, treatment-emergent lymphopenia was reported in one (1%) of 85 patients who received cenerimod 0·5 mg, five (6%) of 85 patients who received 1·0 mg, nine (10%) of 86 patients who received 2·0 mg, 12 (14%) of 84 patients who received 4·0 mg, and no patients who received placebo. Two deaths due to adverse events occurred (both in the cenerimod 1·0 mg group; one due to acute coronary syndrome and the other due to upper gastrointestinal haemorrhage), and were determined to be unrelated to study treatment. INTERPRETATION The primary endpoint was not met. Cenerimod was well tolerated over 12 months. Cenerimod 4·0 mg is being investigated for the treatment of SLE in two ongoing phase 3 trials (NCT05648500, NCT05672576). FUNDING Idorsia Pharmaceuticals.
Collapse
Affiliation(s)
- Anca D Askanase
- Columbia University Lupus Center, Division of Rheumatology, Columbia University College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| | - David D'Cruz
- Department of Medicine, Guy's and St Thomas' Hospital, London, UK
| | - Kenneth Kalunian
- Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Joan T Merrill
- Division of Rheumatology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sandra V Navarra
- Clinical Projects, Arthritis and Clinical Immunology Program, University of Santo Tomas, Manila, Philippines
| | - Clélia Cahuzac
- Data and Statistical Sciences Department, Hoffmann-La Roche Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
5
|
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
|
6
|
Kharel Y, Huang T, Dunnavant K, Foster D, Souza G, Nimchuk KE, Merchak AR, Pavelec CM, Juskiewicz ZJ, Gaultier A, Abbott S, Shin JB, Isakson BE, Xu W, Leitinger N, Santos WL, Lynch KR. Assessing Spns2-dependent S1P Transport as a Prospective Therapeutic Target. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586765. [PMID: 38746194 PMCID: PMC11092524 DOI: 10.1101/2024.03.26.586765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
S1P (sphingosine 1-phosphate) receptor modulator (SRM) drugs interfere with lymphocyte trafficking by downregulating lymphocyte S1P receptors. While the immunosuppressive activity of SRM drugs has proved useful in treating autoimmune diseases such as multiple sclerosis, that drug class is beset by on-target liabilities such as initial dose bradycardia. The S1P that binds to cell surface lymphocyte S1P receptors is provided by S1P transporters. Mice born deficient in one of these, spinster homolog 2 (Spns2), are lymphocytopenic and have low lymph S1P concentrations. Such observations suggest that inhibition of Spns2-mediated S1P transport might provide another therapeutically beneficial method to modulate immune cell positioning. We report here results using a novel S1P transport blocker (STB), SLF80821178, to investigate the consequences of S1P transport inhibition in rodents. We found that SLF80821178 is efficacious in a multiple sclerosis model but - unlike the SRM fingolimod - neither decreases heart rate nor compromises lung endothelial barrier function. Notably, although Spns2 null mice have a sensorineural hearing defect, mice treated chronically with SLF80821178 have normal hearing acuity. STBs such as SLF80821178 evoke a dose-dependent decrease in peripheral blood lymphocyte counts, which affords a reliable pharmacodynamic marker of target engagement. However, the maximal reduction in circulating lymphocyte counts in response to SLF80821178 is substantially less than the response to SRMs such as fingolimod (50% vs. 90%) due to a lesser effect on T lymphocyte sub-populations by SLF80821178. Finally, in contrast to results obtained with Spns2 deficient mice, lymph S1P concentrations were not significantly changed in response to administration of STBs at doses that evoke maximal lymphopenia, which indicates that current understanding of the mechanism of action of S1P transport inhibitors is incomplete.
Collapse
|
7
|
Shao L, Yang M, Sun T, Xia H, Du D, Li X, Jie Z. Role of solute carrier transporters in regulating dendritic cell maturation and function. Eur J Immunol 2024; 54:e2350385. [PMID: 38073515 DOI: 10.1002/eji.202350385] [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: 07/02/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 02/27/2024]
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that initiate and regulate innate and adaptive immune responses. Solute carrier (SLC) transporters mediate diverse physiological functions and maintain cellular metabolite homeostasis. Recent studies have highlighted the significance of SLCs in immune processes. Notably, upon activation, immune cells undergo rapid and robust metabolic reprogramming, largely dependent on SLCs to modulate diverse immunological responses. In this review, we explore the central roles of SLC proteins and their transported substrates in shaping DC functions. We provide a comprehensive overview of recent studies on amino acid transporters, metal ion transporters, and glucose transporters, emphasizing their essential contributions to DC homeostasis under varying pathological conditions. Finally, we propose potential strategies for targeting SLCs in DCs to bolster immunotherapy for a spectrum of human diseases.
Collapse
Affiliation(s)
- Lin Shao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- School of Life Sciences, Fudan University, Shanghai, China
| | - Mengxin Yang
- School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Tao Sun
- Department of Laboratory Medicine, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Haotang Xia
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Dan Du
- Department of Stomatology, School of Medicine, Xiamen University, Xiamen, Fujian, China
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xun Li
- Department of Laboratory Medicine, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zuliang Jie
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
8
|
Ruddle NH. Regulation, Maintenance, and Remodeling of High Endothelial Venules in Homeostasis, Inflammation, and Cancer. CURRENT OPINION IN PHYSIOLOGY 2023; 36:100705. [PMID: 38523879 PMCID: PMC10956444 DOI: 10.1016/j.cophys.2023.100705] [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] [Indexed: 03/26/2024]
Abstract
High endothelial venules (HEVs), high walled cuboidal blood vessels, through their expression of adhesion molecules and chemokines, allow the entrance of lymphoid cells into primary, secondary, and tertiary lymphoid structures (aka tertiary lymphoid organs). HEV heterogeneity exists between various lymphoid organs in their expression of peripheral node addressin (PNAd) and mucosal vascular addressin adhesion molecule 1(MAdCAM-1). Transcriptomic analyses reveal extensive heterogeneity, plasticity, and regulation of HEV gene expression in ontogeny, acute inflammation, and chronic inflammation within and between lymphoid organs. Rules regulating HEV development are flexible in inflammation. HEVs in tumor tertiary lymphoid structures are diagnostic of favorable clinical outcome and response to Immunotherapy, including immune check point blockade. Immunotherapy induces HEVs and provides an entrance for naïve, central memory, and effector cells and a niche for stem like precursor cells. Understanding HEV regulation will permit their exploitation as routes for drug delivery to autoimmune lesions, rejecting organs, and tumors.
Collapse
Affiliation(s)
- Nancy H Ruddle
- Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06520-8034
| |
Collapse
|
9
|
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
|
10
|
Burgio AL, Shrader CW, Kharel Y, Huang T, Salamoun JM, Lynch KR, Santos WL. 2-Aminobenzoxazole Derivatives as Potent Inhibitors of the Sphingosine-1-Phosphate Transporter Spinster Homolog 2 (Spns2). J Med Chem 2023; 66:5873-5891. [PMID: 37010497 PMCID: PMC10167756 DOI: 10.1021/acs.jmedchem.3c00149] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
The S1P1 receptor is the target of four marketed drugs for the treatment of multiple sclerosis and ulcerative colitis. Targeting an S1P exporter, specifically Spns2, that is "upstream" of S1P receptor engagement is an alternate strategy that might recapitulate the efficacy of S1P receptor modulators without cardiac toxicity. We recently reported the first Spns2 inhibitor SLF1081851 (16d) that has modest potency with in vivo activity. To develop more potent compounds, we initiated a structure-activity relationship study that identified 2-aminobenzoxazole as a viable scaffold. Our studies revealed SLB1122168 (33p), which is a potent inhibitor (IC50 = 94 ± 6 nM) of Spns2-mediated S1P release. Administration of 33p to mice and rats resulted in a dose-dependent decrease in circulating lymphocytes, a pharmacodynamic indication of Spns2 inhibition. 33p provides a valuable tool compound to explore both the therapeutic potential of targeting Spns2 and the physiologic consequences of selective S1P export inhibition.
Collapse
Affiliation(s)
- Ariel L. Burgio
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061
| | - Christopher W. Shrader
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061
| | - Yugesh Kharel
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908
| | - Tao Huang
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908
| | - Joseph M. Salamoun
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061
| | - Kevin R. Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908
| | - Webster L. Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061
| |
Collapse
|
11
|
Pezhman L, Hopkin SJ, Begum J, Heising S, Nasteska D, Wahid M, Ed Rainger G, Hodson DJ, Iqbal AJ, Chimen M, McGettrick HM. PEPITEM modulates leukocyte trafficking to reduce obesity-induced inflammation. Clin Exp Immunol 2023; 212:1-10. [PMID: 36891817 PMCID: PMC10081110 DOI: 10.1093/cei/uxad022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/10/2023] [Accepted: 02/13/2023] [Indexed: 03/10/2023] Open
Abstract
Dysregulation of leukocyte trafficking, lipid metabolism, and other metabolic processes are the hallmarks that underpin and drive pathology in obesity. Current clinical management targets alternations in lifestyle choices (e.g. exercise, weight loss) to limit the impact of the disease. Crucially, re-gaining control over the pathogenic cellular and molecular processes may offer an alternative, complementary strategy for obese patients. Here we investigate the impact of the immunopeptide, PEPITEM, on pancreas homeostasis and leukocyte trafficking in mice on high-fed obesogenic diet (HFD). Both prophylactic and therapeutic treatment with PEPITEM alleviated the effects of HFD on the pancreas, reducing pancreatic beta cell size. Moreover, PEPITEM treatment also limited T-cell trafficking (CD4+ T-cells and KLRG1+ CD3+ T-cells) to obese visceral, but not subcutaneous, adipose tissue. Similarly, PEPITEM treatment reduced macrophage numbers within the peritoneal cavity of mice on HFD diet at both 6 and 12 weeks. By contrast, PEPITEM therapy elevated numbers of T and B cells were observed in the secondary lymphoid tissues (e.g. spleen and inguinal lymph node) when compared to the untreated HFD controls. Collectively our data highlights the potential for PEPITEM as a novel therapy to combat the systemic low-grade inflammation experienced in obesity and minimize the impact of obesity on pancreatic homeostasis. Thus, offering an alternative strategy to reduce the risk of developing obesity-related co-morbidities, such as type 2 diabetes mellitus, in individuals at high risk and struggling to control their weight through lifestyle modifications.
Collapse
Affiliation(s)
- Laleh Pezhman
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Sophie J Hopkin
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Jenefa Begum
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Silke Heising
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Daniela Nasteska
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Mussarat Wahid
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - G Ed Rainger
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - David J Hodson
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Asif J Iqbal
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Myriam Chimen
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Helen M McGettrick
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
12
|
Vella G, Hua Y, Bergers G. High endothelial venules in cancer: Regulation, function, and therapeutic implication. Cancer Cell 2023; 41:527-545. [PMID: 36827979 DOI: 10.1016/j.ccell.2023.02.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/12/2023] [Accepted: 02/01/2023] [Indexed: 02/25/2023]
Abstract
The lack of sufficient intratumoral CD8+ T lymphocytes is a significant obstacle to effective immunotherapy in cancer. High endothelial venules (HEVs) are organ-specific and specialized postcapillary venules uniquely poised to facilitate the transmigration of lymphocytes to lymph nodes (LNs) and other secondary lymphoid organs (SLOs). HEVs can also form in human and murine cancer (tumor HEVs [TU-HEVs]) and contribute to the generation of diffuse T cell-enriched aggregates or tertiary lymphoid structures (TLSs), which are commonly associated with a good prognosis. Thus, therapeutic induction of TU-HEVs may provide attractive avenues to induce and sustain the efficacy of immunotherapies by overcoming the major restriction of T cell exclusion from the tumor microenvironment. In this review, we provide current insight into the commonalities and discrepancies of HEV formation and regulation in LNs and tumors and discuss the specific function and significance of TU-HEVs in eliciting, predicting, and aiding anti-tumoral immunity.
Collapse
Affiliation(s)
- Gerlanda Vella
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Yichao Hua
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
13
|
Higashiyama M, Miura S, Hokari R. Modulation by luminal factors on the functions and migration of intestinal innate immunity. Front Immunol 2023; 14:1113467. [PMID: 36860849 PMCID: PMC9968923 DOI: 10.3389/fimmu.2023.1113467] [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: 12/21/2022] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
Luminal antigens, nutrients, metabolites from commensal bacteria, bile acids, or neuropeptides influence the function and trafficking of immune cells in the intestine. Among the immune cells in the gut, innate lymphoid cells, including macrophages, neutrophils, dendritic cells, mast cells, and innate lymphoid cells, play an important role for the maintenance of intestinal homeostasis through a rapid immune response to luminal pathogens. These innate cells are influenced by several luminal factors, possibly leading to dysregulated gut immunity and intestinal disorders such as inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), and intestinal allergy. Luminal factors are sensed by distinct neuro-immune cell units, which also have a strong impact on immunoregulation of the gut. Immune cell trafficking from the blood stream through the lymphatic organ to lymphatics, an essential function for immune responses, is also modulated by luminal factors. This mini-review examines knowledge of luminal and neural factors that regulate and modulate response and migration of leukocytes including innate immune cells, some of which are clinically associated with pathological intestinal inflammation.
Collapse
Affiliation(s)
- Masaaki Higashiyama
- Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan,*Correspondence: Masaaki Higashiyama,
| | - Soichiro Miura
- International University of Health and Welfare, Tokyo, Japan
| | - Ryota Hokari
- Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| |
Collapse
|
14
|
Levesque MV, Hla T. Signal Transduction and Gene Regulation in the Endothelium. Cold Spring Harb Perspect Med 2023; 13:a041153. [PMID: 35667710 PMCID: PMC9722983 DOI: 10.1101/cshperspect.a041153] [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] [Indexed: 01/07/2023]
Abstract
Extracellular signals act on G-protein-coupled receptors (GPCRs) to regulate homeostasis and adapt to stress. This involves rapid intracellular post-translational responses and long-lasting gene-expression changes that ultimately determine cellular phenotype and fate changes. The lipid mediator sphingosine 1-phosphate (S1P) and its receptors (S1PRs) are examples of well-studied GPCR signaling axis essential for vascular development, homeostasis, and diseases. The biochemical cascades involved in rapid S1P signaling are well understood. However, gene-expression regulation by S1PRs are less understood. In this review, we focus our attention to how S1PRs regulate nuclear chromatin changes and gene transcription to modulate vascular and lymphatic endothelial phenotypic changes during embryonic development and adult homeostasis. Because S1PR-targeted drugs approved for use in the treatment of autoimmune diseases cause substantial vascular-related adverse events, these findings are critical not only for general understanding of stimulus-evoked gene regulation in the vascular endothelium, but also for therapeutic development of drugs for autoimmune and perhaps vascular diseases.
Collapse
Affiliation(s)
- Michel V Levesque
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
15
|
Gluschke H, Siegert E, Minich WB, Hackler J, Riemekasten G, Kuebler WM, Simmons S, Schomburg L. Autoimmunity to Sphingosine-1-Phosphate-Receptors in Systemic Sclerosis and Pulmonary Arterial Hypertension. Front Immunol 2022; 13:935787. [PMID: 35860272 PMCID: PMC9289471 DOI: 10.3389/fimmu.2022.935787] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/07/2022] [Indexed: 12/04/2022] Open
Abstract
Context Pulmonary arterial hypertension (PAH) is a frequent extracutaneous manifestation of systemic sclerosis (SSc). PAH is characterized by increased vasomotor tone, progressive remodeling of pulmonary arteries and arterioles, consequentially increased pulmonary vascular resistance, right heart hypertrophy, and eventually right ventricular failure. Autoimmunity against G-protein coupled receptors (GPCRs) has been implicated in the development of SSc-associated PAH. Sphingosine-1-phosphate (S1P) receptors (S1PR) present a potential, yet so far untested antigen for PAH autoimmunity, given the documented role of S1P/S1PR signaling in PAH pathogenesis. Objective We hypothesized that S1P receptors (S1PR) may constitute autoantigens in human patients, and that the prevalence of autoantibodies (aAb) to S1PR1, S1PR2 and S1PR3 is elevated in SSc patients and associated with PAH. Methods For this exploratory study, serum samples from 158 SSc patients, 58 of whom with PAH, along with 333 healthy control subjects were screened for S1PR-aAb. S1PR1-3 were expressed as fusion proteins with luciferase in human embryonic kidney cells and used to establish novel in-vitro assays for detecting and quantifying S1PR-aAb. The fusion proteins were incubated with serum samples, the aAb-S1PR complexes formed were precipitated by protein-A, washed and tested for luciferase activity. Commercial anti-S1PR-antibodies were used to verify specificity of the assays. Results All three assays showed dose-dependent signal intensities when tested with S1PR-subtype specific commercial antibodies. Natural aAb to each S1PR were detected in healthy controls with a prevalence of <10% each, i.e., 2.7% for S1PR1-aAb, 3.6% for S1PR2-aAb, and 8.3% for S1PR3. The respective prevalence was higher in the cohort of SSc patients without PAH, with 17.1% for S1PR1-aAb, 19.0% for S1PR2-aAb, and 21.5% for S1PR3. In the subgroup of SSc patients with PAH, prevalence of aAb to S1PR2 and S1PR3 was further elevated to 25.9% for S1PR2-aAb, and 27.6% for S1PR3. Notably, the majority of patients with positive S1PR2-aAb (60.7%) or S1PR3-aAb (71.9%) displayed interstitial lung disease. Conclusion S1PR1–3 can constitute autoantigens in humans, particularly in SSC patients with PAH. The potential pathophysiological significance for the etiology of the disease is currently unknown, but the elevated prevalence of S1PR2-aAb and S1PR3-aAb in SSC patients with PAH merits further mechanistic investigations.
Collapse
Affiliation(s)
- Hans Gluschke
- Institute for Experimental Endocrinology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Elise Siegert
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Waldemar B. Minich
- Institute for Experimental Endocrinology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Julian Hackler
- Institute for Experimental Endocrinology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Gabriela Riemekasten
- Department of Rheumatology, University Medical Center Schleswig Holstein, Lübeck, Germany
| | - Wolfgang M. Kuebler
- Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Deutschs Zentrum für Herz-Kreislauf-Forschung e.V. (DZHK) (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Szandor Simmons
- Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Deutschs Zentrum für Herz-Kreislauf-Forschung e.V. (DZHK) (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- *Correspondence: Szandor Simmons, ; Lutz Schomburg,
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- *Correspondence: Szandor Simmons, ; Lutz Schomburg,
| |
Collapse
|
16
|
Fritzemeier R, Foster D, Peralta A, Payette M, Kharel Y, Huang T, Lynch KR, Santos WL. Discovery of In Vivo Active Sphingosine-1-phosphate Transporter (Spns2) Inhibitors. J Med Chem 2022; 65:7656-7681. [PMID: 35609189 PMCID: PMC9733493 DOI: 10.1021/acs.jmedchem.1c02171] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a pleiotropic signaling molecule that interacts with five G-protein-coupled receptors (S1P1-5) to regulate cellular signaling pathways. S1P export is facilitated by Mfsd2b and spinster homologue 2 (Spns2). While mouse genetic studies suggest that Spns2 functions to maintain lymph S1P, Spns2 inhibitors are necessary to understand its biology and to learn whether Spns2 is a viable drug target. Herein, we report a structure-activity relationship study that identified the first Spns2 inhibitor 16d (SLF1081851). In vitro studies in HeLa cells demonstrated that 16d inhibited S1P release with an IC50 of 1.93 μM. Administration of 16d to mice and rats drove significant decreases in circulating lymphocyte counts and plasma S1P concentrations, recapitulating the phenotype observed in mice made deficient in Spns2. Thus, 16d has the potential for development and use as a probe to investigate Spns2 biology and to determine the potential of Spns2 as a drug target.
Collapse
Affiliation(s)
- Russell Fritzemeier
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia 24060, United States
| | - Daniel Foster
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia 24060, United States
| | - Ashley Peralta
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia 24060, United States
| | - Michael Payette
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia 24060, United States
| | - Yugesh Kharel
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Tao Huang
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Kevin R Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Webster L Santos
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia 24060, United States
- Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24060, United States
| |
Collapse
|
17
|
Vishwakarma S, Joshi D, Pandey R, Das S, Mukhopadhyay S, Rai R, Singhal R, Kapoor N, Kumar A. Downregulation of Lipid Phosphate Phosphatase 3 Correlates With Tumor-Infiltrating Immune Cells in Oral Cancer. Cureus 2022; 14:e23553. [PMID: 35494957 PMCID: PMC9045791 DOI: 10.7759/cureus.23553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2022] [Indexed: 11/27/2022] Open
Abstract
Background Sphingosine-1-phosphate (S1P) is a potent oncogenic lipid. Intracellular levels of S1P are tightly regulated by eight S1P-metabolizing enzymes. S1P synthesis is catalyzed by two sphingosine kinases, i.e., sphingosine kinase 1 (SphK1) and sphingosine kinase 2 (SphK2). Five lipid phosphatases (two S1P phosphatases and lipid phosphate phosphatases (LPPs) 1, 2, and 3) reversibly convert S1P back to sphingosine. Previously, we have determined the mRNA expression profile of eight S1P-metabolizing enzymes in tumor tissues and adjacent normal tissues from oral squamous cell carcinoma (OSCC) patients. Except for SphK1, the role of S1P-metabolizing enzymes in OSCC has been poorly studied. Methods We have determined the protein expression of four S1P-metabolizing enzymes (SphK1, SphK2, sphingosine-1-phosphate phosphatase 1 (SGPP1), and lipid phosphate phosphatase 3 (LPP3)) by immunohistochemistry (IHC) in tumor tissues of 46 OSCC patients. Six subjects with non-dysplastic oral mucosa were also included in the study. The immunoreactivity score (IRS) was calculated for each protein in every subject. Further, we determined the associations of expression of S1P-metabolizing enzymes with clinicopathological features of OSCC patients. Results We demonstrate the low IRS for SphK2 and LPP3 in OSCC tumors. Importantly, expression of SphK2 and LPP3 was downregulated in malignant epithelial cells compared to non-malignant mucosa. Further, LPP3 expression negatively correlated with tumor‑node‑metastasis (TNM) staging of patients (r = −0.307, p = 0.043). Importantly, expression of LPP3 in tumors was found to be an independent predictor of perinodal extension (b = −0.440, p = 0.009), lymphovascular invasion (b = −0.614, p < 0.001), lymph node ratio (b = 0.336, p = 0.039), and TNM staging (b = −0.364, p = 0.030). Conclusion Taken together, our data show that expression of SphK2 and LPP3 is decreased compared to normal mucosa. Thus, the S1P signaling pathway could represent a potential therapeutic target.
Collapse
|
18
|
Funk-Hilsdorf TC, Behrens F, Grune J, Simmons S. Dysregulated Immunity in Pulmonary Hypertension: From Companion to Composer. Front Physiol 2022; 13:819145. [PMID: 35250621 PMCID: PMC8891568 DOI: 10.3389/fphys.2022.819145] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 01/20/2022] [Indexed: 12/26/2022] Open
Abstract
Pulmonary hypertension (PH) represents a grave condition associated with high morbidity and mortality, emphasizing a desperate need for innovative and targeted therapeutic strategies. Cumulative evidence suggests that inflammation and dysregulated immunity interdependently affect maladaptive organ perfusion and congestion as hemodynamic hallmarks of the pathophysiology of PH. The role of altered cellular and humoral immunity in PH gains increasing attention, especially in pulmonary arterial hypertension (PAH), revealing novel mechanistic insights into the underlying immunopathology. Whether these immunophysiological aspects display a universal character and also hold true for other types of PH (e.g., PH associated with left heart disease, PH-LHD), or whether there are unique immunological signatures depending on the underlying cause of disease are points of consideration and discussion. Inflammatory mediators and cellular immune circuits connect the local inflammatory landscape in the lung and heart through inter-organ communication, involving, e.g., the complement system, sphingosine-1-phosphate (S1P), cytokines and subsets of, e.g., monocytes, macrophages, natural killer (NK) cells, dendritic cells (DCs), and T- and B-lymphocytes with distinct and organ-specific pro- and anti-inflammatory functions in homeostasis and disease. Perivascular macrophage expansion and monocyte recruitment have been proposed as key pathogenic drivers of vascular remodeling, the principal pathological mechanism in PAH, pinpointing toward future directions of anti-inflammatory therapeutic strategies. Moreover, different B- and T-effector cells as well as DCs may play an important role in the pathophysiology of PH as an imbalance of T-helper-17-cells (TH17) activated by monocyte-derived DCs, a potentially protective role of regulatory T-cells (Treg) and autoantibody-producing plasma cells occur in diverse PH animal models and human PH. This article highlights novel aspects of the innate and adaptive immunity and their interaction as disease mediators of PH and its specific subtypes, noticeable inflammatory mediators and summarizes therapeutic targets and strategies arising thereby.
Collapse
Affiliation(s)
- Teresa C. Funk-Hilsdorf
- Junior Research Group “Immunodynamics”, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Felix Behrens
- Junior Research Group “Immunodynamics”, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Jana Grune
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Szandor Simmons
- Junior Research Group “Immunodynamics”, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- *Correspondence: Szandor Simmons,
| |
Collapse
|
19
|
Sasset L, Di Lorenzo A. Sphingolipid Metabolism and Signaling in Endothelial Cell Functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:87-117. [PMID: 35503177 DOI: 10.1007/978-981-19-0394-6_8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The endothelium, inner layer of blood vessels, constitutes a metabolically active paracrine, endocrine, and autocrine organ, able to sense the neighboring environment and exert a variety of biological functions important to preserve the health of vasculature, tissues, and organs. Sphingolipids are both fundamental structural components of the eukaryotic membranes and signaling molecules regulating a variety of biological functions. Ceramide and sphingosine-1-phosphate (S1P), bioactive sphingolipids, have emerged as important regulators of cardiovascular functions in health and disease. In this review we discuss recent insights into the role of ceramide and S1P biosynthesis and signaling in regulating endothelial cell functions, in health and diseases. We also highlight advances into the mechanisms regulating serine palmitoyltransferase, the first and rate-limiting enzyme of de novo sphingolipid biosynthesis, with an emphasis on its inhibitors, ORMDL and NOGO-B. Understanding the molecular mechanisms regulating the sphingolipid de novo biosynthesis may provide the foundation for therapeutic modulation of this pathway in a variety of conditions, including cardiovascular diseases, associated with derangement of this pathway.
Collapse
Affiliation(s)
- Linda Sasset
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Feil Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Feil Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
20
|
Tatsumi N, Codrington AL, El-Fenej J, Phondge V, Kumamoto Y. Effective CD4 T cell priming requires repertoire scanning by CD301b + migratory cDC2 cells upon lymph node entry. Sci Immunol 2021; 6:eabg0336. [PMID: 34890253 DOI: 10.1126/sciimmunol.abg0336] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Naoya Tatsumi
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA.,Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Alicia L Codrington
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA.,Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Jihad El-Fenej
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA.,Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Varoon Phondge
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA.,Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Yosuke Kumamoto
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA.,Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
21
|
Menzel L, Zschummel M, Crowley T, Franke V, Grau M, Ulbricht C, Hauser A, Siffrin V, Bajénoff M, Acton SE, Akalin A, Lenz G, Willimsky G, Höpken UE, Rehm A. Lymphocyte access to lymphoma is impaired by high endothelial venule regression. Cell Rep 2021; 37:109878. [PMID: 34706240 PMCID: PMC8567313 DOI: 10.1016/j.celrep.2021.109878] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/27/2021] [Accepted: 10/01/2021] [Indexed: 12/15/2022] Open
Abstract
Blood endothelial cells display remarkable plasticity depending on the demands of a malignant microenvironment. While studies in solid tumors focus on their role in metabolic adaptations, formation of high endothelial venules (HEVs) in lymph nodes extends their role to the organization of immune cell interactions. As a response to lymphoma growth, blood vessel density increases; however, the fate of HEVs remains elusive. Here, we report that lymphoma causes severe HEV regression in mouse models that phenocopies aggressive human B cell lymphomas. HEV dedifferentiation occurrs as a consequence of a disrupted lymph-carrying conduit system. Mechanosensitive fibroblastic reticular cells then deregulate CCL21 migration paths, followed by deterioration of dendritic cell proximity to HEVs. Loss of this crosstalk deprives HEVs of lymphotoxin-β-receptor (LTβR) signaling, which is indispensable for their differentiation and lymphocyte transmigration. Collectively, this study reveals a remodeling cascade of the lymph node microenvironment that is detrimental for immune cell trafficking in lymphoma.
Collapse
Affiliation(s)
- Lutz Menzel
- Translational Tumorimmunology, Max-Delbrück-Center for Molecular Medicine Berlin, Germany, 13125 Berlin, Germany
| | - Maria Zschummel
- Microenvironmental Regulation in Autoimmunity and Cancer, Max-Delbrück-Center for Molecular Medicine Berlin, 13125 Berlin, Germany
| | - Tadhg Crowley
- Neuroimmunology Laboratory, Max-Delbrück-Center for Molecular Medicine Berlin, Germany, 13125 Berlin, Germany
| | - Vedran Franke
- Bioinformatics & Omics Data Science Platform, BIMSB at Max-Delbrück-Center for Molecular Medicine Berlin, 13125 Berlin, Germany
| | - Michael Grau
- Medical Department A for Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Carolin Ulbricht
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, and Immune Dynamics, Deutsches Rheumaforschungszentrum Berlin, 10117 Berlin, Germany
| | - Anja Hauser
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, and Immune Dynamics, Deutsches Rheumaforschungszentrum Berlin, 10117 Berlin, Germany
| | - Volker Siffrin
- Neuroimmunology Laboratory, Max-Delbrück-Center for Molecular Medicine Berlin, Germany, 13125 Berlin, Germany; Neuroimmunology Laboratory, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Marc Bajénoff
- Aix Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, 13288 Marseille, France
| | - Sophie E Acton
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, WC1E 6BT London, UK
| | - Altuna Akalin
- Bioinformatics & Omics Data Science Platform, BIMSB at Max-Delbrück-Center for Molecular Medicine Berlin, 13125 Berlin, Germany
| | - Georg Lenz
- Medical Department A for Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Gerald Willimsky
- Institute of Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13125 Berlin, Germany; German Cancer Research Center, 69120 Heidelberg, Germany; German Cancer Consortium, partner site Berlin, Germany
| | - Uta E Höpken
- Microenvironmental Regulation in Autoimmunity and Cancer, Max-Delbrück-Center for Molecular Medicine Berlin, 13125 Berlin, Germany
| | - Armin Rehm
- Translational Tumorimmunology, Max-Delbrück-Center for Molecular Medicine Berlin, Germany, 13125 Berlin, Germany.
| |
Collapse
|
22
|
Vella G, Guelfi S, Bergers G. High Endothelial Venules: A Vascular Perspective on Tertiary Lymphoid Structures in Cancer. Front Immunol 2021; 12:736670. [PMID: 34484246 PMCID: PMC8416033 DOI: 10.3389/fimmu.2021.736670] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 07/30/2021] [Indexed: 01/22/2023] Open
Abstract
High endothelial venules (HEVs) are specialized postcapillary venules composed of cuboidal blood endothelial cells that express high levels of sulfated sialomucins to bind L-Selectin/CD62L on lymphocytes, thereby facilitating their transmigration from the blood into the lymph nodes (LN) and other secondary lymphoid organs (SLO). HEVs have also been identified in human and murine tumors in predominantly CD3+T cell-enriched areas with fewer CD20+B-cell aggregates that are reminiscent of tertiary lymphoid-like structures (TLS). While HEV/TLS areas in human tumors are predominantly associated with increased survival, tumoral HEVs (TU-HEV) in mice have shown to foster lymphocyte-enriched immune centers and boost an immune response combined with different immunotherapies. Here, we discuss the current insight into TU-HEV formation, function, and regulation in tumors and elaborate on the functional implication, opportunities, and challenges of TU-HEV formation for cancer immunotherapy.
Collapse
Affiliation(s)
- Gerlanda Vella
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, Vlaams Instituut voor Biotechnologie (VIB)-Center for Cancer Biology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Sophie Guelfi
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, Vlaams Instituut voor Biotechnologie (VIB)-Center for Cancer Biology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, Vlaams Instituut voor Biotechnologie (VIB)-Center for Cancer Biology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium.,Department of Neurological Surgery, UCSF Comprehensive Cancer Center, University of California San Francisco (UCSF), San Francisco, CA, United States
| |
Collapse
|
23
|
Akhter MZ, Chandra Joshi J, Balaji Ragunathrao VA, Maienschein-Cline M, Proia RL, Malik AB, Mehta D. Programming to S1PR1 + Endothelial Cells Promotes Restoration of Vascular Integrity. Circ Res 2021; 129:221-236. [PMID: 33926208 PMCID: PMC8273089 DOI: 10.1161/circresaha.120.318412] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 04/28/2021] [Indexed: 12/31/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Md Zahid Akhter
- Pharmacology and Regenerative Medicine and Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago (M.Z.A., J.C.J., V.A.B.R., A.B.M., D.M.)
| | - Jagdish Chandra Joshi
- Pharmacology and Regenerative Medicine and Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago (M.Z.A., J.C.J., V.A.B.R., A.B.M., D.M.)
| | - Vijay Avin Balaji Ragunathrao
- Pharmacology and Regenerative Medicine and Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago (M.Z.A., J.C.J., V.A.B.R., A.B.M., D.M.)
| | | | - Richard L Proia
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD (R.L.P.)
| | - Asrar B Malik
- Pharmacology and Regenerative Medicine and Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago (M.Z.A., J.C.J., V.A.B.R., A.B.M., D.M.)
| | - Dolly Mehta
- Pharmacology and Regenerative Medicine and Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago (M.Z.A., J.C.J., V.A.B.R., A.B.M., D.M.)
| |
Collapse
|
24
|
Blanchard L, Girard JP. High endothelial venules (HEVs) in immunity, inflammation and cancer. Angiogenesis 2021; 24:719-753. [PMID: 33956259 PMCID: PMC8487881 DOI: 10.1007/s10456-021-09792-8] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022]
Abstract
High endothelial venules (HEVs) are specialized blood vessels mediating lymphocyte trafficking to lymph nodes (LNs) and other secondary lymphoid organs. By supporting high levels of lymphocyte extravasation from the blood, HEVs play an essential role in lymphocyte recirculation and immune surveillance for foreign invaders (bacterial and viral infections) and alterations in the body’s own cells (neoantigens in cancer). The HEV network expands during inflammation in immune-stimulated LNs and is profoundly remodeled in metastatic and tumor-draining LNs. HEV-like blood vessels expressing high levels of the HEV-specific sulfated MECA-79 antigens are induced in non-lymphoid tissues at sites of chronic inflammation in many human inflammatory and allergic diseases, including rheumatoid arthritis, Crohn’s disease, allergic rhinitis and asthma. Such vessels are believed to contribute to the amplification and maintenance of chronic inflammation. MECA-79+ tumor-associated HEVs (TA-HEVs) are frequently found in human tumors in CD3+ T cell-rich areas or CD20+ B-cell rich tertiary lymphoid structures (TLSs). TA-HEVs have been proposed to play important roles in lymphocyte entry into tumors, a process essential for successful antitumor immunity and lymphocyte-mediated cancer immunotherapy with immune checkpoint inhibitors, vaccines or adoptive T cell therapy. In this review, we highlight the phenotype and function of HEVs in homeostatic, inflamed and tumor-draining lymph nodes, and those of HEV-like blood vessels in chronic inflammatory diseases. Furthermore, we discuss the role and regulation of TA-HEVs in human cancer and mouse tumor models.
Collapse
Affiliation(s)
- Lucas Blanchard
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
25
|
A Bioassay Using a Pentadecanal Derivative to Measure S1P Lyase Activity. Int J Mol Sci 2021; 22:ijms22031438. [PMID: 33535437 PMCID: PMC7867068 DOI: 10.3390/ijms22031438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 01/02/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a unique lipid ligand binding to S1P receptors to transduce various cell survival or proliferation signals via small G proteins. S1P lyase (S1PL) is the specific enzyme that degrades S1P to phosphoethanolamine and (2E)-hexadecenal and therefore regulates S1P levels. S1PL also degrades dihydrosphingosine-1-phosphate (Sa1P), with a higher affinity to produce hexadecanal. Here, we developed a newly designed assay using a C17-Sa1P substrate that degrades into pentadecanal and phosphoethanolamine. For higher sensitivity in pentadecanal analysis, we developed a quantitative protocol as well as a 5,5-dimethyl cyclohexanedione (5,5-dimethyl CHD) derivatization method. The derivatization conditions were optimized for the reaction time, temperature, and concentrations of the 5,5-dimethyl CHD reagent, acetic acid, and ammonium acetate. The S1PL reaction in the cell lysate after spiking 20 µM of C17-Sa1P for 20 min was linear to the total protein concentrations of 50 µg. The S1PL levels (4 pmol/mg/min) were readily detected in this HPLC with fluorescence detection (λex = 366 nm, λem = 455 nm). The S1PL-catalyzed reaction was linear over 30 min and yielded a Km value of 2.68 μM for C17-Sa1P. This new method was validated to measure the S1PL activity of mouse embryonal carcinoma cell lines of the standard cell (F9-0), S1PL knockdown cells (F9-2), and S1PL-overexpressed cells (F9-4). Furthermore, we treated F9-4 cells with different S1PL inhibitors such as FTY720, 4-deoxypyridoxine (DOP), and the deletion of pyridoxal-5-phosphate (P5P), an essential cofactor for S1PL activity, and observed a significant decrease in pentadecanal relative to the untreated cells. In conclusion, we developed a highly sensitive S1PL assay using a C17-Sa1P substrate for pentadecanal quantification for application in the characterization of S1PL activity in vitro.
Collapse
|
26
|
Engelbrecht E, MacRae CA, Hla T. Lysolipids in Vascular Development, Biology, and Disease. Arterioscler Thromb Vasc Biol 2020; 41:564-584. [PMID: 33327749 DOI: 10.1161/atvbaha.120.305565] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Membrane phospholipid metabolism forms lysophospholipids, which possess unique biochemical and biophysical properties that influence membrane structure and dynamics. However, lysophospholipids also function as ligands for G-protein-coupled receptors that influence embryonic development, postnatal physiology, and disease. The 2 most well-studied species-lysophosphatidic acid and S1P (sphingosine 1-phosphate)-are particularly relevant to vascular development, physiology, and cardiovascular diseases. This review summarizes the role of lysophosphatidic acid and S1P in vascular developmental processes, endothelial cell biology, and their roles in cardiovascular disease processes. In addition, we also point out the apparent connections between lysophospholipid biology and the Wnt (int/wingless family) pathway, an evolutionarily conserved fundamental developmental signaling system. The discovery that components of the lysophospholipid signaling system are key genetic determinants of cardiovascular disease has warranted current and future research in this field. As pharmacological approaches to modulate lysophospholipid signaling have entered the clinical sphere, new findings in this field promise to influence novel therapeutic strategies in cardiovascular diseases.
Collapse
Affiliation(s)
- Eric Engelbrecht
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery (E.E., T.H.), Harvard Medical School, Boston, MA
| | - Calum A MacRae
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Department of Medicine (C.A.M.), Harvard Medical School, Boston, MA
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery (E.E., T.H.), Harvard Medical School, Boston, MA
| |
Collapse
|
27
|
Engelbrecht E, Levesque MV, He L, Vanlandewijck M, Nitzsche A, Niazi H, Kuo A, Singh SA, Aikawa M, Holton K, Proia RL, Kono M, Pu WT, Camerer E, Betsholtz C, Hla T. Sphingosine 1-phosphate-regulated transcriptomes in heterogenous arterial and lymphatic endothelium of the aorta. eLife 2020; 9:52690. [PMID: 32091396 PMCID: PMC7054001 DOI: 10.7554/elife.52690] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 02/22/2020] [Indexed: 12/17/2022] Open
Abstract
Despite the medical importance of G protein-coupled receptors (GPCRs), in vivo cellular heterogeneity of GPCR signaling and downstream transcriptional responses are not understood. We report the comprehensive characterization of transcriptomes (bulk and single-cell) and chromatin domains regulated by sphingosine 1-phosphate receptor-1 (S1PR1) in adult mouse aortic endothelial cells. First, S1PR1 regulates NFκB and nuclear glucocorticoid receptor pathways to suppress inflammation-related mRNAs. Second, S1PR1 signaling in the heterogenous endothelial cell (EC) subtypes occurs at spatially-distinct areas of the aorta. For example, a transcriptomically distinct arterial EC population at vascular branch points (aEC1) exhibits ligand-independent S1PR1/ß-arrestin coupling. In contrast, circulatory S1P-dependent S1PR1/ß-arrestin coupling was observed in non-branch point aEC2 cells that exhibit an inflammatory gene expression signature. Moreover, S1P/S1PR1 signaling regulates the expression of lymphangiogenic and inflammation-related transcripts in an adventitial lymphatic EC (LEC) population in a ligand-dependent manner. These insights add resolution to existing concepts of endothelial heterogeneity, GPCR signaling and S1P biology.
Collapse
Affiliation(s)
- Eric Engelbrecht
- Vascular Biology Program, Boston Children's Hospital, Deapartment of Surgery, Harvard Medical School, Boston, United States
| | - Michel V Levesque
- Vascular Biology Program, Boston Children's Hospital, Deapartment of Surgery, Harvard Medical School, Boston, United States
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.,Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Karolinska Institutet, Huddinge, Sweden
| | - Michael Vanlandewijck
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.,Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Karolinska Institutet, Huddinge, Sweden
| | - Anja Nitzsche
- Université de Paris, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Hira Niazi
- Université de Paris, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Andrew Kuo
- Vascular Biology Program, Boston Children's Hospital, Deapartment of Surgery, Harvard Medical School, Boston, United States
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Kristina Holton
- Harvard Medical School Research Computing, Boston, United States
| | - Richard L Proia
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Mari Kono
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, United States.,Harvard Stem Cell Institute, Harvard University, Cambridge, United States
| | - Eric Camerer
- Université de Paris, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.,Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Karolinska Institutet, Huddinge, Sweden
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital, Deapartment of Surgery, Harvard Medical School, Boston, United States
| |
Collapse
|
28
|
Druggable Sphingolipid Pathways: Experimental Models and Clinical Opportunities. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1274:101-135. [PMID: 32894509 DOI: 10.1007/978-3-030-50621-6_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Intensive research in the field of sphingolipids has revealed diverse roles in cell biological responses and human health and disease. This immense molecular family is primarily represented by the bioactive molecules ceramide, sphingosine, and sphingosine 1-phosphate (S1P). The flux of sphingolipid metabolism at both the subcellular and extracellular levels provides multiple opportunities for pharmacological intervention. The caveat is that perturbation of any single node of this highly regulated flux may have effects that propagate throughout the metabolic network in a dramatic and sometimes unexpected manner. Beginning with S1P, the receptors for which have thus far been the most clinically tractable pharmacological targets, this review will describe recent advances in therapeutic modulators targeting sphingolipids, their chaperones, transporters, and metabolic enzymes.
Collapse
|