1
|
Ruffenach G, Le Ribeuz H, Dutheil M, El Jekmek K, Dumont F, Willer AS, Humbert M, Capuano V, Medzikovic L, Eghbali M, Montani D, Antigny F. Transcriptome analyses reveal common immune system dysregulation in PAH patients and Kcnk3-deficient rats. Pulm Circ 2024; 14:e12434. [PMID: 39444497 PMCID: PMC11497494 DOI: 10.1002/pul2.12434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/26/2024] [Accepted: 08/15/2024] [Indexed: 10/25/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe disease caused by progressive distal pulmonary artery obstruction. One cause of PAH are loss-of-function mutations in the potassium channel subfamily K member 3 (KCNK3). KCNK3 encodes a two-pore domain potassium channel, which is crucial for pulmonary circulation homeostasis. However, our understanding of the pathophysiological mechanisms underlying KCNK3 dysfunction in PAH is still incomplete. Taking advantage of unique Kcnk3-deficient rats, we analyzed the transcriptomic changes in the lungs from homozygous Kcnk3-deficient rats and wild-type (WT) littermates and compared them to PAH patient transcriptomic data. Transcriptome analysis of lung tissue obtained from WT and Kcnk3-deficient rats identified 1915 down- or upregulated genes. In addition, despite limited similarities at the gene level, we found a strong common signature at the pathway level in PAH patients and Kcnk3-deficient rat lungs, especially for immune response. Using the dysregulated genes involved in the immune response, we identified Spleen Associated Tyrosine Kinase (SYK), a significantly downregulated gene in human PAH patients and Kcnk3-deficient rats, as a hub gene. Our data suggests that the altered immune system response observed in PAH patients may be partly explained by KCNK3 dysfunction through the alteration of SYK expression.
Collapse
Affiliation(s)
- Grégoire Ruffenach
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesUSA
| | - Hélène Le Ribeuz
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
| | - Mary Dutheil
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
- Groupe Hospitalier Paris Saint‐Joseph, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
| | - Kristell El Jekmek
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
| | - Florent Dumont
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- UMS Ingénierie et Plateformes au Service de l'Innovation ThérapeutiqueUniversité Paris‐SaclayOrsayFrance
| | - Anaïs Saint‐Martin Willer
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
| | - Marc Humbert
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
- Assistance Publique—Hôpitaux de Paris (AP‐HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin‐BicêtreLe Kremlin‐BicêtreFrance
| | - Véronique Capuano
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
- Groupe Hospitalier Paris Saint‐Joseph, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
| | - Lejla Medzikovic
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesUSA
| | - Mansoureh Eghbali
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesUSA
| | - David Montani
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
- Groupe Hospitalier Paris Saint‐Joseph, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
- Assistance Publique—Hôpitaux de Paris (AP‐HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin‐BicêtreLe Kremlin‐BicêtreFrance
| | - Fabrice Antigny
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
| |
Collapse
|
2
|
Chaurasiya V, Nidhina Haridas PA, Olkkonen VM. Adipocyte-endothelial cell interplay in adipose tissue physiology. Biochem Pharmacol 2024; 222:116081. [PMID: 38408682 DOI: 10.1016/j.bcp.2024.116081] [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: 10/31/2023] [Revised: 02/02/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
Adipose tissue (AT) expansion through hyperplasia or hypertrophy requires vascular remodeling that involves angiogenesis. There is quite some evidence that obese white AT (WAT) displays altered vasculature. Some studies suggest that this is associated with hypoxia, which is thought to play a role in inducing inflammatory activation of the excessively expanding WAT. Increasing evidence, based on genetic manipulations or treatments with inhibitory or activator pharmaceuticals, demonstrates that AT angiogenesis is crucial for AT metabolic function, and thereby for whole body metabolism and metabolic health. Despite some contradiction between studies, disturbance of WAT angiogenesis in obesity could be an important factor driving WAT dysfunction and the comorbidities of obesity. Endothelial cells (ECs) contribute to healthy WAT metabolism via transport of fatty acids and other plasma components, secretory signaling molecules, and extracellular vesicles (EVs). This communication is crucial for adipocyte metabolism and underscores the key role that the AT endothelium plays in systemic energy homeostasis and healthy metabolism. Adipocytes communicate towards the neighboring endothelium through several mechanisms. The pro-inflammatory status of hypertrophic adipocytes in obesity is reflected in ECs activation, which promotes chronic inflammation. On the other hand, adiponectin secreted by the adipocytes is important for healthy endothelial function, and adipocytes also secrete other pro- or anti-angiogenic effector molecules and a wealth of EVs - however, their detailed roles in signaling towards the endothelium are yet poorly understood. To conclude, targeting AT angiogenesis and promoting the healthy communication between adipocytes and ECs represent potentially promising strategies to treat obesity and its comorbidities.
Collapse
Affiliation(s)
- Vaishali Chaurasiya
- Minerva Foundation Institute for Medical Research, Helsinki, Finland, and Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland
| | - P A Nidhina Haridas
- Minerva Foundation Institute for Medical Research, Helsinki, Finland, and Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland, and Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland.
| |
Collapse
|
3
|
Van Daele M, Kilpatrick LE, Woolard J, Hill SJ. Characterisation of tyrosine kinase inhibitor-receptor interactions at VEGFR2 using sunitinib-red and nanoBRET. Biochem Pharmacol 2023:115672. [PMID: 37406966 DOI: 10.1016/j.bcp.2023.115672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/13/2023] [Accepted: 06/26/2023] [Indexed: 07/07/2023]
Abstract
Vascular endothelial growth factor (VEGF) is an important mediator of angiogenesis, proliferation and migration of vascular endothelial cells. It is well known that cardiovascular safety liability for a wide range of small molecule tyrosine kinase inhibitors (TKIs) can result from interference with the VEGFR2 signalling system. In this study we have developed a ligand-binding assay using a fluorescent analogue of sunitinib (sunitinib-red) and full length VEGFR2 tagged on its C-terminus with the bioluminescent protein nanoluciferase to monitor ligand-binding to VEGFR2 using bioluminescence resonance energy transfer (BRET). This NanoBRET assay is a proximity-based assay (requiring the fluorescent and bioluminescent components to be within 10nm of each other) that can monitor the binding of ligands to the kinase domain of VEGFR2. Sunitinib-red was not membrane permeable but was able to monitor the binding affinity and kinetics of a range of TKIs in cell lysates. Kinetic studies showed that sunitinib-red bound rapidly to VEGFR2 at 25 °C and that cediranib had slower binding kinetics with an average residence time of 112 min. Comparison between the log Ki values for inhibition of binding of sunitinib-red and log IC50 values for attenuation of VEGF165a-stimulated NFAT responses showed very similar values for compounds that inhibited sunitinib-red binding. However, two compounds that failed to inhibit sunitinib-red binding (dasatinib and entospletinib) were still able to attenuate VEGFR2-mediated NFAT signalling through inhibition of downstream signalling events. These results suggest that these compounds may still exhibit cardiovascular liabilities as a result of interference with downstream VEGFR2 signalling.
Collapse
Affiliation(s)
- Marieke Van Daele
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK
| | - Laura E Kilpatrick
- Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK; Division of Bimolecular Science and Medicinal Chemistry, School of Pharmacy, Biodiscovery Institute, University of Nottingham, NG7 2RD, UK
| | - Jeanette Woolard
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK
| | - Stephen J Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK.
| |
Collapse
|
4
|
Lawler J. Counter Regulation of Tumor Angiogenesis by Vascular Endothelial Growth Factor and Thrombospondin-1. Semin Cancer Biol 2022; 86:126-135. [PMID: 36191900 DOI: 10.1016/j.semcancer.2022.09.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 10/31/2022]
Abstract
Considerable progress has been made in our understanding of the process of angiogenesis in the context of normal and tumor tissue over the last fifty years. Angiogenesis, like most physiological processes, is carefully controlled by dynamic and opposing effects of positive factors, such as vascular endothelial growth factor (VEGF), and negative factors, such as thrombospondin-1. In most cases, the progression of a small mass of cancerous cells to a life-threatening tumor depends upon the initiation of angiogenesis and involves the dysregulation of the angiogenic balance. Whereas our newfound appreciation for the role of angiogenesis in cancer has opened up new avenues for treatment, the success of these treatments, which have focused almost exclusively on antagonizing the VEGF pathway, has been limited to date. It is anticipated that this situation will improve as more therapeutics that target other pathways are developed, more strategies for combination therapies are advanced, more detailed stratification of patient populations occurs, and a better understanding of resistance to anti-angiogenic therapy is gained.
Collapse
Affiliation(s)
- Jack Lawler
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, The Center for Vascular Biology Research, 99 Brookline Ave, Boston MA 02215, United States.
| |
Collapse
|
5
|
Immune Thrombocytopenia: Recent Advances in Pathogenesis and Treatments. Hemasphere 2021; 5:e574. [PMID: 34095758 PMCID: PMC8171374 DOI: 10.1097/hs9.0000000000000574] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/08/2021] [Indexed: 11/26/2022] Open
Abstract
Immune thrombocytopenia (ITP) is a rare autoimmune disease due to both a peripheral destruction of platelets and an inappropriate bone marrow production. Although the primary triggering factors of ITP remain unknown, a loss of immune tolerance-mostly represented by a regulatory T-cell defect-allows T follicular helper cells to stimulate autoreactive splenic B cells that differentiate into antiplatelet antibody-producing plasma cells. Glycoprotein IIb/IIIa is the main target of antiplatelet antibodies leading to platelet phagocytosis by splenic macrophages, through interactions with Fc gamma receptors (FcγRs) and complement receptors. This allows macrophages to activate autoreactive T cells by their antigen-presenting functions. Moreover, the activation of the classical complement pathway participates to platelet opsonization and also to their destruction by complement-dependent cytotoxicity. Platelet destruction is also mediated by a FcγR-independent pathway, involving platelet desialylation that favors their binding to the Ashwell-Morell receptor and their clearance in the liver. Cytotoxic T cells also contribute to ITP pathogenesis by mediating cytotoxicity against megakaryocytes and peripheral platelets. The deficient megakaryopoiesis resulting from both the humoral and the cytotoxic immune responses is sustained by inappropriate levels of thrombopoietin, the major growth factor of megakaryocytes. The better understanding of ITP pathogenesis has provided important therapeutic advances. B cell-targeting therapies and thrombopoietin-receptor agonists (TPO-RAs) have been used for years. New emerging therapeutic strategies that inhibit FcγR signaling, the neonatal Fc receptor or the classical complement pathway, will deeply modify the management of ITP in the near future.
Collapse
|
6
|
Morandi V, Petrik J, Lawler J. Endothelial Cell Behavior Is Determined by Receptor Clustering Induced by Thrombospondin-1. Front Cell Dev Biol 2021; 9:664696. [PMID: 33869231 PMCID: PMC8044760 DOI: 10.3389/fcell.2021.664696] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/10/2021] [Indexed: 11/13/2022] Open
Abstract
The thrombospondins (TSPs) are a family of multimeric extracellular matrix proteins that dynamically regulate cellular behavior and response to stimuli. In so doing, the TSPs directly and indirectly affect biological processes such as embryonic development, wound healing, immune response, angiogenesis, and cancer progression. Many of the direct effects of Thrombospondin 1 (TSP-1) result from the engagement of a wide range of cell surface receptors including syndecans, low density lipoprotein receptor-related protein 1 (LRP1), CD36, integrins, and CD47. Different or even opposing outcomes of TSP-1 actions in certain pathologic contexts may occur, depending on the structural/functional domain involved. To expedite response to external stimuli, these receptors, along with vascular endothelial growth factor receptor 2 (VEGFR2) and Src family kinases, are present in specific membrane microdomains, such as lipid rafts or tetraspanin-enriched microdomains. The molecular organization of these membrane microdomains and their constituents is modulated by TSP-1. In this review, we will describe how the presence of TSP-1 at the plasma membrane affects endothelial cell signal transduction and angiogenesis.
Collapse
Affiliation(s)
| | - Jim Petrik
- University of Guelph, Guelph, ON, Canada
| | - Jack Lawler
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
7
|
Emerging Therapies in Immune Thrombocytopenia. J Clin Med 2021; 10:jcm10051004. [PMID: 33801294 PMCID: PMC7958340 DOI: 10.3390/jcm10051004] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/11/2022] Open
Abstract
Immune thrombocytopenia (ITP) is a rare autoimmune disorder caused by peripheral platelet destruction and inappropriate bone marrow production. The management of ITP is based on the utilization of steroids, intravenous immunoglobulins, rituximab, thrombopoietin receptor agonists (TPO-RAs), immunosuppressants and splenectomy. Recent advances in the understanding of its pathogenesis have opened new fields of therapeutic interventions. The phagocytosis of platelets by splenic macrophages could be inhibited by spleen tyrosine kinase (Syk) or Bruton tyrosine kinase (BTK) inhibitors. The clearance of antiplatelet antibodies could be accelerated by blocking the neonatal Fc receptor (FcRn), while new strategies targeting B cells and/or plasma cells could improve the reduction of pathogenic autoantibodies. The inhibition of the classical complement pathway that participates in platelet destruction also represents a new target. Platelet desialylation has emerged as a new mechanism of platelet destruction in ITP, and the inhibition of neuraminidase could dampen this phenomenon. T cells that support the autoimmune B cell response also represent an interesting target. Beyond the inhibition of the autoimmune response, new TPO-RAs that stimulate platelet production have been developed. The upcoming challenges will be the determination of predictive factors of response to treatments at a patient scale to optimize their management.
Collapse
|
8
|
Akil A, Gutiérrez-García AK, Guenter R, Rose JB, Beck AW, Chen H, Ren B. Notch Signaling in Vascular Endothelial Cells, Angiogenesis, and Tumor Progression: An Update and Prospective. Front Cell Dev Biol 2021; 9:642352. [PMID: 33681228 PMCID: PMC7928398 DOI: 10.3389/fcell.2021.642352] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
The Notch signaling pathway plays an essential role in a wide variety of biological processes including cell fate determination of vascular endothelial cells and the regulation of arterial differentiation and angiogenesis. The Notch pathway is also an essential regulator of tumor growth and survival by functioning as either an oncogene or a tumor suppressor in a context-dependent manner. Crosstalk between the Notch and other signaling pathways is also pivotal in tumor progression by promoting cancer cell growth, migration, invasion, metastasis, tumor angiogenesis, and the expansion of cancer stem cells (CSCs). In this review, we provide an overview and update of Notch signaling in endothelial cell fate determination and functioning, angiogenesis, and tumor progression, particularly in the development of CSCs and therapeutic resistance. We further summarize recent studies on how endothelial signaling crosstalk with the Notch pathway contributes to tumor angiogenesis and the development of CSCs, thereby providing insights into vascular biology within the tumor microenvironment and tumor progression.
Collapse
Affiliation(s)
- Abdellah Akil
- Department of Surgery, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ana K. Gutiérrez-García
- Department of Surgery, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rachael Guenter
- Department of Surgery, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - J. Bart Rose
- Department of Surgery, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Adam W. Beck
- Department of Surgery, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Herbert Chen
- Department of Surgery, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Bin Ren
- Department of Surgery, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
9
|
Abumrad NA, Cabodevilla AG, Samovski D, Pietka T, Basu D, Goldberg IJ. Endothelial Cell Receptors in Tissue Lipid Uptake and Metabolism. Circ Res 2021; 128:433-450. [PMID: 33539224 DOI: 10.1161/circresaha.120.318003] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lipid uptake and metabolism are central to the function of organs such as heart, skeletal muscle, and adipose tissue. Although most heart energy derives from fatty acids (FAs), excess lipid accumulation can cause cardiomyopathy. Similarly, high delivery of cholesterol can initiate coronary artery atherosclerosis. Hearts and arteries-unlike liver and adrenals-have nonfenestrated capillaries and lipid accumulation in both health and disease requires lipid movement from the circulation across the endothelial barrier. This review summarizes recent in vitro and in vivo findings on the importance of endothelial cell receptors and uptake pathways in regulating FAs and cholesterol uptake in normal physiology and cardiovascular disease. We highlight clinical and experimental data on the roles of ECs in lipid supply to tissues, heart, and arterial wall in particular, and how this affects organ metabolism and function. Models of FA uptake into ECs suggest that receptor-mediated uptake predominates at low FA concentrations, such as during fasting, whereas FA uptake during lipolysis of chylomicrons may involve paracellular movement. Similarly, in the setting of an intact arterial endothelial layer, recent and historic data support a role for receptor-mediated processes in the movement of lipoproteins into the subarterial space. We conclude with thoughts on the need to better understand endothelial lipid transfer for fuller comprehension of the pathophysiology of hyperlipidemia, and lipotoxic diseases such as some forms of cardiomyopathy and atherosclerosis.
Collapse
Affiliation(s)
- Nada A Abumrad
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Ainara G Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| | - Dmitri Samovski
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Terri Pietka
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| |
Collapse
|
10
|
Haugen MH, Lingjærde OC, Hedenfalk I, Garred Ø, Borgen E, Loman N, Hatschek T, Børresen-Dale AL, Naume B, Mills GB, Mælandsmo GM, Engebraaten O. Protein Signature Predicts Response to Neoadjuvant Treatment With Chemotherapy and Bevacizumab in HER2-Negative Breast Cancers. JCO Precis Oncol 2021; 5:PO.20.00086. [PMID: 34036235 PMCID: PMC8140811 DOI: 10.1200/po.20.00086] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 10/21/2020] [Accepted: 12/07/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Antiangiogenic therapy using bevacizumab has proven effective for a number of cancers; however, in breast cancer (BC), there is an unmet need to identify patients who benefit from such treatment. PATIENTS AND METHODS In the NeoAva phase II clinical trial, patients (N = 132) with large (≥ 25 mm) human epidermal growth factor receptor 2 (HER2)-negative primary tumors were randomly assigned 1:1 to treatment with neoadjuvant chemotherapy (CTx) alone or in combination with bevacizumab (Bev plus CTx). The ratio of the tumor size after relative to before treatment was calculated into a continuous response scale. Tumor biopsies taken prior to neoadjuvant treatment were analyzed by reverse-phase protein arrays (RPPA) for expression levels of 210 BC-relevant (phospho-) proteins. Lasso regression was used to derive a predictor of tumor shrinkage from the expression of selected proteins prior to treatment. RESULTS We identified a nine-protein signature score named vascular endothelial growth factor inhibition response predictor (ViRP) for use in the Bev plus CTx treatment arm able to predict with accuracy pathologic complete response (pCR) (area under the curve [AUC] = 0.85; 95% CI, 0.74 to 0.97) and low residual cancer burden (RCB 0/I) (AUC = 0.80; 95% CI, 0.68 to 0.93). The ViRP score was significantly lower in patients with pCR (P < .001) and in patients with low RCB (P < .001). The ViRP score was internally validated on mRNA data and the resultant surrogate mRNA ViRP score significantly separated the pCR patients (P = .016). Similarly, the mRNA ViRP score was validated (P < .001) in an independent phase II clinical trial (PROMIX). CONCLUSION Our ViRP score, integrating the expression of nine proteins and validated on mRNA data both internally and in an independent clinical trial, may be used to increase the likelihood of benefit from treatment with bevacizumab combined with chemotherapy in patients with HER2-negative BC.
Collapse
Affiliation(s)
- Mads H Haugen
- Department of Tumor Biology, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Ole Christian Lingjærde
- Department of Genetics, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Department of Informatics-Biomedical Informatics, University of Oslo, Oslo, Norway.,K.G. Jebsen-Centre for B Cell Malignancies, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingrid Hedenfalk
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Øystein Garred
- Division of Laboratory Medicine-Pathology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Elin Borgen
- Division of Laboratory Medicine-Pathology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Niklas Loman
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden.,Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Skåne, Sweden
| | - Thomas Hatschek
- Department of Oncology-Pathology, Karolinska University Hospital, Stockholm, Sweden
| | - Anne-Lise Børresen-Dale
- Department of Genetics, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Bjørn Naume
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Oncology, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| | - Gordon B Mills
- Department of Cell, Developmental and Cancer Biology, School of Medicine, Oregon Health Science University, Portland, OR
| | - Gunhild M Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Institute for Medical Biology, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, Tromsø, Norway
| | - Olav Engebraaten
- Department of Tumor Biology, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Oncology, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
11
|
Lian C, Zhao L, Qiu J, Wang Y, Chen R, Liu Z, Cui J, Zhu X, Wen X, Wang S, Wang J. miR-25-3p promotes endothelial cell angiogenesis in aging mice via TULA-2/SYK/VEGFR-2 downregulation. Aging (Albany NY) 2020; 12:22599-22613. [PMID: 33201836 PMCID: PMC7746355 DOI: 10.18632/aging.103834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/14/2020] [Indexed: 12/21/2022]
Abstract
In aging, the regulation of angiogenesis is a dynamic and complex process. We aimed to identify and characterize microRNAs that regulate angiogenesis during aging. We showed that, in response to vascular endothelial senescence, microRNA-25-3p (miR-25-3p) plays the role of an angiogenic microRNA by targeting TULA-2 (T-cell ubiquitin ligand-2)/SYK (spleen tyrosine kinase)/VEGFR-2 (vascular endothelial growth factor receptor 2) signaling in vitro and in vivo. Mechanistic studies demonstrated that miR-25-3p inhibits a TULA-2/SYK/VEGFR-2 signaling pathway in endothelial cells. In old endothelial cells (OECs), upregulation of miR-25-3p inhibited the expression of TULA-2, which caused downregulation of the interaction between TULA-2 and SYK and increased phosphorylation of SYK Y323. The increased SYK Y323 phosphorylation level upregulated the phosphorylation of VEGFR-2 Y1175, which plays a vital role in angiogenesis, while miR-25-3p downregulation in YECs showed opposite effects. Finally, a salvage study showed that miR-25-3p upregulation promoted capillary regeneration and hindlimb blood flow recovery in aging mice with hindlimb ischemia. These findings suggest that miR-25-3p acts as an agonist of TULA-2/SYK/VEGFR-2 and mediates the endothelial cell angiogenesis response, which shows that the miR-25-3p/TULA-2 pathway may be potential therapeutic targets for angiogenesis during aging.
Collapse
Affiliation(s)
- Chong Lian
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, China
| | - Lei Zhao
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, China
| | - Jiacong Qiu
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, China
| | - Yang Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, China
| | - Rencong Chen
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, China
| | - Zhen Liu
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, China
| | - Jin Cui
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, China
| | - Xiaonan Zhu
- Department of Pharmacology Laboratory, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xuejun Wen
- Institute for Engineering and Medicine, Department of Biomedical Engineering, Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Shenming Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, China
| | - Jinsong Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, China
| |
Collapse
|
12
|
Su X, Sun ZH, Ren Q, Liu JR, Yin L, Liang N, Meng L, Sun RX. The effect of spleen tyrosine kinase inhibitor R406 on diabetic retinopathy in experimental diabetic rats. Int Ophthalmol 2020; 40:2371-2383. [PMID: 32462561 DOI: 10.1007/s10792-020-01422-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE To investigate the effect of spleen tyrosine kinase (Syk) inhibitor R406 on diabetic retinopathy (DR) in diabetic mellitus (DM) rats. METHODS Rats were randomized into Normal, DM, DM + 5 mg/kg R406 and DM + 10 mg/kg R406 groups. DM rats were established via injection of streptozotocin (STZ). One week after model establishment, rats in treatment groups received 5 mg/kg or 10 mg/kg R406 by gavage administration for 12 weeks consecutively, followed by the detection with hematoxylin-eosin (HE) staining, Evans blue angiography, retinal trypsin digestion assay, Western blotting, immunohistochemistry, TUNEL assay, immunofluorescence assay and quantitative reverse transcriptase real-time polymerase chain reaction (qRT-PCR). RESULTS The retina of DM rats presented different degree of edema, disordered and loose structure, swollen cells with enlarged intercellular space, and dilated and congested capillaries. Besides, the retinal vessels of DM rats showed high fluorescence leakage. However, R406 alleviated the above-mentioned conditions, which was much better with high concentration of R406 (10 mg/kg). R406 also reversed the down-regulations of occludin, claudin-5, ZO-1 and the up-regulation of and VEGF in retinal tissues of DM rats; inhibited retinal cell apoptosis; strengthened retinal cell proliferation; and reduced expressions of IL-1β, IL-6, TNF-α and nuclear p65 NF-κB in retinal tissues. The improvement in all these indexes was much more significant in rats of DM + 10 mg/kg R406 group than in rats of DM + 5 mg/kg R406 group. CONCLUSION Syk inhibitor R406 could attenuate retinal inflammation in DR rats via the repression of NF-κB activation.
Collapse
Affiliation(s)
- Xian Su
- Department of Ophthalmology, The First Hospital of Shijiazhuang City, No. 12, Pingan North Street, Shijiazhuang, 050000, Hebei Province, China
| | - Zhao-Hui Sun
- Department of Ophthalmology, The First Hospital of Shijiazhuang City, No. 12, Pingan North Street, Shijiazhuang, 050000, Hebei Province, China
| | - Qian Ren
- Department of Ophthalmology, The First Hospital of Shijiazhuang City, No. 12, Pingan North Street, Shijiazhuang, 050000, Hebei Province, China
| | - Jun-Ru Liu
- Department of Ophthalmology, The Third Hospital of Shijiazhuang City, Shijiazhuang, 050011, Hebei Province, China
| | - Li Yin
- Department of Ophthalmology, The First Hospital of Shijiazhuang City, No. 12, Pingan North Street, Shijiazhuang, 050000, Hebei Province, China
| | - Na Liang
- Department of Ophthalmology, The First Hospital of Shijiazhuang City, No. 12, Pingan North Street, Shijiazhuang, 050000, Hebei Province, China
| | - Ling Meng
- Department of Ophthalmology, The First Hospital of Shijiazhuang City, No. 12, Pingan North Street, Shijiazhuang, 050000, Hebei Province, China
| | - Rui-Xue Sun
- Department of Ophthalmology, The First Hospital of Shijiazhuang City, No. 12, Pingan North Street, Shijiazhuang, 050000, Hebei Province, China.
| |
Collapse
|
13
|
Ren B, Rose JB, Liu Y, Jaskular-Sztul R, Contreras C, Beck A, Chen H. Heterogeneity of Vascular Endothelial Cells, De Novo Arteriogenesis and Therapeutic Implications in Pancreatic Neuroendocrine Tumors. J Clin Med 2019; 8:jcm8111980. [PMID: 31739580 PMCID: PMC6912347 DOI: 10.3390/jcm8111980] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/08/2019] [Accepted: 11/12/2019] [Indexed: 02/07/2023] Open
Abstract
Arteriogenesis supplies oxygen and nutrients in the tumor microenvironment (TME), which may play an important role in tumor growth and metastasis. Pancreatic neuroendocrine tumors (pNETs) are the second most common pancreatic malignancy and are frequently metastatic on presentation. Nearly a third of pNETs secrete bioactive substances causing debilitating symptoms. Current treatment options for metastatic pNETs are limited. Importantly, these tumors are highly vascularized and heterogeneous neoplasms, in which the heterogeneity of vascular endothelial cells (ECs) and de novo arteriogenesis may be critical for their progression. Current anti-angiogenetic targeted treatments have not shown substantial clinical benefits, and they are poorly tolerated. This review article describes EC heterogeneity and heterogeneous tumor-associated ECs (TAECs) in the TME and emphasizes the concept of de novo arteriogenesis in the TME. The authors also emphasize the challenges of current antiangiogenic therapy in pNETs and discuss the potential of tumor arteriogenesis as a novel therapeutic target. Finally, the authors prospect the clinical potential of targeting the FoxO1-CD36-Notch pathway that is associated with both pNET progression and arteriogenesis and provide insights into the clinical implications of targeting plasticity of cancer stem cells (CSCs) and vascular niche, particularly the arteriolar niche within the TME in pNETs, which will also provide insights into other types of cancer, including breast cancer, lung cancer, and malignant melanoma.
Collapse
Affiliation(s)
- Bin Ren
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.B.R.); (R.J.-S.); (C.C.); (A.B.); (H.C.)
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Nutrition & Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Diabetes Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Graduate Biomedical Science Program of the Graduate School, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Correspondence:
| | - J. Bart Rose
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.B.R.); (R.J.-S.); (C.C.); (A.B.); (H.C.)
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yehe Liu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Renata Jaskular-Sztul
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.B.R.); (R.J.-S.); (C.C.); (A.B.); (H.C.)
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Carlo Contreras
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.B.R.); (R.J.-S.); (C.C.); (A.B.); (H.C.)
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Adam Beck
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.B.R.); (R.J.-S.); (C.C.); (A.B.); (H.C.)
| | - Herbert Chen
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.B.R.); (R.J.-S.); (C.C.); (A.B.); (H.C.)
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Graduate Biomedical Science Program of the Graduate School, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
14
|
Li D, Finley SD. The impact of tumor receptor heterogeneity on the response to anti-angiogenic cancer treatment. Integr Biol (Camb) 2019; 10:253-269. [PMID: 29623971 DOI: 10.1039/c8ib00019k] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Multiple promoters and inhibitors mediate angiogenesis, the formation of new blood vessels, and these factors represent potential targets for impeding vessel growth in tumors. Vascular endothelial growth factor (VEGF) is a potent angiogenic factor targeted in anti-angiogenic cancer therapies. In addition, thrombospondin-1 (TSP1) is a major endogenous inhibitor of angiogenesis, and TSP1 mimetics are being developed as an alternative type of anti-angiogenic agent. The combination of bevacizumab, an anti-VEGF agent, and ABT-510, a TSP1 mimetic, has been tested in clinical trials to treat advanced solid tumors. However, the patients' responses are highly variable and show disappointing outcomes. To obtain mechanistic insight into the effects of this combination anti-angiogenic therapy, we have constructed a novel whole-body systems biology model including the VEGF and TSP1 reaction networks. Using this molecular-detailed model, we investigated how the combination anti-angiogenic therapy changes the amounts of pro-angiogenic and anti-angiogenic complexes in cancer patients. We particularly focus on answering the question of how the effect of the combination therapy is influenced by tumor receptor expression, one aspect of patient-to-patient variability. Overall, this model complements the clinical administration of combination anti-angiogenic therapy, highlights the role of tumor receptor variability in the heterogeneous responses to anti-angiogenic therapy, and identifies the tumor receptor profiles that correlate with a high likelihood of a positive response to the combination therapy. Our model provides novel understanding of the VEGF-TSP1 balance in cancer patients at the systems-level and could be further used to optimize combination anti-angiogenic therapy.
Collapse
Affiliation(s)
- Ding Li
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, California 90089, USA.
| | | |
Collapse
|
15
|
Cifarelli V, Eichmann A. The Intestinal Lymphatic System: Functions and Metabolic Implications. Cell Mol Gastroenterol Hepatol 2018; 7:503-513. [PMID: 30557701 DOI: 10.1016/j.jcmgh.2018.12.002'||'] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 01/29/2024]
Abstract
The lymphatic system of the gut plays important roles in the transport of dietary lipids, as well as in immunosurveillance and removal of interstitial fluid. Historically, despite its crucial functions in intestinal homeostasis, the lymphatic system has been poorly studied. In the last 2 decades, identification of specific molecular mediators of lymphatic endothelial cells (LECs) growth together with novel genetic approaches and intravital imaging techniques, have advanced our understanding of the mechanisms regulating intestinal lymphatic physiology in health and disease. As its metabolic implications are gaining recognition, intestinal lymphatic biology is currently experiencing a surge in interest. This review describes current knowledge related to molecular control of intestinal lymphatic vessel structure and function. We discuss regulation of chylomicron entry into lymphatic vessels by vascular endothelial growth factors (VEGFs), hormones, transcription factors and the specific signaling pathways involved. The information covered supports the emerging role of intestinal lymphatics in etiology of the metabolic syndrome and their potential as a therapeutic target.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri.
| | - Anne Eichmann
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut; INSERM U970, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
16
|
Cifarelli V, Eichmann A. The Intestinal Lymphatic System: Functions and Metabolic Implications. Cell Mol Gastroenterol Hepatol 2018; 7:503-513. [PMID: 30557701 DOI: 10.1016/j.jcmgh.2018.12.002" and 2*3*8=6*8 and "tkbp"="tkbp] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 01/29/2024]
Abstract
The lymphatic system of the gut plays important roles in the transport of dietary lipids, as well as in immunosurveillance and removal of interstitial fluid. Historically, despite its crucial functions in intestinal homeostasis, the lymphatic system has been poorly studied. In the last 2 decades, identification of specific molecular mediators of lymphatic endothelial cells (LECs) growth together with novel genetic approaches and intravital imaging techniques, have advanced our understanding of the mechanisms regulating intestinal lymphatic physiology in health and disease. As its metabolic implications are gaining recognition, intestinal lymphatic biology is currently experiencing a surge in interest. This review describes current knowledge related to molecular control of intestinal lymphatic vessel structure and function. We discuss regulation of chylomicron entry into lymphatic vessels by vascular endothelial growth factors (VEGFs), hormones, transcription factors and the specific signaling pathways involved. The information covered supports the emerging role of intestinal lymphatics in etiology of the metabolic syndrome and their potential as a therapeutic target.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri.
| | - Anne Eichmann
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut; INSERM U970, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
17
|
Cifarelli V, Eichmann A. The Intestinal Lymphatic System: Functions and Metabolic Implications. Cell Mol Gastroenterol Hepatol 2018; 7:503-513. [PMID: 30557701 DOI: 10.1016/j.jcmgh.2018.12.002' and 2*3*8=6*8 and 'gakc'='gakc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 01/29/2024]
Abstract
The lymphatic system of the gut plays important roles in the transport of dietary lipids, as well as in immunosurveillance and removal of interstitial fluid. Historically, despite its crucial functions in intestinal homeostasis, the lymphatic system has been poorly studied. In the last 2 decades, identification of specific molecular mediators of lymphatic endothelial cells (LECs) growth together with novel genetic approaches and intravital imaging techniques, have advanced our understanding of the mechanisms regulating intestinal lymphatic physiology in health and disease. As its metabolic implications are gaining recognition, intestinal lymphatic biology is currently experiencing a surge in interest. This review describes current knowledge related to molecular control of intestinal lymphatic vessel structure and function. We discuss regulation of chylomicron entry into lymphatic vessels by vascular endothelial growth factors (VEGFs), hormones, transcription factors and the specific signaling pathways involved. The information covered supports the emerging role of intestinal lymphatics in etiology of the metabolic syndrome and their potential as a therapeutic target.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri.
| | - Anne Eichmann
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut; INSERM U970, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
18
|
Cifarelli V, Eichmann A. The Intestinal Lymphatic System: Functions and Metabolic Implications. Cell Mol Gastroenterol Hepatol 2018; 7:503-513. [PMID: 30557701 DOI: 10.1016/j.jcmgh.2018.12.002����%2527%2522\'\"] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 01/29/2024]
Abstract
The lymphatic system of the gut plays important roles in the transport of dietary lipids, as well as in immunosurveillance and removal of interstitial fluid. Historically, despite its crucial functions in intestinal homeostasis, the lymphatic system has been poorly studied. In the last 2 decades, identification of specific molecular mediators of lymphatic endothelial cells (LECs) growth together with novel genetic approaches and intravital imaging techniques, have advanced our understanding of the mechanisms regulating intestinal lymphatic physiology in health and disease. As its metabolic implications are gaining recognition, intestinal lymphatic biology is currently experiencing a surge in interest. This review describes current knowledge related to molecular control of intestinal lymphatic vessel structure and function. We discuss regulation of chylomicron entry into lymphatic vessels by vascular endothelial growth factors (VEGFs), hormones, transcription factors and the specific signaling pathways involved. The information covered supports the emerging role of intestinal lymphatics in etiology of the metabolic syndrome and their potential as a therapeutic target.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri.
| | - Anne Eichmann
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut; INSERM U970, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
19
|
Cifarelli V, Eichmann A. The Intestinal Lymphatic System: Functions and Metabolic Implications. Cell Mol Gastroenterol Hepatol 2018; 7:503-513. [PMID: 30557701 DOI: 10.1016/j.jcmgh.2018.12.002'"] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 01/29/2024]
Abstract
The lymphatic system of the gut plays important roles in the transport of dietary lipids, as well as in immunosurveillance and removal of interstitial fluid. Historically, despite its crucial functions in intestinal homeostasis, the lymphatic system has been poorly studied. In the last 2 decades, identification of specific molecular mediators of lymphatic endothelial cells (LECs) growth together with novel genetic approaches and intravital imaging techniques, have advanced our understanding of the mechanisms regulating intestinal lymphatic physiology in health and disease. As its metabolic implications are gaining recognition, intestinal lymphatic biology is currently experiencing a surge in interest. This review describes current knowledge related to molecular control of intestinal lymphatic vessel structure and function. We discuss regulation of chylomicron entry into lymphatic vessels by vascular endothelial growth factors (VEGFs), hormones, transcription factors and the specific signaling pathways involved. The information covered supports the emerging role of intestinal lymphatics in etiology of the metabolic syndrome and their potential as a therapeutic target.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri.
| | - Anne Eichmann
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut; INSERM U970, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
20
|
Cifarelli V, Eichmann A. The Intestinal Lymphatic System: Functions and Metabolic Implications. Cell Mol Gastroenterol Hepatol 2018; 7:503-513. [PMID: 30557701 DOI: 10.1016/j.jcmgh.2018.12.002%' and 2*3*8=6*8 and 'htng'!='htng%] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 01/29/2024]
Abstract
The lymphatic system of the gut plays important roles in the transport of dietary lipids, as well as in immunosurveillance and removal of interstitial fluid. Historically, despite its crucial functions in intestinal homeostasis, the lymphatic system has been poorly studied. In the last 2 decades, identification of specific molecular mediators of lymphatic endothelial cells (LECs) growth together with novel genetic approaches and intravital imaging techniques, have advanced our understanding of the mechanisms regulating intestinal lymphatic physiology in health and disease. As its metabolic implications are gaining recognition, intestinal lymphatic biology is currently experiencing a surge in interest. This review describes current knowledge related to molecular control of intestinal lymphatic vessel structure and function. We discuss regulation of chylomicron entry into lymphatic vessels by vascular endothelial growth factors (VEGFs), hormones, transcription factors and the specific signaling pathways involved. The information covered supports the emerging role of intestinal lymphatics in etiology of the metabolic syndrome and their potential as a therapeutic target.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri.
| | - Anne Eichmann
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut; INSERM U970, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
21
|
Cifarelli V, Eichmann A. The Intestinal Lymphatic System: Functions and Metabolic Implications. Cell Mol Gastroenterol Hepatol 2018; 7:503-513. [PMID: 30557701 DOI: 10.1016/j.jcmgh.2018.12.002mueybbdd] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 01/29/2024]
Abstract
The lymphatic system of the gut plays important roles in the transport of dietary lipids, as well as in immunosurveillance and removal of interstitial fluid. Historically, despite its crucial functions in intestinal homeostasis, the lymphatic system has been poorly studied. In the last 2 decades, identification of specific molecular mediators of lymphatic endothelial cells (LECs) growth together with novel genetic approaches and intravital imaging techniques, have advanced our understanding of the mechanisms regulating intestinal lymphatic physiology in health and disease. As its metabolic implications are gaining recognition, intestinal lymphatic biology is currently experiencing a surge in interest. This review describes current knowledge related to molecular control of intestinal lymphatic vessel structure and function. We discuss regulation of chylomicron entry into lymphatic vessels by vascular endothelial growth factors (VEGFs), hormones, transcription factors and the specific signaling pathways involved. The information covered supports the emerging role of intestinal lymphatics in etiology of the metabolic syndrome and their potential as a therapeutic target.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri.
| | - Anne Eichmann
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut; INSERM U970, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
22
|
Cifarelli V, Eichmann A. The Intestinal Lymphatic System: Functions and Metabolic Implications. Cell Mol Gastroenterol Hepatol 2018; 7:503-513. [PMID: 30557701 PMCID: PMC6396433 DOI: 10.1016/j.jcmgh.2018.12.002] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 12/26/2022]
Abstract
The lymphatic system of the gut plays important roles in the transport of dietary lipids, as well as in immunosurveillance and removal of interstitial fluid. Historically, despite its crucial functions in intestinal homeostasis, the lymphatic system has been poorly studied. In the last 2 decades, identification of specific molecular mediators of lymphatic endothelial cells (LECs) growth together with novel genetic approaches and intravital imaging techniques, have advanced our understanding of the mechanisms regulating intestinal lymphatic physiology in health and disease. As its metabolic implications are gaining recognition, intestinal lymphatic biology is currently experiencing a surge in interest. This review describes current knowledge related to molecular control of intestinal lymphatic vessel structure and function. We discuss regulation of chylomicron entry into lymphatic vessels by vascular endothelial growth factors (VEGFs), hormones, transcription factors and the specific signaling pathways involved. The information covered supports the emerging role of intestinal lymphatics in etiology of the metabolic syndrome and their potential as a therapeutic target.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri,Correspondence Address correspondence to: Vincenza Cifarelli, PhD, Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, Campus box 8031, 660 Euclid Avenue, St. Louis, Missouri 63110. fax: (314) 362-8230.
| | - Anne Eichmann
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut,INSERM U970, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
23
|
Samovski D, Dhule P, Pietka T, Jacome-Sosa M, Penrose E, Son NH, Flynn CR, Shoghi KI, Hyrc KL, Goldberg IJ, Gamazon ER, Abumrad NA. Regulation of Insulin Receptor Pathway and Glucose Metabolism by CD36 Signaling. Diabetes 2018; 67:1272-1284. [PMID: 29748289 PMCID: PMC6014550 DOI: 10.2337/db17-1226] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 04/26/2018] [Indexed: 12/19/2022]
Abstract
During reduced energy intake, skeletal muscle maintains homeostasis by rapidly suppressing insulin-stimulated glucose utilization. Loss of this adaptation is observed with deficiency of the fatty acid transporter CD36. A similar loss is also characteristic of the insulin-resistant state where CD36 is dysfunctional. To elucidate what links CD36 to muscle glucose utilization, we examined whether CD36 signaling might influence insulin action. First, we show that CD36 deletion specific to skeletal muscle reduces expression of insulin signaling and glucose metabolism genes. It decreases muscle ceramides but impairs glucose disposal during a meal. Second, depletion of CD36 suppresses insulin signaling in primary-derived human myotubes, and the mechanism is shown to involve functional CD36 interaction with the insulin receptor (IR). CD36 promotes tyrosine phosphorylation of IR by the Fyn kinase and enhances IR recruitment of P85 and downstream signaling. Third, pretreatment for 15 min with saturated fatty acids suppresses CD36-Fyn enhancement of IR phosphorylation, whereas unsaturated fatty acids are neutral or stimulatory. These findings define mechanisms important for muscle glucose metabolism and optimal insulin responsiveness. Potential human relevance is suggested by genome-wide analysis and RNA sequencing data that associate genetically determined low muscle CD36 expression to incidence of type 2 diabetes.
Collapse
Affiliation(s)
- Dmitri Samovski
- Departments of Medicine and Cell Biology, Washington University in St. Louis, St. Louis, MO
| | - Pallavi Dhule
- Departments of Medicine and Cell Biology, Washington University in St. Louis, St. Louis, MO
| | - Terri Pietka
- Departments of Medicine and Cell Biology, Washington University in St. Louis, St. Louis, MO
| | - Miriam Jacome-Sosa
- Departments of Medicine and Cell Biology, Washington University in St. Louis, St. Louis, MO
| | - Eric Penrose
- Departments of Medicine and Cell Biology, Washington University in St. Louis, St. Louis, MO
| | - Ni-Huiping Son
- Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York, NY
| | | | - Kooresh I Shoghi
- Department of Radiology, Washington University in St. Louis, St. Louis, MO
| | - Krzysztof L Hyrc
- Alafi Neuroimaging Laboratory, Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York, NY
| | - Eric R Gamazon
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Clare Hall, University of Cambridge, Cambridge, U.K
| | - Nada A Abumrad
- Departments of Medicine and Cell Biology, Washington University in St. Louis, St. Louis, MO
| |
Collapse
|
24
|
Karimzadeh M, Jandaghi P, Papadakis AI, Trainor S, Rung J, Gonzàlez-Porta M, Scelo G, Vasudev NS, Brazma A, Huang S, Banks RE, Lathrop M, Najafabadi HS, Riazalhosseini Y. Aberration hubs in protein interaction networks highlight actionable targets in cancer. Oncotarget 2018; 9:25166-25180. [PMID: 29861861 PMCID: PMC5982744 DOI: 10.18632/oncotarget.25382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 04/24/2018] [Indexed: 01/08/2023] Open
Abstract
Despite efforts for extensive molecular characterization of cancer patients, such as the international cancer genome consortium (ICGC) and the cancer genome atlas (TCGA), the heterogeneous nature of cancer and our limited knowledge of the contextual function of proteins have complicated the identification of targetable genes. Here, we present Aberration Hub Analysis for Cancer (AbHAC) as a novel integrative approach to pinpoint aberration hubs, i.e. individual proteins that interact extensively with genes that show aberrant mutation or expression. Our analysis of the breast cancer data of the TCGA and the renal cancer data from the ICGC shows that aberration hubs are involved in relevant cancer pathways, including factors promoting cell cycle and DNA replication in basal-like breast tumors, and Src kinase and VEGF signaling in renal carcinoma. Moreover, our analysis uncovers novel functionally relevant and actionable targets, among which we have experimentally validated abnormal splicing of spleen tyrosine kinase as a key factor for cell proliferation in renal cancer. Thus, AbHAC provides an effective strategy to uncover novel disease factors that are only identifiable by examining mutational and expression data in the context of biological networks.
Collapse
Affiliation(s)
- Mehran Karimzadeh
- Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada
- McGill University and Genome Quebec Innovation Centre, Montreal, QC H3A 0G1, Canada
| | - Pouria Jandaghi
- Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada
- McGill University and Genome Quebec Innovation Centre, Montreal, QC H3A 0G1, Canada
| | - Andreas I. Papadakis
- Department of Biochemistry, The Rosalind and Morris Goodman Cancer Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Sebastian Trainor
- Leeds Institute of Cancer and Pathology, University of Leeds, Cancer Research Building, St James's University Hospital, Leeds, LS9 7TF, UK
| | - Johan Rung
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Wellcome Trust Genome Campus, Hinxton, CB10 1SD, UK
| | - Mar Gonzàlez-Porta
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Wellcome Trust Genome Campus, Hinxton, CB10 1SD, UK
| | - Ghislaine Scelo
- International Agency for Research on Cancer (IARC), Lyon, 69008, France
| | - Naveen S. Vasudev
- Leeds Institute of Cancer and Pathology, University of Leeds, Cancer Research Building, St James's University Hospital, Leeds, LS9 7TF, UK
| | - Alvis Brazma
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Wellcome Trust Genome Campus, Hinxton, CB10 1SD, UK
| | - Sidong Huang
- Department of Biochemistry, The Rosalind and Morris Goodman Cancer Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Rosamonde E. Banks
- Leeds Institute of Cancer and Pathology, University of Leeds, Cancer Research Building, St James's University Hospital, Leeds, LS9 7TF, UK
| | - Mark Lathrop
- Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada
- McGill University and Genome Quebec Innovation Centre, Montreal, QC H3A 0G1, Canada
| | - Hamed S. Najafabadi
- Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada
- McGill University and Genome Quebec Innovation Centre, Montreal, QC H3A 0G1, Canada
| | - Yasser Riazalhosseini
- Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada
- McGill University and Genome Quebec Innovation Centre, Montreal, QC H3A 0G1, Canada
| |
Collapse
|
25
|
Ponticelli C, Anders HJ. Thrombospondin immune regulation and the kidney. Nephrol Dial Transplant 2018; 32:1084-1089. [PMID: 28088772 DOI: 10.1093/ndt/gfw431] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 11/14/2016] [Indexed: 12/17/2022] Open
Abstract
Most therapeutic attempts to prevent the progression of kidney diseases have been based on interventions to inhibit the production of transforming growth factor-β (TGF-β). Thrombospondins (TSPs) play an important role in activating TGF-β. In the healthy kidney, two TSPs are expressed, TSP1 and TSP2, which exert contrasting effects. While TSP1 is a major activator of TGF-β in renal cells and exerts pro-inflammatory effects both in vitro and in vivo, TSP2 lacks the ability for TGF-β activation but regulates matrix remodeling and inflammation in experimental kidney disease. The effects of TSPs in the kidney have been mostly investigated by using the murine model of unilateral ureteral obstruction. In this model, TSP1 expression is increased along with the development of interstitial fibrosis and TGF-β. Relief of the obstruction gradually improves renal function and decreases the expression in TSP1 and TGF-β1. Several inhibitors of TSP1 prevented progressive interstitial fibrosis in murine models of ureteral obstruction, suggesting that control of latent TGF-β activation by inhibiting TSP1 might represent a novel potential target for preventing renal interstitial fibrosis. However, further studies are needed to assess whether TSP1-mediated TGF-β activation can be safely used in humans. In fact, TSPs normally act to suppress tumors in vivo. Moreover, TGF-β can exert a pivotal function in the immune system, as it may induce the production of regulatory T cells and suppress B cell responses. Knowledge of the molecular mechanisms involved in TGF-β regulation may help in finding effective treatments of tissue fibrosis, cancer and autoimmune disease.
Collapse
Affiliation(s)
- Claudio Ponticelli
- Renal Unit, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Hans-Joachim Anders
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| |
Collapse
|
26
|
Kazerounian S, Lawler J. Integration of pro- and anti-angiogenic signals by endothelial cells. J Cell Commun Signal 2017; 12:171-179. [PMID: 29264709 DOI: 10.1007/s12079-017-0433-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 11/02/2017] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis or neovascularization is a complex multi-step physiological process that occurs throughout life both in normal tissues and in disease. It is tightly regulated by the balance between pro-angiogenic and anti-angiogenic factors. The angiogenic switch has been identified as the key step during tumor progression in which the balance between pro-angiogenic and anti-angiogenic factors leans toward pro-angiogenic stimuli promoting the progression of tumors from dormancy to dysplasia and ultimately malignancy. This event can be described as either the outcome of a genetic event occurring in cancer cells themselves, or the positive and negative cross-talk between tumor-associated endothelial cells and other cellular components of the tumor microenvironment. In recent years, the mechanisms underlying the angiogenic switch have been extensively investigated in particular to identify therapeutic targets that can lead to development of effective therapies. In this review, we will discuss the current findings on the regulatory pathways in endothelial cells that are involved in the angiogenic switch with an emphasis on the role of anti-angiogenic protein, thrombospondin-1 (TSP-1) and pro-angiogenic factor, vascular endothelial growth factor (VEGF).
Collapse
Affiliation(s)
- Shideh Kazerounian
- The Department of Pediatrics, Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jack Lawler
- The Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
27
|
Benon A, Ya C, Martin L, Watrin C, Chounlamountri N, Jaaoini I, Honnorat J, Pellier-Monnin V, Noraz N. The Syk kinases orchestrate cerebellar granule cell tangential migration. Neuroscience 2017; 360:230-239. [DOI: 10.1016/j.neuroscience.2017.07.057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 07/21/2017] [Accepted: 07/21/2017] [Indexed: 01/03/2023]
|
28
|
Hammoud L, Adams JR, Loch AJ, Marcellus RC, Uehling DE, Aman A, Fladd C, McKee TD, Jo CEB, Al-Awar R, Egan SE, Rossant J. Identification of RSK and TTK as Modulators of Blood Vessel Morphogenesis Using an Embryonic Stem Cell-Based Vascular Differentiation Assay. Stem Cell Reports 2016; 7:787-801. [PMID: 27618721 PMCID: PMC5063585 DOI: 10.1016/j.stemcr.2016.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 08/04/2016] [Accepted: 08/05/2016] [Indexed: 11/05/2022] Open
Abstract
Blood vessels are formed through vasculogenesis, followed by remodeling of the endothelial network through angiogenesis. Many events that occur during embryonic vascular development are recapitulated during adult neoangiogenesis, which is critical to tumor growth and metastasis. Current antiangiogenic tumor therapies, based largely on targeting the vascular endothelial growth factor pathway, show limited clinical benefits, thus necessitating the discovery of alternative targets. Here we report the development of a robust embryonic stem cell-based vascular differentiation assay amenable to small-molecule screens to identify novel modulators of angiogenesis. In this context, RSK and TTK were identified as angiogenic modulators. Inhibition of these pathways inhibited angiogenesis in embryoid bodies and human umbilical vein endothelial cells. Furthermore, inhibition of RSK and TTK reduced tumor growth, vascular density, and improved survival in an in vivo Lewis lung carcinoma mouse model. Our study suggests that RSK and TTK are potential targets for antiangiogenic therapy, and provides an assay system for further pathway screens. Development of ESC-based vascular differentiation assay amenable to drug screening Screening a kinase library identified RSK and TTK as angiogenic modulators RSK and TTK inhibition disrupted angiogenesis in vitro RSK and TTK inhibition inhibited Lewis lung tumor growth and angiogenesis in vivo
Collapse
Affiliation(s)
- Lamis Hammoud
- Program in Developmental and Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Jessica R Adams
- Program in Developmental and Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Amanda J Loch
- Program in Developmental and Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Richard C Marcellus
- Drug Discovery Department, Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - David E Uehling
- Drug Discovery Department, Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Ahmed Aman
- Drug Discovery Department, Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Christopher Fladd
- SPARC BioCentre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Trevor D McKee
- Radiation Medicine Program, STTARR Innovation Centre, Princess Margaret Cancer Centre, Toronto, ON M5G 1L7, Canada
| | - Christine E B Jo
- Program in Developmental and Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Rima Al-Awar
- Drug Discovery Department, Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Sean E Egan
- Program in Developmental and Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Janet Rossant
- Program in Developmental and Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
29
|
Identification of TAX2 peptide as a new unpredicted anti-cancer agent. Oncotarget 2016; 6:17981-8000. [PMID: 26046793 PMCID: PMC4627230 DOI: 10.18632/oncotarget.4025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 05/09/2015] [Indexed: 11/25/2022] Open
Abstract
The multi-modular glycoprotein thrombospondin-1 (TSP-1) is considered as a key actor within the tumor microenvironment. Besides, TSP-1 binding to CD47 is widely reported to regulate cardiovascular function as it promotes vasoconstriction and angiogenesis limitation. Therefore, many studies focused on targeting TSP-1:CD47 interaction, aiming for up-regulation of physiological angiogenesis to enhance post-ischemia recovery or to facilitate engraftment. Thus, we sought to identify an innovative selective antagonist for TSP-1:CD47 interaction. Protein-protein docking and molecular dynamics simulations were conducted to design a novel CD47-derived peptide, called TAX2. TAX2 binds TSP-1 to prevent TSP-1:CD47 interaction, as revealed by ELISA and co-immunoprecipitation experiments. Unexpectedly, TAX2 inhibits in vitro and ex vivo angiogenesis features in a TSP-1-dependent manner. Consistently, our data highlighted that TAX2 promotes TSP-1 binding to CD36-containing complexes, leading to disruption of VEGFR2 activation and downstream NO signaling. Such unpredicted results prompted us to investigate TAX2 potential in tumor pathology. A multimodal imaging approach was conducted combining histopathological staining, MVD, MRI analysis and μCT monitoring for tumor angiography longitudinal follow-up and 3D quantification. TAX2 in vivo administrations highly disturb syngeneic melanoma tumor vascularization inducing extensive tumor necrosis and strongly inhibit growth rate and vascularization of human pancreatic carcinoma xenografts in nude mice.
Collapse
|
30
|
Vance DT, Dufresne J, Florentinus-Mefailoski A, Tucholska M, Trimble W, Grinstein S, Marshall JG. A phagocytosis assay for oxidized low-density lipoprotein versus immunoglobulin G-coated microbeads in human U937 macrophages. Anal Biochem 2016; 500:24-34. [DOI: 10.1016/j.ab.2016.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 12/24/2015] [Accepted: 01/11/2016] [Indexed: 10/22/2022]
|
31
|
Noraz N, Jaaoini I, Charoy C, Watrin C, Chounlamountri N, Benon A, Malleval C, Boudin H, Honnorat J, Castellani V, Pellier-Monnin V. Syk kinases are required for spinal commissural axon repulsion at the midline via the ephrin/Eph pathway. Development 2016; 143:2183-93. [PMID: 27122172 DOI: 10.1242/dev.128629] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 04/15/2016] [Indexed: 12/26/2022]
Abstract
In the hematopoietic system, Syk family tyrosine kinases are essential components of immunoreceptor ITAM-based signaling. While there is increasing data indicating the involvement of immunoreceptors in neural functions, the contribution of Syk kinases remains obscure. Previously, we identified phosphorylated forms of Syk kinases in specialized populations of migrating neurons or projecting axons. Moreover, we identified ephrin/Eph as guidance molecules utilizing the ITAM-bearing CD3zeta (Cd247) and associated Syk kinases for the growth cone collapse response induced in vitro Here, we show that in the developing spinal cord, Syk is phosphorylated in navigating commissural axons. By analyzing axon trajectories in open-book preparations of Syk(-/-); Zap70(-/-) mouse embryos, we show that Syk kinases are dispensable for attraction towards the midline but confer growth cone responsiveness to repulsive signals that expel commissural axons from the midline. Known to serve a repulsive function at the midline, ephrin B3/EphB2 are obvious candidates for driving the Syk-dependent repulsive response. Indeed, Syk kinases were found to be required for ephrin B3-induced growth cone collapse in cultured commissural neurons. In fragments of commissural neuron-enriched tissues, Syk is in a constitutively phosphorylated state and ephrin B3 decreased its level of phosphorylation. Direct pharmacological inhibition of Syk kinase activity was sufficient to induce growth cone collapse. In conclusion, Syk kinases act as a molecular switch of growth cone adhesive and repulsive responses.
Collapse
Affiliation(s)
- Nelly Noraz
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Iness Jaaoini
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Camille Charoy
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Chantal Watrin
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Naura Chounlamountri
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Aurélien Benon
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Céline Malleval
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Hélène Boudin
- INSERM U1064, Institut de Transplantation Urologie-Néphrologie, Nantes F-44035, France
| | - Jérôme Honnorat
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France Hospices Civils de Lyon, Lyon F-69000, France
| | - Valérie Castellani
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Véronique Pellier-Monnin
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| |
Collapse
|
32
|
Ren B, Best B, Ramakrishnan DP, Walcott BP, Storz P, Silverstein RL. LPA/PKD-1-FoxO1 Signaling Axis Mediates Endothelial Cell CD36 Transcriptional Repression and Proangiogenic and Proarteriogenic Reprogramming. Arterioscler Thromb Vasc Biol 2016; 36:1197-208. [PMID: 27013613 DOI: 10.1161/atvbaha.116.307421] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 03/10/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE CD36 is a scavenger and antiangiogenic receptor that is important in atherothrombotic diseases, diabetes mellitus, cancer, and obesity. Lysophosphatidic acid, a phospholipid signaling mediator, abolishes endothelial cell responses to antiangiogenic proteins containing thrombospondin type 1 homology domains by downregulating endothelial CD36 transcription via protein kinase D1 (PKD-1) signaling. We aimed to understand mechanisms by which lysophosphatidic acid-mediated angiogenic signaling is integrated to regulate CD36 transcription and endothelial cell function via a nuclear transcriptional complex. APPROACH AND RESULTS Microvascular endothelial cells expressing CD36 were used for studying angiogenic signaling and CD36 transcription. Gene transfection and transduction, RT-qPCR, avidin-biotin-conjugated DNA-binding assay, chromatin immunoprecipitation assay, co-immunoprecipitation, proximal ligation assay, and immunofluorescence microscopy showed that lysophosphatidic acid-mediated CD36 transcriptional repression involved PKD-1 signaling mediated formation of forkhead box protein O1-histone deacetylase 7 complex in the nucleus. Unexpectedly, turning off CD36 transcription initiated reprogramming microvascular endothelial cells to express ephrin B2, a critical molecular signature involved in angiogenesis and arteriogenesis. Spheroid-based angiogenesis and in vivo Matrigel angiogenesis assays indicated that angiogenic branching morphogenesis and in vivo angiogenesis were dependent on PKD-1 signaling. A mouse tumor angiogenesis model revealed enhanced PKD-1 signaling and expression of ephrin B2 and smooth muscle actin in neovessels of Lewis Lung Carcinomas, along with low-CD36 expression or CD36 deficiency. CONCLUSIONS Lysophosphatidic acid/PKD-1 signaling leads to nuclear accumulation of histone deacetylase 7, where it interacts with forkhead box protein O1 to suppress endothelial CD36 transcription and mediates silencing of antiangiogenic switch, resulting in proangiogenic and proarteriogenic reprogramming. Targeting this signaling cascade could be a novel approach for ischemic cardiovascular disease and cancer.
Collapse
Affiliation(s)
- Bin Ren
- From the Department of Medicine, Medical College of Wisconsin, Milwaukee (B.R., R.L.S.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (B.R., B.B., D.P.R., R.L.S.); Department of Neurological Surgery, Cardiovascular Research Center, Massachusetts General Hospital & Harvard Medical School, Boston (B.P.W.); and Department of Cancer Biology, Mayo Clinic, Jacksonville, FL (P.S.)
| | - Brad Best
- From the Department of Medicine, Medical College of Wisconsin, Milwaukee (B.R., R.L.S.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (B.R., B.B., D.P.R., R.L.S.); Department of Neurological Surgery, Cardiovascular Research Center, Massachusetts General Hospital & Harvard Medical School, Boston (B.P.W.); and Department of Cancer Biology, Mayo Clinic, Jacksonville, FL (P.S.)
| | - Devi Prasadh Ramakrishnan
- From the Department of Medicine, Medical College of Wisconsin, Milwaukee (B.R., R.L.S.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (B.R., B.B., D.P.R., R.L.S.); Department of Neurological Surgery, Cardiovascular Research Center, Massachusetts General Hospital & Harvard Medical School, Boston (B.P.W.); and Department of Cancer Biology, Mayo Clinic, Jacksonville, FL (P.S.)
| | - Brian P Walcott
- From the Department of Medicine, Medical College of Wisconsin, Milwaukee (B.R., R.L.S.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (B.R., B.B., D.P.R., R.L.S.); Department of Neurological Surgery, Cardiovascular Research Center, Massachusetts General Hospital & Harvard Medical School, Boston (B.P.W.); and Department of Cancer Biology, Mayo Clinic, Jacksonville, FL (P.S.)
| | - Peter Storz
- From the Department of Medicine, Medical College of Wisconsin, Milwaukee (B.R., R.L.S.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (B.R., B.B., D.P.R., R.L.S.); Department of Neurological Surgery, Cardiovascular Research Center, Massachusetts General Hospital & Harvard Medical School, Boston (B.P.W.); and Department of Cancer Biology, Mayo Clinic, Jacksonville, FL (P.S.)
| | - Roy L Silverstein
- From the Department of Medicine, Medical College of Wisconsin, Milwaukee (B.R., R.L.S.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (B.R., B.B., D.P.R., R.L.S.); Department of Neurological Surgery, Cardiovascular Research Center, Massachusetts General Hospital & Harvard Medical School, Boston (B.P.W.); and Department of Cancer Biology, Mayo Clinic, Jacksonville, FL (P.S.).
| |
Collapse
|
33
|
Deng GM, Kyttaris VC, Tsokos GC. Targeting Syk in Autoimmune Rheumatic Diseases. Front Immunol 2016; 7:78. [PMID: 27014261 PMCID: PMC4779881 DOI: 10.3389/fimmu.2016.00078] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/16/2016] [Indexed: 02/03/2023] Open
Abstract
Spleen tyrosine kinase (Syk) is a member of the Src family of non-receptor tyrosine kinases, which associates directly with surface receptors, including B-cell receptor and Fcγ receptor, and is involved in a variety of signal transduction pathways. Rheumatoid arthritis (RA) and systemic lupus erythematosus are autoimmune diseases in which autoantibodies, immune complexes, and autoreactive T cells account for the expression of tissue inflammation and damage. Syk inhibitors efficiently suppress RA in patients albeit in the expression of unwanted side effects, including gastrointestinal effects, hypertension, and neutropenia. Syk inhibitors also inhibit clinical manifestations in lupus-prone mice. Here, we review the evidence that supports the use of Syk inhibitors to treat rheumatic and other autoimmune diseases.
Collapse
Affiliation(s)
- Guo-Min Deng
- Key Laboratory of Antibody Techniques of Ministry of Health, Nanjing Medical University, Nanjing, China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | | | - George C Tsokos
- Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
34
|
Lopez-Dee ZP, Chittur SV, Patel H, Chinikaylo A, Lippert B, Patel B, Lawler J, Gutierrez LS. Thrombospondin-1 in a Murine Model of Colorectal Carcinogenesis. PLoS One 2015; 10:e0139918. [PMID: 26461935 PMCID: PMC4603676 DOI: 10.1371/journal.pone.0139918] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 09/19/2015] [Indexed: 12/12/2022] Open
Abstract
Colorectal Cancer (CRC) is one of the late complications observed in patients suffering from inflammatory bowel diseases (IBD). Carcinogenesis is promoted by persistent chronic inflammation occurring in IBD. Understanding the mechanisms involved is essential in order to ameliorate inflammation and prevent CRC. Thrombospondin 1 (TSP-1) is a multidomain glycoprotein with important roles in angiogenesis. The effects of TSP-1 in colonic tumor formation and growth were analyzed in a model of inflammation-induced carcinogenesis. WT and TSP-1 deficient mice (TSP-1-/-) of the C57BL/6 strain received a single injection of azoxymethane (AOM) and multiple cycles of dextran sodium sulfate (DSS) to induce chronic inflammation-related cancers. Proliferation and angiogenesis were histologically analyzed in tumors. The intestinal transcriptome was also analyzed using a gene microarray approach. When the area containing tumors was compared with the entire colonic area of each mouse, the tumor burden was decreased in AOM/DSS-treated TSP-1-/- versus wild type (WT) mice. However, these lesions displayed more angiogenesis and proliferation rates when compared with the WT tumors. AOM-DSS treatment of TSP-1-/- mice resulted in significant deregulation of genes involved in transcription, canonical Wnt signaling, transport, defense response, regulation of epithelial cell proliferation and metabolism. Microarray analyses of these tumors showed down-regulation of 18 microRNAs in TSP-1-/- tumors. These results contribute new insights on the controversial role of TSP-1 in cancer and offer a better understanding of the genetics and pathogenesis of CRC.
Collapse
Affiliation(s)
- Zenaida P. Lopez-Dee
- Department of Biology, Wilkes University, Wilkes Barre, Pennsylvania, United States of America
| | - Sridar V. Chittur
- Center for Functional Genomics, University of Albany, State University of New York, Renssaeler, New York, United States of America
| | - Hiral Patel
- Department of Biology, Wilkes University, Wilkes Barre, Pennsylvania, United States of America
| | - Aleona Chinikaylo
- Department of Biology, Wilkes University, Wilkes Barre, Pennsylvania, United States of America
| | - Brittany Lippert
- Department of Biology, Wilkes University, Wilkes Barre, Pennsylvania, United States of America
| | - Bhumi Patel
- Department of Biology, Wilkes University, Wilkes Barre, Pennsylvania, United States of America
| | - Jack Lawler
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Linda S. Gutierrez
- Department of Biology, Wilkes University, Wilkes Barre, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
35
|
Koerber RM, Held SAE, Heine A, Kotthoff P, Daecke SN, Bringmann A, Brossart P. Analysis of the anti-proliferative and the pro-apoptotic efficacy of Syk inhibition in multiple myeloma. Exp Hematol Oncol 2015; 4:21. [PMID: 26251761 PMCID: PMC4526421 DOI: 10.1186/s40164-015-0016-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 07/20/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a clonal B cell malignancy characterized by proliferation of malignant plasma cells in the bone marrow. Despite high-dose melphalan therapy with autologous stem cell transplantation (ASCT) and the introduction of immunomodulatory drugs like bortezomib or lenalidomide, that have been associated with improved survival, MM is still incurable and new treatment options are needed. In B cell malignancies such as chronic lymphocytic leukaemia (CLL) or diffuse large B cell lymphoma (DLBCL), Syk (spleen tyrosine kinase) inhibitors have shown promising in vitro and first clinical results. In our study, we analyzed the potential of Syk as a target in MM. METHODS The MM cell lines AMO-1, U266 and RPMI8226 and primary MM cells were treated with the Syk inhibitors BAY61-3606, R406 or Piceatannol and proliferation, migration and apoptosis induction were analyzed. Effects on involved intracellular signaling cascades were determined by Western blotting. Furthermore, we analyzed synergistic and additive effects of Syk inhibitors in combination with established anti-myeloma drugs and experimental inhibitors (e.g. PI-3-Kinase inhibitor NVP-BEZ235). RESULTS Incubation of MM cell lines as well as primary MM cells with Syk inhibitors resulted in a reduced proliferation and stromal cell-derived factor-1 alpha (SDF-1 alpha) induced migration that was accompanied by a concentration dependent inhibition of the MAP-Kinase, characterized by reduced phosphorylation of ERK an p38 molecules, and NF-kappaB signalling pathways. Furthermore, Syk inhibition induced apoptosis in MM cells in a dose-dependent manner, characterized by reduced expression of pro-caspase 3, increased PARP-1 cleavage and enhanced release of cytochrome c. In addition combined treatment of MM cells with Syk inhibitors and NVP-BEZ235 (dual PI3-kinase/mTOR inhibitor) or MAPK inhibitors (PD98059, SP600125, U0126, SB203580) resulted in increased apoptotic activity of the drugs. CONCLUSIONS Our results show that Syk inhibition might represent a promising new treatment option in MM with an increased efficacy when combined with MAP kinase inhibitors. Furthermore, our study strongly underlines the potency of Syk inhibitors as a potential therapeutic treatment option for MM patients.
Collapse
Affiliation(s)
- Ruth-Miriam Koerber
- Medical Clinic III, Department of Hematology and Oncology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Stefanie Andrea Erika Held
- Medical Clinic III, Department of Hematology and Oncology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Annkristin Heine
- Medical Clinic III, Department of Hematology and Oncology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Philipp Kotthoff
- Medical Clinic III, Department of Hematology and Oncology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Solveig Nora Daecke
- Medical Clinic III, Department of Hematology and Oncology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Anita Bringmann
- Medical Clinic III, Department of Hematology and Oncology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Peter Brossart
- Medical Clinic III, Department of Hematology and Oncology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| |
Collapse
|
36
|
Shoni M, Lui KO, Vavvas DG, Muto MG, Berkowitz RS, Vlahos N, Ng SW. Protein kinases and associated pathways in pluripotent state and lineage differentiation. Curr Stem Cell Res Ther 2015; 9:366-87. [PMID: 24998240 DOI: 10.2174/1574888x09666140616130217] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 06/07/2014] [Accepted: 06/12/2014] [Indexed: 02/06/2023]
Abstract
Protein kinases (PKs) mediate the reversible conversion of substrate proteins to phosphorylated forms, a key process in controlling intracellular signaling transduction cascades. Pluripotency is, among others, characterized by specifically expressed PKs forming a highly interconnected regulatory network that culminates in a finely-balanced molecular switch. Current high-throughput phosphoproteomic approaches have shed light on the specific regulatory PKs and their function in controlling pluripotent states. Pluripotent cell-derived endothelial and hematopoietic developments represent an example of the importance of pluripotency in cancer therapeutics and organ regeneration. This review attempts to provide the hitherto known kinome profile and the individual characterization of PK-related pathways that regulate pluripotency. Elucidating the underlying intrinsic and extrinsic signals may improve our understanding of the different pluripotent states, the maintenance or induction of pluripotency, and the ability to tailor lineage differentiation, with a particular focus on endothelial cell differentiation for anti-cancer treatment, cell-based tissue engineering, and regenerative medicine strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shu-Wing Ng
- 221 Longwood Avenue, BLI- 449A, Boston MA 02115, USA.
| |
Collapse
|
37
|
Schlich R, Lamers D, Eckel J, Sell H. Adipokines enhance oleic acid-induced proliferation of vascular smooth muscle cells by inducing CD36 expression. Arch Physiol Biochem 2015; 121:81-7. [PMID: 26135380 DOI: 10.3109/13813455.2015.1045520] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Adipose tissue is not only releasing lipids but also various adipokines that are both dysregulated in the obese state and may contribute to obesity-associated vascular dysfunction and cardiovascular risk. We have previously shown that the combination of adipocyte-conditioned medium (CM) and oleic acid (OA) increases proliferation of human vascular smooth muscle cells (VSMC) in a synergistic way. We identified vascular endothelial growth factor (VEGF) as a component within CM that is responsible for most of the observed effects. In this study, we investigate novel mechanisms that underlie the combined effects of adipokine and oleic acid-induced proliferation of VSMC. Oleic acid leads to significant lipid accumulation in VSMC that is further enhanced by the combined treatment with CM. Accordingly CM stimulates CD36 expression in VSMC while OA is not affecting CD36. Silencing of CD36 was established and prevents lipid accumulation in all tested conditions. CD36 silencing also abrogates CM- and OA-induced proliferation and considerably reduces proliferation induced by the combination of CM and OA. At the same time, VEGF secretion and VEGF-receptor 1 (VEGF-R1) by VSMC was not affected by CD36 silencing. However, VEGF was not able to induce any proliferation in VSMC after CD36 silencing that also blunted VEGF-induced extracellular signal-regulated kinase (ERK) activation. Finally, combined silencing of CD36 together with a blocking antibody against VEGF prevented most of CMOA-induced proliferation. In conclusion, our results demonstrate that CD36 is mediating CM-induced proliferation of VSMC. Induction of CD36 by adipokines enhances the response of VSMC towards VEGF and OA.
Collapse
MESH Headings
- Adipocytes/cytology
- Adipocytes/metabolism
- Adipokines/metabolism
- Adipokines/pharmacology
- Adipose Tissue/cytology
- Adipose Tissue/metabolism
- Adult
- Antibodies, Neutralizing/pharmacology
- CD36 Antigens/antagonists & inhibitors
- CD36 Antigens/genetics
- CD36 Antigens/metabolism
- Cell Proliferation/drug effects
- Coronary Vessels/cytology
- Coronary Vessels/drug effects
- Coronary Vessels/metabolism
- Culture Media, Conditioned/chemistry
- Culture Media, Conditioned/pharmacology
- Female
- Gene Expression Regulation
- Humans
- Male
- Middle Aged
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/genetics
- Mitogen-Activated Protein Kinase 3/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Oleic Acid/pharmacology
- Primary Cell Culture
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor Receptor-1/genetics
- Vascular Endothelial Growth Factor Receptor-1/metabolism
Collapse
Affiliation(s)
- Raphaela Schlich
- Paul-Langerhans-Group for Integrative Physiology, German Diabetes Center , Düsseldorf , Germany
| | | | | | | |
Collapse
|
38
|
Small-molecule therapeutics in rheumatoid arthritis: scientific rationale, efficacy and safety. Best Pract Res Clin Rheumatol 2014; 28:605-24. [PMID: 25481553 DOI: 10.1016/j.berh.2014.10.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Rheumatoid arthritis (RA) remains a formidable clinical challenge. This is despite remarkable recent advances in our understanding of pathogenesis and the introduction of a variety of novel agents, particularly biologic therapeutics that are potent inhibitors of extracellular immune pathways. Whereas the latter have brought substantial improvements in efficacy and thus outcomes, there remain significant numbers of non- or partial responders to current standard of care. The discovery of key intracellular pathways, particularly kinases that subserve the function of these pivotal cytokine and immune cell receptors implicated in RA pathogenesis, has facilitated the advent of a new phase of RA drug development. Thus, a range of kinase inhibitors has entered clinical trials and one agent has been licenced for use in some regions. Herein we summarise the chequered history of kinase inhibitor development in RA, describing successes and failures alike, and thereafter examine future trends in this exciting field.
Collapse
|
39
|
Prospection of genomic regions divergently selected in racing line of Quarter Horses in relation to cutting line. Animal 2014; 8:1754-64. [PMID: 25032727 DOI: 10.1017/s1751731114001761] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Selection of Quarter Horses for different purposes has led to the formation of lines, including racing and cutting horses. The objective of this study was to identify genomic regions divergently selected in racing line of Quarter Horses in relation to cutting line applying relative extended haplotype homozygosity (REHH) analysis, an extension of extended haplotype homozygosity (EHH) analysis, and the fixation index (F ST) statistic. A total of 188 horses of both sexes, born between 1985 and 2009 and registered at the Brazilian Association of Quarter Horse Breeders, including 120 of the racing line and 68 of the cutting line, were genotyped using single nucleotide polymorphism arrays. On the basis of 27 genomic regions identified as selection signatures by REHH and F ST statistics, functional annotations of genes were made in order to identify those that could have been important during formation of the racing line and that could be used subsequently for the development of selection tools. Genes involved in muscle growth (n=8), skeletal growth (n=10), muscle energy metabolism (n=15), cardiovascular system (n=14) and nervous system (n=23) were identified, including the FKTN, INSR, GYS1, CLCN1, MYLK, SYK, ANG, CNTFR and HTR2B.
Collapse
|
40
|
Murphy-Ullrich JE, Sage EH. Revisiting the matricellular concept. Matrix Biol 2014; 37:1-14. [PMID: 25064829 PMCID: PMC4379989 DOI: 10.1016/j.matbio.2014.07.005] [Citation(s) in RCA: 299] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 07/07/2014] [Accepted: 07/08/2014] [Indexed: 12/16/2022]
Abstract
The concept of a matricellular protein was first proposed by Paul Bornstein in the mid-1990s to account for the non-lethal phenotypes of mice with inactivated genes encoding thrombospondin-1, tenascin-C, or SPARC. It was also recognized that these extracellular matrix proteins were primarily counter or de-adhesive. This review reappraises the matricellular concept after nearly two decades of continuous investigation. The expanded matricellular family as well as the diverse and often unexpected functions, cellular location, and interacting partners/receptors of matricellular proteins are considered. Development of therapeutic strategies that target matricellular proteins are discussed in the context of pathology and regenerative medicine.
Collapse
Affiliation(s)
- Joanne E Murphy-Ullrich
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, United States.
| | | |
Collapse
|
41
|
Duquette M, Nadler M, Okuhara D, Thompson J, Shuttleworth T, Lawler J. Members of the thrombospondin gene family bind stromal interaction molecule 1 and regulate calcium channel activity. Matrix Biol 2014; 37:15-24. [PMID: 24845346 PMCID: PMC4502920 DOI: 10.1016/j.matbio.2014.05.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 05/06/2014] [Accepted: 05/06/2014] [Indexed: 01/24/2023]
Abstract
The thrombospondins (TSPs) are a family of matricellular proteins that regulate cellular phenotype through interactions with a myriad of other proteins and proteoglycans. We have identified a novel interaction of the members of the TSP gene family with stromal interaction molecule 1 (STIM1). This association is robust since it is preserved in Triton X-100, can be detected with multiple anti-TSP-1 and anti-STIM1 antibodies, and is detected in a wide range of cell types. We have also found that STIM1 co-immunoprecipitates with TSP-4 and cartilage oligomeric matrix protein (COMP), and that a recombinant version of the N-terminal domain of STIM1 binds to the signature domain of TSP-1 and COMP. The association of the TSPs with STIM1 is observed in both the presence and absence of calcium indicating that the calcium-dependent conformation of the signature domain of TSPs is not required for binding. Thus, this interaction could occur in the ER under conditions of normal or low calcium concentration. Furthermore, we observed that the expression of COMP in HEK 293 cells decreases STIM1-mediated calcium release activated calcium (CRAC) channel currents and increases arachidonic acid calcium (ARC) channel currents. These data indicate that the TSPs regulate STIM1 function and participate in the reciprocal regulation of two channels that mediate calcium entry into the cell.
Collapse
Affiliation(s)
- Mark Duquette
- The Division of Experimental Pathology, Department of Pathology, Beth Israel Deaconess Medical School, Harvard Medical School, 99 Brookline Ave., Boston, MA 02215, United States
| | - Monica Nadler
- The Division of Experimental Pathology, Department of Pathology, Beth Israel Deaconess Medical School, Harvard Medical School, 99 Brookline Ave., Boston, MA 02215, United States
| | - Dayne Okuhara
- Synta Pharmaceuticals, 45 Hartwell Avenue, Lexington, MA 02421, United States
| | - Jill Thompson
- The Department of Pharmacology and Physiology, Box 711, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - Trevor Shuttleworth
- The Department of Pharmacology and Physiology, Box 711, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - Jack Lawler
- The Division of Experimental Pathology, Department of Pathology, Beth Israel Deaconess Medical School, Harvard Medical School, 99 Brookline Ave., Boston, MA 02215, United States.
| |
Collapse
|
42
|
Scott IC, Scott DL. Spleen Tyrosine Kinase Inhibitors for Rheumatoid Arthritis: Where Are We Now? Drugs 2014; 74:415-22. [DOI: 10.1007/s40265-014-0193-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
43
|
Resovi A, Pinessi D, Chiorino G, Taraboletti G. Current understanding of the thrombospondin-1 interactome. Matrix Biol 2014; 37:83-91. [PMID: 24476925 DOI: 10.1016/j.matbio.2014.01.012] [Citation(s) in RCA: 195] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 01/20/2014] [Accepted: 01/20/2014] [Indexed: 12/24/2022]
Abstract
The multifaceted action of thrombospondin-1 (TSP-1) depends on its ability to physically interact with different ligands, including structural components of the extracellular matrix, other matricellular proteins, cell receptors, growth factors, cytokines and proteases. Through this network, TSP-1 regulates the ligand activity, availability and structure, ultimately tuning the cell response to environmental stimuli in a context-dependent manner, contributing to physiological and pathological processes. Complete mapping of the TSP-1 interactome is needed to understand its diverse functions and to lay the basis for the rational design of TSP-1-based therapeutic approaches. So far, large-scale approaches to identify TSP-1 ligands have been rarely used, but many interactions have been identified in small-scale studies in defined biological systems. This review, based on information from protein interaction databases and the literature, illustrates current knowledge of the TSP-1 interactome map.
Collapse
Affiliation(s)
- Andrea Resovi
- Tumor Angiogenesis Unit, Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 24126 Bergamo, Italy
| | - Denise Pinessi
- Tumor Angiogenesis Unit, Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 24126 Bergamo, Italy
| | - Giovanna Chiorino
- Fondo Edo ed Elvo Tempia Valenta, Laboratory of Cancer Genomics, 13900 Biella, Italy
| | - Giulia Taraboletti
- Tumor Angiogenesis Unit, Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 24126 Bergamo, Italy.
| |
Collapse
|
44
|
Gomez-Puerta JA, Mócsai A. Tyrosine kinase inhibitors for the treatment of rheumatoid arthritis. Curr Top Med Chem 2014; 13:760-73. [PMID: 23574525 PMCID: PMC3796894 DOI: 10.2174/15680266113139990094] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 10/26/2012] [Accepted: 11/09/2012] [Indexed: 12/12/2022]
Abstract
Tyrosine kinases (TK) are enzymes capable of transferring phosphate groups to tyrosine residues in cytoplasmic proteins or the intracellular domains of transmembrane receptors. TK play critical roles in diverse biological functions including cellular processes such as adhesion, motility, proliferation, cell cycle control, cell death, as well as biological functions at the whole-organism level such as growth and development, metabolism or immune defense. TK inhibitors including spleen TK (fostamatinib) and Janus kinases (tofacitinib) inhibitors are two novel oral therapies that have demonstrated short-term good clinical responses in active rheumatoid arthritis patients with and inadequate responses to methotrexate or other traditional (non-biologic) disease-modifying antirheumatic drugs (DMARDs). Those responses are comparable to responses rates from pivotal trials of TNF inhibitors. TK inhibitors are generally well tolerated but not free of adverse effects. Several side effects had been described including gastrointestinal symptoms, neutropenia, hypertension, elevated liver function test and lipid alterations among others. Owing to the limited duration of follow-up of patients treated with TK inhibitors, the long term safety profile of these drugs are unknown.
Collapse
Affiliation(s)
- Jose A Gomez-Puerta
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | | |
Collapse
|
45
|
Lin IC, Sheen JM, Tain YL, Chou MH, Huang LT, Yang KD. Vascular Endothelial Growth Factor-A in <i>Lactobacillus Casei </i>Cell Wall Extract-Induced Coronary Arteritis of a Murine Model. Circ J 2014; 78:752-762. [DOI: 10.1253/circj.cj-13-0612] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- I-Chun Lin
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine
| | - Jiunn-Ming Sheen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine
| | - Ming-Huei Chou
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University
| | - Li-Tung Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine
| | - Kuender D. Yang
- Department of Medical Research, Show Chwan Memorial Hospital in Chang Bing
| |
Collapse
|
46
|
Wang JM, Isenberg JS, Billiar TR, Chen AF. Thrombospondin-1/CD36 pathway contributes to bone marrow-derived angiogenic cell dysfunction in type 1 diabetes via Sonic hedgehog pathway suppression. Am J Physiol Endocrinol Metab 2013; 305:E1464-72. [PMID: 24148348 PMCID: PMC3882377 DOI: 10.1152/ajpendo.00516.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 10/21/2013] [Indexed: 01/30/2023]
Abstract
Refractory wounds in diabetic patients present a significant clinical problem. Sonic hedgehog (SHH), a morphogenic protein central to wound repair, is deficient in diabetes. Regulation of SHH in wound healing is poorly understood. We hypothesize that thrombospondin-1 (TSP-1), through its receptor CD36, contributes to the SHH signaling defect in bone marrow-derived angiogenic cells (BMACs) in type 1 diabetic mice. Isolated BMACs from TSP-1-knockout mice demonstrated improved tube formation, migration, and adhesion in parallel with active SHH signaling. BMACs from STZ-induced type 1 diabetic mice showed significantly impaired Matrigel tube formation (n = 5; P < 0.05 vs. control), which was rescued by TSP-1 depletion (n = 5; P < 0.05 STZ-TSP-1(-/-) vs. STZ-WT) or exogenous SHH (20 mg/l, 24 h, n = 4; P < 0.05 vs. STZ-control). The expression of CD36 was elevated in BMACs from STZ mice (n = 4; P < 0.05). SHH signaling was significantly higher in BMACs from TSP-1(-/-) mice and TSP-1 receptor CD36-knockout mice (n = 6; P < 0.05 vs. WT) but not CD47-knockout mice (n = 3; P > 0.05 vs. WT). The impairment of recombinant human TSP-1 (2.2 nM, 24 h) on BMAC Matrigel tube formation was delayed significantly by CD36 deletion (n = 5; P < 0.05). CD36(-/-) BMACs demonstrated better tube formation under both normal and diabetic conditions with active SHH signaling (n = 4; P < 0.05 vs. WT BMACs). In conclusion, The TSP-1/CD36 pathway contributes to the SHH signaling defect, resulting in BMAC dysfunction in type 1 diabetic mice.
Collapse
MESH Headings
- Animals
- Bone Marrow Cells/physiology
- CD36 Antigens/physiology
- Cells, Cultured
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/physiopathology
- Diabetic Angiopathies/etiology
- Diabetic Angiopathies/physiopathology
- Endothelial Cells/physiology
- Gene Silencing
- Hedgehog Proteins/antagonists & inhibitors
- Hedgehog Proteins/genetics
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/physiology
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neovascularization, Physiologic/drug effects
- Neovascularization, Physiologic/genetics
- Signal Transduction
- Streptozocin
- Thrombospondin 1/physiology
Collapse
Affiliation(s)
- Jie-Mei Wang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | | | | |
Collapse
|
47
|
Berre S, Gaudin R, Cunha de Alencar B, Desdouits M, Chabaud M, Naffakh N, Rabaza-Gairi M, Gobert FX, Jouve M, Benaroch P. CD36-specific antibodies block release of HIV-1 from infected primary macrophages and its transmission to T cells. ACTA ACUST UNITED AC 2013; 210:2523-38. [PMID: 24145510 PMCID: PMC3832921 DOI: 10.1084/jem.20130566] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
HIV-1-infected macrophages likely represent viral reservoirs, as they accumulate newly formed virions in internal virus-containing compartments (VCCs). However, the nature and biogenesis of VCCs remain poorly defined. We show that upon HIV-1 infection of primary human macrophages, Gag is recruited to preexisting compartments containing the scavenger receptor CD36, which then become VCCs. Silencing of CD36 in HIV-1-infected macrophages decreases the amount of virions released. Strikingly, soluble anti-CD36 antibodies, but not the natural ligands of CD36, inhibit release of virions from HIV-1-infected macrophages and the transmission of virus to CD4(+) T cells. The effect of the antibodies is potent, rapid, and induces the retention of virions within VCCs. Ectopic expression of CD36 in HeLa cells renders them susceptible to the inhibitory effect of the anti-CD36 mAb upon HIV-1 infection. We show that the anti-CD36 mAb inhibits HIV-1 release by clustering newly formed virions at their site of budding, and that signaling via CD36 is not required. Thus, HIV-1 reservoirs in macrophages may be tackled therapeutically using anti-CD36 antibodies to prevent viral dissemination.
Collapse
Affiliation(s)
- Stefano Berre
- Institut Curie, Centre de Recherche, F-75248 Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Rios FJO, Ferracini M, Pecenin M, Koga MM, Wang Y, Ketelhuth DFJ, Jancar S. Uptake of oxLDL and IL-10 production by macrophages requires PAFR and CD36 recruitment into the same lipid rafts. PLoS One 2013; 8:e76893. [PMID: 24130805 PMCID: PMC3793910 DOI: 10.1371/journal.pone.0076893] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 08/28/2013] [Indexed: 01/24/2023] Open
Abstract
Macrophage interaction with oxidized low-density lipoprotein (oxLDL) leads to its differentiation into foam cells and cytokine production, contributing to atherosclerosis development. In a previous study, we showed that CD36 and the receptor for platelet-activating factor (PAFR) are required for oxLDL to activate gene transcription for cytokines and CD36. Here, we investigated the localization and physical interaction of CD36 and PAFR in macrophages stimulated with oxLDL. We found that blocking CD36 or PAFR decreases oxLDL uptake and IL-10 production. OxLDL induces IL-10 mRNA expression only in HEK293T expressing both receptors (PAFR and CD36). OxLDL does not induce IL-12 production. The lipid rafts disruption by treatment with βCD reduces the oxLDL uptake and IL-10 production. OxLDL induces co-immunoprecipitation of PAFR and CD36 with the constitutive raft protein flotillin-1, and colocalization with the lipid raft-marker GM1-ganglioside. Finally, we found colocalization of PAFR and CD36 in macrophages from human atherosclerotic plaques. Our results show that oxLDL induces the recruitment of PAFR and CD36 into the same lipid rafts, which is important for oxLDL uptake and IL-10 production. This study provided new insights into how oxLDL interact with macrophages and contributing to atherosclerosis development.
Collapse
Affiliation(s)
- Francisco J O Rios
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil ; BHF-Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
49
|
CD36 recruits α₅β₁ integrin to promote cytoadherence of P. falciparum-infected erythrocytes. PLoS Pathog 2013; 9:e1003590. [PMID: 24009511 PMCID: PMC3757042 DOI: 10.1371/journal.ppat.1003590] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 07/15/2013] [Indexed: 12/15/2022] Open
Abstract
The adhesion of Plasmodium falciparum-infected erythrocytes (IRBC) to receptors on different host cells plays a divergent yet critical role in determining the progression and outcome of the infection. Based on our ex vivo studies with clinical parasite isolates from adult Thai patients, we have previously proposed a paradigm for IRBC cytoadherence under physiological shear stress that consists of a recruitment cascade mediated largely by P-selectin, ICAM-1 and CD36 on primary human dermal microvascular endothelium (HDMEC). In addition, we detected post-adhesion signaling events involving Src family kinases and the adaptor protein p130CAS in endothelial cells that lead to CD36 clustering and cytoskeletal rearrangement which enhance the magnitude of the adhesive strength, allowing adherent IRBC to withstand shear stress of up to 20 dynes/cm2. In this study, we addressed whether CD36 supports IRBC adhesion as part of an assembly of membrane receptors. Using a combination of flow chamber assay, atomic force and confocal microscopy, we showed for the first time by loss- and gain-of function assays that in the resting state, the integrin α5β1 does not support adhesive interactions between IRBC and HDMEC. Upon IRBC adhesion to CD36, the integrin is recruited either passively as part of a molecular complex with CD36, or actively to the site of IRBC attachment through phosphorylation of Src family kinases, a process that is Ca2+-dependent. Clustering of β1 integrin is associated with an increase in IRBC recruitment as well as in adhesive strength after attachment (∼40% in both cases). The adhesion of IRBC to a multimolecular complex on the surface of endothelial cells could be of critical importance in enabling adherent IRBC to withstand the high shear stress in the microcirculations. Targeting integrins may provide a novel approach to decrease IRBC cytoadherence to microvascular endothelium. Of the several species of malaria parasites that infect humans, disease caused by Plasmodium falciparum is responsible for most of the deaths. The unique pathological finding of this infection is the intense adhesion of infected red blood cells (IRBC) in the microcirculation, resulting in obstruction to blood flow and organ dysfunction. The focus of our research is to identify the molecules on host endothelial cells that support the adhesion as potential therapeutic targets. In this report, we showed for the first time a functional association between CD36, a well studied adhesion molecule for parasite adhesion, and α5β1, a member of the integrin family of adhesion molecules that are important for adhesion of leukocytes to blood vessels and cell adhesion to the extracellular matrix. We found that by itself, α5β1 integrin does not support IRBC adhesion. When IRBC adhere to CD36, the integrin is recruited to the site of adhesion through activation of the Src family kinase signaling pathway. As a result, both the number of adherent IRBC and the strength with which they adhere is greatly increased. These results demonstrate that IRBC adhesion is a dynamic and complex process. We need to identify each of the functional components to design anti-adhesive therapy.
Collapse
|
50
|
Thrombospondin-1 modulates VEGF signaling via CD36 by recruiting SHP-1 to VEGFR2 complex in microvascular endothelial cells. Blood 2013; 122:1822-32. [PMID: 23896411 DOI: 10.1182/blood-2013-01-482315] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Thrombospondin-1 (TSP-1) inhibits growth factor signaling at the receptor level in microvascular endothelial cells (MVEC), and CD36 has been suggested to be involved in this inhibition, but the mechanisms are not known. We hypothesized that CD36-TSP-1 interaction recruits Src homology 2 domain-containing protein tyrosine phosphatase (SHP)-1 to the vascular endothelial growth factor receptor 2 (VEGFR2) signaling complex and attenuates vascular endothelial growth factor (VEGF) signaling. Western blots of anti-CD36 and anti-VEGFR2 immunoprecipitates from VEGF-treated MVEC showed that exposure of the cells to a recombinant protein containing the CD36 binding domain of thrombospondin-1 (known as the TSR domain) induced association of SHP-1 with the VEGFR2/CD36 signaling complex and thereby suppressed VEGFR2 phosphorylation. SHP-1 phosphatase activity was increased in immunoprecipitated VEGFR2 complexes from TSR-treated cells. Silencing CD36 expression in MVEC by small interfering RNA (siRNA) or genetic deletion of cd36 in mice showed that TSR-induced SHP-1/VEGFR2 complex formation required CD36 in vitro and in vivo. Silencing SHP-1 expression in MVEC by siRNA abrogated TSR-mediated inhibition of VEGFR2 phosphorylation as well as TSR-mediated inhibition of VEGF-induced endothelial cell migration and tube formation. These studies reveal a SHP-1-mediated antiangiogenic pathway induced by CD36-TSP-1 interaction that inhibits VEGFR2 signaling and they provide a novel target to modulate angiogenesis therapeutically.
Collapse
|