1
|
Isakova AA, Artykov AA, Plotnikova EA, Trunova GV, Khokhlova VА, Pankratov AA, Shuvalova ML, Mazur DV, Antipova NV, Shakhparonov MI, Dolgikh DA, Kirpichnikov MP, Gasparian ME, Yagolovich AV. Dual targeting of DR5 and VEGFR2 molecular pathways by multivalent fusion protein significantly suppresses tumor growth and angiogenesis. Int J Biol Macromol 2024; 255:128096. [PMID: 37972835 DOI: 10.1016/j.ijbiomac.2023.128096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/31/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
Destroying tumor vasculature is a relevant therapeutic strategy due to its involvement in tumor progression. However, adaptive resistance to approved antiangiogenic drugs targeting VEGF/VEGFR pathway requires the recruitment of additional targets. In this aspect, targeting TRAIL pathway is promising as it is an important component of the immune system involved in tumor immunosurveillance. For dual targeting of malignant cells and tumor vascular microenvironment, we designed a multivalent fusion protein SRH-DR5-B-iRGD with antiangiogenic VEGFR2-specific peptide SRH at the N-terminus and a tumor-targeting and -penetrating peptide iRGD at the C-terminus of receptor-selective TRAIL variant DR5-B. SRH-DR5-B-iRGD obtained high affinity for DR5, VEGFR2 and αvβ3 integrin in nanomolar range. Fusion of DR5-B with effector peptides accelerated DR5 receptor internalization rate upon ligand binding. Antitumor efficacy was evaluated in vitro in human tumor cell lines and primary patient-derived glioblastoma neurospheres, and in vivo in xenograft mouse model of human glioblastoma. Multivalent binding of SRH-DR5-B-iRGD fusion efficiently stimulated DR5-mediated tumor cell death via caspase-dependent mechanism, suppressed xenograft tumor growth by >80 %, doubled the lifespan of xenograft animals, and inhibited tumor vascularization. Therefore, targeting DR5 and VEGFR2 molecular pathways with SRH-DR5-B-iRGD protein may provide a novel therapeutic approach for treatment of solid tumors.
Collapse
Affiliation(s)
- Alina A Isakova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia; Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Artem A Artykov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Ekaterina A Plotnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia; P.А. Hertsen Moscow Oncology Research Institute - branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 125284 Moscow, Russia
| | - Galina V Trunova
- P.А. Hertsen Moscow Oncology Research Institute - branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 125284 Moscow, Russia
| | - Varvara А Khokhlova
- P.А. Hertsen Moscow Oncology Research Institute - branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 125284 Moscow, Russia
| | - Andrey A Pankratov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia; P.А. Hertsen Moscow Oncology Research Institute - branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 125284 Moscow, Russia
| | - Margarita L Shuvalova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia; Laboratory of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Diana V Mazur
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia; Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Nadezhda V Antipova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | | | - Dmitry A Dolgikh
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia; Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Mikhail P Kirpichnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia; Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Marine E Gasparian
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia; Manebio LLC, 115280 Moscow, Russia.
| | - Anne V Yagolovich
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia; Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia; Manebio LLC, 115280 Moscow, Russia.
| |
Collapse
|
2
|
Kastana P, Ntenekou D, Mourkogianni E, Enake MK, Xanthopoulos A, Choleva E, Marazioti A, Nikou S, Akwii RG, Papadaki E, Gramage E, Herradón G, Stathopoulos GT, Mikelis CM, Papadimitriou E. Genetic deletion or tyrosine phosphatase inhibition of PTPRZ1 activates c-Met to up-regulate angiogenesis and lung adenocarcinoma growth. Int J Cancer 2023; 153:1051-1066. [PMID: 37260355 PMCID: PMC10524925 DOI: 10.1002/ijc.34564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/12/2023] [Accepted: 04/28/2023] [Indexed: 06/02/2023]
Abstract
Protein tyrosine phosphatase receptor zeta 1 (PTPRZ1) is a transmembrane tyrosine phosphatase (TP) expressed in endothelial cells and required for stimulation of cell migration by vascular endothelial growth factor A165 (VEGFA165 ) and pleiotrophin (PTN). It is also over or under-expressed in various tumor types. In this study, we used genetically engineered Ptprz1-/- and Ptprz1+/+ mice to study mechanistic aspects of PTPRZ1 involvement in angiogenesis and investigate its role in lung adenocarcinoma (LUAD) growth. Ptprz1-/- lung microvascular endothelial cells (LMVEC) have increased angiogenic features compared with Ptprz1+/+ LMVEC, in line with the increased lung angiogenesis and the enhanced chemically induced LUAD growth in Ptprz1-/- compared with Ptprz1+/+ mice. In LUAD cells isolated from the lungs of urethane-treated mice, PTPRZ1 TP inhibition also enhanced proliferation and migration. Expression of beta 3 (β3 ) integrin is decreased in Ptprz1-/- LMVEC, linked to enhanced VEGF receptor 2 (VEGFR2), c-Met tyrosine kinase (TK) and Akt kinase activities. However, only c-Met and Akt seem responsible for the enhanced endothelial cell activation in vitro and LUAD growth and angiogenesis in vivo in Ptprz1-/- mice. A selective PTPRZ1 TP inhibitor, VEGFA165 and PTN also activate c-Met and Akt in a PTPRZ1-dependent manner in endothelial cells, and their stimulatory effects are abolished by the c-Met TK inhibitor (TKI) crizotinib. Altogether, our data suggest that low PTPRZ1 expression is linked to worse LUAD prognosis and response to c-Met TKIs and uncover for the first time the role of PTPRZ1 in mediating c-Met activation by VEGFA and PTN.
Collapse
Affiliation(s)
- Pinelopi Kastana
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Greece
| | - Despoina Ntenekou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Greece
| | - Eleni Mourkogianni
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Greece
| | - Michaela-Karina Enake
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Greece
| | | | - Effrosyni Choleva
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Greece
| | - Antonia Marazioti
- Laboratory of Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Greece
| | - Sophia Nikou
- Department of Anatomy, Faculty of Medicine, University of Patras, Greece
| | - Racheal G. Akwii
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Centre, Amarillo, TX, USA
| | - Eleni Papadaki
- Department of Anatomy, Faculty of Medicine, University of Patras, Greece
| | - Esther Gramage
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Gonzalo Herradón
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Georgios T. Stathopoulos
- Laboratory of Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Greece
| | - Constantinos M. Mikelis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Greece
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Centre, Amarillo, TX, USA
| | | |
Collapse
|
3
|
Abstract
The endothelium is a dynamic, semipermeable layer lining all blood vessels, regulating blood vessel formation and barrier function. Proper composition and function of the endothelial barrier are required for fluid homeostasis, and clinical conditions characterized by barrier disruption are associated with severe morbidity and high mortality rates. Endothelial barrier properties are regulated by cell-cell junctions and intracellular signaling pathways governing the cytoskeleton, but recent insights indicate an increasingly important role for integrin-mediated cell-matrix adhesion and signaling in endothelial barrier regulation. Here, we discuss diseases characterized by endothelial barrier disruption, and provide an overview of the composition of endothelial cell-matrix adhesion complexes and associated signaling pathways, their crosstalk with cell-cell junctions, and with other receptors. We further present recent insights into the role of cell-matrix adhesions in the developing and mature/adult endothelium of various vascular beds, and discuss how the dynamic regulation and turnover of cell-matrix adhesions regulates endothelial barrier function in (patho)physiological conditions like angiogenesis, inflammation and in response to hemodynamic stress. Finally, as clinical conditions associated with vascular leak still lack direct treatment, we focus on how understanding of endothelial cell-matrix adhesion may provide novel targets for treatment, and discuss current translational challenges and future perspectives.
Collapse
Affiliation(s)
- Jurjan Aman
- Department of Pulmonology, Amsterdam University Medical Center, the Netherlands (J.A.)
| | - Coert Margadant
- Department of Medical Oncology, Amsterdam University Medical Center, the NetherlandsInstitute of Biology, Leiden University, the Netherlands (C.M.)
| |
Collapse
|
4
|
Chen JR, Zhao JT, Xie ZZ. Integrin-mediated cancer progression as a specific target in clinical therapy. Biomed Pharmacother 2022; 155:113745. [DOI: 10.1016/j.biopha.2022.113745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 11/15/2022] Open
|
5
|
Nuclear Molecular Imaging of Cardiac Remodeling after Myocardial Infarction. Pharmaceuticals (Basel) 2022; 15:ph15020183. [PMID: 35215296 PMCID: PMC8875369 DOI: 10.3390/ph15020183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 12/03/2022] Open
Abstract
The role of molecular imaging technologies in detecting, evaluating, and monitoring cardiovascular disease and their treatment is expanding rapidly. Gradually replacing the conventional anatomical or physiological approaches, molecular imaging strategies using biologically targeted markers provide unique insight into pathobiological processes at molecular and cellular levels and allow for cardiovascular disease evaluation and individualized therapy. This review paper will discuss currently available and developing molecular-based single-photon emission computed tomography (SPECT) and positron emission tomography (PET) imaging strategies to evaluate post-infarction cardiac remodeling. These approaches include potential targeted methods of evaluating critical biological processes, such as inflammation, angiogenesis, and scar formation.
Collapse
|
6
|
Kossatz S, Beer AJ, Notni J. It's Time to Shift the Paradigm: Translation and Clinical Application of Non-αvβ3 Integrin Targeting Radiopharmaceuticals. Cancers (Basel) 2021; 13:cancers13235958. [PMID: 34885066 PMCID: PMC8657165 DOI: 10.3390/cancers13235958] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Cancer cells often present a different set of proteins on their surface than normal cells. This also applies to integrins, a class of 24 cell surface receptors which mainly are responsible for physically anchoring cells in tissues, but also fulfil a plethora of other functions. If a certain integrin is found on tumor cells but not on normal ones, radioactive molecules (named tracers) that specifically bind to this integrin will accumulate in the cancer lesion if injected into the blood stream. The emitted radiation can be detected from outside the body and allows for localization and thus, diagnosis, of cancer. Only one of the 24 integrins, the subtype αvβ3, has hitherto been thoroughly investigated in this context. We herein summarize the most recent, pertinent research on other integrins, and argue that some of these approaches might ultimately improve the clinical management of the most lethal cancers, such as pancreatic carcinoma. Abstract For almost the entire period of the last two decades, translational research in the area of integrin-targeting radiopharmaceuticals was strongly focused on the subtype αvβ3, owing to its expression on endothelial cells and its well-established role as a biomarker for, and promoter of, angiogenesis. Despite a large number of translated tracers and clinical studies, a clinical value of αvβ3-integrin imaging could not be defined yet. The focus of research has, thus, been moving slowly but steadily towards other integrin subtypes which are involved in a large variety of tumorigenic pathways. Peptidic and non-peptidic radioligands for the integrins α5β1, αvβ6, αvβ8, α6β1, α6β4, α3β1, α4β1, and αMβ2 were first synthesized and characterized preclinically. Some of these compounds, targeting the subtypes αvβ6, αvβ8, and α6β1/β4, were subsequently translated into humans during the last few years. αvβ6-Integrin has arguably attracted most attention because it is expressed by some of the cancers with the worst prognosis (above all, pancreatic ductal adenocarcinoma), which substantiates a clinical need for the respective theranostic agents. The receptor furthermore represents a biomarker for malignancy and invasiveness of carcinomas, as well as for fibrotic diseases, such as idiopathic pulmonary fibrosis (IPF), and probably even for Sars-CoV-2 (COVID-19) related syndromes. Accordingly, the largest number of recent first-in-human applications has been reported for radiolabeled compounds targeting αvβ6-integrin. The results indicate a substantial clinical value, which might lead to a paradigm change and trigger the replacement of αvβ3 by αvβ6 as the most popular integrin in theranostics.
Collapse
Affiliation(s)
- Susanne Kossatz
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, 81675 Munich, Germany;
- Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | | | - Johannes Notni
- Department of Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- TRIMT GmbH, 01454 Radeberg, Germany
- Correspondence: ; Tel.: +49-89-4140-6075; Fax: +49-89-4140-6949
| |
Collapse
|
7
|
Newport EL, Pedrosa AR, Njegic A, Hodivala-Dilke KM, Muñoz-Félix JM. Improved Immunotherapy Efficacy by Vascular Modulation. Cancers (Basel) 2021; 13:5207. [PMID: 34680355 PMCID: PMC8533721 DOI: 10.3390/cancers13205207] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 12/26/2022] Open
Abstract
Several strategies have been developed to modulate the tumour vasculature for cancer therapy including anti-angiogenesis and vascular normalisation. Vasculature modulation results in changes to the tumour microenvironment including oxygenation and immune cell infiltration, therefore lending itself to combination with cancer therapy. The development of immunotherapies has led to significant improvements in cancer treatment. Particularly promising are immune checkpoint blockade and CAR T cell therapies, which use antibodies against negative regulators of T cell activation and T cells reprogrammed to better target tumour antigens, respectively. However, while immunotherapy is successful in some patients, including those with advanced or metastatic cancers, only a subset of patients respond. Therefore, better predictors of patient response and methods to overcome resistance warrant investigation. Poor, or periphery-limited, T cell infiltration in the tumour is associated with poor responses to immunotherapy. Given that (1) lymphocyte recruitment requires leucocyte-endothelial cell adhesion and (2) the vasculature controls tumour oxygenation and plays a pivotal role in T cell infiltration and activation, vessel targeting strategies including anti-angiogenesis and vascular normalisation in combination with immunotherapy are providing possible new strategies to enhance therapy. Here, we review the progress of vessel modulation in enhancing immunotherapy efficacy.
Collapse
Affiliation(s)
- Emma L. Newport
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (E.L.N.); (A.R.P.); (A.N.); (K.M.H.-D.)
| | - Ana Rita Pedrosa
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (E.L.N.); (A.R.P.); (A.N.); (K.M.H.-D.)
| | - Alexandra Njegic
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (E.L.N.); (A.R.P.); (A.N.); (K.M.H.-D.)
| | - Kairbaan M. Hodivala-Dilke
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (E.L.N.); (A.R.P.); (A.N.); (K.M.H.-D.)
| | - José M. Muñoz-Félix
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (E.L.N.); (A.R.P.); (A.N.); (K.M.H.-D.)
- Department of Biochemistry and Molecular Biology, Institute of Biomedical Research of Salamanca (IBSAL), Universidad de Salamanca Spain, 37007 Salamanca, Spain
| |
Collapse
|
8
|
Steiger K, Quigley NG, Groll T, Richter F, Zierke MA, Beer AJ, Weichert W, Schwaiger M, Kossatz S, Notni J. There is a world beyond αvβ3-integrin: Multimeric ligands for imaging of the integrin subtypes αvβ6, αvβ8, αvβ3, and α5β1 by positron emission tomography. EJNMMI Res 2021; 11:106. [PMID: 34636990 PMCID: PMC8506476 DOI: 10.1186/s13550-021-00842-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND In the context of nuclear medicine and theranostics, integrin-related research and development was, for most of the time, focused predominantly on 'RGD peptides' and the subtype αvβ3-integrin. However, there are no less than 24 known integrins, and peptides without the RGD sequence as well as non-peptidic ligands play an equally important role as selective integrin ligands. On the other hand, multimerization is a well-established method to increase the avidity of binding structures, but multimeric radiopharmaceuticals have not made their way into clinics yet. In this review, we describe how these aspects have been interwoven in the framework of the German Research Foundation's multi-group interdisciplinary funding scheme CRC 824, yielding a series of potent PET imaging agents for selective imaging of various integrin subtypes. RESULTS The gallium-68 chelator TRAP was utilized to elaborate symmetrical trimers of various peptidic and non-peptidic integrin ligands. Preclinical data suggested a high potential of the resulting Ga-68-tracers for PET-imaging of the integrins α5β1, αvβ8, αvβ6, and αvβ3. For the first three, we provide some additional immunohistochemistry data in human cancers, which suggest several future clinical applications. Finally, application of αvβ3- and αvβ6-integrin tracers in pancreatic carcinoma patients revealed that unlike αvβ3-targeted PET, αvβ6-integrin PET is not characterized by off-target uptake and thus, enables a substantially improved imaging of this type of cancer. CONCLUSIONS Novel radiopharmaceuticals targeting a number of different integrins, above all, αvβ6, have proven their clinical potential and will play an increasingly important role in future theranostics.
Collapse
Affiliation(s)
- Katja Steiger
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Neil Gerard Quigley
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Tanja Groll
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Frauke Richter
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | | | | | - Wilko Weichert
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Markus Schwaiger
- Klinik Für Nuklearmedizin Und Zentralinstitut Für Translationale Krebsforschung (TranslaTUM), Klinikum Rechts Der Isar der Technischen Universität München, Munich, Germany
| | - Susanne Kossatz
- Klinik Für Nuklearmedizin Und Zentralinstitut Für Translationale Krebsforschung (TranslaTUM), Klinikum Rechts Der Isar der Technischen Universität München, Munich, Germany
| | - Johannes Notni
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany. .,Experimental Radiopharmacy, Clinic for Nuclear Medicine, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany.
| |
Collapse
|
9
|
Peláez R, Ochoa R, Pariente A, Villanueva-Martínez Á, Pérez-Sala Á, Larráyoz IM. Sterculic Acid Alters Adhesion Molecules Expression and Extracellular Matrix Compounds to Regulate Migration of Lung Cancer Cells. Cancers (Basel) 2021; 13:cancers13174370. [PMID: 34503180 PMCID: PMC8431022 DOI: 10.3390/cancers13174370] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/19/2021] [Accepted: 08/25/2021] [Indexed: 12/11/2022] Open
Abstract
Sterculic acid (SA) is a cyclopropenoid fatty acid isolated from Sterculia foetida seeds. This molecule is a well-known inhibitor of SCD1 enzyme, also known as ∆9-desaturase, which main function is related to lipid metabolism. However, recent studies have demonstrated that it also modifies many other pathways and the underlying gene expression. SCD overexpression, or up-regulated activity, has been associated with tumor aggressiveness and poor prognosis in many cancer types. Scd1 down-regulation, with different inhibitors or molecular strategies, reduces tumor cell survival and cell proliferation, as well as the chemoresistance associated with cancer stem cell presence. However, SA effects over cancer cell migration and extracellular matrix or adhesion molecules have not been described in cancer cells up to now. We used different migration assays and qPCR gene expression analysis to evaluate the effects of SA treatment in cancer cells. The results reveal that SA induces tumoral cell death at high doses, but we also observed that lower SA-treatments induce cell adhesion-migration capacity reduction as a result of modifications in the expression of genes related to integrins and extracellular matrix compounds. Overall, the functional and transcriptomic findings suggest that SA could represent a new inhibitor activity of epithelial to mesenchymal transition.
Collapse
Affiliation(s)
- Rafael Peláez
- Correspondence: (R.P.); (I.M.L.); Tel.: +34-941-278-770 ((ext. 84866) (R.P.) & (ext. 89878) (I.M.L.))
| | | | | | | | | | - Ignacio M. Larráyoz
- Correspondence: (R.P.); (I.M.L.); Tel.: +34-941-278-770 ((ext. 84866) (R.P.) & (ext. 89878) (I.M.L.))
| |
Collapse
|
10
|
Hu M, Varier KM, Li Z, Qin X, Rao Q, Song J, Hu A, Hang Y, Yuan C, Gajendran B, Shu L, Wen M, Li Y, Liu H. A natural acylphloroglucinol triggered antiproliferative possessions in HEL cells by impeding STAT3 signaling and attenuating angiogenesis in transgenic zebrafish model. Biomed Pharmacother 2021; 141:111877. [PMID: 34323693 DOI: 10.1016/j.biopha.2021.111877] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/07/2021] [Accepted: 06/28/2021] [Indexed: 01/20/2023] Open
Abstract
Leukemia is responsible for a reason of death, globally. Even though there are several treatment regimens available in the clinics against this disease, a perfect chemotherapeutic agent for the same is still under investigation. Natural plant-derived secondary metabolites are used in clinics to treat leukemia for better benefits with reduced side-effects. Likely, several bioactive compounds from Callistemon sp. were reported for their bioactive benefits. Furthermore, acylphloroglucinol derivatives from Callistemon salignus, showed both antimicrobial and cytotoxic activities in various adherent human cancer cell lines. Thus, in the present study, a natural acylphloroglucinol (2,6-dihydroxy-4-methoxyisobutyrophenone, L72) was tested for its antiproliferative efficacy in HEL cells. The MTT and the cell cycle analysis study revealed that L72 treatment can offer antiproliferative effects, both time and dose-dependent manner, causing G2/M cell cycle arrest. The western blot analysis revealed that L72 treatment triggered intrinsic apoptotic machinery and activated p21. Likewise, L72 could downregulate the gene expressions of XIAP, FLT3, IDH2, and SOD2, which was demonstrated by qPCR analysis, thus promoting its antiproliferative action. The L72 could impede STAT3 expression, which was evidenced by insilico autodock analysis and western blot analysis using STAT3 inhibitor, Pimozide. The treatment of transgenic (Flk-1+/egfr+) zebrafish embryos resulted in the STAT3 gene inhibition, proving its anti-angiogenic effect, as well. Thus, the study revealed that L72 could act as an antiproliferative agent, by triggering caspase-dependent intrinsic apoptosis, reducing cell proliferation by attenuating STAT3, and activating an anti-angiogenic pathway via Flk-1inhibition.
Collapse
Affiliation(s)
- Mi Hu
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China; Key Laboratory of Regenerative Medicine of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou, PR China
| | - Krishnapriya M Varier
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China; School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Zhicao Li
- Key Laboratory of Regenerative Medicine of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou, PR China
| | - Xujie Qin
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650204, PR China
| | - Qing Rao
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Jingrui Song
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Anling Hu
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Yubing Hang
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Chunmao Yuan
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Babu Gajendran
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China; School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou, PR China.
| | - Liping Shu
- Key Laboratory of Regenerative Medicine of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou, PR China.
| | - Min Wen
- Key Laboratory of Regenerative Medicine of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou, PR China.
| | - Yanmei Li
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China.
| | - Haiyang Liu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650204, PR China.
| |
Collapse
|
11
|
Dzobo K. Integrins Within the Tumor Microenvironment: Biological Functions, Importance for Molecular Targeting, and Cancer Therapeutics Innovation. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:417-430. [PMID: 34191612 DOI: 10.1089/omi.2021.0069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Many cellular functions important for solid tumor initiation and progression are mediated by members of the integrin family, a diverse family of cell attachment receptors. With recent studies emphasizing the role of the tumor microenvironment (TME) in tumor initiation and progression, it is not surprising that considerable attention is being paid to integrins. Several integrin antagonists are under clinical trials, with many demonstrating promising activity in patients with different cancers. A deeper knowledge of the functions of integrins within the TME is still required and might lead to better inhibitors being discovered. Integrin expression is commonly dysregulated in many tumors with integrins playing key roles in signaling as well as promotion of tumor cell invasion and migration. Integrins also play a major role in adhesion of circulating tumor cells to new sites and the resulting formation of secondary tumors. Furthermore, integrins have demonstrated the ability to promoting stem cell-like properties in tumor cells as well as drug resistance. Anti-integrin therapies rely heavily on the doses or concentrations used as these determine whether the drugs act as antagonists or as integrin agonists. This expert review offers the latest synthesis in terms of the current knowledge of integrins functions within the TME and as potential molecular targets for cancer therapeutics innovation.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
12
|
Notohamiprodjo S, Varasteh Z, Beer AJ, Niu G, Chen X(S, Weber W, Schwaiger M. Tumor Vasculature. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00090-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
13
|
Bhavya B, Easwer HV, Vilanilam GC, Anand CR, Sreelakshmi K, Urulangodi M, Rajalakshmi P, Neena I, Padmakrishnan CJ, Menon GR, Krishnakumar K, Deepti AN, Gopala S. MutT Homolog1 has multifaceted role in glioma and is under the apparent orchestration by Hypoxia Inducible factor1 alpha. Life Sci 2020; 264:118673. [PMID: 33130078 DOI: 10.1016/j.lfs.2020.118673] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/15/2020] [Accepted: 10/22/2020] [Indexed: 11/17/2022]
Abstract
AIMS The study focused on the expression and role of a recent potential cancer therapeutic target protein, MutT Homolog1 (MTH1). MTH1 gets activated in an increased reactive oxygen species (ROS) environment and removes the oxidized nucleotides from the cell. The study aimed to check the role of MTH1 in DNA damage and apoptosis, migration and angiogenesis and also to examine its regulation in glioma. MAIN METHODS The experiments were carried out in human glioma tissue samples and brain tissues of epilepsy patients (non-tumor control). We used two human glioblastomas cell lines, U87MG and U251MG cells. In order to study the role of MTH1 in glioma and to analyze the relation of MTH1 with Hif1α, we have used MTH1 siRNA and Hif1α siRNA respectively. KEY FINDINGS We found an increased expression of MTH1 in glioma tissues compared to the non-tumor brain tissues. Correlation analysis revealed that those samples showing reduced expression of MTH1 also had high levels of DNA damage and apoptotic markers, while diminished expression of angiogenesis regulators and levels of migration. MTH1 knockdown in vitro by siRNA in tumor cell lines corroborates the above observation. This justifies the emergence of MTH1 inhibitors as potential first-in-class drugs. Mechanistically, our observations suggest that Hif1α may modulate MTH1 expression. SIGNIFICANCE We found elevated MTH1 expression in glioma irrespective of their grades, while its inhibition affects multiple tumor progression pathways, and that targeting Hif1α could simulate the same.
Collapse
Affiliation(s)
- Bharathan Bhavya
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India
| | - H V Easwer
- Department of Neurosurgery, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India
| | - G C Vilanilam
- Department of Neurosurgery, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India
| | - C R Anand
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India
| | - K Sreelakshmi
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India
| | - Madhusoodanan Urulangodi
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India
| | - P Rajalakshmi
- Department of Pathology, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India
| | - Issac Neena
- Department of Pathology, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India
| | - C J Padmakrishnan
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India
| | - Girish R Menon
- Department of Neurosurgery, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India
| | - K Krishnakumar
- Department of Neurosurgery, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India
| | - A N Deepti
- Department of Pathology, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India
| | - Srinivas Gopala
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India.
| |
Collapse
|
14
|
Pleiotrophin selectively binds to vascular endothelial growth factor receptor 2 and inhibits or stimulates cell migration depending on α νβ 3 integrin expression. Angiogenesis 2020; 23:621-636. [PMID: 32681389 DOI: 10.1007/s10456-020-09733-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 07/06/2020] [Indexed: 12/28/2022]
Abstract
Pleiotrophin (PTN) has a moderate stimulatory effect on endothelial cell migration through ανβ3 integrin, while it decreases the stimulatory effect of vascular endothelial growth factor A (VEGFA) and inhibits cell migration in the absence of ανβ3 through unknown mechanism(s). In the present work, by using a multitude of experimental approaches, we show that PTN binds to VEGF receptor type 2 (VEGFR2) with a KD of 11.6 nM. Molecular dynamics approach suggests that PTN binds to the same VEGFR2 region with VEGFA through its N-terminal domain. PTN inhibits phosphorylation of VEGFR2 at Tyr1175 and still stimulates endothelial cell migration in the presence of a selective VEGFR2 tyrosine kinase inhibitor. VEGFR2 downregulation by siRNA or an anti-VEGFR2 antibody that binds to the ligand-binding VEGFR2 domain also induce endothelial cell migration, which is abolished by a function-blocking antibody against ανβ3 or the peptide PTN112-136 that binds ανβ3 and inhibits PTN binding. In cells that do not express ανβ3, PTN decreases both VEGFR2 Tyr1175 phosphorylation and cell migration in a VEGFR2-dependent manner. Collectively, our data identify VEGFR2 as a novel PTN receptor involved in the regulation of cell migration by PTN and contribute to the elucidation of the mechanism of activation of endothelial cell migration through the interplay between VEGFR2 and ανβ3.
Collapse
|
15
|
Wong PP, Muñoz-Félix JM, Hijazi M, Kim H, Robinson SD, De Luxán-Delgado B, Rodríguez-Hernández I, Maiques O, Meng YM, Meng Q, Bodrug N, Dukinfield MS, Reynolds LE, Elia G, Clear A, Harwood C, Wang Y, Campbell JJ, Singh R, Zhang P, Schall TJ, Matchett KP, Henderson NC, Szlosarek PW, Dreger SA, Smith S, Jones JL, Gribben JG, Cutillas PR, Meier P, Sanz-Moreno V, Hodivala-Dilke KM. Cancer Burden Is Controlled by Mural Cell-β3-Integrin Regulated Crosstalk with Tumor Cells. Cell 2020; 181:1346-1363.e21. [PMID: 32473126 DOI: 10.1016/j.cell.2020.02.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 11/21/2019] [Accepted: 01/31/2020] [Indexed: 02/07/2023]
Abstract
Enhanced blood vessel (BV) formation is thought to drive tumor growth through elevated nutrient delivery. However, this observation has overlooked potential roles for mural cells in directly affecting tumor growth independent of BV function. Here we provide clinical data correlating high percentages of mural-β3-integrin-negative tumor BVs with increased tumor sizes but no effect on BV numbers. Mural-β3-integrin loss also enhances tumor growth in implanted and autochthonous mouse tumor models with no detectable effects on BV numbers or function. At a molecular level, mural-cell β3-integrin loss enhances signaling via FAK-p-HGFR-p-Akt-p-p65, driving CXCL1, CCL2, and TIMP-1 production. In particular, mural-cell-derived CCL2 stimulates tumor cell MEK1-ERK1/2-ROCK2-dependent signaling and enhances tumor cell survival and tumor growth. Overall, our data indicate that mural cells can control tumor growth via paracrine signals regulated by β3-integrin, providing a previously unrecognized mechanism of cancer growth control.
Collapse
Affiliation(s)
- Ping-Pui Wong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Centre for Tumor Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK.
| | - José M Muñoz-Félix
- Centre for Tumor Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK.
| | - Maruan Hijazi
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Hyojin Kim
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Mary-Jean Mitchell Green Building, Chester Beatty Laboratories, 237 Fulham Road, London SW3 6JB, UK
| | - Stephen D Robinson
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK; School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Beatriz De Luxán-Delgado
- Centre for Tumor Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Irene Rodríguez-Hernández
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Oscar Maiques
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Ya-Ming Meng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Qiong Meng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Natalia Bodrug
- Centre for Tumor Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Matthew Scott Dukinfield
- Centre for Tumor Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Louise E Reynolds
- Centre for Tumor Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - George Elia
- Centre for Tumor Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Andrew Clear
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Catherine Harwood
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Yu Wang
- ChemoCentryx Inc., 850 Maude Ave., Mountain View, CA 94043, USA
| | | | - Rajinder Singh
- ChemoCentryx Inc., 850 Maude Ave., Mountain View, CA 94043, USA
| | - Penglie Zhang
- ChemoCentryx Inc., 850 Maude Ave., Mountain View, CA 94043, USA
| | - Thomas J Schall
- ChemoCentryx Inc., 850 Maude Ave., Mountain View, CA 94043, USA
| | - Kylie P Matchett
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Peter W Szlosarek
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Sally A Dreger
- Centre for Tumor Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Sally Smith
- Centre for Tumor Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - J Louise Jones
- Centre for Tumor Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - John G Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Pedro R Cutillas
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Pascal Meier
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Mary-Jean Mitchell Green Building, Chester Beatty Laboratories, 237 Fulham Road, London SW3 6JB, UK
| | - Victoria Sanz-Moreno
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Kairbaan M Hodivala-Dilke
- Centre for Tumor Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
16
|
Reciprocal integrin/integrin antagonism through kindlin-2 and Rho GTPases regulates cell cohesion and collective migration. Matrix Biol 2020; 93:60-78. [PMID: 32450218 DOI: 10.1016/j.matbio.2020.05.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 05/12/2020] [Accepted: 05/12/2020] [Indexed: 02/07/2023]
Abstract
Collective cell behaviour during embryogenesis and tissue repair requires the coordination of intercellular junctions, cytoskeleton-dependent shape changes controlled by Rho GTPases, and integrin-dependent cell-matrix adhesion. Many different integrins are simultaneously expressed during wound healing, embryonic development, and sprouting angiogenesis, suggesting that there is extensive integrin/integrin cross-talk to regulate cell behaviour. Here, we show that fibronectin-binding β1 and β3 integrins do not act synergistically, but rather antagonize each other during collective cell processes in neuro-epithelial cells, placental trophoblasts, and endothelial cells. Reciprocal β1/β3 antagonism controls RhoA activity in a kindlin-2-dependent manner, balancing cell spreading, contractility, and intercellular adhesion. In this way, reciprocal β1/β3 antagonism controls cell cohesion and cellular plasticity to switch between extreme and opposing states, including epithelial versus mesenchymal-like phenotypes and collective versus individual cell migration. We propose that integrin/integrin antagonism is a universal mechanism to effectuate social cellular interactions, important for tissue morphogenesis, endothelial barrier function, trophoblast invasion, and sprouting angiogenesis.
Collapse
|
17
|
Are Integrins Still Practicable Targets for Anti-Cancer Therapy? Cancers (Basel) 2019; 11:cancers11070978. [PMID: 31336983 PMCID: PMC6678560 DOI: 10.3390/cancers11070978] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/27/2019] [Accepted: 07/09/2019] [Indexed: 01/01/2023] Open
Abstract
Correlative clinical evidence and experimental observations indicate that integrin adhesion receptors, in particular those of the αV family, are relevant to cancer cell features, including proliferation, survival, migration, invasion, and metastasis. In addition, integrins promote events in the tumor microenvironment that are critical for tumor progression and metastasis, including tumor angiogenesis, matrix remodeling, and the recruitment of immune and inflammatory cells. In spite of compelling preclinical results demonstrating that the inhibition of integrin αVβ3/αVβ5 and α5β1 has therapeutic potential, clinical trials with integrin inhibitors targeting those integrins have repeatedly failed to demonstrate therapeutic benefits in cancer patients. Here, we review emerging integrin functions and their proposed contribution to tumor progression, discuss preclinical evidence of therapeutic significance, revisit clinical trial results, and consider alternative approaches for their therapeutic targeting in oncology, including targeting integrins in the other cells of the tumor microenvironment, e.g., cancer-associated fibroblasts and immune/inflammatory cells. We conclude that integrins remain a valid target for cancer therapy; however, agents with better pharmacological properties, alternative models for their preclinical evaluation, and innovative combination strategies for clinical testing (e.g., together with immuno-oncology agents) are needed.
Collapse
|
18
|
Rauchman M, Griggs D. Emerging strategies to disrupt the central TGF-β axis in kidney fibrosis. Transl Res 2019; 209:90-104. [PMID: 31085163 PMCID: PMC6850218 DOI: 10.1016/j.trsl.2019.04.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/27/2019] [Accepted: 04/08/2019] [Indexed: 12/26/2022]
Abstract
Chronic kidney disease (CKD) affects more than 20 million people in the United States and the global burden of this disorder is increasing. Many affected individuals will progress to end stage kidney disease necessitating dialysis or transplantation. CKD is also a major independent contributor to the risk of cardiovascular morbidity and mortality. Tubulointerstitial fibrosis is a final common pathway for most causes of progressive CKD. Currently, there are no clinically available therapies targeting fibrosis that can slow the decline in kidney function. Although it has long been known that TGF-β signaling is a critical mediator of kidney fibrosis, translating this knowledge to the clinic has been challenging. In this review, we highlight some recent insights into the mechanisms of TGF-β signaling that target activation of this cytokine at the site of injury or selectively inhibit pro-fibrotic gene expression. Molecules directed at these targets hold the promise of attaining therapeutic efficacy while limiting toxicity seen with global inhibition of TGF-β. Kidney injury has profound epigenetic effects leading to altered expression of more than a thousand genes. We discuss how drugs targeting epigenetic modifications, some of which are in use for cancer therapy, have the potential to reprogram gene regulatory networks to favor adaptive repair and prevent fibrosis. The lack of reliable biomarkers of kidney fibrosis is a major limitation in designing clinical trials for testing CKD treatments. We conclude by reviewing recent advances in fibrosis biomarker development.
Collapse
Affiliation(s)
- Michael Rauchman
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri; VA St. Louis Health Care System, Saint Louis, Missouri.
| | - David Griggs
- Department of Molecular Microbiology and Immunology, Edward A. Doisy Research Center, Saint Louis University, Saint Louis, Missouri.
| |
Collapse
|
19
|
Łasiñska I, Mackiewicz J. Integrins as A New Target for Cancer Treatment. Anticancer Agents Med Chem 2019; 19:580-586. [DOI: 10.2174/1871520618666181119103413] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/16/2018] [Accepted: 11/10/2018] [Indexed: 12/19/2022]
Abstract
:Despite the great progress in the development of targeted therapies for different types of cancer utilizing monoclonal antibodies (e.g., cetuximab for colorectal cancer and head and neck cancer therapy), kinase inhibitors (e.g., sorafenib for kidney cancer and gastrointestinal stromal tumours therapy), and immunomodulatory treatments (e.g., nivolumab and pembrolizumab for melanoma therapy and lung cancer therapy), there is still a need to search for new, more effective treatments.:Integrins are responsible for intercellular adhesion and interaction with the cellular matrix. The function of integrins is related to the transduction of intracellular signals associated with adhesion, migration, cell proliferation, differentiation, and apoptosis. Molecules targeting integrins that lead to cancer cell death have been developed. The most advanced molecules studied in clinical trials are abituzumab, intetumumab and cilengitide. There are different groups of anti-integrin drugs: monoclonal antibodies (e.g., abituzumab) and other such as cilengitide, E7820 and MK-0429. These drugs have been evaluated in various cancer types. However, they have shown modest efficacy, and none of them have yet been approved for cancer treatment. Studies have shown that patient selection using biomarkers might improve the efficacy of anti-integrin cancer treatment. Many preclinical models have demonstrated promising results using integrin visualization for cancer detection and treatment efficacy monitoring; however, these strategies require further evaluation in humans.
Collapse
Affiliation(s)
- Izabela Łasiñska
- Department of Medical and Experimental Oncology, Heliodor Swiecicki University Hospital, Poznan University of Medical Sciences, Poznan, Poland
| | - Jacek Mackiewicz
- Department of Medical and Experimental Oncology, Heliodor Swiecicki University Hospital, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
20
|
Zeriouh M, Sabashnikov A, Tenbrock A, Neef K, Merkle J, Eghbalzadeh K, Weber C, Liakopoulos OJ, Deppe AC, Stamm C, Cowan DB, Wahlers T, Choi YH. Dysregulation of proangiogeneic factors in pressure-overload left-ventricular hypertrophy results in inadequate capillary growth. Ther Adv Cardiovasc Dis 2019; 13:1753944719841795. [PMID: 31088231 PMCID: PMC6535753 DOI: 10.1177/1753944719841795] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Background: Pressure-overload left-ventricular hypertrophy (LVH) is an increasingly prevalent pathological condition of the myocardial muscle and an independent risk factor for a variety of cardiac diseases. We investigated changes in expression levels of proangiogeneic genes in a small animal model of LVH. Methods: Myocardial hypertrophy was induced by transaortic constriction (TAC) in C57BL/6 mice and compared with sham-operated controls. The myocardial expression levels of vascular endothelial growth factor (VEGF), its receptors (KDR and FLT-1), stromal-cell-derived factor 1 (SDF1) and the transcription factors hypoxia-inducible factor-1 and 2 (HIF1 and HIF2) were analyzed by quantitative polymerase chain reaction over the course of 25 weeks. Histological sections were stained for caveolin-1 to visualize endothelial cells and determine the capillary density. The left-ventricular morphology and function were assessed weekly by electrocardiogram-gated magnetic resonance imaging. Results: The heart weight of TAC animals increased significantly from week 4 to 25 (p = 0.005) compared with sham-treated animals. At 1 day after TAC, the expression of VEGF and SDF1 also increased, but was downregulated again after 1 week. The expression of HIF2 was significantly downregulated after 1 week and remained at a lower level in the subsequent weeks. The expression level of FLT-1 was also significantly decreased 1 week after TAC. HIF-1 and KDR showed similar changes compared with sham-operated animals. However, the expression levels of HIF1 after 4 and 8 weeks were significantly decreased compared with day 1. KDR changes were significantly decreased after 1, 2, 4, 8 and 25 weeks compared with week 3. After 4 weeks post-TAC, the size of the capillary vessels increased (p = 0.005) while the capillary density itself decreased (TAC: 2143 ± 293 /mm2versus sham: 2531 ± 321 /mm2; p = 0.021). Starting from week 4, the left-ventricular ejection fraction decreased compared with controls (p = 0.049). Conclusions: The decrease in capillary density in the hypertrophic myocardium appears to be linked to the dysregulation in the expression of proangiogeneic factors. The results suggest that overcoming this dysregulation may lead to reconstitution of capillary density in the hypertrophic heart, and thus be beneficial for cardiac function and survival.
Collapse
Affiliation(s)
- Mohamed Zeriouh
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - Anton Sabashnikov
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - Arne Tenbrock
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - Klaus Neef
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - Julia Merkle
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - Kaveh Eghbalzadeh
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - Carolyn Weber
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | | | | | - Christof Stamm
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | - Douglas B Cowan
- Department of Anesthesiology, Perioperative and Pain Medicine, Children's Hospital Boston and Harvard Medical School, Boston, MA, USA
| | - Thorsten Wahlers
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany.,Center of Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Yeong-Hoon Choi
- Center of Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
21
|
Bianchini F, De Santis A, Portioli E, Russo Krauss I, Battistini L, Curti C, Peppicelli S, Calorini L, D'Errico G, Zanardi F, Sartori A. Integrin-targeted AmpRGD sunitinib liposomes as integrated antiangiogenic tools. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 18:135-145. [PMID: 30849548 DOI: 10.1016/j.nano.2019.02.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 02/08/2019] [Accepted: 02/19/2019] [Indexed: 01/01/2023]
Abstract
We report here the preparation, physico-chemical characterization, and biological evaluation of a new liposome formulation as a tool for tumor angiogenesis inhibition. Liposomes are loaded with sunitinib, a tyrosine kinase inhibitor, and decorated with cyclo-aminoprolineRGD units (cAmpRGD), efficient and selective ligands for integrin αVβ3. The RGD units play multiple roles since they target the nanovehicles at the integrin αVβ3-overexpressing cells (e.g. activated endothelial cells), favor their active cell internalization, providing drug accumulation in the cytoplasm, and likely take part in the angiogenesis inhibition by interfering in the αVβ3-VEGFR2 cross-talk. Both in vitro and in vivo studies show a better efficacy of this integrated antiangiogenic tool with respect to the free sunitinib and untargeted sunitinib-loaded liposomes. This system could allow a lower administration of the drug and, by increasing the vector specificity, reduce side-effects in a prolonged antiangiogenic therapy.
Collapse
Affiliation(s)
- Francesca Bianchini
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Firenze, Italy
| | - Augusta De Santis
- Dipartimento di Scienze Chimiche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Elisabetta Portioli
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | - Irene Russo Krauss
- Dipartimento di Scienze Chimiche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Lucia Battistini
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | - Claudio Curti
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | - Silvia Peppicelli
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Firenze, Italy
| | - Lido Calorini
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Firenze, Italy
| | - Gerardino D'Errico
- Dipartimento di Scienze Chimiche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Franca Zanardi
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy.
| | - Andrea Sartori
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy.
| |
Collapse
|
22
|
Thoreau F, Vanwonterghem L, Henry M, Coll JL, Boturyn D. Design of RGD-ATWLPPR peptide conjugates for the dual targeting of α Vβ 3 integrin and neuropilin-1. Org Biomol Chem 2019; 16:4101-4107. [PMID: 29774910 DOI: 10.1039/c8ob00669e] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Targeting the tumour microenvironment is a promising strategy to detect and/or treat cancer. The design of selective compounds that co-target several receptors frequently overexpressed in solid tumours may allow a reliable and selective detection of tumours. Here we report the modular synthesis of compounds encompassing ligands of αVβ3 integrin and neuropilin-1 that are overexpressed in the tumour microenvironment. These compounds were then evaluated through cellular experiments and imaging of tumours in mice. We observed that the peptide that displays both ligands is more specifically accumulating in the tumours than in controls. Simultaneous interaction with αVβ3 integrin and NRP1 induces NRP1 stabilization at the cell membrane surface which is not observed with the co-injection of the controls.
Collapse
Affiliation(s)
- Fabien Thoreau
- Univ. Grenoble Alpes, CNRS, Department of Molecular Chemistry, UMR 5250, F-38000 Grenoble, France.
| | | | | | | | | |
Collapse
|
23
|
Counterbalance: modulation of VEGF/VEGFR activities by TNFSF15. Signal Transduct Target Ther 2018; 3:21. [PMID: 30101034 PMCID: PMC6085396 DOI: 10.1038/s41392-018-0023-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/23/2018] [Accepted: 05/31/2018] [Indexed: 01/11/2023] Open
Abstract
Vascular hyperpermeability occurs in angiogenesis and several pathobiological conditions, producing elevated interstitial fluid pressure and lymphangiogenesis. How these closely related events are modulated is a fundamentally important question regarding the maintenance of vascular homeostasis and treatment of disease conditions such as cancer, stroke, and myocardial infarction. Signals mediated by vascular endothelial growth factor receptors, noticeably VEGFR-1, −2, and −3, are centrally involved in the promotion of both blood vessel and lymphatic vessel growth. These signaling pathways are counterbalanced or, in the case of VEGFR3, augmented by signals induced by tumor necrosis factor superfamily-15 (TNFSF15). TNFSF15 can simultaneously downregulate membrane-bound VEGFR1 and upregulate soluble VEGFR1, thus changing VEGF/VEGFR1 signals from pro-angiogenic to anti-angiogenic. In addition, TNFSF15 inhibits VEGF-induced VEGFR2 phosphorylation, thereby curbing VEGFR2-mediated enhancement of vascular permeability. Third, and perhaps more interestingly, TNFSF15 is capable of stimulating VEGFR3 gene expression in lymphatic endothelial cells, thus augmenting VEGF-C/D-VEGFR3-facilitated lymphangiogenesis. We discuss the intertwining relationship between the actions of TNFSF15 and VEGF in this review. The ability of tumor necrosis factor superfamily-15 (TNFSF15) protein to balance the actions of vascular endothelial growth factors (VEGFs) highlights new therapeutic strategies for the treatment of diseases that disrupt the circulatory system. Gui-Li Yang at the Tianjin Neurological Institute and Lu-Yuan Li at Nankai University describe the mechanisms through which TNFSF15 inhibits blood vessel growth mediated by VEGF receptor-1 (VEGFR1) and counterbalances the increase in vascular permeability mediated by VEGFR2. Interestingly, TNFSF15 enhances the effects of VEGFR3 on the formation of lymphatic vessels by promoting VEGFR3 gene expression in lymphatic endothelial cells. Further research will determine whether TNFSF15′s unique capacity to regulate the properties of both blood and lymph vessels can be harnessed to improve the treatment of conditions such as cancer, stroke, myocardial infarction and lymphoedema.
Collapse
|
24
|
Atkinson SJ, Gontarczyk AM, Alghamdi AA, Ellison TS, Johnson RT, Fowler WJ, Kirkup BM, Silva BC, Harry BE, Schneider JG, Weilbaecher KN, Mogensen MM, Bass MD, Parsons M, Edwards DR, Robinson SD. The β3-integrin endothelial adhesome regulates microtubule-dependent cell migration. EMBO Rep 2018; 19:embr.201744578. [PMID: 29794156 PMCID: PMC6030693 DOI: 10.15252/embr.201744578] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 04/25/2018] [Accepted: 04/27/2018] [Indexed: 01/11/2023] Open
Abstract
Integrin β3 is seen as a key anti-angiogenic target for cancer treatment due to its expression on neovasculature, but the role it plays in the process is complex; whether it is pro- or anti-angiogenic depends on the context in which it is expressed. To understand precisely β3's role in regulating integrin adhesion complexes in endothelial cells, we characterised, by mass spectrometry, the β3-dependent adhesome. We show that depletion of β3-integrin in this cell type leads to changes in microtubule behaviour that control cell migration. β3-integrin regulates microtubule stability in endothelial cells through Rcc2/Anxa2-driven control of active Rac1 localisation. Our findings reveal that angiogenic processes, both in vitro and in vivo, are more sensitive to microtubule targeting agents when β3-integrin levels are reduced.
Collapse
Affiliation(s)
- Samuel J Atkinson
- School of Biological Sciences, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Aleksander M Gontarczyk
- School of Biological Sciences, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Abdullah Aa Alghamdi
- School of Biological Sciences, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Tim S Ellison
- School of Biological Sciences, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Robert T Johnson
- School of Biological Sciences, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Wesley J Fowler
- School of Biological Sciences, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Benjamin M Kirkup
- School of Biological Sciences, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Bernardo C Silva
- School of Biological Sciences, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Bronwen E Harry
- School of Biological Sciences, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Jochen G Schneider
- Luxembourg Center for Systems Biomedicine (LCSB), Luxembourg & Saarland University Medical Center, Internal Medicine II, University of Luxembourg, Homburg, Germany.,Centre Hospitalier Emily Mayrisch, Esch, Luxembourg
| | - Katherine N Weilbaecher
- Division of Molecular Oncology, Department of Internal Medicine, Washington University in St Louis, St. Louis, MO, USA
| | - Mette M Mogensen
- School of Biological Sciences, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Mark D Bass
- Department of Biomedical Science, Centre for Membrane Interactions and Dynamics, University of Sheffield, Sheffield, UK
| | - Maddy Parsons
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guys Campus, London, UK
| | - Dylan R Edwards
- Faculty of Medicine and Health Sciences, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Stephen D Robinson
- School of Biological Sciences, Norwich Research Park, University of East Anglia, Norwich, UK
| |
Collapse
|
25
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
26
|
Jia T, Choi J, Ciccione J, Henry M, Mehdi A, Martinez J, Eymin B, Subra G, Coll JL. Heteromultivalent targeting of integrin α vβ 3 and neuropilin 1 promotes cell survival via the activation of the IGF-1/insulin receptors. Biomaterials 2017; 155:64-79. [PMID: 29169039 DOI: 10.1016/j.biomaterials.2017.10.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/19/2017] [Accepted: 10/25/2017] [Indexed: 12/29/2022]
Abstract
Angiogenesis strongly depends on the activation of integrins, especially integrin αvβ3, and of neuropilin-1 (NRP-1), a co-receptor of VEGFR2. Dual-targeted molecules that simultaneously block both of them are expected have increased anti-angiogenic and antitumor activity. Toward this goal, we generated bifunctional 40 nm-sized silica nanoparticles (NPs) coated with controlled amounts of cRGD and ATWLPPR peptides and studied their affinity, selectivity and biological activity in HUVECs. Sub-nanomolar concentrations of NPs grafted either with ATWLPPR alone or in combination with cRGD exhibit potent and specific antagonist activity against VEGFR2/AKT signaling. However, a 1 nM concentration of the cRGD/ATWLPPR-heteromultivalent particles (RGD/ATW-NPs) also blocks the phosphorylation of VEGFR2 while co-inducing an unexpected long-lasting activation of AKT via IGF-1R/IR-AKT/GSK3β/eNOS signaling that stimulates cell survival and abrogates the intrinsic toxicity of silica-NPs to serum-starved HUVECs. We also showed that their repeated intravenous administration was associated with the proliferation of human U87MG tumor cells engrafted in nude mice and a dilatation of the tumor blood vessels. We present biochemical evidence for the complex cross-talk generated by the binding of the heteromultivalent NPs with αvβ3-integrin and with NRP1. In particular, we show for the first time that such heteromultivalent NPs can trans-activate IGF-1/insulin receptors and exert dose-dependent pro-survival activity. This study demonstrates the difficulties in designing targeted silica-based NPs for antiangiogenic therapies and the possible risks posed by undesirable side effects.
Collapse
Affiliation(s)
- Tao Jia
- INSERM-UGA U1209, CNRS UMR5309, Institute for Advanced Biosciences, La Tronche, France
| | - Jungyoon Choi
- INSERM-UGA U1209, CNRS UMR5309, Institute for Advanced Biosciences, La Tronche, France
| | - Jéremy Ciccione
- Institut des Biomolécules Max Mousseron (IBMM), UMR5247 CNRS, Université de Montpellier, ENSCM, Montpellier, France
| | - Maxime Henry
- INSERM-UGA U1209, CNRS UMR5309, Institute for Advanced Biosciences, La Tronche, France
| | - Ahmad Mehdi
- Institut Charles Gerhardt, UMR5253, CNRS, Université de Montpellier, ENSCM, Montpellier Cedex 05, France
| | - Jean Martinez
- Institut des Biomolécules Max Mousseron (IBMM), UMR5247 CNRS, Université de Montpellier, ENSCM, Montpellier, France
| | - Béatrice Eymin
- INSERM-UGA U1209, CNRS UMR5309, Institute for Advanced Biosciences, La Tronche, France
| | - Gilles Subra
- Institut des Biomolécules Max Mousseron (IBMM), UMR5247 CNRS, Université de Montpellier, ENSCM, Montpellier, France
| | - Jean-Luc Coll
- INSERM-UGA U1209, CNRS UMR5309, Institute for Advanced Biosciences, La Tronche, France.
| |
Collapse
|
27
|
Raab-Westphal S, Marshall JF, Goodman SL. Integrins as Therapeutic Targets: Successes and Cancers. Cancers (Basel) 2017; 9:E110. [PMID: 28832494 PMCID: PMC5615325 DOI: 10.3390/cancers9090110] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 08/11/2017] [Accepted: 08/14/2017] [Indexed: 12/12/2022] Open
Abstract
Integrins are transmembrane receptors that are central to the biology of many human pathologies. Classically mediating cell-extracellular matrix and cell-cell interaction, and with an emerging role as local activators of TGFβ, they influence cancer, fibrosis, thrombosis and inflammation. Their ligand binding and some regulatory sites are extracellular and sensitive to pharmacological intervention, as proven by the clinical success of seven drugs targeting them. The six drugs on the market in 2016 generated revenues of some US$3.5 billion, mainly from inhibitors of α4-series integrins. In this review we examine the current developments in integrin therapeutics, especially in cancer, and comment on the health economic implications of these developments.
Collapse
Affiliation(s)
- Sabine Raab-Westphal
- Translational In Vivo Pharmacology, Translational Innovation Platform Oncology, Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany.
| | - John F Marshall
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| | - Simon L Goodman
- Translational and Biomarkers Research, Translational Innovation Platform Oncology, Merck KGaA, 64293 Darmstadt, Germany.
| |
Collapse
|
28
|
Nurden AT. Should studies on Glanzmann thrombasthenia not be telling us more about cardiovascular disease and other major illnesses? Blood Rev 2017; 31:287-299. [PMID: 28395882 DOI: 10.1016/j.blre.2017.03.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/23/2017] [Indexed: 12/17/2022]
Abstract
Glanzmann thrombasthenia (GT) is a rare inherited bleeding disorder caused by loss of αIIbβ3 integrin function in platelets. Most genetic variants of β3 also affect the widely expressed αvβ3 integrin. With brief mention of mouse models, I now look at the consequences of disease-causing ITGA2B and ITGB3 mutations on the non-hemostatic functions of platelets and other cells. Reports of arterial thrombosis in GT patients are rare, but other aspects of cardiovascular disease do occur including deep vein thrombosis and congenital heart defects. Thrombophilic and other risk factors for thrombosis and lessons from heterozygotes and variant forms of GT are discussed. Assessed for GT patients are reports of leukemia and cancer, loss of fertility, bone pathology, inflammation and wound repair, infections, kidney disease, autism and respiratory disease. This survey shows an urgent need for a concerted international effort to better determine how loss of αIIbβ3 and αvβ3 influences health and disease.
Collapse
Affiliation(s)
- Alan T Nurden
- Institut de Rhythmologie et de Modélisation Cardiaque, Plateforme Technologique d'Innovation Biomédicale, Hôpital Xavier Arnozan, Pessac, France.
| |
Collapse
|
29
|
Randi AM, Laffan MA. Von Willebrand factor and angiogenesis: basic and applied issues. J Thromb Haemost 2017; 15:13-20. [PMID: 27778439 DOI: 10.1111/jth.13551] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Indexed: 12/11/2022]
Abstract
The recent discovery that von Willebrand factor (VWF) regulates blood vessel formation has opened a novel perspective on the function of this complex protein. VWF was discovered as a key component of hemostasis, capturing platelets at sites of endothelial damage and synthesized in megakaryocytes and endothelial cells (EC). In recent years, novel functions and binding partners have been identified for VWF. The finding that loss of VWF in EC results in enhanced, possibly dysfunctional, angiogenesis is consistent with the clinical observations that in some patients with von Willebrand disease (VWD), vascular malformations can cause severe gastrointestinal (GI) bleeding. In vitro and in vivo studies indicate that VWF can regulate angiogenesis through multiple pathways, both intracellular and extracellular, although their relative importance is still unclear. Investigation of these pathways has been greatly facilitated by the ability to isolate EC from progenitors circulating in the peripheral blood of normal controls and patients with VWD. In the next few years, these will yield further evidence on the molecular pathways controlled by VWF and shed light on this novel and fascinating area of vascular biology. In this article, we will review the evidence supporting a role for VWF in blood vessel formation, the link between VWF dysfunction and vascular malformations causing GI bleeding and how they may be causally related. Finally, we will discuss how these findings point to novel therapeutic approaches to bleeding refractory to VWF replacement therapy in VWD.
Collapse
Affiliation(s)
- A M Randi
- National Heart and Lung Institute, Imperial College, London, UK
| | - M A Laffan
- Department of Haematology, Imperial College, London, UK
| |
Collapse
|
30
|
Sartori A, Portioli E, Battistini L, Calorini L, Pupi A, Vacondio F, Arosio D, Bianchini F, Zanardi F. Synthesis of Novel c(AmpRGD)-Sunitinib Dual Conjugates as Molecular Tools Targeting the α vβ 3 Integrin/VEGFR2 Couple and Impairing Tumor-Associated Angiogenesis. J Med Chem 2016; 60:248-262. [PMID: 27997164 DOI: 10.1021/acs.jmedchem.6b01266] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
On the basis of a previously discovered anti-αVβ3 integrin peptidomimetic (c(AmpRGD)) and the clinically approved antiangiogenic kinase inhibitor sunitinib, three novel dual conjugates were synthesized (compounds 1-3), featuring the covalent and robust linkage between these two active modules. In all conjugates, the ligand binding competence toward αVβ3 (using both isolated receptors and αVβ3-overexpressing endothelial progenitor EP cells) and the kinase inhibitory activity (toward both isolated kinases and EPCs) remained almost untouched and comparable to the activity of the single active units. Compounds 1-3 showed interesting antiangiogenesis properties in an in vitro tubulogenic assay; furthermore, dimeric-RGD conjugate 3 strongly inhibited in vivo angiogenesis in Matrigel plug assays in FVB mice. These results offer proof-of-concept of how the covalent conjugation of two angiogenesis-related small modules may result in novel and stable molecules, which impair tumor-related angiogenesis with equal or even superior ability as compared to the single modules or their simple combinations.
Collapse
Affiliation(s)
- Andrea Sartori
- Dipartimento di Farmacia, Università degli Studi di Parma , Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Elisabetta Portioli
- Dipartimento di Farmacia, Università degli Studi di Parma , Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Lucia Battistini
- Dipartimento di Farmacia, Università degli Studi di Parma , Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Lido Calorini
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze , Viale G. B. Morgagni 50, 50134 Firenze, Italy
| | - Alberto Pupi
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze , Viale G. B. Morgagni 50, 50134 Firenze, Italy.,Centro Interdipartimentale per lo Sviluppo Preclinico dell'Imaging Molecolare (CISPIM), Università degli Studi di Firenze , Viale G. B. Morgagni 50, 50134 Firenze, Italy
| | - Federica Vacondio
- Dipartimento di Farmacia, Università degli Studi di Parma , Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Daniela Arosio
- Istituto di Scienze e Tecnologie Molecolari, Consiglio Nazionale delle Ricerche , Via Golgi 19, 20133 Milano, Italy
| | - Francesca Bianchini
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze , Viale G. B. Morgagni 50, 50134 Firenze, Italy.,Centro Interdipartimentale per lo Sviluppo Preclinico dell'Imaging Molecolare (CISPIM), Università degli Studi di Firenze , Viale G. B. Morgagni 50, 50134 Firenze, Italy
| | - Franca Zanardi
- Dipartimento di Farmacia, Università degli Studi di Parma , Parco Area delle Scienze 27A, 43124 Parma, Italy
| |
Collapse
|
31
|
Lu Q, Wang J, Jiang J, Wang S, Jia Q, Wang Y, Li W, Zhou Q, Lv L, Li Q. rLj-RGD3, a Novel Recombinant Toxin Protein from Lampetra japonica, Protects against Cerebral Reperfusion Injury Following Middle Cerebral Artery Occlusion Involving the Integrin-PI3K/Akt Pathway in Rats. PLoS One 2016; 11:e0165093. [PMID: 27768719 PMCID: PMC5074578 DOI: 10.1371/journal.pone.0165093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 08/31/2016] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The RGD-toxin protein Lj-RGD3 is a naturally occurring 118 amino acid peptide that can be obtained from the salivary gland of the Lampetra japonica fish. This unique peptide contains 3 RGD (Arg-Gly-Asp) motifs in its primary structure. Lj-RGD3 is available in recombinant form (rLj-RGD3) and can be produced in large quantities using DNA recombination techniques. The pharmacology of the three RGD motif-containing peptides has not been studied. This study investigated the protective effects of rLj-RGD3, a novel polypeptide, against ischemia/reperfusion-induced damage to the brain caused by middle cerebral artery occlusion (MCAO) in a rat stroke model. We also explored the mechanism by which rLj-RGD3 acts by measuring protein and mRNA expression levels, with an emphasis on the FAK and integrin-PI3K/Akt anti-apoptosis pathways. METHODS rLj-RGD3 was obtained from the buccal secretions of Lampetra japonica using gene recombination technology. Sprague Dawley (SD) rats were randomly divided into the following seven groups: a sham group; a vehicle-treated (VT) group; 100.0 μg·kg-1, 50.0 μg·kg-1 and 25.0 μg·kg-1 dose rLj-RGD3 groups; and two positive controls, including 1.5 mg·kg-1 Edaravone (ED) and 100.0 μg·kg-1 Eptifibatide (EP). MCAO was induced using a model consisting of 2 h of ischemia and 24 h of reperfusion. Behavioral changes were observed in the normal and operation groups after focal cerebral ischemia/reperfusion was applied. In addition, behavioral scores were evaluated at 4 and 24 h after reperfusion. Brain infarct volumes were determined based on 2,3,5-triphenyltetrazolium chloride (TTC) staining. Pathological changes in brain tissues were observed using hematoxylin and eosin (H&E) staining. Moreover, neuronal apoptosis was detected using terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling (TUNEL) assays. We determined the expression levels of focal adhesion kinase (FAK), phosphatidyl inositol 3-kinase (PI3K), protein kinase B (Akt, PKB), caspase-3 and Bcl-2 in the brain using western blot analysis and RT-PCR assays. The research protocol was approved by the Institutional Ethics Committee of Dalian Medical University. RESULTS The behavioral scores and cerebral infarct volumes of the rLj-RGD3 groups were markedly lower at 4 and 24 h/RF. The rLj-RGD3 protein significantly ameliorated pathological changes in the brain and reduced the number of apoptotic neurons. Furthermore, the FAK and PI3K/Akt pathways were activated. rLj-RGD3 significantly increased the expression of FAK, p-FAK and Bcl-2 proteins. In contrast, caspase-3 expression was inhibited. CONCLUSION/SIGNIFICANCE We conclude that recombinant Lampetra japonica RGD-peptide (rLj-RGD3) exerts a protective effect against cerebral ischemia/reperfusion injury in the brain. In addition, the mechanism of this protection is associated with the activation of the integrin-PI3K/Akt pathway. These results provide a theoretical foundation and an experimental basis for using RGD peptides as novel drugs for treating ischemic cerebral vascular diseases in addition to promoting the research and development of marine biotechnology drugs.
Collapse
Affiliation(s)
- Qian Lu
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning Province, China, 116044
- College of Basic Medicine, Jilin Medical College, Jilin, Jilin Province, China, 132013
| | - Jihong Wang
- School of Life Sciences, Liaoning Normal University, Dalian, Liaoning Province, China, 116029
- Key Laboratory of Biotechnology and Drug Discovery of Liaoning Province, Dalian, Liaoning Province, China, 116029
| | - Junshu Jiang
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning Province, China, 116044
| | - Shengnan Wang
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning Province, China, 116044
| | - Qilan Jia
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning Province, China, 116044
| | - Yue Wang
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning Province, China, 116044
| | - Weiping Li
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning Province, China, 116044
| | - Qin Zhou
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning Province, China, 116044
| | - Li Lv
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning Province, China, 116044
- School of Life Sciences, Liaoning Normal University, Dalian, Liaoning Province, China, 116029
| | - Qingwei Li
- School of Life Sciences, Liaoning Normal University, Dalian, Liaoning Province, China, 116029
- Key Laboratory of Biotechnology and Drug Discovery of Liaoning Province, Dalian, Liaoning Province, China, 116029
| |
Collapse
|
32
|
Expression Profiling of Genes Related to Endothelial Cells Biology in Patients with Type 2 Diabetes and Patients with Prediabetes. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1845638. [PMID: 27781209 PMCID: PMC5066000 DOI: 10.1155/2016/1845638] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/18/2016] [Accepted: 08/30/2016] [Indexed: 12/31/2022]
Abstract
Endothelial dysfunction appears to be an early sign indicating vascular damage and predicts the progression of atherosclerosis and cardiovascular disorders. Extensive clinical and experimental evidence suggests that endothelial dysfunction occurs in Type 2 Diabetes Mellitus (T2DM) and prediabetes patients. This study was carried out with an aim to appraise the expression levels in the peripheral blood of 84 genes related to endothelial cells biology in patients with diagnosed T2DM or prediabetes, trying to identify new genes whose expression might be changed under these pathological conditions. The study covered a total of 45 participants. The participants were divided into three groups: group 1, patients with T2DM; group 2, patients with prediabetes; group 3, control group. The gene expression analysis was performed using the Endothelial Cell Biology RT2 Profiler PCR Array. In the case of T2DM, 59 genes were found to be upregulated, and four genes were observed to be downregulated. In prediabetes patients, increased expression was observed for 49 genes, with two downregulated genes observed. Our results indicate that diabetic and prediabetic conditions change the expression levels of genes related to endothelial cells biology and, consequently, may increase the risk for occurrence of endothelial dysfunction.
Collapse
|
33
|
Mandic L, Traxler D, Gugerell A, Zlabinger K, Lukovic D, Pavo N, Goliasch G, Spannbauer A, Winkler J, Gyöngyösi M. Molecular Imaging of Angiogenesis in Cardiac Regeneration. CURRENT CARDIOVASCULAR IMAGING REPORTS 2016; 9:27. [PMID: 27683600 PMCID: PMC5018257 DOI: 10.1007/s12410-016-9389-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Myocardial infarction (MI) leading to heart failure displays an important cause of death worldwide. Adequate restoration of blood flow to prevent this transition is a crucial factor to improve long-term morbidity and mortality. Novel regenerative therapies have been thoroughly investigated within the past decades. RECENT FINDINGS Increased angiogenesis in infarcted myocardium has shown beneficial effects on the prognosis of MI; therefore, the proangiogenic capacity of currently tested treatments is of specific interest. Molecular imaging to visualize formation of new blood vessels in vivo displays a promising option to monitor proangiogenic effects of regenerative substances. SUMMARY Based on encouraging results in preclinical models, molecular angiogenesis imaging has recently been applied in a small set of patients. This article reviews recent literature on noninvasive in vivo molecular imaging of angiogenesis after MI as an integral part of cardiac regeneration.
Collapse
Affiliation(s)
- Ljubica Mandic
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Denise Traxler
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Alfred Gugerell
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Katrin Zlabinger
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Dominika Lukovic
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Noemi Pavo
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Georg Goliasch
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Andreas Spannbauer
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Johannes Winkler
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
34
|
The role of integrins in glaucoma. Exp Eye Res 2016; 158:124-136. [PMID: 27185161 DOI: 10.1016/j.exer.2016.05.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/06/2016] [Accepted: 05/10/2016] [Indexed: 01/04/2023]
Abstract
Integrins are a family of heterodimeric transmembrane receptors that mediate adhesion to the extracellular matrix (ECM). In addition to their role as adhesion receptors, integrins can act as ''bidirectional signal transducers'' that coordinate a large number of cellular activities in response to the extracellular environment and intracellular signaling events. This bidirectional signaling helps maintain tissue homeostasis. Dysregulated bidirectional signaling, however, could trigger the propagation of feedback loops that can lead to the establishment of a disease state such as glaucoma. Here we discuss the role of integrins and bidirectional signaling as they relate to the glaucomatous phenotype with special emphasis on the αvβ3 integrin. We present evidence that this particular integrin may have a significant impact on the pathogenesis of glaucoma.
Collapse
|
35
|
Zhao J, Niu H, Li A, Nie L. Acetylbritannilactone Modulates Vascular Endothelial Growth Factor Signaling and Regulates Angiogenesis in Endothelial Cells. PLoS One 2016; 11:e0148968. [PMID: 26863518 PMCID: PMC4749253 DOI: 10.1371/journal.pone.0148968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 01/26/2016] [Indexed: 11/19/2022] Open
Abstract
The present study was conducted to determine the effects of 1-O-acetylbritannilactone (ABL), a compound extracted from Inula britannica L., on vascular endothelial growth factor (VEGF) signaling and angiogenesis in endothelial cells (ECs). We showed that ABL promotes VEGF-induced cell proliferation, growth, migration, and tube formation in cultured human ECs. Furthermore, the modulatory effect of ABL on VEGF-induced Akt, MAPK p42/44, and p38 phosphorylation, as well as on upstream VEGFR-2 phosphorylation, were associated with VEGF-dependent Matrigel angiogenesis in vivo. In addition, animals treated with ABL (26 mg/kg/day) recovered blood flow significantly earlier than control animals, suggesting that ABL affects ischemia-mediated angiogenesis and arteriogenesis in vivo. Finally, we demonstrated that ABL strongly reduced the levels of VEGFR-2 on the cell surface, enhanced VEGFR-2 endocytosis, which consistent with inhibited VE-cadherin, a negative regulator of VEGF signaling associated with VEGFR-2 complex formation, but did not alter VE-cadherin or VEGFR-2 expression in ECs. Our results suggest that ABL may serve as a novel therapeutic intervention for various cardiovascular diseases, including chronic ischemia, by regulating VEGF signaling and modulating angiogenesis.
Collapse
Affiliation(s)
- Jingshan Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Honglin Niu
- Department of Nephrology, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- Key Laboratory of Kidney Diseases of Hebei Province, Shijiazhuang, 050071, China
| | - Aiying Li
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Lei Nie
- Key Laboratory of Medical Biotechnology of Hebei Province and Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
- * E-mail:
| |
Collapse
|
36
|
Notni J, Steiger K, Hoffmann F, Reich D, Kapp TG, Rechenmacher F, Neubauer S, Kessler H, Wester HJ. Complementary, Selective PET Imaging of Integrin Subtypes α5β1 and αvβ3 Using 68Ga-Aquibeprin and 68Ga-Avebetrin. J Nucl Med 2015; 57:460-6. [DOI: 10.2967/jnumed.115.165720] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
37
|
Ellison TS, Atkinson SJ, Steri V, Kirkup BM, Preedy MEJ, Johnson RT, Ruhrberg C, Edwards DR, Schneider JG, Weilbaecher K, Robinson SD. Suppression of β3-integrin in mice triggers a neuropilin-1-dependent change in focal adhesion remodelling that can be targeted to block pathological angiogenesis. Dis Model Mech 2015; 8:1105-19. [PMID: 26159543 PMCID: PMC4582102 DOI: 10.1242/dmm.019927] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 07/07/2015] [Indexed: 12/12/2022] Open
Abstract
Anti-angiogenic treatments against αvβ3-integrin fail to block tumour growth in the long term, which suggests that the tumour vasculature escapes from angiogenesis inhibition through αvβ3-integrin-independent mechanisms. Here, we show that suppression of β3-integrin in mice leads to the activation of a neuropilin-1 (NRP1)-dependent cell migration pathway in endothelial cells via a mechanism that depends on NRP1's mobilisation away from mature focal adhesions following VEGF-stimulation. The simultaneous genetic targeting of both molecules significantly impairs paxillin-1 activation and focal adhesion remodelling in endothelial cells, and therefore inhibits tumour angiogenesis and the growth of already established tumours. These findings provide a firm foundation for testing drugs against these molecules in combination to treat patients with advanced cancers. Summary: Targeting both β3-integrin and neuropilin-1 prevents anti-angiogenic treatment escape.
Collapse
Affiliation(s)
- Tim S Ellison
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Samuel J Atkinson
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Veronica Steri
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Benjamin M Kirkup
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Michael E J Preedy
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Robert T Johnson
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | | | - Dylan R Edwards
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Jochen G Schneider
- Luxembourg Center for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg & Saarland University Medical Center, Internal Medicine II, L-4362 Homburg, Germany
| | - Katherine Weilbaecher
- Department of Internal Medicine, Division of Molecular Oncology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Stephen D Robinson
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| |
Collapse
|
38
|
Abstract
For nearly two decades now, the RGD (Arg-Gly-Asp)-binding αvβ3-integrin has been a focus of anti-angiogenic drug design. These inhibitors are well-tolerated, but have shown only limited success in patients. Over the years, studies in β3-integrin-knockout mice have shed some light on possible explanations for disappointing clinical outcomes. However, studying angiogenesis in β3-integrin-knockout mice is a blunt tool to investigate β3-integrin's role in pathological angiogenesis. Since establishing our laboratory at University of East Anglia (UEA), we have adopted more refined models of genetically manipulating the expression of the β3-integrin subunit. The present review will highlight some of our findings from these models and describe how data from them have forced us to rethink how targeting αvβ3-integrin expression affects tumour angiogenesis and cancer progression. Revisiting the fundamental biology behind how this integrin regulates tumour growth and angiogenesis, we believe, is the key not only to understanding how angiogenesis is normally co-ordinated, but also in success with drugs directed against it.
Collapse
|
39
|
Inhibition of tumor-associated αvβ3 integrin regulates the angiogenic switch by enhancing expression of IGFBP-4 leading to reduced melanoma growth and angiogenesis in vivo. Angiogenesis 2014; 18:31-46. [PMID: 25249331 DOI: 10.1007/s10456-014-9445-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 09/16/2014] [Indexed: 12/20/2022]
Abstract
A more complete understanding of the mechanisms that regulate the angiogenic switch, which contributes to the conversion of small dormant tumors to actively growing malignancies, is important for the development of more effective anti-angiogenic strategies for cancer therapy. While significant progress has been made in understanding the complex mechanisms by which integrin αvβ3 expressed in endothelial cells governs angiogenesis, less is known concerning the ability of αvβ3 expressed within the tumor cell compartment to modulate the angiogenic output of a tumor. Here we provide evidence that αvβ3 expressed in melanoma cells may contribute to the suppression of IGFBP-4, an important negative regulator of IGF-1 signaling. Given the multiple context-dependent roles for αvβ3 in angiogenesis and tumor progression, our novel findings provide additional molecular insight into how αvβ3 may govern the angiogenic switch by a mechanism associated with a p38 MAPK and matrix metalloproteinases-dependent regulation of the endogenous angiogenesis inhibitor IGFBP-4.
Collapse
|
40
|
Hutchins ED, Markov GJ, Eckalbar WL, George RM, King JM, Tokuyama MA, Geiger LA, Emmert N, Ammar MJ, Allen AN, Siniard AL, Corneveaux JJ, Fisher RE, Wade J, DeNardo DF, Rawls JA, Huentelman MJ, Wilson-Rawls J, Kusumi K. Transcriptomic analysis of tail regeneration in the lizard Anolis carolinensis reveals activation of conserved vertebrate developmental and repair mechanisms. PLoS One 2014; 9:e105004. [PMID: 25140675 PMCID: PMC4139331 DOI: 10.1371/journal.pone.0105004] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/17/2014] [Indexed: 01/09/2023] Open
Abstract
Lizards, which are amniote vertebrates like humans, are able to lose and regenerate a functional tail. Understanding the molecular basis of this process would advance regenerative approaches in amniotes, including humans. We have carried out the first transcriptomic analysis of tail regeneration in a lizard, the green anole Anolis carolinensis, which revealed 326 differentially expressed genes activating multiple developmental and repair mechanisms. Specifically, genes involved in wound response, hormonal regulation, musculoskeletal development, and the Wnt and MAPK/FGF pathways were differentially expressed along the regenerating tail axis. Furthermore, we identified 2 microRNA precursor families, 22 unclassified non-coding RNAs, and 3 novel protein-coding genes significantly enriched in the regenerating tail. However, high levels of progenitor/stem cell markers were not observed in any region of the regenerating tail. Furthermore, we observed multiple tissue-type specific clusters of proliferating cells along the regenerating tail, not localized to the tail tip. These findings predict a different mechanism of regeneration in the lizard than the blastema model described in the salamander and the zebrafish, which are anamniote vertebrates. Thus, lizard tail regrowth involves the activation of conserved developmental and wound response pathways, which are potential targets for regenerative medical therapies.
Collapse
Affiliation(s)
- Elizabeth D. Hutchins
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Glenn J. Markov
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Walter L. Eckalbar
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Rajani M. George
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Jesse M. King
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Minami A. Tokuyama
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Lauren A. Geiger
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Nataliya Emmert
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Michael J. Ammar
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - April N. Allen
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Ashley L. Siniard
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Jason J. Corneveaux
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Rebecca E. Fisher
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States of America
| | - Juli Wade
- Departments of Psychology and Zoology, Program in Neuroscience, Michigan State University, East Lansing, Michigan, United States of America
| | - Dale F. DeNardo
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - J. Alan Rawls
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Matthew J. Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Jeanne Wilson-Rawls
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Kenro Kusumi
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States of America
| |
Collapse
|
41
|
Winograd-Katz SE, Fässler R, Geiger B, Legate KR. The integrin adhesome: from genes and proteins to human disease. Nat Rev Mol Cell Biol 2014; 15:273-88. [PMID: 24651544 DOI: 10.1038/nrm3769] [Citation(s) in RCA: 445] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The adhesive interactions of cells with their environment through the integrin family of transmembrane receptors have key roles in regulating multiple aspects of cellular physiology, including cell proliferation, viability, differentiation and migration. Consequently, failure to establish functional cell adhesions, and thus the assembly of associated cytoplasmic scaffolding and signalling networks, can have severe pathological effects. The roles of specific constituents of integrin-mediated adhesions, which are collectively known as the 'integrin adhesome', in diverse pathological states are becoming clear. Indeed, the prominence of mutations in specific adhesome molecules in various human diseases is now appreciated, and experimental as well as in silico approaches provide insights into the molecular mechanisms underlying these pathological conditions.
Collapse
Affiliation(s)
- Sabina E Winograd-Katz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Benjamin Geiger
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Kyle R Legate
- 1] Department of Molecular Medicine, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany. [2] Center for Nanosciences, Department of Applied Physics, Ludwig-Maximilians University, 80799 Munich, Germany
| |
Collapse
|
42
|
Steri V, Ellison TS, Gontarczyk AM, Weilbaecher K, Schneider JG, Edwards D, Fruttiger M, Hodivala-Dilke KM, Robinson SD. Acute depletion of endothelial β3-integrin transiently inhibits tumor growth and angiogenesis in mice. Circ Res 2014; 114:79-91. [PMID: 24103390 DOI: 10.1161/circresaha.114.301591] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Accepted: 10/08/2013] [Indexed: 11/16/2022]
Abstract
RATIONALE The dramatic upregulation of αvβ3-integrin that occurs in the vasculature during tumor growth has long suggested that the endothelial expression of this molecule is an ideal target for antiangiogenic therapy to treat cancer. This discovery led to the development of small-molecule inhibitors directed against αvβ3-integrin that are currently in clinical trials. In 2002, we reported that β3-integrin-knockout mice exhibit enhanced tumor growth and angiogenesis. However, as β3-integrin is expressed by a wide variety of cells, endothelial cell-specific contributions to tumor angiogenesis are muddied by the use of a global knockout of β3-integrin function. OBJECTIVE Our aim was to examine the endothelial-specific contribution β3-integrin makes to tumor growth and angiogenesis. METHODS AND RESULTS We have crossed β3-integrin-floxed (β3-floxed) mice to 2 endothelial-specific Cre models and examined angiogenic responses in vivo, ex vivo, and in vitro. We show that acute depletion of endothelial β3-integrin inhibits tumor growth and angiogenesis preventatively, but not in already established tumors. However, the effects are transient, and long-term depletion of the molecule is ineffective. Furthermore, long-term depletion of the molecule correlates with many molecular changes, such as reduced levels of focal adhesion kinase expression and a misbalance in focal adhesion kinase phosphorylation, which may lead to a release from the inhibitory effects of decreased endothelial β3-integrin expression. CONCLUSIONS Our findings imply that timing and length of inhibition are critical factors that need to be considered when targeting the endothelial expression of β3-integrin to inhibit tumor growth and angiogenesis.
Collapse
Affiliation(s)
- Veronica Steri
- From the School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, United Kingdom (V.S., T.S.E., A.M.G., D.E., S.D.R.); Department of Internal Medicine, Division of Molecular Oncology, Washington University in St Louis, MO (K.W.); Luxembourg Center for Systems Biomedicine, University of Luxembourg, Luxembourg and Saarland University Medical Center, Internal Medicine II, Homburg, Germany (J.G.S.); UCL Institute of Ophthalmology, University College London, London, United Kingdom (M.F.); Barts Cancer Institute - a Cancer Research UK Centre of Excellence, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London, United Kingdom (K.M.H.-D.)
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Abstract
During angiogenesis, αv integrins are overexpressed on the endothelial cell surface to facilitate the growth and survival of newly forming vessels. Accordingly, blocking αv integrin function by disrupting ligand binding can produce an antiangiogenic effect. Although the integrin ectodomain regulates ligand binding specificity, the short cytoplasmic tail facilitates intracellular signaling pathways through the recruitment and activation of specific kinases and signaling intermediates. This in turn controls endothelial cell adhesion, morphology, migration, invasion, proliferation, and survival. These same integrin-mediated signaling pathways are exploited in cancer to promote the invasiveness and survival of tumor cells and to manipulate the host microenvironment to provide ample blood vessel and stromal resources to support tumor growth and metastatic spread. Because expression of αv integrins on distinct cell types contributes to cancer growth, αv integrin antagonists have the potential to disrupt multiple aspects of disease progression.
Collapse
Affiliation(s)
- Sara M Weis
- Moores UCSD Cancer Center, and University of California, San Diego, La Jolla, California 92093-0803, USA; Department of Pathology, University of California, San Diego, La Jolla, California 92093-0803, USA
| | | |
Collapse
|
45
|
Han SW, Jung YK, Lee EJ, Park HR, Kim GW, Jeong JH, Han MS, Choi JY. DICAM inhibits angiogenesis via suppression of AKT and p38 MAP kinase signalling. Cardiovasc Res 2013; 98:73-82. [DOI: 10.1093/cvr/cvt019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
46
|
Jones DT, Lechertier T, Reynolds LE, Mitter R, Robinson SD, Kirn-Safran CB, Hodivala-Dilke KM. Endogenous ribosomal protein L29 (RPL29): a newly identified regulator of angiogenesis in mice. Dis Model Mech 2013; 6:115-24. [PMID: 23118343 PMCID: PMC3529344 DOI: 10.1242/dmm.009183] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 06/29/2012] [Indexed: 01/03/2023] Open
Abstract
Cellular ribosomal protein L29 (RPL29) is known to be important in protein synthesis, but its function during angiogenesis has never been described before. We have shown previously that mice lacking β3-integrins support enhanced tumour angiogenesis and, therefore, deletion of endothelial αvβ3 can provide a method for discovery of novel regulators of tumour angiogenesis. Here, we describe significant upregulation of RPL29 in β3-null endothelial cells at both the mRNA and protein level. Ex vivo, we show that VEGF-stimulated microvessel sprouting was reduced significantly in Rpl29-heterozygous and Rpl29-null aortic ring assays compared with wild-type controls. Moreover, we provide in vivo evidence that RPL29 can regulate tumour angiogenesis. Tumour blood vessel density in subcutaneously grown Lewis lung carcinomas was reduced significantly in Rpl29-mutant mice. Additionally, depletion of Rpl29 using RNA interference inhibited VEGF-induced aortic ring sprouting, suggesting that anti-RPL29 strategies might have anti-angiogenic potential. Overall, our results identify that loss or depletion of RPL29 can reduce angiogenesis in vivo and ex vivo.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/metabolism
- Endothelial Cells/metabolism
- Gene Expression
- Integrin alphaVbeta3/deficiency
- Integrin alphaVbeta3/genetics
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/prevention & control
- Neovascularization, Physiologic/genetics
- RNA Interference
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA-Binding Proteins
- Ribosomal Proteins/antagonists & inhibitors
- Ribosomal Proteins/deficiency
- Ribosomal Proteins/genetics
- Ribosomal Proteins/physiology
- Vascular Endothelial Growth Factor A/pharmacology
Collapse
Affiliation(s)
- Dylan T Jones
- Centre for Tumour Biology, Barts Cancer Institute-a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK.
| | | | | | | | | | | | | |
Collapse
|
47
|
Xu S, Olenyuk BZ, Okamoto CT, Hamm-Alvarez SF. Targeting receptor-mediated endocytotic pathways with nanoparticles: rationale and advances. Adv Drug Deliv Rev 2013; 65:121-38. [PMID: 23026636 PMCID: PMC3565049 DOI: 10.1016/j.addr.2012.09.041] [Citation(s) in RCA: 298] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 09/13/2012] [Accepted: 09/20/2012] [Indexed: 12/22/2022]
Abstract
Targeting of drugs and their carrier systems by using receptor-mediated endocytotic pathways was in its nascent stages 25 years ago. In the intervening years, an explosion of knowledge focused on design and synthesis of nanoparticulate delivery systems as well as elucidation of the cellular complexity of what was previously-termed receptor-mediated endocytosis has now created a situation when it has become possible to design and test the feasibility of delivery of highly specific nanoparticle drug carriers to specific cells and tissue. This review outlines the mechanisms governing the major modes of receptor-mediated endocytosis used in drug delivery and highlights recent approaches using these as targets for in vivo drug delivery of nanoparticles. The review also discusses some of the inherent complexity associated with the simple shift from a ligand-drug conjugate versus a ligand-nanoparticle conjugate, in terms of ligand valency and its relationship to the mode of receptor-mediated internalization.
Collapse
Affiliation(s)
- Shi Xu
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA, USA 90033
| | - Bogdan Z. Olenyuk
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA, USA 90033
| | - Curtis T. Okamoto
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA, USA 90033
| | - Sarah F. Hamm-Alvarez
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA, USA 90033
| |
Collapse
|
48
|
Elevated VEGF Levels in Pulmonary Edema Fluid and PBMCs from Patients with Acute Hantavirus Pulmonary Syndrome. Adv Virol 2012; 2012:674360. [PMID: 22956954 PMCID: PMC3432326 DOI: 10.1155/2012/674360] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 07/11/2012] [Indexed: 12/19/2022] Open
Abstract
Hantavirus pulmonary syndrome is characterized by vascular permeability, hypoxia, and acute pulmonary edema. Vascular endothelial growth factor (VEGF) is induced by hypoxia, potently induces vascular permeability, and is associated with high-altitude-induced pulmonary edema. Hantaviruses alter the normal regulation of β3 integrins that restrict VEGF-directed permeability and hantavirus infected endothelial cells are hyperresponsive to the permeabilizing effects of VEGF. However, the role of VEGF in acute pulmonary edema observed in HPS patients remains unclear. Here we retrospectively evaluate VEGF levels in pulmonary edema fluid (PEF), plasma, sera, and PBMCs from 31 HPS patients. VEGF was elevated in HPS patients PEF compared to controls with the highest levels observed in PEF samples from a fatal HPS case. VEGF levels were highest in PBMC samples during the first five days of hospitalization and diminished during recovery. Significantly increased PEF and PBMC VEGF levels are consistent with acute pulmonary edema observed in HPS patients and HPS disease severity. We observed substantially lower VEGF levels in a severe HPS disease survivor after extracorporeal membrane oxygenation. These findings suggest the importance of patients' VEGF levels during HPS, support the involvement of VEGF responses in HPS pathogenesis, and suggest targeting VEGF responses as a potential therapeutic approach.
Collapse
|
49
|
Nurden AT. Is there a redundancy of β3 and other platelet receptors in the brain and central nervous system? Platelets 2012; 24:170-2. [DOI: 10.3109/09537104.2012.678427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
50
|
Nam JO, Son HN, Jun E, Cha K, Lee BH, Park RW, Kim IS. FAS1 domain protein inhibits VEGF165-induced angiogenesis by targeting the interaction between VEGFR-2 and αvβ3 integrin. Mol Cancer Res 2012; 10:1010-20. [PMID: 22710795 DOI: 10.1158/1541-7786.mcr-11-0600] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It is known that VEGF receptors (VEGFR) and integrins interact with each other to regulate angiogenesis. We reported previously that the fasciclin 1 (FAS1) domain-containing protein, TGFBIp/βig-h3 (TGF-β-induced protein) is an angiogenesis regulator that inhibits both endothelial cell migration and growth via αvβ3 integrin. In an attempt to target the interaction between VEGFR-2 and αvβ3 integrin, we determined whether the FAS1 domain region of TGFBIp/βig-h3 (FAS1 domain protein) can block the interaction between the two receptors, leading to the suppression of angiogenesis. In this study, we showed that FAS1 domain protein inhibits VEGF165-induced endothelial cell proliferation and migration via αvβ3 integrin, resulting in the inhibition of VEGF165-induced angiogenesis. We also defined a molecular mechanism by which FAS1 domain protein blocks the association between αvβ3 integrin and VEGFR-2, showing that it binds to αvβ3 integrin but not to VEGFR-2. Blocking the association of these major angiogenic receptors with FAS1 domain protein inhibits signaling pathways downstream of VEGFR-2. Collectively, our results indicate that FAS1 domain protein, in addition to its inhibitory effect on αvβ3 integrin-mediated angiogenesis, also inhibits VEGF165-induced angiogenesis. Thus, FAS1 domain protein can be further developed into a potent anticancer drug that targets two principal angiogenic pathways.
Collapse
Affiliation(s)
- Ju-Ock Nam
- Department of Ecological Environment Conservation, Kyungpook National University, Gyeongsangbuk-do, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|