1
|
Ntekoumes D, Song J, Liu H, Amelung C, Guan Y, Gerecht S. Acute Three-Dimensional Hypoxia Regulates Angiogenesis. Adv Healthc Mater 2025; 14:e2403860. [PMID: 39623803 PMCID: PMC11729260 DOI: 10.1002/adhm.202403860] [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: 10/06/2024] [Indexed: 01/15/2025]
Abstract
Hypoxia elicits a multitude of tissue responses depending on the severity and duration of the exposure. While chronic hypoxia is shown to impact development, regeneration, and cancer, the understanding of the threats of acute (i.e., short-term) hypoxia is limited mainly due to its transient nature. Here, a novel gelatin-dextran (Gel-Dex) hydrogel is established that decouples hydrogel formation and oxygen consumption and thus facilitates 3D sprouting from endothelial spheroids and, subsequently, induces hypoxia "on-demand." The Gel-Dex platform rapidly achieves acute moderate hypoxic conditions without compromising its mechanical properties. Acute exposure to hypoxia leads to increased endothelial cell migration and proliferation, promoting the total length and number of vascular sprouts. This work finds that the enhanced angiogenic response is mediated by reactive oxygen species, independently of hypoxia-inducible factors. Reactive oxygen species-dependent matrix metalloproteinases activity mediated angiogenic sprouting is observed following acute hypoxia. Overall, the Gel-Dex hydrogel offers a novel platform to study how "on-demand" acute moderate hypoxia impacts angiogenesis, with broad applicability to the development of novel sensing technologies.
Collapse
Affiliation(s)
- Dimitris Ntekoumes
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Jiyeon Song
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Haohao Liu
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Connor Amelung
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Ya Guan
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Sharon Gerecht
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| |
Collapse
|
2
|
Landau S, Zhao Y, Hamidzada H, Kent GM, Okhovatian S, Lu RXZ, Liu C, Wagner KT, Cheung K, Shawky SA, Vosoughi D, Beroncal EL, Fernandes I, Cummins CL, Andreazza AC, Keller GM, Epelman S, Radisic M. Primitive macrophages enable long-term vascularization of human heart-on-a-chip platforms. Cell Stem Cell 2024; 31:1222-1238.e10. [PMID: 38908380 PMCID: PMC11297673 DOI: 10.1016/j.stem.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 04/12/2024] [Accepted: 05/30/2024] [Indexed: 06/24/2024]
Abstract
The intricate anatomical structure and high cellular density of the myocardium complicate the bioengineering of perfusable vascular networks within cardiac tissues. In vivo neonatal studies highlight the key role of resident cardiac macrophages in post-injury regeneration and angiogenesis. Here, we integrate human pluripotent stem-cell-derived primitive yolk-sac-like macrophages within vascularized heart-on-chip platforms. Macrophage incorporation profoundly impacted the functionality and perfusability of microvascularized cardiac tissues up to 2 weeks of culture. Macrophages mitigated tissue cytotoxicity and the release of cell-free mitochondrial DNA (mtDNA), while upregulating the secretion of pro-angiogenic, matrix remodeling, and cardioprotective cytokines. Bulk RNA sequencing (RNA-seq) revealed an upregulation of cardiac maturation and angiogenesis genes. Further, single-nuclei RNA sequencing (snRNA-seq) and secretome data suggest that macrophages may prime stromal cells for vascular development by inducing insulin like growth factor binding protein 7 (IGFBP7) and hepatocyte growth factor (HGF) expression. Our results underscore the vital role of primitive macrophages in the long-term vascularization of cardiac tissues, offering insights for therapy and advancing heart-on-a-chip technologies.
Collapse
Affiliation(s)
- Shira Landau
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Yimu Zhao
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Homaira Hamidzada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Gregory M Kent
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Sargol Okhovatian
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Rick Xing Ze Lu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Chuan Liu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Karl T Wagner
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Krisco Cheung
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Sarah A Shawky
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Daniel Vosoughi
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Erika Leigh Beroncal
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Ian Fernandes
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada; Banting and Best Diabetes Centre, Toronto, ON, Canada; The Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, ON, Canada
| | - Ana C Andreazza
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Gordon M Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada; Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada; Terrence Donnelly Centre for Cellular & Biomolecular Research, 160 College St, Toronto, ON M5S 3E1, Canada.
| |
Collapse
|
3
|
Sharma S, Ehrlich M, Zhang M, Blobe GC, Henis YI. NRP1 interacts with endoglin and VEGFR2 to modulate VEGF signaling and endothelial cell sprouting. Commun Biol 2024; 7:112. [PMID: 38242992 PMCID: PMC10799020 DOI: 10.1038/s42003-024-05798-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 01/09/2024] [Indexed: 01/21/2024] Open
Abstract
Endothelial cells express neuropilin 1 (NRP1), endoglin (ENG) and vascular endothelial growth factor receptor 2 (VEGFR2), which regulate VEGF-A-mediated vascular development and angiogenesis. However, the link between complex formation among these receptors with VEGF-A-induced signaling and biology is yet unclear. Here, we quantify surface receptor interactions by IgG-mediated immobilization of one receptor, and fluorescence recovery after photobleaching (FRAP) measurements of the mobility of another coexpressed receptor. We observe stable ENG/NRP1, ENG/VEGFR2, and NRP1/VEGFR2 complexes, which are enhanced by VEGF-A. ENG augments NRP1/VEGFR2 interactions, suggesting formation of tripartite complexes bridged by ENG. Effects on signaling are measured in murine embryonic endothelial cells expressing (MEEC+/+) or lacking (MEEC-/-) ENG, along with NRP1 and/or ENG overexpression or knockdown. We find that optimal VEGF-A-mediated phosphorylation of VEGFR2 and Erk1/2 requires ENG and NRP1. ENG or NRP1 increase VEGF-A-induced sprouting, becoming optimal in cells expressing all three receptors, and both processes are inhibited by a MEK1/2 inhibitor. We propose a model where the maximal potency of VEGF-A involves a tripartite complex where ENG bridges VEGFR2 and NRP1, providing an attractive therapeutic target for modulation of VEGF-A signaling and biological responses.
Collapse
Affiliation(s)
- Swati Sharma
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Manqi Zhang
- Department of Medicine, Duke University Medical Center, Durham, NC, 27708, USA
| | - Gerard C Blobe
- Department of Medicine, Duke University Medical Center, Durham, NC, 27708, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Yoav I Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel.
| |
Collapse
|
4
|
Cerdà P, Castillo SD, Aguilera C, Iriarte A, Rocamora JL, Larrinaga AM, Viñals F, Graupera M, Riera-Mestre A. New genetic drivers in hemorrhagic hereditary telangiectasia. Eur J Intern Med 2024; 119:99-108. [PMID: 37689549 DOI: 10.1016/j.ejim.2023.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/11/2023]
Abstract
BACKGROUND Hereditary hemorrhagic telangiectasia (HHT) is a rare vascular disease inherited in an autosomal dominant manner. Disease-causing variants in endoglin (ENG) and activin A receptor type II-like 1 (ACVRL1) genes are detected in around 90% of the patients; also 2% of patients harbor pathogenic variants at SMAD4 and GDF2. Importantly, the genetic cause of 8% of patients with clinical HHT remains unknown. Here, we present new putative genetic drivers of HHT. METHODS To identify new HHT genetic drivers, we performed exome sequencing of 19 HHT patients and relatives with unknown HHT genetic etiology. We applied a multistep filtration strategy to catalog deleterious variants and prioritize gene candidates based on their known relevance in endothelial cell biology. Additionally, we performed in vitro validation of one of the identified variants. RESULTS We identified variants in the INHA, HIF1A, JAK2, DNM2, POSTN, ANGPTL4, FOXO1 and SMAD6 genes as putative drivers in HHT. We have identified the SMAD6 p.(Glu407Lys) variant in one of the families; this is a loss-of-function variant leading to the activation of the BMP/TGFβ signaling in endothelial cells. CONCLUSIONS Variants in these genes should be considered for genetic testing in patients with HHT phenotype and negative for ACVRL1/ENG mutations.
Collapse
Affiliation(s)
- Pau Cerdà
- HHT Unit, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain; Internal Medicine Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain; Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Sandra D Castillo
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute, Badalona, Spain
| | - Cinthia Aguilera
- HHT Unit, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain; Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Genetics Laboratory, Laboratori Clínic Territorial Metropolitana Sud, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain
| | - Adriana Iriarte
- HHT Unit, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain; Internal Medicine Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain; Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - José Luis Rocamora
- HHT Unit, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain; Molecular Signaling Group, Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Institut d'Investigacio Biomedica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ane M Larrinaga
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute, Badalona, Spain
| | - Francesc Viñals
- Molecular Signaling Group, Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Institut d'Investigacio Biomedica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Department of Physiological Sciences, Faculty of Medicine and Health Sciences, Universitat de Barcelona (UB), L'Hospitalet de Llobregat, Spain; Program Against Cancer Therapeutic Resistance (ProCURE), Institut Catala d'Oncologia (ICO), L'Hospitalet de Llobregat, Spain
| | - Mariona Graupera
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute, Badalona, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain; ICREA, Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Antoni Riera-Mestre
- HHT Unit, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain; Internal Medicine Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain; Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Department of Clinical Sciences, Faculty of Medicine and Health Sciences, Universitat de Barcelona (UB), L'Hospitalet de Llobregat, Spain.
| |
Collapse
|
5
|
Oladejo M, Nguyen HM, Wood L. CD105 in the progression and therapy of renal cell carcinoma. Cancer Lett 2023; 570:216327. [PMID: 37499740 DOI: 10.1016/j.canlet.2023.216327] [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: 04/18/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 07/29/2023]
Abstract
Molecular biomarkers that interact with the vascular and immune compartments play an important role in the progression of solid malignancies. CD105, which is a component of the transforming growth factor beta (TGF β) signaling cascade, has long been studied for its role in potentiating angiogenesis in numerous cancers. In renal cell carcinoma (RCC), the role of CD105 is more complicated due to its diverse expression profile on the tumor cells, tumor vasculature, and the components of the immune system. Since its discovery, its angiogenic role has overshadowed other potential functions, especially in cancers. In this review, we aim to summarize the recent evidence and findings of the multifunctional roles of CD105 in angiogenesis and immunomodulation in the context of the various subtypes of RCC, with a specific emphasis on the clear cell RCC subtype. Since CD105 is an established biomarker and tumor antigen, we also provide an update on the preclinical and clinical applications of CD105 as a therapeutic platform in RCC.
Collapse
Affiliation(s)
- Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Hong-My Nguyen
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Laurence Wood
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA.
| |
Collapse
|
6
|
Effects of apoptosis by 20α-hydroxysteroid dehydrogenase activity on corpus luteum formation during early pregnancy in cattle. CZECH JOURNAL OF ANIMAL SCIENCE 2023. [DOI: 10.17221/116/2022-cjas] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
|
7
|
Litwiniuk-Kosmala M, Makuszewska M, Czesak M. Endoglin in head and neck neoplasms. Front Med (Lausanne) 2023; 10:1115212. [PMID: 36844233 PMCID: PMC9950573 DOI: 10.3389/fmed.2023.1115212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 01/27/2023] [Indexed: 02/12/2023] Open
Abstract
Tumors of the head and neck region form a heterogeneous group of pathologies, including various benign lesions and malignant neoplasms. Endoglin, also known as CD105, is an accessory receptor for transforming growth factor beta (TGF-β), that regulates angiogenesis, both under physiological and pathological conditions. It is highly expressed in proliferating endothelial cells. Therefore, it is considered as a marker of tumor-related angiogenesis. In this review we discuss the role of endoglin as a possible marker of carcinogenesis, as well as a potential target for antibody-based therapies in the neoplasms of the head and neck region.
Collapse
Affiliation(s)
| | - Maria Makuszewska
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Małgorzata Czesak
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
8
|
Bastos AGP, Carvalho B, Silva R, Leitão D, Linhares P, Vaz R, Lima J. Endoglin (CD105) and proliferation index in recurrent glioblastoma treated with anti-angiogenic therapy. Front Oncol 2022; 12:910196. [PMID: 36147918 PMCID: PMC9486379 DOI: 10.3389/fonc.2022.910196] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/15/2022] [Indexed: 11/15/2022] Open
Abstract
Introduction CD105 is an angiogenic biomarker that is useful to determine the microvessel density (MVD) within a tumor, namely, in highly vascularized tumors like glioblastoma (GBM). However, its expression has shown inconsistent associations with the prognosis of GBM patients. The aim of this study was to evaluate the value of MVD-CD105 (microvessel density assessed with anti-CD105 antibody) and Ki-67 (proliferation index marker) as prognostic and therapy response biomarkers, specifically in primary tumors and in recurrent tumoral specimens of a cohort of GBM patients treated with bevacizumab upon recurrence. Materials and methods We conducted a retrospective study of 102 consecutive GBM patients treated with bevacizumab upon recurrence at CHUSJ between 2010 and 2017. Demographic, clinical, and survival data of all patients were collected and analyzed. The tissue expression of MVD-CD105 and Ki-67 in primary and recurrent specimens was correlated with progression-free survival after temozolomide (PFS-1), progression-free survival after bevacizumab (PFS-2), and overall survival (OS). Results The immunohistochemical expression score for MVD-CD105 was similar in primary and recurrent tumoral specimens (mean scores of 15 and 16, respectively). Likewise, the mean Ki-67 expression was similar in primary (mean of 31% of tumor cells) and recurrent tumoral specimens (mean of 29% of tumor cells). MVD-CD105 expression in primary tumors had no impact on PFS-1, PFS-2, or OS. At recurrence, patients whose tumors showed increased MVD-CD105 had worse median PFS-2 (2 vs. 8 months, p = 0.045) and OS (17 vs. 26 months, p = 0.007) compared to those whose tumors showed lower MVD-CD105. CD105 tumoral pattern and localization had no impact on prognosis. Ki-67 expression was not associated with differences in survival outcomes. Conclusion In this study, higher MVD-CD105 expression in recurrent GBM patients seems to be associated with a worse PFS-2 and OS while portending no prognostic significance in the primary tumors. This highlights the importance of keeping track of the molecular evolution of the tumor over the course of the disease.
Collapse
Affiliation(s)
| | - Bruno Carvalho
- Faculty of Medicine of the University of Porto, Porto, Portugal
- Department of Neurosurgery, Centro Hospitalar Universitário S. João, Porto, Portugal
- Institute for Research and Innovation in Health (i3S), R. Alfredo Allen Porto, Porto, Portugal
- *Correspondence: Bruno Carvalho,
| | - Roberto Silva
- Faculty of Medicine of the University of Porto, Porto, Portugal
- Department of Pathology, Centro Hospitalar Universitário S. João, Porto, Portugal
| | - Dina Leitão
- Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Paulo Linhares
- Faculty of Medicine of the University of Porto, Porto, Portugal
- Department of Neurosurgery, Centro Hospitalar Universitário S. João, Porto, Portugal
- Neurosciences Center-CUF Hospital, Porto, Portugal
| | - Rui Vaz
- Faculty of Medicine of the University of Porto, Porto, Portugal
- Department of Neurosurgery, Centro Hospitalar Universitário S. João, Porto, Portugal
- Neurosciences Center-CUF Hospital, Porto, Portugal
| | - Jorge Lima
- Faculty of Medicine of the University of Porto, Porto, Portugal
- Institute for Research and Innovation in Health (i3S), R. Alfredo Allen Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (Ipatimup), Porto, Portugal
| |
Collapse
|
9
|
Haarmann A, Zimmermann L, Bieber M, Silwedel C, Stoll G, Schuhmann MK. Regulation and Release of Vasoactive Endoglin by Brain Endothelium in Response to Hypoxia/Reoxygenation in Stroke. Int J Mol Sci 2022; 23:ijms23137085. [PMID: 35806090 PMCID: PMC9267030 DOI: 10.3390/ijms23137085] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
In large vessel occlusion stroke, recanalization to restore cerebral perfusion is essential but not necessarily sufficient for a favorable outcome. Paradoxically, in some patients, reperfusion carries the risk of increased tissue damage and cerebral hemorrhage. Experimental and clinical data suggest that endothelial cells, representing the interface for detrimental platelet and leukocyte responses, likely play a crucial role in the phenomenon referred to as ischemia/reperfusion (I/R)-injury, but the mechanisms are unknown. We aimed to determine the role of endoglin in cerebral I/R-injury; endoglin is a membrane-bound protein abundantly expressed by endothelial cells that has previously been shown to be involved in the maintenance of vascular homeostasis. We investigated the expression of membranous endoglin (using Western blotting and RT-PCR) and the generation of soluble endoglin (using an enzyme-linked immunosorbent assay of cell culture supernatants) after hypoxia and subsequent reoxygenation in human non-immortalized brain endothelial cells. To validate these in vitro data, we additionally examined endoglin expression in an intraluminal monofilament model of permanent and transient middle cerebral artery occlusion in mice. Subsequently, the effects of recombinant human soluble endoglin were assessed by label-free impedance-based measurement of endothelial monolayer integrity (using the xCELLigence DP system) and immunocytochemistry. Endoglin expression is highly inducible by hypoxia in human brain endothelial monolayers in vitro, and subsequent reoxygenation induced its shedding. These findings were corroborated in mice during MCAO; an upregulation of endoglin was displayed in the infarcted hemispheres under occlusion, whereas endoglin expression was significantly diminished after transient MCAO, which is indicative of shedding. Of note is the finding that soluble endoglin induced an inflammatory phenotype in endothelial monolayers. The treatment of HBMEC with endoglin resulted in a decrease in transendothelial resistance and the downregulation of VE-cadherin. Our data establish a novel mechanism in which hypoxia triggers the initial endothelial upregulation of endoglin and subsequent reoxygenation triggers its release as a vasoactive mediator that, when rinsed into adjacent vascular beds after recanalization, can contribute to cerebral reperfusion injury.
Collapse
Affiliation(s)
- Axel Haarmann
- Department of Neurology, University of Würzburg, 97080 Würzburg, Germany; (L.Z.); (M.B.); (G.S.)
- Correspondence: (A.H.); (M.K.S.)
| | - Lena Zimmermann
- Department of Neurology, University of Würzburg, 97080 Würzburg, Germany; (L.Z.); (M.B.); (G.S.)
| | - Michael Bieber
- Department of Neurology, University of Würzburg, 97080 Würzburg, Germany; (L.Z.); (M.B.); (G.S.)
| | - Christine Silwedel
- University Children’s Hospital, University of Würzburg, 97080 Würzburg, Germany;
| | - Guido Stoll
- Department of Neurology, University of Würzburg, 97080 Würzburg, Germany; (L.Z.); (M.B.); (G.S.)
| | - Michael K. Schuhmann
- Department of Neurology, University of Würzburg, 97080 Würzburg, Germany; (L.Z.); (M.B.); (G.S.)
- Correspondence: (A.H.); (M.K.S.)
| |
Collapse
|
10
|
Bronchoalveolar-Lavage-Derived Fibroblast Cell Line (B-LSDM7) as a New Protocol for Investigating the Mechanisms of Idiopathic Pulmonary Fibrosis. Cells 2022; 11:cells11091441. [PMID: 35563747 PMCID: PMC9103910 DOI: 10.3390/cells11091441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 12/27/2022] Open
Abstract
Background: The use of BAL to study ILDs has improved our understanding of IPF pathogenesis. BAL fluid is routinely collected and can be considered a clinical and research tool. The procedure is well tolerated and minimally invasive. No specific cell lines from BAL or immortalized cell lines from IPF patients are available commercially. A method to quickly isolate and characterize fibroblasts from BAL is an unmet research need. Materials and methods: Here we describe a new protocol by which we isolated a cell line from IPF. The cell line was expanded in vitro and characterized phenotypically, morphologically and functionally. Results: This culture showed highly filamentous cells with an evident central nucleus. From the phenotypic point of view, this cell line displays fibroblast/myofibroblast-like features including expression of alpha-SMA, vimentin, collagen type-1 and fibronectin. The results showed high expression of ROS in these cells. Oxidative stress invariably promotes extracellular matrix expression in lung diseases directly or through over-production of pro-fibrotic growth factors. Conclusions: Our protocol makes it possible to obtain fibroblasts BAL that is a routine non-invasive method that offers the possibility of having a large sample of patients. Standardized culture methods are important for a reliable model for testing molecules and eventual novel development therapeutic targets.
Collapse
|
11
|
Ebian HF, Issa DR, Al-Karamany AS, Etewa RL, El Maghraby HM, Hussein S. Evaluation of CDCP1 (CD318) and endoglin (CD105) expression as prognostic markers in acute myeloid leukemia. Cancer Biomark 2021; 34:285-296. [DOI: 10.3233/cbm-210346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: The most commonly used prognostic factors in acute myeloid leukemia (AML) are cytogenetic, molecular, and morphological markers. However, AML prognosis is still unfavorable particularly in adults. So, further reliable markers are urgently needed to improve the risk stratification and treatment decisions. CUB domain-containing protein 1 (CDCP1; CD318) and endoglin (CD105) are new markers correlated with poor prognosis in different solid tumors, but their role in AML prognosis is not fully evaluated. OBJECTIVES: This work aimed to evaluate the prognostic role of CD318 and CD105 in AML and their impact on the outcomes. METHODS: Sixty-five newly diagnosed AML patients were included in this study. CD318 and CD105 expression was assessed by quantitative real-time polymerase chain reaction. Patients were followed up for ∼ 2 years to evaluate the prognostic impact of gene expression on the outcomes. RESULTS: Patients with high CD318 and CD105 showed higher white blood cell (WBC) count, M2 subtype, poor cytogenetic risk, reduced complete remission, and a greater number of deaths compared to low CD318 and CD105. CD318 was correlated with CD105, and both were correlated with WBC count, bone marrow blasts, and peripheral blood blasts. After a follow-up period of up to 24 months, relapse-free survival for high CD318 and CD105 was significantly different (42.1% and 52.6% vs. 64.5% and 58.1% for low CD318 and CD105, respectively). Survival was worse in patients with high CD318 and CD105, as the mean survival time was 13.9 and 13.3 months compared to 24 and 22.7 months in low CD318 and CD105, respectively. CONCLUSIONS: CD318 and CD105 are upregulated in AML patients. Their overexpression was associated with poor response to treatment and poor outcomes. Therefore, CD318 and CD105 can be useful prognostic markers in AML.
Collapse
Affiliation(s)
- Huda F. Ebian
- Clinical Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Dina R. Issa
- Internal Medicine Department, Faculty of Medicine, Helwan University, Egypt
| | - Amira S. Al-Karamany
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Rasha L. Etewa
- Pathology Department College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Hanaa M. El Maghraby
- Medical Microbiology and Immunology Department, Faculty of Medicine, Zagazig University, Egypt
| | - Samia Hussein
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
12
|
Guo K, Xiao N, Liu Y, Wang Z, Tóth J, Gyenis J, Thakur VK, Oyane A, Shubhra QT. Engineering polymer nanoparticles using cell membrane coating technology and their application in cancer treatments: Opportunities and challenges. NANO MATERIALS SCIENCE 2021. [DOI: 10.1016/j.nanoms.2021.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
13
|
Dilated microvessel with endothelial cell proliferation involves intraplaque hemorrhage in unstable carotid plaque. Acta Neurochir (Wien) 2021; 163:1777-1785. [PMID: 32995934 DOI: 10.1007/s00701-020-04595-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/22/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND The purpose of the present study was to clarify the characteristics of endothelial cell (EC) proliferation in intraplaque microvessels in vulnerable plaques and impact on clinical results. METHODS The present study included 76 patients who underwent carotid endarterectomy. Patients were classified into three groups based on their symptoms: asymptomatic, symptomatic without recurrent ischemic event, and symptomatic with recurrent ischemic event. MR plaque imaging was performed and surgical specimens underwent immunohistochemical analysis. The number of CD31+ microvessels, and Ki67+ and CD105+ ECs in the carotid plaques was quantified, as measurements of maximum CD31+ microvessel diameter. RESULTS MR plaque imaging yielded 41 subjects (54.0%) diagnosed with plaque with intraplaque hemorrhage (IPH), 14 subjects (18.4%) diagnosed with fibrous plaques, and 21 (27.6%) subjects diagnosed with lipid-rich plaques. The average largest diameter of microvessel in fibrous plaques, lipid-rich plaques, and plaque with IPH was 12.7 ± 4.1 μm, 31.3 ± 9.3 μm, and 56.4 ± 10.0 μm, respectively (p < 0.01). Dilated microvessels (>40 μm) were observed in 9.6% of plaques with IPH but only in 2.8% of lipid-rich plaques and 0% of fibrous plaques (p < 0.01). Ki67+/CD31+ ECs were identified in 2.8 ± 1.2% of fibrous plaques, 9.6 ± 6.9% of lipid-rich plaques, and in 19.5 ± 5.9% of plaques with IPH (p < 0.01). The average largest diameter of microvessels in the asymptomatic group was 17.1 ± 8.7 μm, 32.3 ± 10.8 μm in the symptomatic without recurrence group, and 55.2 ± 13.2 μm in the symptomatic with recurrence group (p < 0.01). CONCLUSION Dilated microvessels with proliferative ECs may play a key role in IPH pathogenesis. Furthermore, dilated microvessels are likely related to clinical onset and the recurrence of ischemic events. The purpose of the present study was to clarify the characteristics of EC proliferation in intraplaque microvessels in vulnerable plaques and their impact on clinical results, focusing on dilated intraplaque microvessels.
Collapse
|
14
|
Parvathala P, Baghirath PV, Reddy CN, Vinay BH, Krishna AB, Naishadham PP. Horoscopic role of CD105 (Endoglin) in progression of oral lichen planus: An immunohistochemical study. J Oral Maxillofac Pathol 2021; 25:37-45. [PMID: 34349409 PMCID: PMC8272491 DOI: 10.4103/jomfp.jomfp_82_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 10/19/2020] [Accepted: 01/19/2021] [Indexed: 11/04/2022] Open
Abstract
Context Role of CD105(Endoglin) in Pathogenesis and progression of OLP. Aim To assess the role of neoangiogenesis in the progression of OLP by determining the expression of CD105 in OLP and normal mucosa. Settings and Design The present study includes a total of 70 formalin-fixed paraffin-embedded blocks of which the study group comprises 50 tissue sections histopathologically confirmed as OLP. They were subdivided into two groups - Group I (Reticular OLP) and Group II (Erosive OLP) - 25 each. The control group (designated as Group III) included 20 sections of normal mucosa. Materials and Methods All the sections were 4 μm thick and stained with CD105 antibodies. After identifying areas of highest vascularity (hot spots) in low power (×10) magnification, individual microvessels were counted manually at high power (×40) magnification. Statistical Analysis Used Analysis of variance test was used to determine the difference of microvessel density (MVD) between variants of OLP and normal mucosa and Cohen's kappa statistic was used to check interobserver variability. Results CD105 staining showed a mean MVD of 1.31 ± 1.8 in the normal mucosa compared to 1.68 ± 1.4 and 4.14 ± 2.7 in the reticular and erosive variants, respectively, with a P = 0.000*, which is statistically significant (*P < 0.05 is statistically significant). Conclusion Based on our observations, it is evident that compared to normal mucosa, MVD is greater in lichen planus. Within the two variants of OLP, MVD is higher in Erosive variant compared with Reticular variant, foreshadowing the role of neoangiogenesis in the progression of OLP and its possible malignant transformation.
Collapse
Affiliation(s)
- Poornima Parvathala
- CKS Theja Institute of Dental Sciences and Research, Tirupati, Andhra Pradesh, India
| | - P Venkat Baghirath
- Panineeya Mahavidyalaya Institute of Dental Sciences and Research Centre, Hyderabad, Telangana, India
| | - C Narendra Reddy
- Department of Surgical Oncology, SVIMS, Tirupati, Andhra Pradesh, India
| | - B Hari Vinay
- Panineeya Mahavidyalaya Institute of Dental Sciences and Research Centre, Hyderabad, Telangana, India
| | - A Bhargavi Krishna
- Panineeya Mahavidyalaya Institute of Dental Sciences and Research Centre, Hyderabad, Telangana, India
| | - Parameshwar P Naishadham
- Panineeya Mahavidyalaya Institute of Dental Sciences and Research Centre, Hyderabad, Telangana, India
| |
Collapse
|
15
|
Kwon HS, Ha J, Kim JY, Park HH, Lee EH, Choi H, Lee KY, Lee YJ, Koh SH. Telmisartan Inhibits the NLRP3 Inflammasome by Activating the PI3K Pathway in Neural Stem Cells Injured by Oxygen-Glucose Deprivation. Mol Neurobiol 2021; 58:1806-1818. [PMID: 33404978 DOI: 10.1007/s12035-020-02253-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 12/08/2020] [Indexed: 01/01/2023]
Abstract
Angiotensin II receptor blockers (ARBs) have been shown to exert neuroprotective effects by suppressing inflammatory and apoptotic responses. In the present study, the effects of the ARB telmisartan on the NLRP3 inflammasome induced by oxygen-glucose deprivation (OGD) in neural stem cells (NSCs) were investigated, as well as their possible association with the activation of the PI3K pathway. Cultured NSCs were treated with different concentrations of telmisartan and subjected to various durations of OGD. Cell counting, lactate dehydrogenase, bromodeoxyuridine, and colony-forming unit assays were performed to measure cell viability and proliferation. In addition, the activity of intracellular signaling pathways associated with the PI3K pathway and NLRP3 inflammasome was evaluated. Telmisartan alone did not affect NSCs up to a concentration of 10 μM under normal conditions but showed toxicity at a concentration of 100 μM. Moreover, OGD reduced the viability of NSCs in a time-dependent manner. Nevertheless, treatment with telmisartan increased the viability and proliferation of OGD-injured NSCs. Furthermore, telmisartan promoted the expression of survival-related proteins and mRNA while inhibiting the expression of death-related proteins induced by OGD. In particular, telmisartan attenuated OGD-dependent expression of the NLRP3 inflammasome and its related signaling proteins. These beneficial effects of telmisartan were blocked by a PI3K inhibitor. Together, these results indicate that telmisartan attenuated the activation of the NLRP3 inflammasome by triggering the PI3K pathway, thereby contributing to neuroprotection.
Collapse
Affiliation(s)
- Hyuk Sung Kwon
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea
| | - Jungsoon Ha
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea
- GemVax & Kael Co., Ltd, Seongnam-si, Republic of Korea
| | - Ji Young Kim
- Department of Nuclear Medicine, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Hyun-Hee Park
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea
| | - Eun-Hye Lee
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Republic of Korea
| | - Hojin Choi
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea
| | - Kyu-Yong Lee
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea
| | - Young Joo Lee
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea.
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Endoglin in the Spotlight to Treat Cancer. Int J Mol Sci 2021; 22:ijms22063186. [PMID: 33804796 PMCID: PMC8003971 DOI: 10.3390/ijms22063186] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/06/2021] [Accepted: 03/17/2021] [Indexed: 01/02/2023] Open
Abstract
A spotlight has been shone on endoglin in recent years due to that fact of its potential to serve as both a reliable disease biomarker and a therapeutic target. Indeed, endoglin has now been assigned many roles in both physiological and pathological processes. From a molecular point of view, endoglin mainly acts as a co-receptor in the canonical TGFβ pathway, but also it may be shed and released from the membrane, giving rise to the soluble form, which also plays important roles in cell signaling. In cancer, in particular, endoglin may contribute to either an oncogenic or a non-oncogenic phenotype depending on the cell context. The fact that endoglin is expressed by neoplastic and non-neoplastic cells within the tumor microenvironment suggests new possibilities for targeted therapies. Here, we aimed to review and discuss the many roles played by endoglin in different tumor types, as well as the strong evidence provided by pre-clinical and clinical studies that supports the therapeutic targeting of endoglin as a novel clinical strategy.
Collapse
|
17
|
Abstract
Microvascular proliferation is a key feature of glioblastoma and neovascularization has been implicated in tumor progression. Glioblastomas use pro-angiogenic factors such as vascular endothelial growth factor (VEGF) for new blood vessel formation. Yet, anti-VEGF therapy does not prolong overall survival so that alternative angiogenic pathways may need to be explored as drug targets. Both glioma cells and glioma-associated endothelial cells produce TGF-β superfamily ligands which bind TGF-β receptors (TGF-βR). The TGF-βR type III endoglin (CD105), is a marker of proliferating endothelium that has already been studied as a potential therapeutic target. We studied endoglin expression in glioblastoma tissue and in glioma-associated endothelial cells in a cohort of 52 newly diagnosed and 10 recurrent glioblastoma patients by immunohistochemistry and by ex vivo single-cell gene expression profiling of 6 tumors. Endoglin protein levels were similar in tumor stroma and endothelium and correlated within tumors. Similarly, endoglin mRNA determined by ex vivo single-cell gene expression profiling was expressed in both compartments. There was positive correlation between endoglin and proteins of TGF-β superfamily signaling. No prognostic role of endoglin expression in either compartment was identified. Endoglin gene silencing in T98G glioma cells and in human cerebral microvascular endothelial cells (hCMEC) did not affect constitutive or exogenous TGF-β superfamily ligand-dependent signaling, except for a minor facilitation of pSmad1/5 signaling in hCMEC. These observations challenge the notion that endoglin might become a promising therapeutic target in glioblastoma.
Collapse
|
18
|
Potential Second-Hits in Hereditary Hemorrhagic Telangiectasia. J Clin Med 2020; 9:jcm9113571. [PMID: 33167572 PMCID: PMC7694477 DOI: 10.3390/jcm9113571] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant genetic disorder that presents with telangiectases in skin and mucosae, and arteriovenous malformations (AVMs) in internal organs such as lungs, liver, and brain. Mutations in ENG (endoglin), ACVRL1 (ALK1), and MADH4 (Smad4) genes account for over 95% of HHT. Localized telangiectases and AVMs are present in different organs, with frequencies which differ among affected individuals. By itself, HHT gene heterozygosity does not account for the focal nature and varying presentation of the vascular lesions leading to the hypothesis of a “second-hit” that triggers the lesions. Accumulating research has identified a variety of triggers that may synergize with HHT gene heterozygosity to generate the vascular lesions. Among the postulated second-hits are: mechanical trauma, light, inflammation, vascular injury, angiogenic stimuli, shear stress, modifier genes, and somatic mutations in the wildtype HHT gene allele. The aim of this review is to summarize these triggers, as well as the functional mechanisms involved.
Collapse
|
19
|
Corliss BA, Ray HC, Doty RW, Mathews C, Sheybani N, Fitzgerald K, Prince R, Kelly-Goss MR, Murfee WL, Chappell J, Owens GK, Yates PA, Peirce SM. Pericyte Bridges in Homeostasis and Hyperglycemia. Diabetes 2020; 69:1503-1517. [PMID: 32321760 PMCID: PMC7306121 DOI: 10.2337/db19-0471] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 04/17/2020] [Indexed: 12/29/2022]
Abstract
Diabetic retinopathy is a potentially blinding eye disease that threatens the vision of one-ninth of patients with diabetes. Progression of the disease has long been attributed to an initial dropout of pericytes that enwrap the retinal microvasculature. Revealed through retinal vascular digests, a subsequent increase in basement membrane bridges was also observed. Using cell-specific markers, we demonstrate that pericytes rather than endothelial cells colocalize with these bridges. We show that the density of bridges transiently increases with elevation of Ang-2, PDGF-BB, and blood glucose; is rapidly reversed on a timescale of days; and is often associated with a pericyte cell body located off vessel. Cell-specific knockout of KLF4 in pericytes fully replicates this phenotype. In vivo imaging of limbal vessels demonstrates pericyte migration off vessel, with rapid pericyte filopodial-like process formation between adjacent vessels. Accounting for off-vessel and on-vessel pericytes, we observed no pericyte loss relative to nondiabetic control retina. These findings reveal the possibility that pericyte perturbations in location and process formation may play a role in the development of pathological vascular remodeling in diabetic retinopathy.
Collapse
Affiliation(s)
- Bruce A Corliss
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - H Clifton Ray
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Richard W Doty
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Corbin Mathews
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Natasha Sheybani
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Kathleen Fitzgerald
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Remi Prince
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Molly R Kelly-Goss
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Walter L Murfee
- Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - John Chappell
- Fralin Biomedical Research Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Gary K Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA
| | - Paul A Yates
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
- Department of Ophthalmometry, University of Virginia School of Medicine, Charlottesville, VA
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
- Department of Ophthalmometry, University of Virginia School of Medicine, Charlottesville, VA
| |
Collapse
|
20
|
CD105 (endoglin) as risk marker in AML patients undergoing stem cell transplantation. Int J Hematol 2020; 112:57-64. [PMID: 32266669 DOI: 10.1007/s12185-020-02875-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/27/2020] [Accepted: 03/29/2020] [Indexed: 10/24/2022]
Abstract
Several genetic and molecular markers are general predictors of outcome in acute myeloid leukemia (AML), but only few are predictive of outcomes after allogeneic hematopoietic stem cell transplantation (HSCT). Novel markers are needed to improve treatment decisions regarding HSCT. CD105 (endoglin) is a type I transmembrane protein capable of activating endothelial cells. Moreover, CD105 mediates stem cell properties of hematopoietic stem cells and enables repopulation within the bone marrow. Expression of CD105 on vessels of solid tumors and on AML blasts is correlated with poor prognosis. We recently demonstrated that CD105 expression is an independent predictor of overall survival in AML. However, its role in patients receiving intensive treatment with consecutive allogenic transplantation has not been assessed. Using flow cytometry, we analyzed primary samples of 41 patients who underwent HSCT. Using the previously defined SFI cut-off of 5.2, we identified differences in CD105 expression regarding FAB classification and NCCN risk score. Moreover, we detected differences regarding transplant indication and WHO classification with regards to CD105 surface levels. In patients undergoing HSCT high CD105 expression correlated significantly with poor overall survival. We identify CD105 expression in AML as prognostic marker for outcome after HSCT in AML.
Collapse
|
21
|
Abstract
While several genetic and morphological markers are established and serve to guide therapy of acute myeloid leukaemia (AML), there is still profound need to identify additional markers to better stratify patients. CD105 (Endoglin) is a type I transmembrane protein reported to induce activation and proliferation of endothelial cells. In addition, CD105 is expressed in haematological malignancies and the vessels of solid tumours. Here, CD105 associates with unfavourable disease course, but so far no data are available on the prognostic relevance of CD105 in haematological malignancies. We here generated a novel CD105 antibody for analysis of expression and prognostic relevance of CD105 in a cohort of 62 AML patients. Flow cytometric analysis revealed substantial expression in the various AML FAB types, with FAB M3 type displaying significantly lower surface levels. Next we established a cut-off specific fluorescence level of 5.22 using receiver-operating characteristics, which allowed to group patients in cases with CD105lo and CD105hi surface expression and revealed that high CD105 expression correlated significantly with poor overall and progression free survival. In conclusion, we here identify CD105 expression as a novel prognostic marker in AML, which may serve to optimize follow up and treatment decisions for AML patients.
Collapse
|
22
|
Choi H, Choi NY, Park HH, Lee KY, Lee YJ, Koh SH. Sublethal Doses of Zinc Protect Rat Neural Stem Cells Against Hypoxia Through Activation of the PI3K Pathway. Stem Cells Dev 2019; 28:769-780. [PMID: 30896367 DOI: 10.1089/scd.2018.0138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cerebral infarction is one of the major causes of severe morbidity and mortality, and thus, research has focused on developing treatment options for this condition. Zinc (Zn) is an essential element in the central nervous system and has several neuroprotective effects in the brain. In this study, we examined the neuroprotective effects of Zn on neural stem cells (NSCs) exposed to hypoxia. After treatment with several concentrations of Zn, the viability of NSCs under hypoxic conditions was measured by a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, Trypan blue staining, and a lactate dehydrogenase assay. To evaluate the effect of Zn on the proliferation of NSCs, bromodeoxyuridine/5-bromo-2'-deoxyuridine (BrdU) labeling and colony formation assays were performed. Apoptosis was also examined in NSCs exposed to hypoxia with and without Zn treatment. In addition, a western blot analysis was performed to evaluate the effect of Zn on intracellular signaling proteins. NSC viability and proliferation were decreased under hypoxic conditions, but treatment with sublethal doses of Zn restored viability and proliferation. Sublethal doses of Zn reduced apoptosis caused by hypoxia, increased the expression levels of proteins related to the phosphatidylinositol-3 kinase (PI3K) pathway, and decreased the expression levels of proteins associated with neuronal cell death. These findings confirm that in vivo, sublethal doses of Zn protect NSCs against hypoxia through the activation of the PI3K pathway. Thus, Zn could be employed as a therapeutic option to protect NSCs in ischemic stroke.
Collapse
Affiliation(s)
- Hojin Choi
- 1 Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
| | - Na-Young Choi
- 1 Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
| | - Hyun-Hee Park
- 1 Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
| | - Kyu-Yong Lee
- 1 Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
| | - Young Joo Lee
- 1 Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
| | - Seong-Ho Koh
- 1 Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
- 2 Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Korea
| |
Collapse
|
23
|
De Vilder EYG, Cardoen S, Hosen MJ, Le Saux O, De Zaeytijd J, Leroy BP, De Reuck J, Coucke PJ, De Paepe A, Hemelsoet D, Vanakker OM. Pathogenic variants in the ABCC6 gene are associated with an increased risk for ischemic stroke. Brain Pathol 2019; 28:822-831. [PMID: 29722917 DOI: 10.1111/bpa.12620] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/29/2018] [Indexed: 12/11/2022] Open
Abstract
Ischemic stroke causes a high mortality and morbidity worldwide. It results from a complex interplay of incompletely known environmental and genetic risk factors. We investigated the ABCC6 gene as a candidate risk factor for ischemic stroke because of the increased ischemic stroke incidence in the autosomal recessive disorder pseudoxanthoma elasticum, caused by biallelic pathogenic ABCC6 variants, the higher cardiovascular risk in heterozygous carriers and the established role of ABCC6 dysfunction in myocardial ischemia. We established segregation of a known pathogenic ABCC6 variant (p.[Arg1314Gln]) in 11/19 family members of an ischemic stroke patient in a large multigenerational family suffering from ischemic stroke and/or cardiovascular disease at a relatively young age. In an independent case-control study in 424 ischemic stroke patients and 250 healthy controls, pathogenic ABCC6 variants were 4.9 times more frequent (P = 0.036; 95% CI 1.11-21.33) in the ischemic stroke patient cohort. To study cellular consequences of ABCC6 deficiency in the brain, immunostaining of brain sections in Abcc6-deficient mice and wild-type controls were performed. An upregulation of Bmp4 and Eng and a downregulation of Alk2 was identified in Abcc6-/- mice, suggesting an increase in apoptosis and angiogenesis. As both of these processes are induced in ischemia, we propose that a pro-ischemic state may explain the higher risk to suffer from ischemic stroke in patients carrying a pathogenic ABCC6 variant, as this may lower the threshold to develop acute ischemic events in these patients. In conclusion, this study identified heterozygous ABCC6 variants as a risk factor for ischemic stroke. Further, dysregulation of Bmp (Bmp4, Alk2) and Tgfβ (Eng) signaling in the brain of Abcc6-/- mice could lead to a pro-ischemic state, lowering the threshold to develop acute ischemic events. These data demonstrate the importance of a molecular analysis of the ABCC6 gene in patients diagnosed with cryptogenic ischemic stroke.
Collapse
Affiliation(s)
- Eva Y G De Vilder
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium.,Department of Ophthalmology, Ghent University Hospital, Ghent, Belgium.,Research Foundation - Flanders, Brussels, Belgium
| | - Stefanie Cardoen
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Mohammad J Hosen
- Department of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Olivier Le Saux
- Department of Cell and Molecular Biology, The John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI
| | - Julie De Zaeytijd
- Department of Ophthalmology, Ghent University Hospital, Ghent, Belgium
| | - Bart P Leroy
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium.,Department of Ophthalmology, Ghent University Hospital, Ghent, Belgium.,Division of Ophthalmology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Jacques De Reuck
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Paul J Coucke
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Anne De Paepe
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | | | | |
Collapse
|
24
|
|
25
|
Atorvastatin Rejuvenates Neural Stem Cells Injured by Oxygen–Glucose Deprivation and Induces Neuronal Differentiation Through Activating the PI3K/Akt and ERK Pathways. Mol Neurobiol 2018; 56:2964-2977. [DOI: 10.1007/s12035-018-1267-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 07/18/2018] [Indexed: 01/01/2023]
|
26
|
Qian H, Yang L, Zhao W, Chen H, He S. A comparison of CD105 and CD31 expression in tumor vessels of hepatocellular carcinoma by tissue microarray and flow cytometry. Exp Ther Med 2018; 16:2881-2888. [PMID: 30214510 PMCID: PMC6125829 DOI: 10.3892/etm.2018.6553] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 07/19/2018] [Indexed: 02/06/2023] Open
Abstract
Tumor endothelial cells (TECs) have been isolated from solid tumors by using immunological magnetic beads and magnetic active cell sorting, and lead to a more precise way to investigate tumor angiogenesis as well as screening of vascular targeting drugs. However, the question of which endothelial marker is a stable molecular signature in TECs and can be used for the isolation of TECs from tumor tissues remains unclear. In this study, we investigated the endothelial markers CD105 and CD31 in the tumor vessels from 90 patients with hepatocellular carcinoma (HCC) by tissue microarray, in addition to their expression in TECs isolated from fresh tissues resected from 11 patients with HCC by flow cytometry and confocal microscopy. The results revealed that among 90 cases of TMA, all tumor vessels were CD31 positive whereas 39 cases (43.3%) had little or no CD105 expression in tumors and their vessels but not peritumoral tissue spots, and that among these 39, 29 cases (74.4%) were poor-differentiated HCC. These findings were further verified by flow cytometry and confocal analysis of TECs isolated from HCC. Overall, the results suggested that CD105 may not be expressed in TECs derived from poor-differentiated HCC cases. In addition, combined with previous studies in which CD105 is not only expressed in TECs, but also in tumor cells, the results indicated a high risk of contamination with CD105+ tumor cells. Thus, there is a limitation to the use CD105 as an endothelial marker for the isolation of TECs.
Collapse
Affiliation(s)
- Hongyan Qian
- Key Laboratory of Cancer Research Center Nantong, Tumor Hospital Affiliated to Nantong University, Nantong, Jiangsu 226361, P.R. China
| | - Liping Yang
- Key Laboratory of Cancer Research Center Nantong, Tumor Hospital Affiliated to Nantong University, Nantong, Jiangsu 226361, P.R. China
| | - Wenjing Zhao
- Key Laboratory of Cancer Research Center Nantong, Tumor Hospital Affiliated to Nantong University, Nantong, Jiangsu 226361, P.R. China
| | - Haizhen Chen
- Key Laboratory of Cancer Research Center Nantong, Tumor Hospital Affiliated to Nantong University, Nantong, Jiangsu 226361, P.R. China
| | - Song He
- Key Laboratory of Cancer Research Center Nantong, Tumor Hospital Affiliated to Nantong University, Nantong, Jiangsu 226361, P.R. China
| |
Collapse
|
27
|
Badmus OO, Michael OS, Rabiu S, Olatunji LA. Gestational glucocorticoid exposure disrupts glucose homeostasis that is accompanied by increased endoglin and DPP-4 activity instead of GSK-3 in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 60:66-75. [PMID: 29677638 DOI: 10.1016/j.etap.2018.04.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 05/27/2023]
Abstract
Gestational glucocorticoid (GC) treatment has been associated with cardiometabolic disorder (CMD) in offspring's in later life. Elevated dipeptidyl peptidase-4 (DPP-4) activity, endoglin and glycogen synthase kinase-3 (GSK-3) has also been implicated in the development of insulin resistance (IR) and/or vascular inflammation. We aimed to investigate the impact of GC exposure on glucose metabolism and the circulating levels of inflammatory biomarkers, DPP-4 activity and GSK-3 in pregnant rats. Pregnant Wistar rats received either vehicle or dexamethasone (DEX; 0.2 mg/kg; po) between gestational days 14 and 19. Gestational GC exposure resulted in impaired glucose homeostasis that is accompanied with elevated circulating levels of inflammatory biomarkers (endoglin, uric acid, and platelet/lymphocyte ratio), oxidative stress (malondialdehyde), blood viscosity, reduced NO level and increased DPP-4 activity. However, these effects were associated with atherogenic dyslipidemia and reduced GSK-3.We conclude that plasma endoglin, a marker of vascular inflammation, and plasma DPP-4 activity are increased in pregnant rats treated with GC during late gestation. Therefore, glucose deregulation associated with gestational GC exposure is through endoglin-/DPP-4-dependent but GSK-3-independent pathway.
Collapse
Affiliation(s)
- Olufunto O Badmus
- Department of Physiology & Hope Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria; Department of Public Health, Kwara State University, Malete, Nigeria
| | - Olugbenga S Michael
- Department of Physiology & Hope Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria; Department of Physiology, Cardiometabolic Research Unit, College of Health and Medical sciences, Bowen University, Iwo, Nigeria
| | - Saheed Rabiu
- Department of Physiology & Hope Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Lawrence A Olatunji
- Department of Physiology & Hope Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.
| |
Collapse
|
28
|
Wu T, Nie H, Dietz AB, Salek DR, Smith J, van Wijnen AJ, Qu W. Cytotoxic Effects of Nonionic Iodinated Contrast Agent on Human Adipose-Derived Mesenchymal Stem Cells. PM R 2018; 11:S1934-1482(18)30294-6. [PMID: 29860023 PMCID: PMC6358519 DOI: 10.1016/j.pmrj.2018.05.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 05/20/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Transplantation of mesenchymal stem cells (MSCs) is a promising therapy for degenerative spine conditions. However, cell therapy for painful spine degeneration presently requires use of contrast agents during fluoroscopy-guided injections, and the effects of these agents on MSCs represents a gap in knowledge. OBJECTIVE To investigate the biological effects of contrast media (CM) that are coinjected with MSCs. DESIGN Prospective observational study. SETTING Academic medical center. PARTICIPANTS Patient-derived clinical-grade culture expanded MSCs. INTERVENTIONS Iohexol (Omnipaque300) was reduced to 12.5%, 25%, 50%, and 100% of the stock solution and incubated with MSCs for 30 minutes, 4 hours, and 48 hours. We also used complete media and 12.5%, 25%, 50%, 100% of phosphate-buffered saline as a control group. MAIN OUTCOME MEASURES We examined cytotoxicity of iohexol at different concentrations and exposure duration, as well as the potential for recovery over time. Cell counts, mitochondrial activity, and quantitative real time reverse-transcriptase polymerase chain reaction of related genes were analyzed immediately after exposure (day 0) and after 2 days of exposure (day 2). RESULTS Human MSCs exhibit a time- and concentration-dependent cytotoxic response to iodinated CM. A brief, 30-minute exposure did not affect MSCs function and viability. However, extended treatment with iohexol for 4 hours at 50% or higher concentration had a significant impact on both viability and gene expression in MSCs. CONCLUSIONS CM (Omnipaque300) is cytotoxic to MSCs in a time-and concentration-dependent manner. Hence, the concentration of CM that accompanies MSC injections should be carefully considered during MSC therapy for disk-degenerative diseases. LEVEL OF EVIDENCE To be determined.
Collapse
Affiliation(s)
- Tao Wu
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Rochester, MN
| | - Hai Nie
- Department of Orthopedic Surgery, Mayo Clinic Rochester, MN
| | - Allan B. Dietz
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, MN
| | | | - Jay Smith
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Rochester, MN
- Department of Radiology, Mayo Clinic Rochester, MN
- Department of Anatomy, Mayo Clinic Rochester, MN
| | - Andre J. van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic Rochester, MN
- Department of Biochemistry and Molecular Biology, Mayo Clinic Rochester, MN
- Department of Physiology and Biomedical Engineering, Mayo Clinic Rochester, MN
| | - Wenchun Qu
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Rochester, MN
- Division of Pain Medicine, Mayo Clinic Rochester, MN
- Spine Center, Mayo Clinic Rochester, MN
| |
Collapse
|
29
|
Workman A, Zhu L, Keel BN, Smith TPL, Jones C. The Wnt Signaling Pathway Is Differentially Expressed during the Bovine Herpesvirus 1 Latency-Reactivation Cycle: Evidence That Two Protein Kinases Associated with Neuronal Survival, Akt3 and BMPR2, Are Expressed at Higher Levels during Latency. J Virol 2018; 92:e01937-17. [PMID: 29321317 PMCID: PMC5972910 DOI: 10.1128/jvi.01937-17] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/04/2018] [Indexed: 12/20/2022] Open
Abstract
Sensory neurons in trigeminal ganglia (TG) of calves latently infected with bovine herpesvirus 1 (BoHV-1) abundantly express latency-related (LR) gene products, including a protein (ORF2) and two micro-RNAs. Recent studies in mouse neuroblastoma cells (Neuro-2A) demonstrated ORF2 interacts with β-catenin and a β-catenin coactivator, high-mobility group AT-hook 1 (HMGA1) protein, which correlates with increased β-catenin-dependent transcription and cell survival. β-Catenin and HMGA1 are readily detected in a subset of latently infected TG neurons but not TG neurons from uninfected calves or reactivation from latency. Consequently, we hypothesized that the Wnt/β-catenin signaling pathway is differentially expressed during the latency and reactivation cycle and an active Wnt pathway promotes latency. RNA-sequencing studies revealed that 102 genes associated with the Wnt/β-catenin signaling pathway were differentially expressed in TG during the latency-reactivation cycle in calves. Wnt agonists were generally expressed at higher levels during latency, but these levels decreased during dexamethasone-induced reactivation. The Wnt agonist bone morphogenetic protein receptor 2 (BMPR2) was intriguing because it encodes a serine/threonine receptor kinase that promotes neuronal differentiation and inhibits cell death. Another differentially expressed gene encodes a protein kinase (Akt3), which is significant because Akt activity enhances cell survival and is linked to herpes simplex virus 1 latency and neuronal survival. Additional studies demonstrated ORF2 increased Akt3 steady-state protein levels and interacted with Akt3 in transfected Neuro-2A cells, which correlated with Akt3 activation. Conversely, expression of Wnt antagonists increased during reactivation from latency. Collectively, these studies suggest Wnt signaling cooperates with LR gene products, in particular ORF2, to promote latency.IMPORTANCE Lifelong BoHV-1 latency primarily occurs in sensory neurons. The synthetic corticosteroid dexamethasone consistently induces reactivation from latency in calves. RNA sequencing studies revealed 102 genes associated with the Wnt/β-catenin signaling pathway are differentially regulated during the latency-reactivation cycle. Two protein kinases associated with the Wnt pathway, Akt3 and BMPR2, were expressed at higher levels during latency but were repressed during reactivation. Furthermore, five genes encoding soluble Wnt antagonists and β-catenin-dependent transcription inhibitors were induced during reactivation from latency. These findings are important because Wnt, BMPR2, and Akt3 promote neurogenesis and cell survival, processes crucial for lifelong viral latency. In transfected neuroblastoma cells, a viral protein expressed during latency (ORF2) interacts with and enhances Akt3 protein kinase activity. These findings provide insight into how cellular factors associated with the Wnt signaling pathway cooperate with LR gene products to regulate the BoHV-1 latency-reactivation cycle.
Collapse
Affiliation(s)
- Aspen Workman
- United States Department of Agriculture, Agricultural Research Service, U.S. Meat Animal Research Center, Clay Center, Nebraska, USA
| | - Liqian Zhu
- Oklahoma State University Center for Veterinary Health Sciences, Department of Veterinary Pathobiology, Stillwater, Oklahoma, USA
- College of Veterinary Medicine and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
| | - Brittney N Keel
- United States Department of Agriculture, Agricultural Research Service, U.S. Meat Animal Research Center, Clay Center, Nebraska, USA
| | - Timothy P L Smith
- United States Department of Agriculture, Agricultural Research Service, U.S. Meat Animal Research Center, Clay Center, Nebraska, USA
| | - Clinton Jones
- Oklahoma State University Center for Veterinary Health Sciences, Department of Veterinary Pathobiology, Stillwater, Oklahoma, USA
| |
Collapse
|
30
|
Nakamura T. Changes in Expression of Bile Canalicular CD10 and Sinusoidal CD105 (Endoglin) in Peritumoral Hepatic Tissue. TUMORI JOURNAL 2018; 95:495-500. [DOI: 10.1177/030089160909500415] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Aims and background Hepatic tissues, including bile canaliculi and sinusoids, around primary or metastatic tumors are destructed and regenerate associated with tumor growth, and may show some phenotypic changes. The present study was undertaken to examine the expression of CD10 in bile canaliculi [CD10(BC)] and CD105 (endoglin) along hepatic sinusoids [CD105(HS)] in peritumoral hepatic tissue (PTH). Methods Fifty samples of resected liver bearing hepatocellular carcinoma (HCC) or metastatic carcinoma were immunostained for CD10 and CD105. The immunoreactivity for CD10(BC) and CD105(HS) in the background hepatic tissue of tumors and PTH was scored separately. Results CD10(BC) was moderately or markedly expressed in the background hepatic tissue without chronic hepatitis or cirrhosis in most of the cases, and was significantly downregulated in chronic hepatitis and cirrhosis. CD105(HS) was negative or minimally positive in most of the cases of hepatic tissue bearing metastatic carcinoma, and showed a significant increase in chronic hepatitis and cirrhosis. Compared with the background, PTH revealed significantly decreased CD10(BC) staining irrespective of HCC or metastatic carcinoma, and showed belt-like CD105(HS) expression in 66.7% of the cases of metastatic carcinoma and in 88.6% of those with HCC. Conclusions These data indicate that the expression patterns of CD10(BC) and CD105(HS) in PTH are similar to those in chronic hepatitis and cirrhosis, which may be caused by persistent injury and resultant regeneration of hepatic tissue.
Collapse
|
31
|
Tian H, Huang JJ, Golzio C, Gao X, Hector-Greene M, Katsanis N, Blobe GC. Endoglin interacts with VEGFR2 to promote angiogenesis. FASEB J 2018; 32:2934-2949. [PMID: 29401587 DOI: 10.1096/fj.201700867rr] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Endoglin, a TGF-β coreceptor predominantly expressed in endothelial cells, plays an important role in vascular development and tumor-associated angiogenesis. However, the mechanism by which endoglin regulates angiogenesis, especially during tip cell formation, remains largely unknown. In this study, we report that endoglin promoted VEGF-induced tip cell formation. Mechanistically, endoglin interacted with VEGF receptor (VEGFR)-2 in a VEGF-dependent manner, which sustained VEGFR2 on the cell surface and prevented its degradation. Endoglin mutants deficient in the ability to interact with VEGFR2 failed to sustain VEGFR2 on the cell surface and to promote VEGF-induced tip cell formation. Further, an endoglin-targeting monoclonal antibody (mAb), TRC105, cooperated with a VEGF-A targeting mAb, bevacizumab, to inhibit VEGF signaling and tip cell formation in vitro and to inhibit tumor growth, metastasis, and tumor-associated angiogenesis in a murine tumor model. This study demonstrate a novel mechanism by which endoglin initiates and regulates VEGF-driven angiogenesis while providing a rationale for combining anti-VEGF and anti-endoglin therapy in patients with cancer.-Tian, H., Huang, J. J., Golzio, C., Gao, X., Hector-Greene, M., Katsanis, N., Blobe, G. C. Endoglin interacts with VEGFR2 to promote angiogenesis.
Collapse
Affiliation(s)
- Hongyu Tian
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Jennifer J Huang
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Christelle Golzio
- Center for Human Disease Modeling, Duke University Medical Center, Durham, North Carolina, USA
| | - Xia Gao
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Melissa Hector-Greene
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Nicholas Katsanis
- Center for Human Disease Modeling, Duke University Medical Center, Durham, North Carolina, USA
| | - Gerard C Blobe
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
32
|
Abstract
Endoglin (ENG, also known as CD105) is a transforming growth factor β (TGFβ) associated receptor and is required for both vasculogenesis and angiogenesis. Angiogenesis is important in the development of cerebral vasculature and in the pathogenesis of cerebral vascular diseases. ENG is an essential component of the endothelial nitric oxide synthase activation complex. Animal studies showed that ENG deficiency impairs stroke recovery. ENG deficiency also impairs the regulation of vascular tone, which contributes to the pathogenesis of brain arteriovenous malformation (bAVM) and vasospasm. In human, functional haploinsufficiency of ENG gene causes type I hereditary hemorrhagic telangiectasia (HHT1), an autosomal dominant disorder. Compared to normal population, HHT1 patients have a higher prevalence of AVM in multiple organs including the brain. Vessels in bAVM are fragile and tend to rupture, causing hemorrhagic stroke. High prevalence of pulmonary AVM in HHT1 patients are associated with a higher incidence of paradoxical embolism in the cerebral circulation causing ischemic brain injury. Therefore, HHT1 patients are at risk for both hemorrhagic and ischemic stroke. This review summarizes the possible mechanism of ENG in the pathogenesis of cerebrovascular diseases in experimental animal models and in patients.
Collapse
Affiliation(s)
- Wan Zhu
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Li Ma
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Rui Zhang
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Hua Su
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
33
|
Ruiz-Llorente L, Gallardo-Vara E, Rossi E, Smadja DM, Botella LM, Bernabeu C. Endoglin and alk1 as therapeutic targets for hereditary hemorrhagic telangiectasia. Expert Opin Ther Targets 2017; 21:933-947. [PMID: 28796572 DOI: 10.1080/14728222.2017.1365839] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Hereditary Haemorrhagic Telangiectasia (HHT) is as an autosomal dominant trait characterized by frequent nose bleeds, mucocutaneous telangiectases, arteriovenous malformations (AVMs) of the lung, liver and brain, and gastrointestinal bleedings due to telangiectases. HHT is originated by mutations in genes whose encoded proteins are involved in the transforming growth factor β (TGF-β) family signalling of vascular endothelial cells. In spite of the great advances in the diagnosis as well as in the molecular, cellular and animal models of HHT, the current treatments remain just at the palliative level. Areas covered: Pathogenic mutations in genes coding for the TGF-β receptors endoglin (ENG) (HHT1) or the activin receptor-like kinase-1 (ACVRL1 or ALK1) (HHT2), are responsible for more than 80% of patients with HHT. Therefore, ENG and ALK1 are the main potential therapeutic targets for HHT and the focus of this review. The current status of the preclinical and clinical studies, including the anti-angiogenic strategy, have been addressed. Expert opinion: Endoglin and ALK1 are attractive therapeutic targets in HHT. Because haploinsufficiency is the pathogenic mechanism in HHT, several therapeutic approaches able to enhance protein expression and/or function of endoglin and ALK1 are keys to find novel and efficient treatments for the disease.
Collapse
Affiliation(s)
- Lidia Ruiz-Llorente
- a Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) , Madrid , Spain
| | - Eunate Gallardo-Vara
- a Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) , Madrid , Spain
| | - Elisa Rossi
- b Faculté de Pharmacie , Paris Descartes University, Sorbonne Paris Cité and Inserm UMR-S1140 , Paris , France
| | - David M Smadja
- b Faculté de Pharmacie , Paris Descartes University, Sorbonne Paris Cité and Inserm UMR-S1140 , Paris , France
| | - Luisa M Botella
- a Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) , Madrid , Spain
| | - Carmelo Bernabeu
- a Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) , Madrid , Spain
| |
Collapse
|
34
|
Mashreghi M, Azarpara H, Bazaz MR, Jafari A, Masoudifar A, Mirzaei H, Jaafari MR. Angiogenesis biomarkers and their targeting ligands as potential targets for tumor angiogenesis. J Cell Physiol 2017; 233:2949-2965. [DOI: 10.1002/jcp.26049] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/12/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Mohammad Mashreghi
- NanotechnologyResearch Center; Mashhad University of Medical Sciences; Mashhad Iran
- School of Pharmacy; Mashhad University of Medical Sciences; Mashhad Iran
| | - Hassan Azarpara
- School of Medicine; Iran University of Medical Sciences; Tehran Iran
| | - Mahere R. Bazaz
- Division of Biotechnology, Faculty of Veterinary Medicine; Ferdowsi University of Mashhad; Mashhad Iran
| | - Arash Jafari
- School of Medicine; Birjand University of Medical Sciences; Birjand Iran
| | - Aria Masoudifar
- Department of Molecular Biotechnology, Cell Science Research Center; Royan Institute for Biotechnology; ACECR Isfahan Iran
| | - Hamed Mirzaei
- Department of Medical Biotechnology, School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| | - Mahmoud R. Jaafari
- NanotechnologyResearch Center; Mashhad University of Medical Sciences; Mashhad Iran
- School of Pharmacy; Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
35
|
Fu J, Wiraja C, Muhammad HB, Xu C, Wang DA. Improvement of endothelial progenitor outgrowth cell (EPOC)-mediated vascularization in gelatin-based hydrogels through pore size manipulation. Acta Biomater 2017; 58:225-237. [PMID: 28611001 DOI: 10.1016/j.actbio.2017.06.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 06/06/2017] [Accepted: 06/09/2017] [Indexed: 12/11/2022]
Abstract
In addition to chemical compositions, physical properties of scaffolds, such as pore size, can also influence vascularization within the scaffolds. A larger pore has been shown to improve host vascular tissue invasion into scaffolds. However, the influence of pore sizes on vascularization by endothelial cells directly encapsulated in hydrogels remains unknown. In this study, micro-cavitary hydrogels with different pore sizes were created in gelatin-methacrylate hydrogels with dissolvable gelatin microspheres (MS) varying in sizes. The effect of pore sizes on vascular network formation by endothelial progenitor outgrowth cells (EPOCs) encapsulated in hydrogels was then investigated both in vitro and in vivo. When cultured in vitro, vascular networks were formed around pore structures in micro-cavitary hydrogels. The middle pore size supported best differentiation of EPOCs and thus best hydrogel vascularization in vitro. When implantation in vivo, functional connections between encapsulated EPOCs and host vasculature micro-cavitary hydrogels were established. Vascularization in vivo was promoted best in hydrogels with the large pore size due to the increased vascular tissue invasion. These results highlight the difference between in vitro and in vivo culture conditions and indicate that pore sizes shall be designed for in vitro and in vivo hydrogel vascularization respectively. Pore sizes for hydrogel vascularization in vitro shall be middle ones and pore sizes for hydrogel vascularization in vivo shall be large ones. STATEMENT OF SIGNIFICANCE This study reveals that the optimal pore size for hydrogel vascularization in vitro and in vivo is different. The middle pore size supported best differentiation of EPOCs and thus best hydrogel vascularization in vitro, while vascularization in vivo was promoted best in hydrogels with the large pore size due to the increased vascular tissue invasion. These results highlight the difference between in vitro and in vivo culture conditions and indicate that pore sizes shall be designed for in vitro and in vivo hydrogel vascularization respectively. Pore sizes for hydrogel vascularization in vitro shall be middle ones and pore sizes for hydrogel vascularization in vivo shall be large ones.
Collapse
|
36
|
Ollauri-Ibáñez C, López-Novoa JM, Pericacho M. Endoglin-based biological therapy in the treatment of angiogenesis-dependent pathologies. Expert Opin Biol Ther 2017; 17:1053-1063. [PMID: 28656781 DOI: 10.1080/14712598.2017.1346607] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Alterations in the process of angiogenesis, either by excess or by defect, are present in different common pathologies. For this reason, great efforts are being made toward the development of pro- and anti-angiogenic therapies. Since endoglin levels are enhanced in tissues undergoing angiogenesis, and changes in its expression lead to alterations in vessel formation, endoglin has become an ideal target for these types of therapies. Areas covered: In this review, the role of endoglin in angiogenesis is summarized. In addition, the authors review pro- and anti-angiogenic therapies that are currently being used and new approaches that target endoglin. The article includes therapies that are both in preclinical and clinical development. Expert opinion: Endoglin is a very good target for anti-angiogenic therapy, as demonstrated by the positive results obtained with anti-endoglin antibodies. However, although endoglin in pro-angiogenic therapies has been successful in vitro, its use has not yet reached clinical settings. Moreover, the authors believe that establishing the exact role of endoglin in angiogenesis is essential and that this should be the next step in this field in the coming years.
Collapse
Affiliation(s)
- Claudia Ollauri-Ibáñez
- a Department of Physiology and Pharmacology , University of Salamanca , Salamanca , Spain.,b Biomedical Research Institute of Salamanca (IBSAL) , Salamanca , Spain
| | - José M López-Novoa
- a Department of Physiology and Pharmacology , University of Salamanca , Salamanca , Spain.,b Biomedical Research Institute of Salamanca (IBSAL) , Salamanca , Spain
| | - Miguel Pericacho
- a Department of Physiology and Pharmacology , University of Salamanca , Salamanca , Spain.,b Biomedical Research Institute of Salamanca (IBSAL) , Salamanca , Spain
| |
Collapse
|
37
|
Vinayagam V, Bobby Z, Habeebullah S, Chaturvedula L, Bharadwaj SK. Maternal and Cord Blood Plasma sEng and TGF-β1 in Patients with Hypertensive Disorders of Pregnancy: A Pilot Study in a South Indian Population. J Clin Diagn Res 2017; 11:QC32-QC34. [PMID: 28511458 DOI: 10.7860/jcdr/2017/22790.9600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 08/31/2016] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Hypertensive Disorders of Pregnancy (HDP) are one of the most widespread complications of pregnancy that affects both mother and foetus. It has been observed that in Preeclampsia, the release of soluble angiogenic factors from the ischemic placenta into maternal plasma plays a crucial role in the pathogenesis. AIM To assess the plasma Soluble Endoglin (sEng) and Transforming Growth Factor (TGF-β1) levels in various types of HDP and to correlate the levels of these markers with the pregnancy outcome. MATERIALS AND METHODS A total of 128 pregnant women were recruited and the study was carried out for a period of three years. Cord blood and maternal blood plasma levels of sEng and TGF-β1 were analysed by ELISA kits in Control Pregnant Women (CPW), Gestational Hypertension (GH), Early Onset Preeclampsia (EOPE), Late Onset Preeclampsia (LOPE), and Eclampsia (E) during third trimester. The Gestational Age (GA) at the time of delivery and Birth Weight (BW) of the baby also were also evaluated. RESULTS The circulating levels of maternal and cord blood sEng were significantly higher in EOPE and E compared to CPW and GH. However, the maternal and cord blood levels of TGF-β1 were significantly lower in LOPE and E when compared to CPW and GH. The GA and BW of the baby were found to be significantly lower in EOPE and E compared to CPW, GH and LOPE. Also, a negative correlation was observed between sEng levels with pregnancy outcome; GA and BW. And also, a positive correlation was found between TGF-β1 and pregnancy outcome. CONCLUSION A generalised angiogenic imbalance and poor birth outcomes were observed in HDP. There is a spectrum of biochemical derangements related to angiogenesis in GH, EOPE, LOPE and E.
Collapse
Affiliation(s)
- Vickneshwaran Vinayagam
- Ph D Scholar, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
| | - Zachariah Bobby
- Professor and Head, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
| | - Syed Habeebullah
- Professor, Department of Obstetrics and Gynaecology, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
| | - Latha Chaturvedula
- Professor, Department of Obstetrics and Gynaecology, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
| | - Shruthi K Bharadwaj
- Student (DM Neonatology), Department of Neonatology, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
| |
Collapse
|
38
|
Vascular Endothelial Growth Factor Isoform-B Stimulates Neurovascular Repair After Ischemic Stroke by Promoting the Function of Pericytes via Vascular Endothelial Growth Factor Receptor-1. Mol Neurobiol 2017; 55:3611-3626. [PMID: 28290152 DOI: 10.1007/s12035-017-0478-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/01/2017] [Indexed: 12/14/2022]
Abstract
Ischemic stroke triggers endogenous angiogenic mechanisms, which correlates with longer survival in patients. As such, promoting angiogenesis appears to be a promising approach. Experimental studies investigated mostly the potent angiogenic factor vascular endothelial growth factor isoform-A (VEGF-A). However, VEGF-A increases the risk of destabilizing the brain microvasculature, thus hindering the translation of its usage in clinics. An attractive alternative VEGF isoform-B (VEGF-B) was recently reported to act as a survival factor rather than a potent angiogenic factor. In this study, we investigated the therapeutic potential of VEGF-B in ischemic stroke using different in vivo and in vitro approaches. We showed that the delayed intranasal administration of VEGF-B reduced neuronal damage and inflammation. Unexpectedly, VEGF-B stimulated the formation of stable brain microvasculature within the injured region by promoting the interaction between endothelial cells and pericytes. Our data indicate that the effects of VEGF-B were mediated via its specific receptor VEGF receptor-1 (VEGFR-1) that is predominately expressed in brain pericytes. Importantly, VEGF-B promoted the survival of pericytes, and not brain endothelial cells, by inducing expression of the anti-apoptotic protein B-cell lymphoma 2 (Bcl-2) and the main protein involved in energy homeostasis AMP-activated protein kinase α (AMPKα). Moreover, we showed that VEGF-B stimulated the pericytic release of factors stimulating a "reparative angiogenesis" that does not compromise microvasculature stability. Our study unraveled hitherto unknown role of VEGF-B/VEGFR-1 signaling in regulating the function of pericytes. Furthermore, our findings suggest that brain microvasculature stabilization via VEGF-B constitutes a safe therapeutic approach for ischemic stroke.
Collapse
|
39
|
Functional significance of CD105-positive cells in papillary renal cell carcinoma. BMC Cancer 2017; 17:21. [PMID: 28056882 PMCID: PMC5217207 DOI: 10.1186/s12885-016-2985-7] [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] [Received: 06/13/2016] [Accepted: 12/06/2016] [Indexed: 12/20/2022] Open
Abstract
Background CD105 was postulated as a renal cell carcinoma (RCC) stem cell marker, and CD133 as a putative RCC progenitor. Hypoxia, a natural microenvironment that prevails in tumors, was also incorporated into the study, especially in terms of the promotion of hypothetical stem-like cell properties. Methods Within this study, we verify the existence of CD105+ and CD133+ populations in selected papillary subtype RCC (pRCC) cell lines. Both populations were analyzed for correlation with stem-like cell properties, such as stemness gene expression, and sphere and colony formation. For the preliminary analysis, several RCC cell lines were chosen (786-O, SMKT-R2, Caki-2, 796-P, ACHN, RCC6) and the control was human kidney cancer stem cells (HKCSC) and renal cells of embryonic origin (ASE-5063). Four cell lines were chosen for further investigation: Caki-2 (one of the highest numbers of CD105+ cells; primary origin), ACHN (a low number of CD105+ cells; metastatic origin), HKCSC (putative positive control), and ASE-5063 (additional control). Results In 769-P and RCC6, we could not detect a CD105+ population. Hypoxia variously affects pRCC cell growth, and mainly diminishes the stem-like properties of cells. Furthermore, we could not observe the correlation of CD105 and/or CD133 expression with the enhancement of stem-like properties. Conclusions Based on this analysis, CD105/CD133 cannot be validated as cancer stem cell markers of pRCC cell lines.
Collapse
|
40
|
Pang P, Abbott M, Chang SL, Abdi M, Chauhan N, Mistri M, Ghofrani J, Fucci QA, Walker C, Leonardi C, Grady S, Halim A, Hoffman R, Lu T, Cao H, Tullius SG, Malek S, Kumar S, Steele G, Kibel A, Freedman BS, Waikar SS, Siedlecki AM. Human vascular progenitor cells derived from renal arteries are endothelial-like and assist in the repair of injured renal capillary networks. Kidney Int 2017; 91:129-143. [PMID: 27692806 PMCID: PMC5179298 DOI: 10.1016/j.kint.2016.07.037] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 07/26/2016] [Accepted: 07/28/2016] [Indexed: 12/21/2022]
Abstract
Vascular progenitor cells show promise for the treatment of microvasculature endothelial injury. We investigated the function of renal artery progenitor cells derived from radical nephrectomy patients, in animal models of acute ischemic and hyperperfusion injuries. Present in human adventitia, CD34positive/CD105negative cells were clonal and expressed transcription factors Sox2/Oct4 as well as surface markers CXCR4 (CD184)/KDR(CD309) consistent with endothelial progenitor cells. Termed renal artery-derived vascular progenitor cells (RAPC), injected cells were associated with decreased serum creatinine after ischemia/reperfusion, reduced albuminuria after hyperperfusion, and improved blood flow in both models. A small population of RAPC integrated with the renal microvasculature following either experimental injury. At a cellular level, RAPC promoted local endothelial migration in co-culture. Profiling of RAPC microRNA identified high levels of miRNA 218; also found at high levels in exosomes isolated from RAPC conditioned media after cell contact for 24 hours. After hydrogen peroxide-induced endothelial injury, RAPC exosomes harbored Robo-1 transcript; a gene known to be regulated by mir218. Such exosomes enhanced endothelial cell migration in culture in the absence of RAPC. Thus, our work shows the feasibility of pre-emptive pro-angiogenic progenitor cell procurement from a targeted patient population and potential therapeutic use in the form of autologous cell transplantation.
Collapse
Affiliation(s)
- Paul Pang
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Molly Abbott
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Steven L Chang
- Urology Division, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Malyun Abdi
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nikita Chauhan
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Murti Mistri
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Joshua Ghofrani
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Quynh-Anh Fucci
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Colleen Walker
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Corey Leonardi
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Samuel Grady
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Arvin Halim
- Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ryan Hoffman
- Internal Medicine, Touro College of Osteopathic Medicine, New York, NY, USA
| | - Tzongshi Lu
- Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Huixia Cao
- Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefan G Tullius
- Transplant Surgery Division, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sayeed Malek
- Transplant Surgery Division, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sanjaya Kumar
- Transplant Surgery Division, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Graeme Steele
- Urology Division, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Adam Kibel
- Urology Division, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Sushrut S Waikar
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew M Siedlecki
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
41
|
Núñez-Gómez E, Pericacho M, Ollauri-Ibáñez C, Bernabéu C, López-Novoa JM. The role of endoglin in post-ischemic revascularization. Angiogenesis 2016; 20:1-24. [PMID: 27943030 DOI: 10.1007/s10456-016-9535-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
Abstract
Following arterial occlusion, blood vessels respond by forming a new network of functional capillaries (angiogenesis), by reorganizing preexisting capillaries through the recruitment of smooth muscle cells to generate new arteries (arteriogenesis) and by growing and remodeling preexisting collateral arterioles into physiologically relevant arteries (collateral development). All these processes result in the recovery of organ perfusion. The importance of endoglin in post-occlusion reperfusion is sustained by several observations: (1) endoglin expression is increased in vessels showing active angiogenesis/remodeling; (2) genetic endoglin haploinsufficiency in humans causes deficient angiogenesis; and (3) the reduction of endoglin expression by gene disruption or the administration of endoglin-neutralizing antibodies reduces angiogenesis and revascularization. However, the precise role of endoglin in the several processes associated with revascularization has not been completely elucidated and, in some cases, the function ascribed to endoglin by different authors is controversial. The purpose of this review is to organize in a critical way the information available for the role of endoglin in several phenomena (angiogenesis, arteriogenesis and collateral development) associated with post-ischemic revascularization.
Collapse
Affiliation(s)
- Elena Núñez-Gómez
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Miguel Pericacho
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Claudia Ollauri-Ibáñez
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Carmelo Bernabéu
- Centro de Investigaciones Biológicas, Spanish National Research Council (CIB, CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - José M López-Novoa
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain. .,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain.
| |
Collapse
|
42
|
Myszczyszyn A, Czarnecka AM, Matak D, Szymanski L, Lian F, Kornakiewicz A, Bartnik E, Kukwa W, Kieda C, Szczylik C. The Role of Hypoxia and Cancer Stem Cells in Renal Cell Carcinoma Pathogenesis. Stem Cell Rev Rep 2016. [PMID: 26210994 PMCID: PMC4653234 DOI: 10.1007/s12015-015-9611-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The cancer stem cell (CSC) model has recently been approached also in renal cell carcinoma (RCC). A few populations of putative renal tumor-initiating cells (TICs) were identified, but they are indifferently understood; however, the first and most thoroughly investigated are CD105-positive CSCs. The article presents a detailed comparison of all renal CSC-like populations identified by now as well as their presumable origin. Hypoxic activation of hypoxia-inducible factors (HIFs) contributes to tumor aggressiveness by multiple molecular pathways, including the governance of immature stem cell-like phenotype and related epithelial-to-mesenchymal transition (EMT)/de-differentiation, and, as a result, poor prognosis. Due to intrinsic von Hippel-Lindau protein (pVHL) loss of function, clear-cell RCC (ccRCC) develops unique pathological intra-cellular pseudo-hypoxic phenotype with a constant HIF activation, regardless of oxygen level. Despite satisfactory evidence concerning pseudo-hypoxia importance in RCC biology, its influence on putative renal CSC-like largely remains unknown. Thus, the article discusses a current knowledge of HIF-1α/2α signaling pathways in the promotion of undifferentiated tumor phenotype in general, including some experimental findings specific for pseudo-hypoxic ccRCC, mostly dependent from HIF-2α oncogenic functions. Existing gaps in understanding both putative renal CSCs and their potential connection with hypoxia need to be filled in order to propose breakthrough strategies for RCC treatment.
Collapse
Affiliation(s)
- Adam Myszczyszyn
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| | - Anna M Czarnecka
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.
| | - Damian Matak
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.,School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Lukasz Szymanski
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.,Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Fei Lian
- Emory School of Medicine, Atlanta, GA, USA
| | - Anna Kornakiewicz
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.,School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland.,Department of General Surgery and Transplantology, Medical University of Warsaw, Warsaw, Poland
| | - Ewa Bartnik
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.,Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Wojciech Kukwa
- Department of Otolaryngology, Czerniakowski Hospital, Medical University of Warsaw, Warsaw, Poland
| | - Claudine Kieda
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Orléans, France
| | - Cezary Szczylik
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| |
Collapse
|
43
|
Watson EC, Whitehead L, Adams RH, Dewson G, Coultas L. Endothelial cell survival during angiogenesis requires the pro-survival protein MCL1. Cell Death Differ 2016; 23:1371-9. [PMID: 26943318 DOI: 10.1038/cdd.2016.20] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 01/07/2016] [Accepted: 01/29/2016] [Indexed: 01/31/2023] Open
Abstract
Angiogenesis is essential to match the size of blood vessel networks to the metabolic demands of growing tissues. While many genes and pathways necessary for regulating angiogenesis have been identified, those responsible for endothelial cell (EC) survival during angiogenesis remain largely unknown. We have investigated the in vivo role of myeloid cell leukemia 1 (MCL1), a pro-survival member of the BCL2 family, in EC survival during angiogenesis. EC-specific deletion of Mcl1 resulted in a dose-dependent increase in EC apoptosis in the angiogenic vasculature and a corresponding decline in vessel density. Our results suggest this apoptosis was independent of the BH3-only protein BIM. Despite the known link between apoptosis and blood vessel regression, this was not the cause of reduced vessel density observed in the absence of endothelial MCL1. Rather, the reduction in vessel density was linked to ectopic apoptosis in regions of the angiogenic vasculature where EC proliferation and new vessel growth occurs. We have therefore identified MCL1 as an essential survival factor for ECs that is required for blood vessel production during angiogenesis.
Collapse
Affiliation(s)
- E C Watson
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, 1G Royal Parade, VIC, Australia
| | - L Whitehead
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, VIC, Australia.,Systems Biology and Personalised Medicine Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, Australia
| | - R H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Roentgenstrasse 20, Münster D, Germany.,Faculty of Medicine, University of Münster, Münster D, Germany
| | - G Dewson
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, VIC, Australia.,Cell Signalling and Cell Death Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, Australia
| | - L Coultas
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, 1G Royal Parade, VIC, Australia
| |
Collapse
|
44
|
Belik D, Tsang H, Wharton J, Howard L, Bernabeu C, Wojciak-Stothard B. Endothelium-derived microparticles from chronically thromboembolic pulmonary hypertensive patients facilitate endothelial angiogenesis. J Biomed Sci 2016; 23:4. [PMID: 26786759 PMCID: PMC4717540 DOI: 10.1186/s12929-016-0224-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 01/11/2016] [Indexed: 12/26/2022] Open
Abstract
Background Increased circulating levels of endoglin+ endothelial microparticles (EMPs) have been identified in several cardiovascular disorders, related to severity. Endoglin is an auxilary receptor for transforming growth factor β (TGF-β) important in the regulation of vascular structure. Results We quantified the number of microparticles in plasma of six patients with chronic thromboembolic pulmonary hypertension (CTEPH) and age- and sex-matched pulmonary embolic (PE) and healthy controls and investigated the role of microparticle endoglin in the regulation of pulmonary endothelial function in vitro. Results show significantly increased levels of endoglin+ EMPs in CTEPH plasma, compared to healthy and disease controls. Co-culture of human pulmonary endothelial cells with CTEPH microparticles increased intracellular levels of endoglin and enhanced TGF-β-induced angiogenesis and Smad1,5,8 phosphorylation in cells, without affecting BMPRII expression. In an in vitro model, we generated endothelium-derived MPs with enforced membrane localization of endoglin. Co-culture of these MPs with endothelial cells increased cellular endoglin content, improved cell survival and stimulated angiogenesis in a manner similar to the effects induced by overexpressed protein. Conclusions Increased generation of endoglin+ EMPs in CTEPH is likely to represent a protective mechanism supporting endothelial cell survival and angiogenesis, set to counteract the effects of vascular occlusion and endothelial damage. Electronic supplementary material The online version of this article (doi:10.1186/s12929-016-0224-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daria Belik
- Centre for Pharmacology and Therapeutics, Department of Medicine, Imperial College London, London, UK
| | - Hilda Tsang
- Centre for Pharmacology and Therapeutics, Department of Medicine, Imperial College London, London, UK
| | - John Wharton
- Centre for Pharmacology and Therapeutics, Department of Medicine, Imperial College London, London, UK
| | - Luke Howard
- National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust, London, UK
| | - Carmelo Bernabeu
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain
| | - Beata Wojciak-Stothard
- Centre for Pharmacology and Therapeutics, Department of Medicine, Imperial College London, London, UK.
| |
Collapse
|
45
|
Neuroprotective Effects of Acetyl-L-Carnitine Against Oxygen-Glucose Deprivation-Induced Neural Stem Cell Death. Mol Neurobiol 2015; 53:6644-6652. [DOI: 10.1007/s12035-015-9563-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 11/29/2015] [Indexed: 12/22/2022]
|
46
|
Hardy B, Raiter A. GRP78 expression beyond cellular stress: A biomarker for tumor manipulation. World J Immunol 2015; 5:78-85. [DOI: 10.5411/wji.v5.i2.78] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 01/14/2015] [Accepted: 06/08/2015] [Indexed: 02/05/2023] Open
Abstract
Physiological stress takes place in the endoplasmic reticulum (ER) of cells where activation and up-regulation of genes and proteins are primarily induced to enhance pro-survival mechanisms such as the unfolded protein response (UPR). A dominant protein in the UPR response is the heat shock GRP78 protein. Although GRP78 is primarily located in the ER, under certain conditions it is transported to the cell surface, where it acts as a receptor inducing pathways of cell signaling such as proliferation or apoptosis. In the prolonged chronic stress transportation of the GRP78 from the ER to the cell membrane is a major event where in addition to the presentation of the GRP78 as a receptor to various ligands, it also marks the cells that will proceed to apoptotic pathways. In the normal cell that under stress acquires cell surface GRP78 and in the tumor cell that already presents cell surface GRP78, cell surface GRP78 is an apoptotic flag. The internalization of GRP78 from the cell surface in normal cells by ligands such as peptides will enhance cell survival and alleviate cardiovascular ischemic diseases. The absence of cell surface GRP78 in the tumor cells portends proliferative and metastatic tumors. Pharmacological induction of cell surface GRP78 will induce the process of apoptosis and might be used as a therapeutic modality for cancer treatment.
Collapse
|
47
|
Jia ZZ, Gu HM, Zhou XJ, Shi JL, Li MD, Zhou GF, Wu XH. The assessment of immature microvascular density in brain gliomas with dynamic contrast-enhanced magnetic resonance imaging. Eur J Radiol 2015; 84:1805-9. [PMID: 26066470 DOI: 10.1016/j.ejrad.2015.05.035] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 05/19/2015] [Accepted: 05/30/2015] [Indexed: 10/23/2022]
Abstract
PURPOSE This study was designed to quantitatively evaluate the immature microvascular density (MVD) of brain gliomas using the volume transfer constant (K(trans)) and volume of extravascular extracellular space per unit volume of tissue (Ve) from dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) noninvasively. MATERIALS AND METHODS Fifty-seven patients (35 males, 22 females; age range, 14-70, mean age 46±12 years old) with brain glioma were included in this study. The maximal values of K(trans) and Ve of all patients with brain glioma (grade II 24, III 7 and IV 26) were obtained. The CD105-microvascular density (CD105-MVD) of each tumor was measured in surgical specimen. The differences of K(trans), Ve and CD105-MVD between the different grades of gliomas were analyzed using the Mann-Whitney U-test. The Pearman correlation coefficient was determined between K(trans), Ve and CD105-MVD. A P-value of less than 0.05 was considered statistically significant. RESULTS The differences in K(trans), Ve and CD105-MVD were statistically significant between low-grade glioma (LGG) and high-grade glioma (HGG) (P=0.001, P<0.001, P<0.001). The K(trans), Ve and CD105-MVD of grade II were significantly lower than those of grade III and IV. K(trans) and Ve were positively correlated with CD105-MVD in HGG (P<0.001, P<0.001). CONCLUSIONS Our results suggest DCE-MRI plays an important part in noninvasively evaluating the immature MVD of brain gliomas.
Collapse
Affiliation(s)
- Zhong Zheng Jia
- Department of Radiology, Affiliated Hospital of Nantong University, 20 Xisi Road Nantong, 226001 Jiangsu, PR China.
| | - Hong Mei Gu
- Department of Radiology, Affiliated Hospital of Nantong University, 20 Xisi Road Nantong, 226001 Jiangsu, PR China.
| | - Xue Jun Zhou
- Department of Radiology, Affiliated Hospital of Nantong University, 20 Xisi Road Nantong, 226001 Jiangsu, PR China.
| | - Jin Long Shi
- Department of Neurosurgery, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong 226001, Jiangsu, PR China.
| | - Min Da Li
- Department of Radiology, Affiliated Hospital of Nantong University, 20 Xisi Road Nantong, 226001 Jiangsu, PR China.
| | - Guo Feng Zhou
- Department of Radiology, Affiliated Hospital of Nantong University, 20 Xisi Road Nantong, 226001 Jiangsu, PR China.
| | - Xian Hua Wu
- Department of Radiology, Affiliated Hospital of Nantong University, 20 Xisi Road Nantong, 226001 Jiangsu, PR China.
| |
Collapse
|
48
|
Papel de endoglina en los eventos fisiológicos involucrados en la revascularización postisquémica. ANGIOLOGIA 2015. [DOI: 10.1016/j.angio.2014.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
49
|
McKittrick CM, Lawrence CE, Carswell HVO. Mast cells promote blood brain barrier breakdown and neutrophil infiltration in a mouse model of focal cerebral ischemia. J Cereb Blood Flow Metab 2015; 35:638-47. [PMID: 25564235 PMCID: PMC4420882 DOI: 10.1038/jcbfm.2014.239] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 11/13/2014] [Accepted: 11/25/2014] [Indexed: 01/30/2023]
Abstract
Blood brain barrier (BBB) breakdown and neuroinflammation are key events in ischemic stroke morbidity and mortality. The present study investigated the effects of mast cell deficiency and stabilization on BBB breakdown and neutrophil infiltration in mice after transient middle cerebral artery occlusion (tMCAo). Adult male C57BL6/J wild type (WT) and mast cell-deficient (C57BL6/J Kit(Wsh/Wsh) (Wsh)) mice underwent tMCAo and BBB breakdown, brain edema and neutrophil infiltration were examined after 4 hours of reperfusion. Blood brain barrier breakdown, brain edema, and neutrophil infiltration were significantly reduced in Wsh versus WT mice (P<0.05). These results were reproduced pharmacologically using mast cell stabilizer, cromoglycate. Wild-type mice administered cromoglycate intraventricularly exhibited reduced BBB breakdown, brain edema, and neutrophil infiltration versus vehicle (P<0.05). There was no effect of cromoglycate versus vehicle in Wsh mice, validating specificity of cromoglycate on brain mast cells. Proteomic analysis in Wsh versus WT indicated that effects may be via expression of endoglin, endothelin-1, and matrix metalloproteinase-9. Using an in vivo model of mast cell deficiency, this is the first study showing that mast cells promote BBB breakdown in focal ischemia in mice, and opens up future opportunities for using mice to identify specific mechanisms of mast cell-related BBB injury.
Collapse
Affiliation(s)
- Craig M McKittrick
- Strathclyde Institute of Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, UK
| | - Catherine E Lawrence
- Strathclyde Institute of Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, UK
| | - Hilary V O Carswell
- Strathclyde Institute of Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, UK
| |
Collapse
|
50
|
Abstract
The paper gives the data available in the literature on vascularization of hepatocellular carcinoma (HCC). Sinusoidal capillarization and unpaired arteries are shown to play an important role in the development and progression of HCC. The density of microvessels detected by immunohistochemical techniques is a morphological indicator of the degree of angiogenic processes. Higher-grade HCC is followed by changes in its vascularization and concurrent with a progressive increase in the proportion of blood entering along the hepatic artery. The morphological indicators of microvessel density are recommended to use as addi- tional criteria for determining the prognosis of the disease, designing targeted anti-angiogenic drugs, and evaluating the efficiency of performed therapy.
Collapse
Affiliation(s)
- U N Tumanova
- Academician V.I. Kulakov Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of Russia, Moscow
| | - A I Shchegolev
- N.I. Pirogov Russian National Research Medical University, Ministry of Health of Russia, Moscow, Russian Federation
| |
Collapse
|