151
|
Trautmann F, Cojoc M, Kurth I, Melin N, Bouchez LC, Dubrovska A, Peitzsch C. CXCR4 as biomarker for radioresistant cancer stem cells. Int J Radiat Biol 2014; 90:687-99. [PMID: 24650104 DOI: 10.3109/09553002.2014.906766] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE Radioresistance of cancer cells remains a fundamental barrier for maximum efficient radiotherapy. Tumor heterogeneity and the existence of distinct cell subpopulations exhibiting different genotypes and biological behaviors raise difficulties to eradicate all tumorigenic cells. Recent evidence indicates that a distinct population of tumor cells, called cancer stem cells (CSC), is involved in tumor initiation and recurrence and is a putative cause of tumor radioresistance. There is an urgent need to identify the intrinsic molecular mechanisms regulating the generation and maintenance of resistance to radiotherapy, especially within the CSC subset. The chemokine C-X-C motif receptor 4 (CXCR4) has been found to be a prognostic marker in various types of cancer, being involved in chemotaxis, stemness and drug resistance. The interaction of CXCR4 with its ligand, the chemokine C-X-C motif ligand 12 (CXCL12), plays an important role in modulating the tumor microenvironment, angiogenesis and CSC niche. Moreover, the therapeutic inhibition of the CXCR4/CXCL12 signaling pathway is sensitizing the malignant cells to conventional anti-cancer therapy. CONTENT Within this review we are summarizing the role of the CXCR4/CXCL12 axis in the modulation of CSC properties, the regulation of the tumor microenvironment in response to irradiation, therapy resistance and tumor relapse. CONCLUSION In light of recent findings, the inhibition of the CXCR4/CXCL12 signaling pathway is a promising therapeutic option to refine radiotherapy.
Collapse
Affiliation(s)
- Franziska Trautmann
- OncoRay - National Center for Radiation Research in Oncology, Medizinische Fakultät Carl Gustav Carus der Technischen Universität and Helmholtz Zentrum Rossendorf , Dresden
| | | | | | | | | | | | | |
Collapse
|
152
|
Merino JJ, Bellver-Landete V, Oset-Gasque MJ, Cubelos B. CXCR4/CXCR7 Molecular Involvement in Neuronal and Neural Progenitor Migration: Focus in CNS Repair. J Cell Physiol 2014; 230:27-42. [DOI: 10.1002/jcp.24695] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 06/03/2014] [Indexed: 12/13/2022]
Affiliation(s)
- José Joaquín Merino
- Biochemistry and Molecular Biology Dept II; Universidad Complutense de Madrid (UCM); Madrid Spain
- Instituto de Investigación; Neuroquímica (IUIN), UCM; Madrid Spain
| | - Victor Bellver-Landete
- Biochemistry and Molecular Biology Dept II; Universidad Complutense de Madrid (UCM); Madrid Spain
| | - María Jesús Oset-Gasque
- Biochemistry and Molecular Biology Dept II; Universidad Complutense de Madrid (UCM); Madrid Spain
- Instituto de Investigación; Neuroquímica (IUIN), UCM; Madrid Spain
| | - Beatriz Cubelos
- Departamento de Biología Molecular; Centro de Biología Molecular Severo Ochoa (CBMSO); Universidad Autónoma de Madrid; Madrid Spain
| |
Collapse
|
153
|
Cherry AE, Stella N. G protein-coupled receptors as oncogenic signals in glioma: emerging therapeutic avenues. Neuroscience 2014; 278:222-36. [PMID: 25158675 DOI: 10.1016/j.neuroscience.2014.08.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 08/14/2014] [Accepted: 08/15/2014] [Indexed: 12/20/2022]
Abstract
Gliomas are the most common malignant intracranial tumors. Newly developed targeted therapies for these cancers aim to inhibit oncogenic signals, many of which emanate from receptor tyrosine kinases, including the epidermal growth factor receptor (EGFR) and the vascular endothelial growth factor receptor (VEGFR). Unfortunately, the first-generation treatments targeting these oncogenic signals provide little survival benefit in both mouse xenograft models and human patients. The search for new treatment options has uncovered several G protein-coupled receptor (GPCR) candidates and generated a growing interest in this class of proteins as alternative therapeutic targets for the treatment of various cancers, including glioblastoma multiforme (GBM). GPCRs constitute a large family of membrane receptors that influence oncogenic pathways through canonical and non-canonical signaling. Accordingly, evidence indicates that GPCRs display a unique ability to crosstalk with receptor tyrosine kinases, making them important molecular components controlling tumorigenesis. This review summarizes the current research on GPCR functionality in gliomas and explores the potential of modulating these receptors to treat this devastating disease.
Collapse
Affiliation(s)
- A E Cherry
- Department of Pharmacology, University of Washington, 1959 NE Pacific Street, BB1538, Health Sciences Building, Seattle, WA 98195, United States.
| | - N Stella
- Department of Pharmacology, University of Washington, 1959 NE Pacific Street, BB1538, Health Sciences Building, Seattle, WA 98195, United States; Department of Psychiatry & Behavioral Sciences, University of Washington, 1959 NE Pacific Street, BB1538, Health Sciences Building, Seattle, WA 98195, United States.
| |
Collapse
|
154
|
Serum protein profiling reveals baseline and pharmacodynamic biomarker signatures associated with clinical outcome in mCRC patients treated with chemotherapy ± cediranib. Br J Cancer 2014; 111:1590-604. [PMID: 25121956 PMCID: PMC4200086 DOI: 10.1038/bjc.2014.436] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/10/2014] [Accepted: 07/09/2014] [Indexed: 02/07/2023] Open
Abstract
Background: This study evaluated soluble serum proteins as biomarkers to subset patients with metastatic colorectal cancer (mCRC) treated with chemotherapy±cediranib, a vascular endothelial growth factor (VEGF) signalling inhibitor (VEGFi). Exploring biomarkers at pre- and on-treatment may identify patient subgroups showing clinical benefit on cediranib combination. Methods: Two hundred and seven serum proteins were analysed in 588 mCRC patients at pre- and on-treatment with chemotherapy (FOLFOX/CAPOX)±cediranib 20 mg. Patients were enrolled in the phase III trial HORIZON II. We correlated baseline biomarker signatures and pharmacodynamic (PD) biomarkers with PFS and OS. Results: We identified a baseline signature (BS) of 47 biomarkers that included VEGFA, VEGFD, VEGFR2, VEGFR3 and TIE-2, which defined two distinct subgroups of patients. Patients treated with chemotherapy plus cediranib who had ‘high' BS had shorter PFS (HR=1.82, P=0.003) than patients with ‘low' BS. This BS did not correlate with PFS of the patients treated with chemotherapy plus placebo. In addition, we identified a profile of 16 PD proteins on treatment associated with PFS (HR=0.58, P<0.001) and OS (HR=0.52, P<0.001) in patients treated with chemotherapy plus cediranib. This PD profile did not correlate with PFS and OS in patients treated with chemotherapy plus placebo. Conclusions: Serum proteins may represent relevant biomarkers to predict the outcome of patients treated with VEGFi-based therapies. We report a BS and PD biomarkers that may identify mCRC patients showing increased benefit of combining cediranib with chemotherapy. These exploratory findings need to be validated in future prospective studies.
Collapse
|
155
|
von Hundelshausen P, Schmitt MMN. Platelets and their chemokines in atherosclerosis-clinical applications. Front Physiol 2014; 5:294. [PMID: 25152735 PMCID: PMC4126210 DOI: 10.3389/fphys.2014.00294] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 07/22/2014] [Indexed: 12/22/2022] Open
Abstract
The concept of platelets as important players in the process of atherogenesis has become increasingly accepted due to accumulating experimental and clinical evidence. Despite the progress in understanding the molecular details of atherosclerosis, particularly by using animal models, the inflammatory and thrombotic roles of activated platelet s especially in the human system remain difficult to dissect, as often only the complications of atherosclerosis, i.e., stroke and myocardial infarction are definable but not the plaque burden. Platelet indices including platelet count and mean platelet volume (MPV) and soluble mediators released by activated platelets are associated with atherosclerosis. The chemokine CXCL4 has multiple atherogenic activities, e.g., altering the differentiation of T cells and macrophages by inhibiting neutrophil and monocyte apoptosis and by increasing the uptake of oxLDL and synergizing with CCL5. CCL5 is released and deposited on endothelium by activated platelets thereby triggering atherogenic monocyte recruitment, which can be attenuated by blocking the corresponding chemokine receptor CCR5. Atheroprotective and plaque stabilizing properties are attributed to CXCL12, which plays an important role in regenerative processes by attracting progenitor cells. Its release from luminal attached platelets accelerates endothelial healing after injury. Platelet surface molecules GPIIb/IIIa, GP1bα, P-selectin, JAM-A and the CD40/CD40L dyade are crucially involved in the interaction with endothelial cells, leukocytes and matrix molecules affecting atherogenesis. Beyond the effects on the arterial inflammatory infiltrate, platelets affect cholesterol metabolism by binding, modifying and endocytosing LDL particles via their scavenger receptors and contribute to the formation of lipid laden macrophages. Current medical therapies for the prevention of atherosclerotic therapies enable the elucidation of mechanisms linking platelets to inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Philipp von Hundelshausen
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University of Munich Munich, Germany ; German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance Munich, Germany
| | - Martin M N Schmitt
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University of Munich Munich, Germany
| |
Collapse
|
156
|
Ultrasound-triggered disruption and self-healing of reversibly cross-linked hydrogels for drug delivery and enhanced chemotherapy. Proc Natl Acad Sci U S A 2014; 111:9762-7. [PMID: 24961369 DOI: 10.1073/pnas.1405469111] [Citation(s) in RCA: 288] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Biological systems are exquisitely sensitive to the location and timing of physiologic cues and drugs. This spatiotemporal sensitivity presents opportunities for developing new therapeutic approaches. Polymer-based delivery systems are used extensively for attaining localized, sustained release of bioactive molecules. However, these devices typically are designed to achieve a constant rate of release. We hypothesized that it would be possible to create digital drug release, which could be accelerated and then switched back off, on demand, by applying ultrasound to disrupt ionically cross-linked hydrogels. We demonstrated that ultrasound does not permanently damage these materials but enables nearly digital release of small molecules, proteins, and condensed oligonucleotides. Parallel in vitro studies demonstrated that the concept of applying temporally short, high-dose "bursts" of drug exposure could be applied to enhance the toxicity of mitoxantrone toward breast cancer cells. We thus used the hydrogel system in vivo to treat xenograft tumors with mitoxantrone, and found that daily ultrasound-stimulated drug release substantially reduced tumor growth compared with sustained drug release alone. This approach of digital drug release likely will be applicable to a broad variety of polymers and bioactive molecules, and is a potentially useful tool for studying how the timing of factor delivery controls cell fate in vivo.
Collapse
|
157
|
Tsaknis G, Tsangaris I, Ikonomidis I, Tsantes A. Clinical usefulness of novel serum and imaging biomarkers in risk stratification of patients with stable angina. DISEASE MARKERS 2014; 2014:831364. [PMID: 25045198 PMCID: PMC4087263 DOI: 10.1155/2014/831364] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/28/2014] [Accepted: 05/22/2014] [Indexed: 01/17/2023]
Abstract
Inflammatory mediators appear to be the most intriguing yet confusing subject, regarding the management of patients with acute coronary syndromes (ACS). The current inflammatory concept of atherosclerotic coronary artery disease (CAD) led many investigators to concentrate on systemic markers of inflammation, as well as imaging techniques, which may be helpful in risk stratification and prognosis assessment for cardiovascular events. In this review, we try to depict many of the recently studied markers regarding stable angina (SA), their clinical usefulness, and possible future applications in the field.
Collapse
Affiliation(s)
- George Tsaknis
- Department of Respiratory Medicine, Glenfield Hospital, University Hospitals of Leicester, Groby Road, Leicester LE3 9QP, UK
- Second Department of Critical Care Medicine, Attikon University Hospital, University of Athens, Medical School, 1 Rimini Street, Haidari, 12462 Athens, Greece
| | - Iraklis Tsangaris
- Department of Respiratory Medicine, Glenfield Hospital, University Hospitals of Leicester, Groby Road, Leicester LE3 9QP, UK
| | - Ignatios Ikonomidis
- Second Department of Cardiology, Attikon University Hospital, University of Athens, Medical School, 1 Rimini Street, Haidari, 12462 Athens, Greece
| | - Argirios Tsantes
- Laboratory of Haematology and Blood Bank Unit, Attikon University Hospital, University of Athens, Medical School, 1 Rimini Street, Haidari, 12462 Athens, Greece
| |
Collapse
|
158
|
Würth R, Bajetto A, Harrison JK, Barbieri F, Florio T. CXCL12 modulation of CXCR4 and CXCR7 activity in human glioblastoma stem-like cells and regulation of the tumor microenvironment. Front Cell Neurosci 2014; 8:144. [PMID: 24904289 PMCID: PMC4036438 DOI: 10.3389/fncel.2014.00144] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 05/06/2014] [Indexed: 12/16/2022] Open
Abstract
Chemokines are crucial autocrine and paracrine players in tumor development. In particular, CXCL12, through its receptors CXCR4 and CXCR7, affects tumor progression by controlling cancer cell survival, proliferation and migration, and, indirectly, via angiogenesis or recruiting immune cells. Glioblastoma (GBM) is the most prevalent primary malignant brain tumor in adults and despite current multimodal therapies it remains almost incurable. The aggressive and recurrent phenotype of GBM is ascribed to high growth rate, invasiveness to normal brain, marked angiogenesis, ability to escape the immune system and resistance to standard of care therapies. Tumor molecular and cellular heterogeneity severely hinders GBM therapeutic improvement. In particular, a subpopulation of chemo- and radio-therapy resistant tumorigenic cancer stem-like cells (CSCs) is believed to be the main responsible for tumor cell dissemination to the brain. GBM cells display heterogeneous expression levels of CXCR4 and CXCR7 that are overexpressed in CSCs, representing a molecular correlate for the invasive potential of GBM. The microenvironment contribution in GBM development is increasingly emphasized. An interplay exists between CSCs, differentiated GBM cells, and the microenvironment, mainly through secreted chemokines (e.g., CXCL12) causing recruitment of fibroblasts, endothelial, mesenchymal and inflammatory cells to the tumor, via specific receptors such as CXCR4. This review covers recent developments on the role of CXCL12/CXCR4-CXCR7 networks in GBM progression and the potential translational impact of their targeting. The biological and molecular understanding of the heterogeneous GBM cell behavior, phenotype and signaling is still limited. Progress in the identification of chemokine-dependent mechanisms that affect GBM cell survival, trafficking and chemo-attractive functions, opens new perspectives for development of more specific therapeutic approaches that include chemokine-based drugs.
Collapse
Affiliation(s)
- Roberto Würth
- Sezione di Farmacologia, Dipartimento di Medicina Interna, University of Genova Genova, Italy ; Centro di Eccellenza per la Ricerca Biomedica, University of Genova Genova, Italy
| | - Adriana Bajetto
- Sezione di Farmacologia, Dipartimento di Medicina Interna, University of Genova Genova, Italy ; Centro di Eccellenza per la Ricerca Biomedica, University of Genova Genova, Italy
| | - Jeffrey K Harrison
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida Gainesville, FL, USA
| | - Federica Barbieri
- Sezione di Farmacologia, Dipartimento di Medicina Interna, University of Genova Genova, Italy ; Centro di Eccellenza per la Ricerca Biomedica, University of Genova Genova, Italy
| | - Tullio Florio
- Sezione di Farmacologia, Dipartimento di Medicina Interna, University of Genova Genova, Italy ; Centro di Eccellenza per la Ricerca Biomedica, University of Genova Genova, Italy
| |
Collapse
|
159
|
Martin BJ. Inhibiting vasculogenesis after radiation: a new paradigm to improve local control by radiotherapy. Semin Radiat Oncol 2014; 23:281-7. [PMID: 24012342 DOI: 10.1016/j.semradonc.2013.05.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Tumors are supported by blood vessels, and it has long been debated whether their response to irradiation is affected by radiation damage to the vasculature. We have shown in preclinical models that, indeed, radiation is damaging to the tumor vasculature and strongly inhibits tumor angiogenesis. However, the vasculature can recover by colonization from circulating cells, primarily proangiogenenic CD11b+ monocytes or macrophages from the bone marrow. This secondary pathway of blood vessel formation, known as vasculogenesis, thus acts to restore the tumor vasculature and allows the tumor to recur following radiation. The stimulus for the influx of these CD11b+ cells into tumors following irradiation is the increased levels of hypoxia-inducible factor-1 in the tumor due to induced tumor hypoxia secondary to blood vessel loss. This increases tumor levels of the chemokine stromal cell-derived factor-1, which has chemokine receptors CXCR4 and CXCR7 on monocytes and endothelial cells thereby capturing these cells in the tumors. The increase in CD11b+ monocytes in tumors following irradiation can be prevented using antibodies or small molecules that inhibit hypoxia-inducible factor-1 or the interaction of stromal cell-derived factor-1 with its receptors. We show that the effect of inhibiting these chemokine-chemokine receptor interactions is a marked increase in the radiation response of transplanted or chemically induced tumors in mice and rats. This strategy of inhibiting vasculogenesis following tumor irradiation is a new paradigm in radiotherapy and suggests that higher levels of local control of tumors in several sites would be achievable with this strategy.
Collapse
Affiliation(s)
- Brown J Martin
- Department of Radiation Oncology, Stanford University, Stanford, CA.
| |
Collapse
|
160
|
Oversulfated chondroitin sulfate binds to chemokines and inhibits stromal cell-derived factor-1 mediated signaling in activated T cells. PLoS One 2014; 9:e94402. [PMID: 24718687 PMCID: PMC3981801 DOI: 10.1371/journal.pone.0094402] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 03/15/2014] [Indexed: 12/20/2022] Open
Abstract
Oversulfated chondroitin sulfate (OSCS), a member of the glycosaminoglycan (GAG) family, was a contaminant in heparin that was linked to the 2008 heparin adverse events in the US. Because of its highly negative charge, OSCS can interact with many components of the contact and immune systems. We have previously demonstrated that OSCS inhibited the complement classical pathway by binding C1 inhibitor and potentiating its interaction with C1s. In the present study, by using surface plasmon resonance, we found OSCS interacts with T cell chemokines that can impact adaptive immunity. The binding of OSCS to stromal cell-derived factor-1 (SDF-1) chemokines, SDF-1α and SDF-1β, caused a significant change in the secondary structures of these chemokines as detected by far-ultraviolet circular dichroism spectra analysis. Functionally, OSCS binding profoundly inhibited SDF-1-induced calcium mobilization and T cell chemotaxis. Imaging flow cytometry revealed T cell morphological changes mediated by SDF-1α were completely blocked by OSCS. We conclude that the OSCS, a past contaminant in heparin, has broad interactions with the components of the human immune system beyond the contact and complement systems, and that may explain, in part, prior OSCS-related adverse events, while suggesting potentially useful therapeutic applications for related GAGs in the control of inflammation.
Collapse
|
161
|
Moschetta M, Mishima Y, Sahin I, Manier S, Glavey S, Vacca A, Roccaro AM, Ghobrial IM. Role of endothelial progenitor cells in cancer progression. Biochim Biophys Acta Rev Cancer 2014; 1846:26-39. [PMID: 24709008 DOI: 10.1016/j.bbcan.2014.03.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/19/2014] [Accepted: 03/27/2014] [Indexed: 12/12/2022]
Abstract
Tumor-associated neovasculature is a critical therapeutic target; however, despite significant progress made in the clinical efficacy of anti-vessel drugs, the effect of these agents remains transient: over time, most patients develop resistance, which inevitably leads to tumor progression. To develop more effective treatments, it is imperative that we better understand the mechanisms involved in tumor vessel formation, how they participate to the tumor progression and metastasis, and the best way to target them. Several mechanisms contribute to the formation of tumor-associated vasculature: i) neoangiogenesis; ii) vascular co-option; iii) mosaicism; iv) vasculogenic mimicry, and v) postnatal vasculogenesis. These mechanisms can also play a role in the development of resistance to anti-angiogenic drugs, and could serve as targets for designing new anti-vascular molecules to treat solid as well as hematological malignancies. Bone marrow-derived endothelial progenitor cell (EPC)-mediated vasculogenesis represents an important new target, especially at the early stage of tumor growth (when EPCs are critical for promoting the "angiogenic switch"), and during metastasis, when EPCs promote the transition from micro- to macro-metastases. In hematologic malignancies, the EPC population could be related to the neoplastic clone, and both may share a common ontogeny. Thus, characterization of tumor-associated EPCs in blood cancers may provide clues for more specific anti-vascular therapy that has both direct and indirect anti-tumor effects. Here, we review the role of vasculogenesis, mediated by bone marrow-derived EPCs, in the progression of cancer, with a particular focus on the role of these cells in promoting progression of hematological malignancies.
Collapse
Affiliation(s)
- Michele Moschetta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; University of Bari Medical School, Department of Biomedical Sciences and Human Oncology (DIMO), Section of Internal Medicine and Clinical Oncology, Bari, Italy
| | - Yuji Mishima
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ilyas Sahin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Salomon Manier
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Siobhan Glavey
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Angelo Vacca
- University of Bari Medical School, Department of Biomedical Sciences and Human Oncology (DIMO), Section of Internal Medicine and Clinical Oncology, Bari, Italy
| | - Aldo M Roccaro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
162
|
Gong Y, Zhao Y, Li Y, Fan Y, Hoover-Plow J. Plasminogen regulates cardiac repair after myocardial infarction through its noncanonical function in stem cell homing to the infarcted heart. J Am Coll Cardiol 2014; 63:2862-72. [PMID: 24681141 DOI: 10.1016/j.jacc.2013.11.070] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 11/05/2013] [Accepted: 11/26/2013] [Indexed: 10/25/2022]
Abstract
OBJECTIVES The purpose of this study was to investigate the role of plasminogen (Plg) in stem cell-mediated cardiac repair and regeneration after myocardial infarction (MI). BACKGROUND An MI induces irreversible tissue damage, eventually leading to heart failure. Bone marrow (BM)-derived stem cells promote tissue repair and regeneration after MI. Thrombolytic treatment with Plg activators significantly improves the clinical outcome in MI by restoring cardiac perfusion. However, the role of Plg in stem cell-mediated cardiac repair remains unclear. METHODS An MI was induced in Plg-deficient (Plg(-/-)) and wild-type (Plg(+/+)) mice by ligation of the left anterior descending coronary artery. Stem cells were visualized by in vivo tracking of green fluorescent protein (GFP)-expressing BM cells after BM transplantation. Cardiac function, stem cell homing, and signaling pathways downstream of Plg were examined. RESULTS Granulocyte colony-stimulating factor, a stem cell mobilizer, significantly promoted BM-derived stem cell (GFP(+)c-kit(+) cell) recruitment into the infarcted heart and stem cell-mediated cardiac repair in Plg(+/+) mice. However, Plg deficiency markedly inhibited stem cell homing and cardiac repair, suggesting that Plg is critical for stem cell-mediated cardiac repair. Moreover, Plg regulated C-X-C chemokine receptor type 4 (CXCR4) expression in stem cells in vivo and in vitro through matrix metalloproteinase-9. Lentiviral reconstitution of CXCR4 expression in BM cells successfully rescued stem cell homing to the infarcted heart in Plg-deficient mice, indicating that CXCR4 has a critical role in Plg-mediated stem cell homing after MI. CONCLUSIONS These findings have identified a novel role for Plg in stem cell-mediated cardiac repair after MI. Thus, targeting Plg may offer a new therapeutic strategy for stem cell-mediated cardiac repair after MI.
Collapse
Affiliation(s)
- Yanqing Gong
- Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Departments of Cardiovascular Medicine and Molecular Cardiology, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio; Division of Translational Medicine and Human Genetics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania.
| | - Yujing Zhao
- Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Departments of Cardiovascular Medicine and Molecular Cardiology, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Ying Li
- Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Departments of Cardiovascular Medicine and Molecular Cardiology, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Jane Hoover-Plow
- Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Departments of Cardiovascular Medicine and Molecular Cardiology, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| |
Collapse
|
163
|
Mehran R, Nilsson M, Khajavi M, Du Z, Cascone T, Wu HK, Cortes A, Xu L, Zurita A, Schier R, Riedel B, El-Zein R, Heymach JV. Tumor endothelial markers define novel subsets of cancer-specific circulating endothelial cells associated with antitumor efficacy. Cancer Res 2014; 74:2731-41. [PMID: 24626092 DOI: 10.1158/0008-5472.can-13-2044] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Circulating endothelial cells (CEC) are derived from multiple sources, including bone marrow (circulating endothelial progenitors; CEP), and established vasculature (mature CEC). Although CECs have shown promise as a biomarker for patients with cancer, their utility has been limited, in part, by the lack of specificity for tumor vasculature and the different nonmalignant causes that can impact CEC. Tumor endothelial markers (TEM) are antigens enriched in tumor versus nonmalignant endothelia. We hypothesized that TEMs may be detectable on CEC and that these circulating TEM(+) endothelial cells (CTEC) may be a more specific marker for cancer and tumor response than standard CEC. We found that tumor-bearing mice had a relative increase in numbers of circulating CTEC, specifically with increased levels of TEM7 and TEM8 expression. Following treatment with various vascular-targeting agents, we observed a decrease in CTEC that correlated with the reductions in tumor growth. We extended these findings to human clinical samples and observed that CTECs were present in patients with esophageal cancer and non-small cell lung cancer (N = 40), and their levels decreased after surgical resection. These results demonstrate that CTECs are detectable in preclinical cancer models and patients with cancer. Furthermore, they suggest that CTECs offer a novel cancer-associated marker that may be useful as a blood-based surrogate for assessing the presence of tumor vasculature and antiangiogenic drug activity.
Collapse
Affiliation(s)
- Reza Mehran
- Authors' Affiliations: Departments of Thoracic and Cardiovascular Surgery,Thoracic/Head and Neck Medical Oncology, Epidemiology, Genitourinary Medical Oncology, and The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Cologne, Germany; and Department of Anaesthesia and Pain Medicine, Peter MacCallum Cancer Centre and The University of Melbourne, Melbourne, Victoria, Australia
| | - Monique Nilsson
- Authors' Affiliations: Departments of Thoracic and Cardiovascular Surgery,Thoracic/Head and Neck Medical Oncology, Epidemiology, Genitourinary Medical Oncology, and The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Cologne, Germany; and Department of Anaesthesia and Pain Medicine, Peter MacCallum Cancer Centre and The University of Melbourne, Melbourne, Victoria, Australia
| | - Mehrdad Khajavi
- Authors' Affiliations: Departments of Thoracic and Cardiovascular Surgery,Thoracic/Head and Neck Medical Oncology, Epidemiology, Genitourinary Medical Oncology, and The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Cologne, Germany; and Department of Anaesthesia and Pain Medicine, Peter MacCallum Cancer Centre and The University of Melbourne, Melbourne, Victoria, Australia
| | - Zhiqiang Du
- Authors' Affiliations: Departments of Thoracic and Cardiovascular Surgery,Thoracic/Head and Neck Medical Oncology, Epidemiology, Genitourinary Medical Oncology, and The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Cologne, Germany; and Department of Anaesthesia and Pain Medicine, Peter MacCallum Cancer Centre and The University of Melbourne, Melbourne, Victoria, Australia
| | - Tina Cascone
- Authors' Affiliations: Departments of Thoracic and Cardiovascular Surgery,Thoracic/Head and Neck Medical Oncology, Epidemiology, Genitourinary Medical Oncology, and The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Cologne, Germany; and Department of Anaesthesia and Pain Medicine, Peter MacCallum Cancer Centre and The University of Melbourne, Melbourne, Victoria, Australia
| | - Hua Kang Wu
- Authors' Affiliations: Departments of Thoracic and Cardiovascular Surgery,Thoracic/Head and Neck Medical Oncology, Epidemiology, Genitourinary Medical Oncology, and The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Cologne, Germany; and Department of Anaesthesia and Pain Medicine, Peter MacCallum Cancer Centre and The University of Melbourne, Melbourne, Victoria, Australia
| | - Andrea Cortes
- Authors' Affiliations: Departments of Thoracic and Cardiovascular Surgery,Thoracic/Head and Neck Medical Oncology, Epidemiology, Genitourinary Medical Oncology, and The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Cologne, Germany; and Department of Anaesthesia and Pain Medicine, Peter MacCallum Cancer Centre and The University of Melbourne, Melbourne, Victoria, Australia
| | - Li Xu
- Authors' Affiliations: Departments of Thoracic and Cardiovascular Surgery,Thoracic/Head and Neck Medical Oncology, Epidemiology, Genitourinary Medical Oncology, and The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Cologne, Germany; and Department of Anaesthesia and Pain Medicine, Peter MacCallum Cancer Centre and The University of Melbourne, Melbourne, Victoria, Australia
| | - Amado Zurita
- Authors' Affiliations: Departments of Thoracic and Cardiovascular Surgery,Thoracic/Head and Neck Medical Oncology, Epidemiology, Genitourinary Medical Oncology, and The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Cologne, Germany; and Department of Anaesthesia and Pain Medicine, Peter MacCallum Cancer Centre and The University of Melbourne, Melbourne, Victoria, Australia
| | - Robert Schier
- Authors' Affiliations: Departments of Thoracic and Cardiovascular Surgery,Thoracic/Head and Neck Medical Oncology, Epidemiology, Genitourinary Medical Oncology, and The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Cologne, Germany; and Department of Anaesthesia and Pain Medicine, Peter MacCallum Cancer Centre and The University of Melbourne, Melbourne, Victoria, Australia
| | - Bernhard Riedel
- Authors' Affiliations: Departments of Thoracic and Cardiovascular Surgery,Thoracic/Head and Neck Medical Oncology, Epidemiology, Genitourinary Medical Oncology, and The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Cologne, Germany; and Department of Anaesthesia and Pain Medicine, Peter MacCallum Cancer Centre and The University of Melbourne, Melbourne, Victoria, Australia
| | - Randa El-Zein
- Authors' Affiliations: Departments of Thoracic and Cardiovascular Surgery,Thoracic/Head and Neck Medical Oncology, Epidemiology, Genitourinary Medical Oncology, and The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Cologne, Germany; and Department of Anaesthesia and Pain Medicine, Peter MacCallum Cancer Centre and The University of Melbourne, Melbourne, Victoria, Australia
| | - John V Heymach
- Authors' Affiliations: Departments of Thoracic and Cardiovascular Surgery,Thoracic/Head and Neck Medical Oncology, Epidemiology, Genitourinary Medical Oncology, and The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Cologne, Germany; and Department of Anaesthesia and Pain Medicine, Peter MacCallum Cancer Centre and The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
164
|
Cowburn AS, Alexander LEC, Southwood M, Nizet V, Chilvers ER, Johnson RS. Epidermal deletion of HIF-2α stimulates wound closure. J Invest Dermatol 2014; 134:801-808. [PMID: 24037341 PMCID: PMC3877686 DOI: 10.1038/jid.2013.395] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 08/28/2013] [Accepted: 08/28/2013] [Indexed: 12/21/2022]
Abstract
Wound closure requires a complex series of micro-environmentally influenced events. A key aspect of wound closure is the migration of keratinocytes across the open wound. It has been found previously that the response to hypoxia via the HIF-1α transcription factor is a key feature of wound closure. The need for hypoxic response is likely due to interrupted wound vasculature, as well as infection, and in this work we investigated the need for a highly related hypoxic response transcription factor, HIF-2α. This factor was deleted tissue specifically in mice, and the resulting mice were found to have an accelerated rate of wound closure. This is correlated with a reduced bacterial load and inflammatory response in these mice. This indicates that manipulating or reducing the HIF-2α response in keratinocytes could be a useful means to accelerate wound healing and tissue repair.
Collapse
Affiliation(s)
- Andrew S Cowburn
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | | | - Mark Southwood
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Victor Nizet
- Division of Pediatric Pharmacology and Drug Discovery, UCSD School of Medicine, La Jolla, California, USA
| | - Edwin R Chilvers
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Randall S Johnson
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
165
|
Sato T, Amano H, Ito Y, Eshima K, Minamino T, Ae T, Katada C, Ohno T, Hosono K, Suzuki T, Shibuya M, Koizumi W, Majima M. Vascular endothelial growth factor receptor 1 signaling facilitates gastric ulcer healing and angiogenesis through the upregulation of epidermal growth factor expression on VEGFR1+CXCR4 + cells recruited from bone marrow. J Gastroenterol 2014; 49:455-69. [PMID: 23982810 DOI: 10.1007/s00535-013-0869-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 08/01/2013] [Indexed: 02/04/2023]
Abstract
BACKGROUND Angiogenesis is essential for gastric ulcer healing. Recent results suggest that vascular endothelial growth factor receptor 1 (VEGFR1), which binds to VEGF, promotes angiogenesis. In the present study, we investigated the role of VEGFR1 signaling in gastric ulcer healing and angiogenesis. METHODS Gastric ulcers were induced by serosal application of 100 % acetic acid in wild-type (WT) and tyrosine kinase-deficient VEGFR1 mice (VEGFR1 TK(-/-)). Bone marrow transplantation into irradiated WT mice was carried out using bone marrow cells isolated from WT and VEGFR1 TK(-/-) mice. RESULTS Ulcer healing was delayed in VEGFR1 TK(-/-) mice compared to WT mice and this was accompanied by decreased angiogenesis, as evidenced by reduced mRNA levels of CD31 and decreased microvessel density. Recruitment of cells expressing VEGFR1 and C-X-C chemokine receptor type 4 (CXCR4) was suppressed and epidermal growth factor (EGF) expression in ulcer granulation tissue was attenuated. Treatment of WT mice with neutralizing antibodies against VEGF or CXCR4 also delayed ulcer healing. In WT mice transplanted with bone marrow cells from VEGFR1 TK(-/-) mice, ulcer healing and angiogenesis were suppressed, and this was associated with reduced recruitment of bone marrow cells to ulcer granulation tissue. VEGFR1 TK(-/-) bone marrow chimeras also exhibited downregulation of EGF expression on CXCR4(+)VEGFR1(+) cells recruited from the bone marrow into ulcer lesions. CONCLUSION VEGFR1-mediated signaling plays a critical role in gastric ulcer healing and angiogenesis through enhanced EGF expression on VEGFR1(+)CXCR4(+) cells recruited from the bone marrow into ulcer granulation tissue.
Collapse
Affiliation(s)
- Takehito Sato
- Department of Pharmacology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa, 252-0374, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Meisner JK, Annex BH, Price RJ. Despite normal arteriogenic and angiogenic responses, hind limb perfusion recovery and necrotic and fibroadipose tissue clearance are impaired in matrix metalloproteinase 9-deficient mice. J Vasc Surg 2014; 61:1583-94.e1-10. [PMID: 24582703 DOI: 10.1016/j.jvs.2014.01.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 01/17/2014] [Accepted: 01/18/2014] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The relative contributions of arteriogenesis, angiogenesis, and ischemic muscle tissue composition toward reperfusion after arterial occlusion are largely unknown. Differential loss of bone marrow-derived cell (BMC) matrix metalloproteinase 9 (MMP9), which has been implicated in all of these processes, was used to assess the relative contributions of these processes during limb reperfusion. METHODS We compared collateral growth (arteriogenesis), capillary growth (angiogenesis), and ischemic muscle tissue composition after femoral artery ligation in FVB/NJ mice that had been reconstituted with bone marrow from wild-type or MMP9(-/-) mice. RESULTS Laser Doppler perfusion imaging confirmed decreased reperfusion capacity in mice with BMC-specific loss of MMP9; however, collateral arteriogenesis was not affected. Furthermore, when accounting for the fact that muscle tissue composition changes markedly with ischemia (ie, necrotic, fibroadipose, and regenerating tissue regions are present), angiogenesis was also unaffected. Instead, BMC-specific loss of MMP9 caused an increase in the proportion of necrotic and fibroadipose tissue, which showed the strongest correlation with poor perfusion recovery. Similarly, the reciprocal loss of MMP9 from non-BMCs showed similar deficits in perfusion and tissue composition without affecting arteriogenesis. CONCLUSIONS By concurrently analyzing arteriogenesis, angiogenesis, and ischemic tissue composition, we determined that the loss of BMC-derived or non-BMC-derived MMP9 impairs necrotic and fibroadipose tissue clearance after femoral artery ligation, despite normal arteriogenic and angiogenic vascular growth. These findings imply that therapeutic revascularization strategies for treating peripheral arterial disease may benefit from additionally targeting necrotic tissue clearance or skeletal muscle regeneration, or both.
Collapse
Affiliation(s)
- Joshua K Meisner
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Va
| | - Brian H Annex
- Division of Cardiovascular Medicine, University of Virginia, Charlottesville, Va; Cardiovascular Research Center, University of Virginia, Charlottesville, Va
| | - Richard J Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Va; Cardiovascular Research Center, University of Virginia, Charlottesville, Va.
| |
Collapse
|
167
|
Chan KYY, Xiang P, Zhou L, Li K, Ng PC, Wang CC, Zhang L, Deng HY, Pong NH, Zhao H, Chan WY, Sung RYT. Thrombopoietin protects against doxorubicin-induced cardiomyopathy, improves cardiac function, and reversely alters specific signalling networks. Eur J Heart Fail 2014; 13:366-76. [DOI: 10.1093/eurjhf/hfr001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Kathy Yuen-Yee Chan
- Department of Paediatrics; The Chinese University of Hong Kong; Hong Kong China
| | - Ping Xiang
- Department of Cardiology; Children's Hospital of Chongqing Medical University; Chongqing China
| | - Ligang Zhou
- Department of Cardiology; Children's Hospital of Chongqing Medical University; Chongqing China
| | - Karen Li
- Department of Paediatrics; The Chinese University of Hong Kong; Hong Kong China
- Li Ka Shing Institute of Health Sciences; The Chinese University of Hong Kong; Hong Kong
| | - Pak-Cheung Ng
- Department of Paediatrics; The Chinese University of Hong Kong; Hong Kong China
| | - Chi-Chiu Wang
- Li Ka Shing Institute of Health Sciences; The Chinese University of Hong Kong; Hong Kong
- Department of Obstetrics & Gynaecology; The Chinese University of Hong Kong; Hong Kong
| | - Lei Zhang
- Department of Paediatrics; The Chinese University of Hong Kong; Hong Kong China
| | - Hai-Yan Deng
- Department of Cardiology; Children's Hospital of Fudan University; Shanghai China
| | - Nga-Hin Pong
- Department of Paediatrics; The Chinese University of Hong Kong; Hong Kong China
| | - Hailu Zhao
- Department of Medicine and Therapeutics; The Chinese University of Hong Kong; Hong Kong
| | - Wood-Yee Chan
- Department of Anatomy; The Chinese University of Hong Kong; Hong Kong
| | - Rita Yn-Tz Sung
- Department of Paediatrics; The Chinese University of Hong Kong; Hong Kong China
- Li Ka Shing Institute of Health Sciences; The Chinese University of Hong Kong; Hong Kong
| |
Collapse
|
168
|
Berahovich RD, Zabel BA, Lewén S, Walters MJ, Ebsworth K, Wang Y, Jaen JC, Schall TJ. Endothelial expression of CXCR7 and the regulation of systemic CXCL12 levels. Immunology 2014; 141:111-22. [PMID: 24116850 DOI: 10.1111/imm.12176] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 09/13/2013] [Accepted: 09/27/2013] [Indexed: 12/24/2022] Open
Abstract
The concentration of CXCL12/SDF-1 in the bloodstream is tightly regulated, given its central role in leucocyte and stem/progenitor cell egress from bone marrow and recruitment to sites of inflammation or injury. The mechanism responsible for this regulation is unknown. Here we show that both genetic deletion and pharmacological inhibition of CXCR7, a high-affinity CXCL12 receptor, caused pronounced increases in plasma CXCL12 levels. The rise in plasma CXCL12 levels was associated with an impairment in the ability of leucocytes to migrate to a local source of CXCL12. Using a set of complementary and highly sensitive techniques, we found that CXCR7 protein is expressed at low levels in multiple organs in both humans and mice. In humans, CXCR7 was detected primarily on venule endothelium and arteriole smooth muscle cells. CXCR7 expression on venule endothelium was also documented in immunodeficient mice and CXCR7(+/lacZ) mice. The vascular expression of CXCR7 therefore gives it immediate access to circulating CXCL12. These studies suggest that endothelial CXCR7 regulates circulating CXCL12 levels and that CXCR7 inhibitors might be used to block CXCL12-mediated cell migration for therapeutic purposes.
Collapse
|
169
|
Chen F, Liu Q, Zhang ZD, Zhu XH. Co-delivery of G-CSF and EPO released from fibrin gel for therapeutic neovascularization in rat hindlimb ischemia model. Microcirculation 2014; 20:416-24. [PMID: 23294128 DOI: 10.1111/micc.12037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Accepted: 01/03/2013] [Indexed: 01/10/2023]
Abstract
OBJECTIVE G-CSF and EPO have shown a notable capability in neovascularization. However, their use is limited because of untoward leucocytosis, erythrogenesis, and short half-life in the plasma. Herein, we examined whether G-CSF and EPO released from fibrin gel injected into ischemic tissues would synergistically promote neovascularization with limited systematic effects in a rat hindlimb ischemic model. METHODS AND RESULTS In vivo study, group Gel received an intramuscular injection of fibrin gel; group Gel+G-CSF received fibrin gel containing human G-CSF; group Gel+EPO received fibrin gel containing human EPO; group Gel+G-CSF&EPO received fibrin gel containing G-CSF and EPO; group G-CSF&EPO received G-CSF and EPO. Through promoting the expression of SDF-1, local high concentration of EPO could traffic CXCR4+ cells mobilized by G-CSF to enhance neovascularization in ischemic muscle. The treatment with Gel+G-CSF&EPO was superior to the other treatments on blood flow reperfusion, capillary density, and α smooth muscle actin-positive vessel density. And this treatment induced a modest WBC count increase in peripheral blood. CONCLUSIONS G-CSF and EPO released from fibrin gel had a combined effect on postischemia neovascularization. This treatment may be a novel therapeutic modality for ischemic peripheral artery disease.
Collapse
Affiliation(s)
- Feng Chen
- Department of Vascular Surgery, the second affiliated Hospital, Nanchang University, Nanchang, China.
| | | | | | | |
Collapse
|
170
|
Hellevik T, Martinez-Zubiaurre I. Radiotherapy and the tumor stroma: the importance of dose and fractionation. Front Oncol 2014; 4:1. [PMID: 24478982 PMCID: PMC3896881 DOI: 10.3389/fonc.2014.00001] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/03/2014] [Indexed: 01/04/2023] Open
Abstract
Ionizing radiation is a non-specific but highly effective way to kill malignant cells. However, tumor recurrence sustained by a minor fraction of surviving tumor cells is a commonplace phenomenon caused by the activation of both cancer cell intrinsic resistance mechanisms, and also extrinsic intermediaries of therapy resistance, represented by non-malignant cells and structural components of the tumor stroma. The improved accuracy offered by advanced radiotherapy (RT)-technology permits reduced volume of healthy tissue in the irradiated field, and has been triggering an increase in the prescription of high-dose oligo-fractionated regimens in the clinics. Given the remarkable clinical success of high-dose RT and the current therapeutic shift occurring in the field, in this review we revise the existing knowledge on the effects that different radiation regimens exert on the different compartments of the tumor microenvironment, and highlight the importance of anti-tumor immunity and other tumor cell extrinsic mechanisms influencing therapeutic responses to high-dose radiation.
Collapse
Affiliation(s)
- Turid Hellevik
- Department of Oncology, University Hospital of Northern-Norway , Tromsø , Norway ; Translational Cancer Research Group, Department of Clinical Medicine, University of Tromsø , Tromsø , Norway
| | - Iñigo Martinez-Zubiaurre
- Translational Cancer Research Group, Department of Clinical Medicine, University of Tromsø , Tromsø , Norway
| |
Collapse
|
171
|
Fremder E, Munster M, Aharon A, Miller V, Gingis-Velitski S, Voloshin T, Alishekevitz D, Bril R, Scherer SJ, Loven D, Brenner B, Shaked Y. Tumor-derived microparticles induce bone marrow-derived cell mobilization and tumor homing: a process regulated by osteopontin. Int J Cancer 2014; 135:270-81. [PMID: 24347266 DOI: 10.1002/ijc.28678] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 10/31/2013] [Accepted: 12/02/2013] [Indexed: 12/15/2022]
Abstract
Acute chemotherapy can induce rapid bone-marrow derived pro-angiogenic cell (BMDC) mobilization and tumor homing, contributing to tumor regrowth. To study the contribution of tumor cells to tumor regrowth following therapy, we focused on tumor-derived microparticles (TMPs). EMT/6 murine-mammary carcinoma cells exposed to paclitaxel chemotherapy exhibited an increased number of TMPs and significantly altered their angiogenic properties. Similarly, breast cancer patients had increased levels of plasma MUC-1(+) TMPs following chemotherapy. In addition, TMPs from cells exposed to paclitaxel induced higher BMDC mobilization and colonization, but had no increased effect on angiogenesis in Matrigel plugs and tumors than TMPs from untreated cells. Since TMPs abundantly express osteopontin, a protein known to participate in BMDC trafficking, the impact of osteopontin-depleted TMPs on BMDC mobilization, colonization, and tumor angiogenesis was examined. Although EMT/6 tumors grown in mice inoculated with osteopontin-depleted TMPs had lower numbers of BMDC infiltration and microvessel density when compared with EMT/6 tumors grown in mice inoculated with wild-type TMPs, no significant difference in tumor growth was seen between the two groups. However, when BMDCs from paclitaxel-treated mice were injected into wild-type EMT/6-bearing mice, a substantial increase in tumor growth and BMDC infiltration was detected compared to osteopontin-depleted EMT/6-bearing mice injected with BMDCs from paclitaxel-treated mice. Collectively, our results suggest that osteopontin expressed by TMPs play an important role in BMDC mobilization and colonization of tumors, but is not sufficient to enhance the angiogenic activity in tumors.
Collapse
Affiliation(s)
- Ella Fremder
- Department of Molecular Pharmacology, Rappaport Faculty of Medicine and Research Institute, Technion, Haifa, Israel
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Tan L, Lin P, Pezeshkian B, Rehman A, Madlambayan G, Zeng X. Real-time monitoring of cell mechanical changes induced by endothelial cell activation and their subsequent binding with leukemic cell lines. Biosens Bioelectron 2014; 56:151-8. [PMID: 24487102 DOI: 10.1016/j.bios.2014.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 12/18/2013] [Accepted: 01/01/2014] [Indexed: 11/25/2022]
Abstract
Endothelial cell (EC) activation and their subsequent binding with different cells have various mechanical consequences that, if monitored real time, can serve as a functional biomarker of many pathophysiological response mechanisms. This work presents an innovative and facile strategy to conduct such monitoring using quartz crystal microbalance (QCM), thereby relating the shifts in its frequency and motional resistance to morphological changes upon cell-cell and cell-substrate interactions. By activating ECs with TNF-α and then characterizing their binding with HL-60 and KG-1 leukemia cells, we are able to induce the mechanical changes in ECs especially in the region of cell-substrate contact which resulted in dynamically coupled mass and viscoelastic changes representing the extent of both activation and binding. The activated ECs suffered a decrease of cellular contact area, leading to positive frequency shift and decreased motional resistance. The binding of leukemia cells onto pre-activated ECs exerted a mechanical force to regain the cell surface contact which resulted in the obvious QCM responses opposite to that of activation, and proportional to the number of cells added, in spite of the fact that these added cells are extremely outside the extinction boundary of the shear wave generated by QCM. Different cell lines demonstrate different attachment behavior, which was detected by the QCM. Despite these variations are quite subtle, yet the sensitivity of the technique for dynamic changes at the interface makes them detectable. Moreover, the reproducibility of the generated data determined at each step by deviation measurements (<10%) in response plot was very high despite the high possible heterogeneity in cell populations. The results are explained on the basis of simple theoretical and physical models, although, the development of a more quantitative and precise model is underway in our laboratory.
Collapse
Affiliation(s)
- Liang Tan
- Department of Chemistry, Oakland University, Rochester, MI 48309, United States; Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Peiling Lin
- Department of Chemistry, Oakland University, Rochester, MI 48309, United States
| | - Bahareh Pezeshkian
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States
| | - Abdul Rehman
- Department of Chemistry, Oakland University, Rochester, MI 48309, United States
| | - Gerard Madlambayan
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States
| | - Xiangqun Zeng
- Department of Chemistry, Oakland University, Rochester, MI 48309, United States.
| |
Collapse
|
173
|
Jardim-Perassi BV, Arbab AS, Ferreira LC, Borin TF, Varma NRS, Iskander ASM, Shankar A, Ali MM, de Campos Zuccari DAP. Effect of melatonin on tumor growth and angiogenesis in xenograft model of breast cancer. PLoS One 2014; 9:e85311. [PMID: 24416386 PMCID: PMC3887041 DOI: 10.1371/journal.pone.0085311] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 11/26/2013] [Indexed: 12/15/2022] Open
Abstract
As neovascularization is essential for tumor growth and metastasis, controlling angiogenesis is a promising tactic in limiting cancer progression. Melatonin has been studied for their inhibitory properties on angiogenesis in cancer. We performed an in vivo study to evaluate the effects of melatonin treatment on angiogenesis in breast cancer. Cell viability was measured by MTT assay after melatonin treatment in triple-negative breast cancer cells (MDA-MB-231). After, cells were implanted in athymic nude mice and treated with melatonin or vehicle daily, administered intraperitoneally 1 hour before turning the room light off. Volume of the tumors was measured weekly with a digital caliper and at the end of treatments animals underwent single photon emission computed tomography (SPECT) with Technetium-99m tagged vascular endothelial growth factor (VEGF) C to detect in vivo angiogenesis. In addition, expression of pro-angiogenic/growth factors in the tumor extracts was evaluated by membrane antibody array and collected tumor tissues were analyzed with histochemical staining. Melatonin in vitro treatment (1 mM) decreased cell viability (p<0.05). The breast cancer xenografts nude mice treated with melatonin showed reduced tumor size and cell proliferation (Ki-67) compared to control animals after 21 days of treatment (p<0.05). Expression of VEGF receptor 2 decreased significantly in the treated animals compared to that of control when determined by immunohistochemistry (p<0.05) but the changes were not significant on SPECT (p>0.05) images. In addition, there was a decrease of micro-vessel density (Von Willebrand Factor) in melatonin treated mice (p<0.05). However, semiquantitative densitometry analysis of membrane array indicated increased expression of epidermal growth factor receptor and insulin-like growth factor 1 in treated tumors compared to vehicle treated tumors (p<0.05). In conclusion, melatonin treatment showed effectiveness in reducing tumor growth and cell proliferation, as well as in the inhibition of angiogenesis.
Collapse
Affiliation(s)
- Bruna Victorasso Jardim-Perassi
- Department of Biology, Universidade Estadual Paulista, São José do Rio Preto, São Paulo, Brazil
- Laboratório de Investigação Molecular no Câncer, Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
| | - Ali S. Arbab
- Cellular and Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Lívia Carvalho Ferreira
- Department of Biology, Universidade Estadual Paulista, São José do Rio Preto, São Paulo, Brazil
- Laboratório de Investigação Molecular no Câncer, Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
| | - Thaiz Ferraz Borin
- Laboratório de Investigação Molecular no Câncer, Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
- Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
| | - Nadimpalli R. S. Varma
- Cellular and Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - A. S. M. Iskander
- Cellular and Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Adarsh Shankar
- Cellular and Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Meser M. Ali
- Cellular and Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Debora Aparecida Pires de Campos Zuccari
- Laboratório de Investigação Molecular no Câncer, Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
- Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
174
|
HoWangYin KY, Loinard C, Bakker W, Guérin CL, Vilar J, D'Audigier C, Mauge L, Bruneval P, Emmerich J, Lévy BI, Pouysségur J, Smadja DM, Silvestre JS. HIF-Prolyl Hydroxylase 2 Inhibition Enhances the Efficiency of Mesenchymal Stem Cell-Based Therapies for the Treatment of Critical Limb Ischemia. Stem Cells 2014; 32:231-43. [DOI: 10.1002/stem.1540] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 08/05/2013] [Indexed: 12/22/2022]
Affiliation(s)
| | - Céline Loinard
- Institut de Radioprotection et de Sureté Nucléaire; Fontenay aux Roses France
| | | | | | - José Vilar
- INSERM UMRS 970; Fontenay aux Roses France
| | - Clément D'Audigier
- INSERM UMRS 765; Université Paris Descartes; Sorbonne Paris Cité Paris France
- AP-HP; Hôpital Européen Georges Pompidou; Service d'hématologie Biologique Paris France
| | - Laetitia Mauge
- INSERM UMRS 765; Université Paris Descartes; Sorbonne Paris Cité Paris France
- AP-HP; Hôpital Européen Georges Pompidou; Service d'hématologie Biologique Paris France
| | - Patrick Bruneval
- AP-HP; Hôpital Européen Georges Pompidou; Service d'anatomie Pathologique Paris France
| | - Joseph Emmerich
- INSERM UMRS 765; Université Paris Descartes; Sorbonne Paris Cité Paris France
| | | | - Jacques Pouysségur
- University of Nice, Institute of Research on Cancer & Aging (IRCAN)Centre A. Lacassagne, Nice, France and Centre Scientifique de Monaco
| | - David M. Smadja
- INSERM UMRS 765; Université Paris Descartes; Sorbonne Paris Cité Paris France
- AP-HP; Hôpital Européen Georges Pompidou; Service d'hématologie Biologique Paris France
| | | |
Collapse
|
175
|
Rajashekhar G. Mesenchymal stem cells: new players in retinopathy therapy. Front Endocrinol (Lausanne) 2014; 5:59. [PMID: 24795699 PMCID: PMC4006021 DOI: 10.3389/fendo.2014.00059] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/10/2014] [Indexed: 12/20/2022] Open
Abstract
Retinopathies in human and animal models have shown to occur through loss of pericytes resulting in edema formation, excessive immature retinal angiogenesis, and neuronal apoptosis eventually leading to blindness. In recent years, the concept of regenerating terminally differentiated organs with a cell-based therapy has evolved. The cells used in these approaches are diverse and include tissue-specific endogenous stem cells, endothelial progenitor (EPC), embryonic stem cells, induced pluripotent stem cells (iPSC) and mesenchymal stem cells (MSC). Recently, MSC derived from the stromal fraction of adipose tissue have been shown to possess pluripotent differentiation potential in vitro. These adipose stromal cells (ASC) have been differentiated in a number of laboratories to osteogenic, myogenic, vascular, and adipocytic cell phenotypes. In vivo, ASC have been shown to have functional and phenotypic overlap with pericytes lining microvessels in adipose tissues. Furthermore, these cells either in paracrine mode or physical proximity with endothelial cells, promoted angiogenesis, improved ischemia-reperfusion, protected from myocardial infarction, and were neuroprotective. Owing to the easy isolation procedure and abundant supply, fat-derived ASC are a more preferred source of autologous mesenchymal cells compared to bone marrow MSC. In this review, we present evidence that these readily available ASC from minimally invasive liposuction will facilitate translation of ASC research into patients with retinal diseases in the near future.
Collapse
Affiliation(s)
- Gangaraju Rajashekhar
- Indiana Center for Vascular Biology and Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Vascular and Cardiac Center for Adult Stem Cell Therapy, Indiana University School of Medicine, Indianapolis, IN, USA
- VA Center for Regenerative Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- *Correspondence: Gangaraju Rajashekhar, Eugene and Marilyn Glick Eye Institute, Indiana Center for Vascular Biology and Medicine, Vascular and Cardiac Center for Adult Stem Cell Therapy, 975 West, Walnut Street IB442B, Indianapolis, IN 46202, USA e-mail:
| |
Collapse
|
176
|
Ma HC, Shi XL, Ren HZ, Yuan XW, Ding YT. Genetically modified mesenchymal stem cells overexpressing CXCR4 show increased colonization ability and confer better liver regeneration in mice. Shijie Huaren Xiaohua Zazhi 2014; 22:2229. [DOI: 10.11569/wcjd.v22.i16.2229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
177
|
Silvestre JS, Smadja DM, Lévy BI. Postischemic revascularization: from cellular and molecular mechanisms to clinical applications. Physiol Rev 2013; 93:1743-802. [PMID: 24137021 DOI: 10.1152/physrev.00006.2013] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After the onset of ischemia, cardiac or skeletal muscle undergoes a continuum of molecular, cellular, and extracellular responses that determine the function and the remodeling of the ischemic tissue. Hypoxia-related pathways, immunoinflammatory balance, circulating or local vascular progenitor cells, as well as changes in hemodynamical forces within vascular wall trigger all the processes regulating vascular homeostasis, including vasculogenesis, angiogenesis, arteriogenesis, and collateral growth, which act in concert to establish a functional vascular network in ischemic zones. In patients with ischemic diseases, most of the cellular (mainly those involving bone marrow-derived cells and local stem/progenitor cells) and molecular mechanisms involved in the activation of vessel growth and vascular remodeling are markedly impaired by the deleterious microenvironment characterized by fibrosis, inflammation, hypoperfusion, and inhibition of endogenous angiogenic and regenerative programs. Furthermore, cardiovascular risk factors, including diabetes, hypercholesterolemia, hypertension, diabetes, and aging, constitute a deleterious macroenvironment that participates to the abrogation of postischemic revascularization and tissue regeneration observed in these patient populations. Thus stimulation of vessel growth and/or remodeling has emerged as a new therapeutic option in patients with ischemic diseases. Many strategies of therapeutic revascularization, based on the administration of growth factors or stem/progenitor cells from diverse sources, have been proposed and are currently tested in patients with peripheral arterial disease or cardiac diseases. This review provides an overview from our current knowledge regarding molecular and cellular mechanisms involved in postischemic revascularization, as well as advances in the clinical application of such strategies of therapeutic revascularization.
Collapse
|
178
|
Liu SC, Alomran R, Chernikova SB, Lartey F, Stafford J, Jang T, Merchant M, Zboralski D, Zöllner S, Kruschinski A, Klussmann S, Recht L, Brown JM. Blockade of SDF-1 after irradiation inhibits tumor recurrences of autochthonous brain tumors in rats. Neuro Oncol 2013; 16:21-8. [PMID: 24335554 PMCID: PMC3870826 DOI: 10.1093/neuonc/not149] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Tumor irradiation blocks local angiogenesis, forcing any recurrent tumor to form new vessels from circulating cells. We have previously demonstrated that the post-irradiation recurrence of human glioblastomas in the brains of nude mice can be delayed or prevented by inhibiting circulating blood vessel–forming cells by blocking the interaction of CXCR4 with its ligand stromal cell-derived factor (SDF)–1 (CXCL12). In the present study we test this strategy by directly neutralizing SDF-1 in a clinically relevant model using autochthonous brain tumors in immune competent hosts. Methods We used NOX-A12, an l-enantiomeric RNA oligonucleotide that binds and inhibits SDF-1 with high affinity. We tested the effect of this inhibitor on the response to irradiation of brain tumors in rat induced by n-ethyl-N-nitrosourea. Results Rats treated in utero with N-ethyl-N-nitrosourea began to die of brain tumors from approximately 120 days of age. We delivered a single dose of whole brain irradiation (20 Gy) on day 115 of age, began treatment with NOX-A12 immediately following irradiation, and continued with either 5 or 20 mg/kg for 4 or 8 weeks, doses and times equivalent to well-tolerated human exposures. We found a marked prolongation of rat life span that was dependent on both drug dose and duration of treatment. In addition we treated tumors only when they were visible by MRI and demonstrated complete regression of the tumors that was not achieved by irradiation alone or with the addition of temozolomide. Conclusions Inhibition of SDF-1 following tumor irradiation is a powerful way of improving tumor response of glioblastoma multiforme.
Collapse
Affiliation(s)
- Shie-Chau Liu
- Corresponding author: J. Martin Brown, PhD, Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, A246, 1050A Arastradero Rd, Palo Alto, CA 94304-1334.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Bone mesenchymal stem cells contributed to the neointimal formation after arterial injury. PLoS One 2013; 8:e82743. [PMID: 24349351 PMCID: PMC3857273 DOI: 10.1371/journal.pone.0082743] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 10/28/2013] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Recent findings suggest that in response to repair-to-injury bone marrow mesenchymal stem cells (BMSCs) participate in the process of angiogenesis. It is unclear what role BMSCs play in the structure of the vessel wall. In present study, we aimed to determine whether BMSCs had the capacity of endothelial cells (ECs). METHODS BMSCs were separated and cultured. FACS and RT-PCR analysis confirmed the gene expression phenotype. The capacity of migration and adhesion and the ultrastructure of BMSCs were examined. The effect of BMSCs transplantation on the vascular repair was investigated in a murine carotid artery-injured model. RESULTS BMSCs could express some markers and form the tube-like structure. The migration and adhesion capacity of BMSCs increased significantly after stimulated. In addition, BMSCs had the intact cell junction. In vivo the local transfer of BMSCs differentiated into neo-endothelial cells in the injury model for carotid artery and contributed to the vascular remodeling. CONCLUSION These results showed that BMSCs could contribute to neointimal formation for vascular lesion and might be associated with the differentiation into ECs, which indicated the important therapeutic implications for vascular diseases.
Collapse
|
180
|
In situ tissue regeneration through host stem cell recruitment. Exp Mol Med 2013; 45:e57. [PMID: 24232256 PMCID: PMC3849571 DOI: 10.1038/emm.2013.118] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 07/31/2013] [Accepted: 08/06/2013] [Indexed: 02/06/2023] Open
Abstract
The field of tissue engineering has made steady progress in translating various tissue applications. Although the classical tissue engineering strategy, which involves the use of culture-expanded cells and scaffolds to produce a tissue construct for implantation, has been validated, this approach involves extensive cell expansion steps, requiring a lot of time and laborious effort before implantation. To bypass this ex vivo process, a new approach has been introduced. In situ tissue regeneration utilizes the body's own regenerating capacity by mobilizing host endogenous stem cells or tissue-specific progenitor cells to the site of injury. This approach relies on development of a target-specific biomaterial scaffolding system that can effectively control the host microenvironment and mobilize host stem/progenitor cells to target tissues. An appropriate microenvironment provided by implanted scaffolds would facilitate recruitment of host cells that can be guided to regenerating structural and functional tissues.
Collapse
|
181
|
GM-CSF exhibits anti-inflammatory activity on endothelial cells derived from chronic venous disease patients. Mediators Inflamm 2013; 2013:561689. [PMID: 24327798 PMCID: PMC3845402 DOI: 10.1155/2013/561689] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 09/19/2013] [Accepted: 09/27/2013] [Indexed: 11/17/2022] Open
Abstract
Twenty patients affected by chronic venous disease (CVD) in tertiary venous network and/or saphenous vein were analyzed before surgical ablation by echo-color-doppler for the hemodynamic parameters reflux time (RT) and resistance index (RI), a negative and a positive prognostic factor, respectively. RT and RI were next correlated with relevant in vitro parameters of venous endothelial cells (VEC) obtained from surgical specimens, such as cell migration in response to serum gradient, proliferation index, intercellular adhesion molecule (ICAM)-1 and vascular cell adhesion molecule (VCAM)-1 expression, as well as cytokines release. Of interest, ICAM-1 expression in patient-derived VEC cultures correlated positively with RT and negatively with RI. Moreover, RT showed a positive correlation with the baseline osteoprotegerin (OPG) expression by VEC and an inverse correlation with VEC proliferation index. On the other hand, RI correlated positively with TNF-related apoptosis inducing ligand (TRAIL) expression. Among the cytokines released by VEC, GM-CSF showed a positive correlation with VEC proliferation and TRAIL expression and a negative correlation with OPG, ICAM-1 and VCAM-1 expression. Since in vitro recombinant GM-CSF induced VEC proliferation and counteracted the induction of ICAM-1, VCAM-1 and OPG upon exposure to TNF-α, our data suggest an anti-inflammatory activity of GM-CSF on venous endothelial cells.
Collapse
|
182
|
Chatterjee M, Gawaz M. Platelet-derived CXCL12 (SDF-1α): basic mechanisms and clinical implications. J Thromb Haemost 2013; 11:1954-67. [PMID: 24024928 DOI: 10.1111/jth.12404] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Indexed: 12/19/2022]
Abstract
Platelets are a major source of CXCL12 (stromal cell-derived factor -1α, SDF-1α) and store CXCL12 as part of their α-granule secretome. Platelet activation enhances surface expression and release of CXCL12. Platelets and megakaryocytes express CXCR4, the major receptor for CXCL12, and interaction of CXCL12 with CXCR4 regulates megakaryopoiesis and the function of circulating platelets. Platelet-derived CXCL12 also modulates paracrine mechanisms such as chemotaxis, adhesion, proliferation and differentiation of nucleated cells, including progenitor cells. Platelet-derived CXCL12 enhances peripheral recruitment of progenitor cells to the sites of vascular and tissue injury both in vitro and in vivo and thereby promotes repair mechanisms. CXCL12 expression on platelets is elevated in patients with acute myocardial infarction, correlates with the number of circulating progenitor cells, is associated with preservation of myocardial function and is an independent predictor of clinical outcome. Administration of recombinant CXCL12 reduces infarct size following transient ischemia in mice. The present review summarizes the role of platelet-derived CXCL12 in cardiovascular biology and its diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- M Chatterjee
- Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Eberhard Karls Universität, Tübingen, Germany
| | | |
Collapse
|
183
|
Avraham-Davidi I, Yona S, Grunewald M, Landsman L, Cochain C, Silvestre JS, Mizrahi H, Faroja M, Strauss-Ayali D, Mack M, Jung S, Keshet E. On-site education of VEGF-recruited monocytes improves their performance as angiogenic and arteriogenic accessory cells. ACTA ACUST UNITED AC 2013; 210:2611-25. [PMID: 24166715 PMCID: PMC3832929 DOI: 10.1084/jem.20120690] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
VEGF-driven neovascularization transiently recruits Ly6Chigh monocytes, which subsequently alter their phenotype and exert angiogenic function to enlarge small vessels. Adult neovascularization relies on the recruitment of monocytes to the target organ or tumor and functioning therein as a paracrine accessory. The exact origins of the recruited monocytes and the mechanisms underlying their plasticity remain unclear. Using a VEGF-based transgenic system in which genetically tagged monocytes are conditionally summoned to the liver as part of a VEGF-initiated angiogenic program, we show that these recruited cells are derived from the abundant pool of circulating Ly6Chi monocytes. Remarkably, however, upon arrival at the VEGF-induced organ, but not the naive organ, monocytes undergo multiple phenotypic and functional changes, endowing them with enhanced proangiogenic capabilities and, importantly, with a markedly increased capacity to remodel existing small vessels into larger conduits. Notably, monocytes do not differentiate into long-lived macrophages, but rather appear as transient accessory cells. Results from transfers of presorted subpopulations and a novel tandem transfer strategy ruled out selective recruitment of a dedicated preexisting subpopulation or onsite selection, thereby reinforcing active reprogramming as the underlying mechanism for improved performance. Collectively, this study uncovered a novel function of VEGF, namely, on-site education of recruited “standard” monocytes to become angiogenic and arteriogenic professional cells, a finding that may also lend itself for a better design of angiogenic therapies.
Collapse
Affiliation(s)
- Inbal Avraham-Davidi
- Department of Developmental Biology and Cancer Research, the Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Hoh BL, Hosaka K, Downes DP, Nowicki KW, Wilmer EN, Velat GJ, Scott EW. Stromal cell-derived factor-1 promoted angiogenesis and inflammatory cell infiltration in aneurysm walls. J Neurosurg 2013; 120:73-86. [PMID: 24160472 DOI: 10.3171/2013.9.jns122074] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECT A small percentage of cerebral aneurysms rupture, but when they do, the effects are devastating. Current management of unruptured aneurysms consists of surgery, endovascular treatment, or watchful waiting. If the biology of how aneurysms grow and rupture were better known, a novel drug could be developed to prevent unruptured aneurysms from rupturing. Ruptured cerebral aneurysms are characterized by inflammation-mediated wall remodeling. The authors studied the role of stromal cell-derived factor-1 (SDF-1) in inflammation-mediated wall remodeling in cerebral aneurysms. METHODS Human aneurysms, murine carotid artery aneurysms, and murine intracranial aneurysms were studied using immunohistochemistry. Flow cytometry analysis was performed on blood from mice developing carotid or intracranial aneurysms. The effect of SDF-1 on endothelial cells and macrophages was studied by chemotaxis cell migration assay and capillary tube formation assay. Anti-SDF-1 blocking antibody was given to mice and compared with control (vehicle)-administered mice for its effects on the walls of carotid aneurysms and the development of intracranial aneurysms. RESULTS Human aneurysms, murine carotid aneurysms, and murine intracranial aneurysms all expressed SDF-1, and mice with developing carotid or intracranial aneurysms had increased progenitor cells expressing CXCR4, the receptor for SDF-1 (p < 0.01 and p < 0.001, respectively). Human aneurysms and murine carotid aneurysms had endothelial cells, macrophages, and capillaries in the walls of the aneurysms, and the presence of capillaries in the walls of human aneurysms was associated with the presence of macrophages (p = 0.01). Stromal cell-derived factor-1 promoted endothelial cell and macrophage migration (p < 0.01 for each), and promoted capillary tube formation (p < 0.001). When mice were given anti-SDF-1 blocking antibody, there was a significant reduction in endothelial cells (p < 0.05), capillaries (p < 0.05), and cell proliferation (p < 0.05) in the aneurysm wall. Mice given anti-SDF-1 blocking antibody developed significantly fewer intracranial aneurysms (33% vs 89% in mice given control immunoglobulin G, respectively; p < 0.05). CONCLUSIONS These data suggest SDF-1 is associated with angiogenesis and inflammatory cell migration and proliferation in the walls of aneurysms, and may have a role in the development of intracranial aneurysms.
Collapse
|
185
|
Pharmacologically antagonizing the CXCR4-CXCL12 chemokine pathway with AMD3100 inhibits sunlight-induced skin cancer. J Invest Dermatol 2013; 134:1091-1100. [PMID: 24226205 DOI: 10.1038/jid.2013.424] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 08/28/2013] [Accepted: 09/06/2013] [Indexed: 11/08/2022]
Abstract
One way sunlight causes skin cancer is by suppressing anti-tumor immunity. A major mechanism involves altering mast cell migration via the C-X-C motif chemokine receptor 4-C-X-C motif chemokine ligand 12 (CXCR4-CXCL12) chemokine pathway. We have discovered that pharmacologically blocking this pathway with the CXCR4 antagonist AMD3100 prevents both UV radiation-induced immune suppression and skin cancer. The majority of control mice receiving UV-only developed histopathologically confirmed squamous cell carcinomas. In contrast, skin tumor incidence and burden was significantly lower in AMD3100-treated mice. Perhaps most striking was that AMD3100 completely prevented the outgrowth of latent tumors that occurred once UV irradiation ceased. AMD3100 protection from UV immunosuppression and skin cancer was associated with reduced mast cell infiltration into the skin, draining lymph nodes, and the tumor itself. Thus a major target of CXCR4 antagonism was the mast cell. Our results indicate that interfering with UV-induced CXCL12 by antagonizing CXCR4 significantly inhibits skin tumor development by blocking UV-induced effects on mast cells. Hence, the CXCR4-CXCL12 chemokine pathway is a novel therapeutic target in the prevention of UV-induced skin cancer.
Collapse
|
186
|
Iwaya T, Fukagawa T, Suzuki Y, Takahashi Y, Sawada G, Ishibashi M, Kurashige J, Sudo T, Tanaka F, Shibata K, Endo F, Katagiri H, Ishida K, Kume K, Nishizuka S, Iinuma H, Wakabayashi G, Mori M, Sasako M, Mimori K. Contrasting expression patterns of histone mRNA and microRNA 760 in patients with gastric cancer. Clin Cancer Res 2013; 19:6438-49. [PMID: 24097871 DOI: 10.1158/1078-0432.ccr-12-3186] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE Recent studies revealed that both disseminated tumor cells and noncancerous cells contributed to cancer progression cooperatively in the bone marrow. Here, RNA-seq analysis of bone marrow from gastric cancer patients was performed to identify prognostic markers for gastric cancer. EXPERIMENTAL DESIGN Bone marrow samples from eight gastric cancer patients (stages I and IV: n = 4 each) were used for RNA-seq analysis. Results were validated through quantitative real-time PCR (qRT-PCR) analysis of HIST1H3D expression in 175 bone marrow, 92 peripheral blood, and 115 primary tumor samples from gastric cancer patients. miR-760 expression was assayed using qRT-PCR in 105 bone marrow and 96 primary tumor samples. Luciferase reporter assays were performed to confirm whether histone mRNAs were direct targets of miR-760. miR-760 expression was also evaluated in noncancerous cells from gastric cancer patients. RESULTS RNA-seq analysis of bone marrow samples from gastric cancer patients revealed higher expression of multiple histone mRNAs in stage IV patients. HIST1H3D expression in the bone marrow, peripheral blood, and primary tumor of stage IV patients was higher than that in stage I patients (P = 0.0284, 0.0243, and 0.0006, respectively). In contrast, miR-760 was downregulated in the bone marrow and primary tumor of stage IV patients compared with stage I patients (P = 0.0094 and 0.0018, respectively). Histone mRNA and miR-760 interacted directly. Furthermore, miR-760 was downregulated in noncancerous mucosa in stage IV gastric cancer patients. CONCLUSION Histone mRNA was upregulated, whereas miR-760 was downregulated in the bone marrow and primary tumor of advanced gastric cancer patients, suggesting that the histone mRNA/miR-760 axis had a crucial role in the development of gastric cancer.
Collapse
Affiliation(s)
- Takeshi Iwaya
- Authors' Affiliations: Department of Surgery, Kyushu University Beppu Hospital, Beppu; Department of Surgery, Iwate Medical University, Morioka; Department of Gastric Surgery Division, National Cancer Center Hospital; Department of Medical Genome Sciences, Graduate School of Frontier Sciences, University of Tokyo, Kashiwa-shi, Chiba; Department of Surgery, Teikyo University School of Medicine, Tokyo; Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka, University, Suita; and Department of Digestive Surgery, Hyogo Medical College, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Roura S, Gálvez-Montón C, Pujal JM, Casani L, Fernández MA, Astier L, Gastelurrutia P, Domingo M, Prat-Vidal C, Soler-Botija C, Llucià-Valldeperas A, Llorente-Cortés V, Bayes-Genis A. New insights into lipid raft function regulating myocardial vascularization competency in human idiopathic dilated cardiomyopathy. Atherosclerosis 2013; 230:354-64. [DOI: 10.1016/j.atherosclerosis.2013.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 07/26/2013] [Accepted: 08/06/2013] [Indexed: 12/15/2022]
|
188
|
Nakamura Y, Ishikawa H, Kawai K, Tabata Y, Suzuki S. Enhanced wound healing by topical administration of mesenchymal stem cells transfected with stromal cell-derived factor-1. Biomaterials 2013; 34:9393-400. [PMID: 24054847 DOI: 10.1016/j.biomaterials.2013.08.053] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 08/19/2013] [Indexed: 12/15/2022]
Abstract
The objective of this study was to investigate the ability of mesenchymal stem cells (MSC) genetically engineered with stromal cell-derived factor-1 (SDF-1) to heal skin wounds. When transfected with SDF-1 plasmid DNA, MSC which were isolated from the bone marrow of rats, secreted SDF-1 for 7 days. In vitro cell migration assay revealed that the SDF-1-engineered MSC (SDF-MSC) enhanced the migration of MSC and dermal fibroblasts to a significantly greater extent than MSC. The SDF-MSC secreted vascular endothelial growth factor, hepatocyte growth factor, and interleukin 6 at a significantly high level. A skin defect model of rats was prepared and MSC and SDF-MSC were applied to the wound to evaluate wound healing in terms of wound size and histological examinations. The wound size decreased significantly faster with SDF-MSC treatment than with MSC and PBS treatments. The length of the neoepithelium and the number of blood vessels newly formed were significantly larger. A cell-tracing experiment with fluorescently labeled cells demonstrated that the percent survival of SDF-MSC in the tissue treated was significantly high compared with that of MSC. It was concluded that SDF-1 genetic engineering is a promising way to promote the wound healing activity of MSC for a skin defect.
Collapse
Affiliation(s)
- Yoko Nakamura
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Japan; Department of Biomaterials, Institute for Frontier Medical Sciences, Kyoto University, Japan.
| | | | | | | | | |
Collapse
|
189
|
Takyar S, Vasavada H, Zhang JG, Ahangari F, Niu N, Liu Q, Lee CG, Cohn L, Elias JA. VEGF controls lung Th2 inflammation via the miR-1-Mpl (myeloproliferative leukemia virus oncogene)-P-selectin axis. ACTA ACUST UNITED AC 2013; 210:1993-2010. [PMID: 24043765 PMCID: PMC3782056 DOI: 10.1084/jem.20121200] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Asthma, the prototypic Th2-mediated inflammatory disorder of the lung, is an emergent disease worldwide. Vascular endothelial growth factor (VEGF) is a critical regulator of pulmonary Th2 inflammation, but the underlying mechanism and the roles of microRNAs (miRNAs) in this process have not been defined. Here we show that lung-specific overexpression of VEGF decreases miR-1 expression in the lung, most prominently in the endothelium, and a similar down-regulation occurs in lung endothelium in Th2 inflammation models. Intranasal delivery of miR-1 inhibited inflammatory responses to ovalbumin, house dust mite, and IL-13 overexpression. Blocking VEGF inhibited Th2-mediated lung inflammation, and this was restored by antagonizing miR-1. Using mRNA arrays, Argonaute pull-down assays, luciferase expression assays, and mutational analysis, we identified Mpl as a direct target of miR-1 and showed that VEGF controls the expression of endothelial Mpl during Th2 inflammation via the regulation of miR-1. In vivo knockdown of Mpl inhibited Th2 inflammation and indirectly inhibited the expression of P-selectin in lung endothelium. These experiments define a novel VEGF-miR-1-Mpl-P-selectin effector pathway in lung Th2 inflammation and herald the utility of miR-1 and Mpl as potential therapeutic targets for asthma.
Collapse
Affiliation(s)
- Seyedtaghi Takyar
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Kumar S, Arbab AS. Neovascularization in Glioblastoma: Current Pitfall in Anti-angiogenic therapy. ZHONG LIU ZA ZHI 2013; 1:16-19. [PMID: 24976869 PMCID: PMC4073792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor in adults. However, the survival of patients with GBM has been dismal after multi-disciplinary treatment with surgery, radiotherapy, and chemotherapy. In the efforts to improve clinical outcome, anti-angiogenic therapy with bevacizumab (Avastin) was introduced to inhibit vascular endothelial growth factor (VEGF) mediated tumor neovascularization. Unfortunately, the results from clinical trials have not lived up to the initial expectations. Patients either fail to respond to anti-angiogenic therapy or develop resistance following an initial response. The failure of anti-angiogenic therapy has led to a frustration among physicians and research community. Recent evidence indicates that the dogma of tumor neovascularization solely dependent on VEGF pathway to be overly simplistic. A realistic model of tumor neovascularization should include alternative pathways that are independent of VEGF signaling. A better understanding of the underlying processes in tumor neovascularization would help in designing successful anti-angiogenic treatment strategies.
Collapse
Affiliation(s)
- Sanath Kumar
- Radiation Oncology, Henry Ford Hospital, Detroit, MI-48202
| | - Ali S. Arbab
- Cellular and Molecular Imaging Laboratory, Radiology, Henry Ford Hospital
- Radiology, Wayne State University School of Medicine, Detroit, MI
| |
Collapse
|
191
|
Petruzziello-Pellegrini TN, Moslemi-Naeini M, Marsden PA. New insights into Shiga toxin-mediated endothelial dysfunction in hemolytic uremic syndrome. Virulence 2013; 4:556-63. [PMID: 23955166 PMCID: PMC5359733 DOI: 10.4161/viru.26143] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Shiga toxin-producing E. coli represents a significant global health concern, especially as hypervirulent pathogens surface amidst outbreaks of hemolytic uremic syndrome (HUS). Shiga toxin (Stx) is key in the microangiopathic events underlying the disease and its central role is underscored by the unprecedented HUS outbreak in Germany in 2011. The mechanisms of Stx-mediated endothelial dysfunction have been a major focus of research that has contributed to the current understanding of the pathogenic changes in endothelial phenotype leading to HUS. Among the newer concepts are Stx-mediated gene regulation in the absence of protein synthesis inhibition, a potential role for complement activation, and accumulating evidence for detectable serum markers before the onset of the classic clinical features of HUS. Further investigation of newer therapeutic targets and potential prognostic markers is essential to assess their utility in mitigating disease and/or predicting outcomes and will provide an improved overall understanding of HUS pathogenesis.
Collapse
|
192
|
de Boer HC, van Oeveren-Rietdijk AM, Rotmans JI, Dekkers OM, Rabelink TJ, van Zonneveld AJ. Activated platelets correlate with mobilization of naïve CD34(+) cells and generation of CD34(+) /KDR(+) cells in the circulation. A meta-regression analysis. J Thromb Haemost 2013; 11:1583-92. [PMID: 23895310 DOI: 10.1111/jth.12315] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Indexed: 11/27/2022]
Abstract
BACKGROUND Bone marrow-derived circulating CD34(+) progenitor cells participate in remodeling and repair of the vasculature. Coexpression of the kinase-insert domain-containing receptor (KDR) has been proposed to identify the regenerative capacity. Recently, we provided evidence that the major fraction of circulating CD34(+) /KDR(+) cells is not mobilized from bone marrow, but is generated at sites of vascular injury through interaction with platelets. OBJECTIVES To determine the relationship between platelet activation, the recruitment of naïve CD34(+) cells and the generation of CD34(+) /KDR(+) progenitor cells in a broad range of (patho)physiologic conditions, a detailed meta-regression analysis was conducted. METHODS/RESULTS Twenty-eight conditions were found in which the numbers of CD34(+) and/or CD34(+) /KDR(+) cells and the levels of soluble P-selectin, as a marker for in vivo platelet activation, were documented. To combine heterogeneous data from 214 selected articles, results were standardized to a uniform scale by calculating standardized mean differences (SMDs) obtained from patient and control cohorts. Subsequently, a random-effects meta-regression analysis was performed on pooled SMDs. CONCLUSIONS Our systemic survey supports a model in which activated platelets are a determinant for mobilization of CD34(+) cells from the bone marrow and the generation of CD34(+) /KDR(+) cells in the circulation.
Collapse
Affiliation(s)
- H C de Boer
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | |
Collapse
|
193
|
Russell JS, Brown JM. The irradiated tumor microenvironment: role of tumor-associated macrophages in vascular recovery. Front Physiol 2013; 4:157. [PMID: 23882218 PMCID: PMC3713331 DOI: 10.3389/fphys.2013.00157] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 06/11/2013] [Indexed: 12/12/2022] Open
Abstract
Radiotherapy is an important modality used in the treatment of more than 50% of cancer patients in the US. However, despite sophisticated techniques for radiation delivery as well as the combination of radiation with chemotherapy, tumors can recur. Thus, any method of improving the local control of the primary tumor by radiotherapy would produce a major improvement in the curability of cancer patients. One of the challenges in the field is to understand how the tumor vasculature can regrow after radiation in order to support tumor recurrence, as it is unlikely that any of the endothelial cells within the tumor could survive the doses given in a typical radiotherapy regimen. There is now considerable evidence from both preclinical and clinical studies that the tumor vasculature can be restored following radiotherapy from an influx of circulating cells consisting primarily of bone marrow derived monocytes and macrophages. The radiation-induced influx of bone marrow derived cells (BMDCs) into tumors can be prevented through the blockade of various cytokine pathways and such strategies can inhibit tumor recurrence. However, the post-radiation interactions between surviving tumor cells, recruited immune cells, and the remaining stroma remain poorly defined. While prior studies have described the monocyte/macrophage inflammatory response within normal tissues and in the tumor microenvironment, less is known about this response with respect to a tumor after radiation therapy. The goal of this review is to summarize existing research studies to provide an understanding of how the myelomonocytic lineage may influence vascular recovery within the irradiated tumor microenvironment.
Collapse
Affiliation(s)
- Jeffery S Russell
- Department of Medical Oncology, Stanford University School of Medicine Stanford, CA, USA
| | | |
Collapse
|
194
|
Roura S, Gálvez-Montón C, Bayes-Genis A. The challenges for cardiac vascular precursor cell therapy: lessons from a very elusive precursor. J Vasc Res 2013; 50:304-23. [PMID: 23860201 DOI: 10.1159/000353294] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 05/01/2013] [Indexed: 11/19/2022] Open
Abstract
There is compelling evidence that cardiovascular disorders arise and/or progress due mainly to endothelial dysfunction. Novel therapeutic strategies aim to generate new myocardial tissue using cells with regenerative potential, either alone or in combination with biomaterials, cytokines and advanced monitoring devices. Among the human adult progenitor cells used in such methods, those historically termed 'endothelial progenitor cells' show promise for vascular growth and repair. Asahara et al. [Science 1997;275:964-967] initially described putative endothelial cell precursors in 1997. Subsequently, distinct cell populations termed endothelial colony-forming units-Hill, circulating angiogenic cells and endothelial colony-forming cells were identified that varied in terms of phenotype, vascular homeostasis contribution and purity. Notably, most of these cells are not genuine vascular precursor cells belonging to the endothelial lineage. This review provides a broad overview of the main properties of the endothelium, focusing on the basis governing its growth and repair. We discuss efforts to identify true vascular precursors, a matter of debate for the past 15 years, as well as recent methodological advances in identifying new hierarchies of more homogeneous, clonogenic and proliferative vascular endothelial-lineage precursors. Consideration of these issues provides insights that may help develop more effective therapies against human diseases that involve vascular deficits.
Collapse
Affiliation(s)
- Santiago Roura
- ICREC Research Program, Health Research Institute Germans Trias i Pujol-IGTP, University Hospital Germans Trias i Pujol, Badalona, Spain.
| | | | | |
Collapse
|
195
|
Salinas-Souza C, De Oliveira R, Alves MTDS, Garcia Filho RJ, Petrilli AS, Toledo SRC. The metastatic behavior of osteosarcoma by gene expression and cytogenetic analyses. Hum Pathol 2013; 44:2188-98. [PMID: 23845465 DOI: 10.1016/j.humpath.2013.04.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 04/17/2013] [Accepted: 04/22/2013] [Indexed: 10/26/2022]
Abstract
Osteosarcoma is a malignant bone tumor with high metastatic potential. Metastasis at diagnosis is the most significant prognostic factor in predicting the clinical outcome of osteosarcoma. We compared the gene expression of metastases that were present at the time of initial diagnosis to those developed later in the course of the disease. We used quantitative real-time polymerase chain reaction to evaluate the gene expression of MDM2, CXCR4, RANKL, RB1, and OSTERIX in 98 samples of osteosarcoma taken from 47 patients (74 metastases and 24 primary tumors) and 30 nonmalignant lung tissues surrounding osteosarcoma metastases. In addition, we investigated the copy number changes of RB1 and MDM2 genes in 12 primary cultures of pulmonary metastases of osteosarcoma, using interphase fluorescence in situ hybridization. Metastases from metastatic patients at diagnosis were characterized by low expression of RB1 and RANKL (P = .0009 and P = .0109, respectively) and overexpression of CXCR4 and MDM2 (P = .0389 and P = .0325, respectively). The loss of RANKL and gain of CXCR4 could also be detected in the primary tumors of metastatic patients at diagnosis (P = .0121 and P = .0264, respectively). Thus, some early genetic events such as the loss of RANKL and the gain of CXCR4 expressions probably facilitate the metastatic progression concomitant with the primary tumor establishment, supporting the role of the CXCR4 receptor in directing osteosarcoma metastases to the lung. On the other hand, late events such as the loss of RB1 and gain of MDM2, crucial regulators of cell cycle, appear to be related to the final mechanisms contributing to the metastatic establishment of osteosarcoma.
Collapse
Affiliation(s)
- Carolina Salinas-Souza
- Genetics Laboratory, Pediatric Oncology Institute (IOP/GRAACC/UNIFESP), Department of Pediatrics, Federal University of São Paulo, São Paulo 04023-062, Brazil.
| | | | | | | | | | | |
Collapse
|
196
|
Zhong C, Wang J, Li B, Xiang H, Ultsch M, Coons M, Wong T, Chiang NY, Clark S, Clark R, Quintana L, Gribling P, Suto E, Barck K, Corpuz R, Yao J, Takkar R, Lee WP, Damico-Beyer LA, Carano RD, Adams C, Kelley RF, Wang W, Ferrara N. Development and Preclinical Characterization of a Humanized Antibody Targeting CXCL12. Clin Cancer Res 2013; 19:4433-45. [DOI: 10.1158/1078-0432.ccr-13-0943] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
197
|
|
198
|
NSAID, aspirin delays gastric ulcer healing with reduced accumulation of CXCR4(+)VEGFR1(+) cells to the ulcer granulation tissues. Biomed Pharmacother 2013; 67:607-13. [PMID: 23809370 DOI: 10.1016/j.biopha.2013.01.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 01/24/2013] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Ulcer healing is a complex process, which involves cell migration, proliferation, angiogenesis and re-epithelialization. Several growth factors have been implicated in this process but the precise mechanism is not well understood. This study examined the involvement of VEGFR1 signaling in the gastric ulcer healing. METHODS Gastric ulcers were induced by the serosal application of 100% acetic acid, and the areas of the ulcers were measured thereafter. RESULTS The healing of acetic acid induced ulcers and the progenitor cells expressing CXCR4(+)VEGFR1(+) cell were significantly delayed in NSAID treated mice. The areas of the ulcer was significantly suppressed in tyrosine kinase-deficient VEGFR1 mice (VEGFR1TKKO) compared with wild type (WT) mice. The plasma level of SDF-1 and stem cell factor (SCF) and bone marrow level of pro-matrix metallopeptidase 9 (pro-MMP-9) were significantly reduced in VEGFR1TKKO mice. In VEGFR1 TKKOmice, the progenitor cells expressing CXCR4(+)VEGFR1(+) cell from bone marrow and the recruitment of these cells in healing ulcer were suppressed. Furthermore, VEGFR1 TKKO mice treated with NSAID did not suppress gastric ulcer healing compared to vehicle mice. These results suggested that NSAID suppressed VEGFR1 TK signaling plays a critical role in ulcer healing through mobilization of CXCR4(+)VEGFR1(+) cells. CONCLUSION VEGFR1 signaling is required for healing of NSAID induced gastric ulcer and angiogenesis with increased recruitment of CXCR4(+)VEGFR1(+) cells to the ulcerative lesion.
Collapse
|
199
|
Virani S, Edwards AK, Thomas R, Childs T, Tayade C. Blocking of stromal cell-derived factor-1 reduces neoangiogenesis in human endometriosis lesions in a mouse model. Am J Reprod Immunol 2013; 70:386-97. [PMID: 23650939 DOI: 10.1111/aji.12134] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 04/03/2013] [Indexed: 02/03/2023] Open
Abstract
PROBLEM Endometriosis affects 5-10% of women and is characterized by the growth of endometrial tissue outside of the uterus. Establishing new blood supply is a fundamental requirement for endometriosis lesion growth. Endothelial progenitor cells (EPCs), recruited by stromal cell-derived factor-1 (SDF-1), contribute to neoangiogenesis in endometriotic lesions. We hypothesized that SDF-1 is central to the neoangiogenesis and survival of endometriotic lesions, and blocking of SDF-1 will reduce vascularization of lesions in a mouse model. METHOD OF STUDY Using immunohistochemistry, we evaluated SDF-1 and CD34(+) EPCs in human endometriotic lesions and normal endometrium samples. EPCs were co-localized using CD34 and VEGFR2. Effects of SDF-1 blocking on endometriotic lesion survival were assessed in BALB/c-Rag2(-/-) /IL2rγ(-/-) mice engrafted with human endometrium and treated with SDF-1-blocking antibody or an isotype control. Weekly blood samples from experimental mice were analyzed for cytokines and EPCs. RESULTS SDF-1 and CD34(+) EPCs were abundant in human endometriotic lesions compared with eutopic endometrium. In our mouse model, SDF-1-blocking antibody reduced CD31(+) microvessels compared with isotype control. CONCLUSION Blocking SDF-1 reduces neovascularization and survival of lesions in a mouse model of endometriosis.
Collapse
Affiliation(s)
- Sophia Virani
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | | | | | | | | |
Collapse
|
200
|
Barone A, Rubin JB. Opportunities and challenges for successful use of bevacizumab in pediatrics. Front Oncol 2013; 3:92. [PMID: 23641361 PMCID: PMC3638307 DOI: 10.3389/fonc.2013.00092] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 04/05/2013] [Indexed: 11/13/2022] Open
Abstract
Bevacizumab (Avastin) has rapidly gained status as a broadly active agent for malignancies of several different histologies in adults. This activity has spawned a range of uses in pediatrics for both oncologic and non-oncologic indications. Early analyses indicate that pediatric cancers exhibit a spectrum of responses to bevacizumab that suggest its activity may be more limited than in adult oncology. Most exciting, is that for low-grade tumors that threaten vision and hearing, there is not only evidence for objective tumor response but for recovery of lost function as well. In addition to oncological indications, there is a range of uses for non-oncologic disease for which bevacizumab has clear activity. Finally, a number of mechanisms have been identified as contributing to bevacizumab resistance in cancer. Elucidating these mechanisms will guide the development of future clinical trials of bevacizumab in pediatric oncology.
Collapse
Affiliation(s)
- Amy Barone
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, St. Louis Children's Hospital, Washington University School of Medicine St. Louis, MO, USA
| | | |
Collapse
|