Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters











Publication year range
1.
EMBO Mol Med ; 13(7): e14089, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34102002

ABSTRACT

The tyrosine phosphatase SHP2 is oncogenic in cancers driven by receptor-tyrosine-kinases, and SHP2 inhibition reduces tumor growth. Here, we report that SHP2 is an essential promoter of endothelial cell survival and growth in the remodeling tumor vasculature. Using genetic and chemical approaches to inhibit SHP2 activity in endothelial cells, we show that SHP2 inhibits pro-apoptotic STAT3 and stimulates proliferative ERK1/2 signaling. Systemic SHP2 inhibition in mice bearing tumor types selected for SHP2-independent tumor cell growth promotes degeneration of the tumor vasculature and blood extravasation; reduces tumor vascularity and blood perfusion; and increases tumor necrosis. Reduction of tumor growth ensues, independent of SHP2 targeting in the tumor cells, blocking immune checkpoints, or recruiting macrophages. We also show that inhibiting the Angiopoietin/TIE2/AKT cascade magnifies the vascular and anti-tumor effects of SHP2 inhibition by blocking tumor endothelial AKT signaling, not a target of SHP2. Since the SHP2 and Ang2/TIE2 pathways are active in vascular endothelial cells of human melanoma and colon carcinoma, SHP2 inhibitors alone or with Ang2/TIE2 inhibitors hold promise to effectively target the tumor endothelium.


Subject(s)
Neoplasms , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Animals , Endothelial Cells/metabolism , Mice , Neoplasms/drug therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Receptor Protein-Tyrosine Kinases , Signal Transduction
2.
Stem Cell Res Ther ; 12(1): 236, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33849659

ABSTRACT

Erythro-myeloid progenitors (EMP) are found in a population of cells expressing CD31 and CD45 markers (CD31+CD45+). A recent study indicated that EMPs persist until adulthood and can be a source of endothelial cells. We identified two sub-populations of EMP cells, CD31lowCD45low and CD31highCD45+, from peripheral blood that can differentiate into cells of erythroid lineage. Our novel findings add to the current knowledge of hematopoietic lineage commitment, and our sequential, dual-step, in vitro culture model provides a platform for the study of the molecular and cellular mechanisms underlying human hematopoiesis and erythroid differentiation.


Subject(s)
Endothelial Cells , Hematopoietic System , Adult , Cell Differentiation , Erythroid Cells , Hematopoiesis , Humans
3.
Mod Pathol ; 30(11): 1567-1576, 2017 11.
Article in English | MEDLINE | ID: mdl-28752846

ABSTRACT

One of the major challenges in biomarker development is the collection of tumor tissue of adequate quality for analysis. A prospective clinical trial was initiated to collect tissues from triple negative breast cancers prior to and after neoadjuvant chemotherapy in order to study the mechanisms of chemoresistance. Sixty patients had pre-chemotherapy biopsies performed by either a surgeon or a radiologist, while those with residual tumor after chemotherapy had research-only biopsies and/or surgical samples collected in liquid nitrogen, RNA-later and formalin. We examined each core for tumor cellularity, stromal content, and necrosis after which, RNA and DNA extraction was performed. We found that biopsies collected with ultrasound guidance were more likely to contain tumor than those collected by the surgeon. Patient reluctance to undergo research-only biopsies after chemotherapy was not a problem. Pre-chemotherapy tumor biopsies frequently did not contain any tumor cells (15%) or did not have ≥50% tumor content (63%). Indeed, 50% of patients had at least 2 pre-chemotherapy core biopsies with <50% tumor content. After chemotherapy, 30% of biopsy or surgical samples in patients with incomplete response did not contain any tumor. Finally, RNA-later not only made histopathological assessment of tumor content difficult, but yielded less DNA than fresh snap frozen samples. We recommend that high-quality tissue procurement can be best accomplished if at least three image-guided core biopsies be obtained per sample, each of these cores be examined for tumor cellularity and that at least some of them be freshly snap frozen in liquid nitrogen.


Subject(s)
Biomarkers, Tumor/analysis , Image-Guided Biopsy/methods , Triple Negative Breast Neoplasms/drug therapy , Ultrasonography, Interventional/methods , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biopsy/methods , Chemotherapy, Adjuvant , Female , Humans , Middle Aged , Neoadjuvant Therapy
4.
J Clin Invest ; 126(12): 4554-4568, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27820703

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) reside in the bone marrow. Stress signals from cancer and other conditions promote HSPC mobilization into circulation and subsequent homing to tissue microenvironments. HSPC infiltration into tissue microenvironments can influence disease progression; notably, in cancer, HSPCs encourage tumor growth. Here we have uncovered a mutually exclusive distribution of EPHB4 receptors in bone marrow sinusoids and ephrin B2 ligands in hematopoietic cells. We determined that signaling interactions between EPHB4 and ephrin B2 control HSPC mobilization from the bone marrow. In mice, blockade of the EPHB4/ephrin B2 signaling pathway reduced mobilization of HSPCs and other myeloid cells to the circulation. EPHB4/ephrin B2 blockade also reduced HSPC infiltration into tumors as well as tumor progression in murine models of melanoma and mammary cancer. These results identify EPHB4/ephrin B2 signaling as critical to HSPC mobilization from bone marrow and provide a potential strategy for reducing cancer progression by targeting the bone marrow.


Subject(s)
Bone Marrow/metabolism , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Receptor, EphB4/metabolism , Signal Transduction/physiology , Stem Cell Niche/physiology , Animals , Cell Line , Ephrin-B2/genetics , Ephrin-B2/metabolism , Mice , Receptor, EphB4/genetics
5.
Sci Signal ; 9(419): ra28, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26980441

ABSTRACT

Interleukin-23 (IL-23), a heterodimeric cytokine composed of the unique p19 peptide (IL-23p19) and a peptide called IL-12p40, which is shared with IL-12, is implicated in Crohn's disease, rheumatoid arthritis, psoriasis, and other immune-mediated inflammatory diseases. Endothelial cells produce the IL-23p19 peptide in the absence of the IL-12p40 chain and thus do not make heterodimeric IL-23. We found that intercellular IL-23p19 increased the cell surface abundances of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) on endothelial cells, which enhanced the attachment of leukocytes and increased their transendothelial migration. Intracellular p19 associated with the cytokine receptor subunit gp130 and stimulated the gp130-dependent activation of signal transducer and activator of transcription 3 (STAT3) signaling. Proinflammatory factors promoted the generation of IL-23p19 in endothelial cells. The adventitial capillaries of inflamed temporal arteries in patients with giant-cell arteritis (GCA) had endothelial p19 protein associated with gp130, but did not contain the IL-12p40 chain. Because adventitial capillaries are essential for the entry of inflammatory cells into arterial walls, these data suggest that p19 may contribute to GCA disease and could represent a therapeutic target. Our results provide evidence that IL-23p19 is a previously unrecognized endothelial proinflammatory peptide that promotes leukocyte transendothelial migration, advancing our current understanding of the complexities of inflammatory responses.


Subject(s)
Cytokine Receptor gp130/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Inflammation Mediators/metabolism , Interleukin-23 Subunit p19/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Cytokine Receptor gp130/genetics , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Interleukin-23 Subunit p19/genetics , STAT3 Transcription Factor/genetics , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
6.
Nat Commun ; 6: 6576, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25807892

ABSTRACT

Angiogenesis produces primitive vascular networks that need pruning to yield hierarchically organized and functional vessels. Despite the critical importance of vessel pruning to vessel patterning and function, the mechanisms regulating this process are not clear. Here we show that EphrinB2, a well-known player in angiogenesis, is an essential regulator of endothelial cell death and vessel pruning. This regulation depends upon phosphotyrosine-EphrinB2 signalling repressing c-jun N-terminal kinase 3 activity via STAT1. JNK3 activation causes endothelial cell death. In the absence of JNK3, hyaloid vessel physiological pruning is impaired, associated with abnormal persistence of hyaloid vessels, defective retinal vasculature and microphthalmia. This syndrome closely resembles human persistent hyperplastic primary vitreus (PHPV), attributed to failed involution of hyaloid vessels. Our results provide evidence that EphrinB2/STAT1/JNK3 signalling is essential for vessel pruning, and that defects in this pathway may contribute to PHPV.


Subject(s)
Endothelial Cells/metabolism , Ephrin-B2/genetics , Mitogen-Activated Protein Kinase 10/metabolism , Neovascularization, Physiologic/genetics , Retinal Vessels/metabolism , STAT1 Transcription Factor/metabolism , Animals , Cell Proliferation , Cell Survival , Chromatin Immunoprecipitation , Flow Cytometry , Fluorescent Antibody Technique , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Immunoblotting , Immunoprecipitation , In Vitro Techniques , Mice , Mice, Knockout , Neovascularization, Pathologic/genetics , Persistent Hyperplastic Primary Vitreous/genetics , Signal Transduction
7.
Cancer Res ; 74(7): 2038-49, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24520074

ABSTRACT

Myeloid cells that orchestrate malignant progression in the tumor microenvironment offer targets for a generalized strategy to attack solid tumors. Through an analysis of tumor microenvironments, we explored an experimental model of lung cancer that uncovered a network of Dll4/Notch/TGF-ß1 signals that links myeloid cells to cancer progression. Myeloid cells attracted to the tumor microenvironment by the tumor-derived cytokines CCL2 and M-CSF expressed increased levels of the Notch ligand Dll4, thereby activating Notch signaling in the tumor cells and amplifying tumor-intrinsic Notch activation. Heightened Dll4/Notch signaling in tumor cells magnified TGF-ß-induced pSMAD2/3 signaling and was required to sustain TGF-ß-induced tumor cell growth. Conversely, Notch blockade reduced TGF-ß signaling and limited lung carcinoma tumor progression. Corroborating these findings, by interrogating RNAseq results from tumor and adjacent normal tissue in clinical specimens of human head and neck squamous carcinoma, we found evidence that TGF-ß/Notch crosstalk contributed to progression. In summary, the myeloid cell-carcinoma signaling network we describe uncovers novel mechanistic links between the tumor microenvironment and tumor growth, highlighting new opportunities to target tumors where this network is active.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Myeloid Cells/pathology , Neoplasms, Experimental/pathology , Receptors, Notch/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Tumor Microenvironment , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Cell Proliferation , Disease Progression , Female , Male , Mice
8.
PLoS One ; 8(11): e81445, 2013.
Article in English | MEDLINE | ID: mdl-24348920

ABSTRACT

The Eph receptor tyrosine kinases mediate juxtacrine signals by interacting "in trans" with ligands anchored to the surface of neighboring cells via a GPI-anchor (ephrin-As) or a transmembrane segment (ephrin-Bs), which leads to receptor clustering and increased kinase activity. Additionally, soluble forms of the ephrin-A ligands released from the cell surface by matrix metalloproteases can also activate EphA receptor signaling. Besides these trans interactions, recent studies have revealed that Eph receptors and ephrins coexpressed in neurons can also engage in lateral "cis" associations that attenuate receptor activation by ephrins in trans with critical functional consequences. Despite the importance of the Eph/ephrin system in tumorigenesis, Eph receptor-ephrin cis interactions have not been previously investigated in cancer cells. Here we show that in cancer cells, coexpressed ephrin-A3 can inhibit the ability of EphA2 and EphA3 to bind ephrins in trans and become activated, while ephrin-B2 can inhibit not only EphB4 but also EphA3. The cis inhibition of EphA3 by ephrin-B2 implies that in some cases ephrins that cannot activate a particular Eph receptor in trans can nevertheless inhibit its signaling ability through cis association. We also found that an EphA3 mutation identified in lung cancer enhances cis interaction with ephrin-A3. These results suggest a novel mechanism that may contribute to cancer pathogenesis by attenuating the tumor suppressing effects of Eph receptor signaling pathways activated by ephrins in trans.


Subject(s)
Ephrins/metabolism , Receptors, Eph Family/metabolism , Cell Line, Tumor , Ephrins/genetics , Humans , Immunoprecipitation , Protein Binding , Receptor, EphA2/genetics , Receptor, EphA2/metabolism , Receptor, EphA3/genetics , Receptor, EphA3/metabolism , Receptor, EphB4/genetics , Receptor, EphB4/metabolism , Receptors, Eph Family/genetics
9.
Blood ; 120(19): 4104-15, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-23007403

ABSTRACT

Formation of new vessels during development and in the mature mammal generally proceeds through angiogenesis. Although a variety of molecules and signaling pathways are known to underlie endothelial cell sprouting and remodeling during angiogenesis, many aspects of this complex process remain unexplained. Here we show that the transmembrane semaphorin6A (Sema6A) is expressed in endothelial cells, and regulates endothelial cell survival and growth by modulating the expression and signaling of VEGFR2, which is known to maintain endothelial cell viability by autocrine VEGFR signaling. The silencing of Sema6A in primary endothelial cells promotes cell death that is not rescued by exogenous VEGF-A or FGF2, attributable to the loss of prosurvival signaling from endogenous VEGF. Analyses of mouse tissues demonstrate that Sema6A is expressed in angiogenic and remodeling vessels. Mice with null mutations of Sema6A exhibit significant defects in hyaloid vessels complexity associated with increased endothelial cell death, and in retinal vessels development that is abnormally reduced. Adult Sema6A-null mice exhibit reduced tumor, matrigel, and choroidal angiogenesis compared with controls. Sema6A plays important roles in development of the nervous system. Here we show that it also regulates vascular development and adult angiogenesis.


Subject(s)
Neovascularization, Physiologic/genetics , Semaphorins/genetics , Semaphorins/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Choroid/blood supply , Fibroblast Growth Factor 2/metabolism , Gene Silencing , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Knockout , Retina/metabolism , Retina/pathology , Retinal Vessels/metabolism , Retinal Vessels/pathology , Semaphorins/deficiency , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
Adv Cancer Res ; 114: 21-57, 2012.
Article in English | MEDLINE | ID: mdl-22588055

ABSTRACT

Eph receptor tyrosine kinases and their Ephrin ligands represent an important signaling system with widespread roles in cell physiology and disease. Receptors and ligands in this family are anchored to the cell surface; thus Eph/Ephrin interactions mainly occur at sites of cell-to-cell contact. EphB4 and EphrinB2 are the Eph/Ephrin molecules that play essential roles in vascular development and postnatal angiogenesis. Analysis of expression patterns and function has linked EphB4/EphrinB2 to endothelial cell growth, survival, migration, assembly, and angiogenesis. Signaling from these molecules is complex, with the potential for being bidirectional, emanating both from the Eph receptors (forward signaling) and from the Ephrin ligands (reverse signaling). In this review, we describe recent advances on the roles of EphB/EphrinB protein family in endothelial cell function and outline potential approaches to inhibit pathological angiogenesis based on this understanding.


Subject(s)
Endothelial Cells/metabolism , Endothelial Cells/pathology , Ephrins/metabolism , Neovascularization, Physiologic/physiology , Receptors, Eph Family/metabolism , Animals , Endothelial Cells/physiology , Humans , Ligands , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
11.
Haematologica ; 97(6): 818-26, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22271895

ABSTRACT

BACKGROUND: Mobilization of hematopoietic stem/progenitor cells from the bone marrow to the peripheral blood by granulocyte colony-stimulating factor is the primary means to acquire stem cell grafts for hematopoietic cell transplantation. Since hematopoietic stem/progenitor cells represent a minority of all blood cells mobilized by granulocyte colony-stimulating factor, the underlying mechanisms need to be understood in order to develop selective drugs. DESIGN AND METHODS: We analyzed phenotypic, biochemical and genetic changes in bone marrow cell populations from granulocyte colony-stimulating factor-mobilized and control mice, and linked such changes to effective mobilization of hematopoietic stem/progenitor cells. RESULTS: We show that granulocyte colony-stimulating factor indirectly reduces expression of surface vascular cell adhesion molecule 1 on bone marrow hematopoietic stem/progenitor cells, stromal cells and endothelial cells by promoting the accumulation of microRNA-126 (miR126)-containing microvescicles in the bone marrow extracellular compartment. We found that hematopoietic stem/progenitor cells, stromal cells and endothelial cells readily incorporate these miR126-loaded microvescicles, and that miR126 represses vascular cell adhesion molecule 1 expression on bone marrow hematopoietic stem/progenitor cells, stromal cells and endothelial cells. In line with this, miR126-null mice displayed a reduced mobilization response to granulocyte colony-stimulating factor. CONCLUSIONS: Our results implicate miR126 in the regulation of hematopoietic stem/progenitor cell trafficking between the bone marrow and peripheral sites, clarify the role of vascular cell adhesion molecule 1 in granulocyte colony-stimulating factor-mediated mobilization, and have important implications for improved approaches to selective mobilization of hematopoietic stem/progenitor cells.


Subject(s)
Bone Marrow/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/drug effects , MicroRNAs/immunology , Vascular Cell Adhesion Molecule-1/genetics , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Movement/drug effects , Cell Movement/immunology , Cytoplasmic Vesicles/immunology , Cytoplasmic Vesicles/metabolism , Down-Regulation , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Extracellular Space/immunology , Extracellular Space/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/immunology , Vascular Cell Adhesion Molecule-1/immunology
12.
Cancer Res ; 72(5): 1157-69, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22237624

ABSTRACT

Endothelial-to-mesenchymal transition (EndMT) is now widely considered a pivotal contributor to cancer progression. In this study, we show that the Kaposi's sarcoma (KS)-associated herpesvirus (KSHV) is a sufficient cause of EndMT, potentially helping to explain the aggressiveness of KS that occurs commonly in AIDS patients. Upon KSHV infection, primary dermal microvascular endothelial cells lost expression of endothelial markers and acquired expression of mesenchymal markers, displaying new invasive and migratory properties along with increased survival. KSHV activated Notch-induced transcription factors Slug and ZEB1, and canonical Notch signaling was required for KSHV-induced EndMT. In contrast, KSHV did not utilize the TGFß signaling pathway, which has also been linked to EndMT. Within KS lesions, KSHV-infected spindle cells displayed features compatible with KSHV-induced EndMT including a complex phenotype of endothelial and mesenchymal properties, Notch activity, and nuclear ZEB1 expression. Our results show that KSHV engages the EndMT program to increase the invasiveness and survival of infected endothelial cells, traits that likely contribute to viral persistence and malignant progression. One important implication of our findings is that therapeutic approaches to disrupt the Notch pathway may offer novel approaches for KS treatment.


Subject(s)
Epithelial-Mesenchymal Transition , Herpesvirus 8, Human/pathogenicity , Sarcoma, Kaposi/pathology , Skin Neoplasms/pathology , Cell Survival , Humans , Neoplasm Invasiveness , Receptors, Notch/metabolism , Sarcoma, Kaposi/virology , Signal Transduction , Transforming Growth Factor beta/metabolism
13.
PLoS One ; 6(12): e28611, 2011.
Article in English | MEDLINE | ID: mdl-22194865

ABSTRACT

The EphB4 receptor tyrosine kinase together with its preferred ligand, ephrin-B2, regulates a variety of physiological and pathological processes, including tumor progression, pathological forms of angiogenesis, cardiomyocyte differentiation and bone remodeling. We previously reported the identification of TNYL-RAW, a 15 amino acid-long peptide that binds to the ephrin-binding pocked of EphB4 with low nanomolar affinity and inhibits ephrin-B2 binding. Although ephrin-B2 interacts promiscuously with all the EphB receptors, the TNYL-RAW peptide is remarkably selective and only binds to EphB4. Therefore, this peptide is a useful tool for studying the biological functions of EphB4 and for imaging EphB4-expressing tumors. Furthermore, TNYL-RAW could be useful for treating pathologies involving EphB4-ephrin-B2 interaction. However, the peptide has a very short half-life in cell culture and in the mouse blood circulation due to proteolytic degradation and clearance by the kidneys and reticuloendothelial system. To overcome these limitations, we have modified TNYL-RAW by fusion with the Fc portion of human IgG1, complexation with streptavidin or covalent coupling to a 40 KDa branched polyethylene glycol (PEG) polymer. These modified forms of TNYL-RAW all have greatly increased stability in cell culture, while retaining high binding affinity for EphB4. Furthermore, PEGylation most effectively increases peptide half-life in vivo. Consistent with increased stability, submicromolar concentrations of PEGylated TNYL-RAW effectively impair EphB4 activation by ephrin-B2 in cultured B16 melanoma cells as well as capillary-like tube formation and capillary sprouting in co-cultures of endothelial and epicardial mesothelial cells. Therefore, PEGylated TNYL-RAW may be useful for inhibiting pathological forms of angiogenesis through a novel mechanism involving disruption of EphB4-ephrin-B2 interactions between endothelial cells and supporting perivascular mesenchymal cells. Furthermore, the PEGylated peptide is suitable for other cell culture and in vivo applications requiring prolonged EphB4 receptor targeting.


Subject(s)
Microchemistry , Peptides/pharmacology , Polyethylene Glycols/metabolism , Receptor, EphB4/antagonists & inhibitors , Animals , Cell Line , Coculture Techniques , Culture Media/pharmacology , Ephrin-B2/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Half-Life , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Neovascularization, Physiologic/drug effects , Peptides/blood , Pericardium/cytology , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Binding/drug effects , Protein Stability/drug effects , Receptor, EphB4/metabolism
14.
Blood ; 118(8): 2105-15, 2011 08 25.
Article in English | MEDLINE | ID: mdl-21715308

ABSTRACT

A precise identification of adult human hemangioblast is still lacking. To identify circulating precursors having the developmental potential of the hemangioblast, we established a new ex vivo long-term culture model supporting the differentiation of both hematopoietic and endothelial cell lineages. We identified from peripheral blood a population lacking the expression of CD34, lineage markers, CD45 and CD133 (CD34⁻Lin⁻CD45⁻CD133⁻ cells), endowed with the ability to differentiate after a 6-week culture into both hematopoietic and endothelial lineages. The bilineage potential of CD34⁻Lin⁻CD45⁻CD133⁻ cells was determined at the single-cell level in vitro and was confirmed by transplantation into NOD/SCID mice. In vivo, CD34⁻Lin⁻CD45⁻CD133⁻ cells showed the ability to reconstitute hematopoietic tissue and to generate functional endothelial cells that contribute to new vessel formation during tumor angiogenesis. Molecular characterization of CD34⁻Lin⁻D45⁻CD133⁻ cells unveiled a stem cell profile compatible with both hematopoietic and endothelial potentials, characterized by the expression of c-Kit and CXCR4 as well as EphB4, EphB2, and ephrinB2. Further molecular and functional characterization of CD34⁻Lin⁻CD45⁻CD133⁻ cells will help dissect their physiologic role in blood and blood vessel maintenance and repair in adult life.


Subject(s)
Adult Stem Cells/cytology , Endothelial Cells/cytology , Hemangioblasts/cytology , AC133 Antigen , Adult , Adult Stem Cells/immunology , Adult Stem Cells/metabolism , Animals , Antigens, CD/metabolism , Antigens, CD34/metabolism , Cell Differentiation/immunology , Cell Differentiation/physiology , Cell Lineage/immunology , Cell Lineage/physiology , Coculture Techniques , Endothelial Cells/immunology , Endothelial Cells/metabolism , Glycoproteins/metabolism , Hemangioblasts/immunology , Hemangioblasts/metabolism , Hematopoiesis/immunology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , Leukocyte Common Antigens/metabolism , Mice , Neovascularization, Physiologic/immunology , Neovascularization, Physiologic/physiology , Peptides/metabolism
15.
Int J Cancer ; 129(1): 225-32, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-20830712

ABSTRACT

CXCR4 is a chemokine receptor implicated in the homing of cancer cells to target metastatic organs, which overexpress its ligand, stromal cell-derived factor (SDF)-1. To determine the efficacy of targeting CXCR4 on primary tumor growth and metastasis, we used a peptide inhibitor of CXCR4, CTCE-9908, that was administered in a clinically relevant approach using a transgenic breast cancer mouse model. We first performed a dosing experiment of CTCE-9908 in the PyMT mouse model, testing 25, 50 and 100 mg/kg versus the scrambled peptide in groups of 8-16 mice. We then combined CTCE-9908 with docetaxel or DC101 (an anti-VEGFR2 monoclonal antibody). We found that increasing doses of CTCE-9908 alone slowed the rate of tumor growth, with a 45% inhibition of primary tumor growth at 3.5 weeks of treatment with 50 mg/kg of CTCE-9908 (p = 0.005). Expression levels of VEGF were also found to be reduced by 42% with CTCE-9908 (p = 0.01). In combination with docetaxel, CTCE-9908 administration decreased tumor volume by 38% (p = 0.02), an effect that was greater than that observed with docetaxel alone. In combination with DC101, CTCE-9908 also demonstrated an enhanced effect compared to DC101 alone, with a 37% decrease in primary tumor volume (p = 0.01) and a 75% reduction in distant metastasis (p = 0.009). In combination with docetaxel or an anti-angiogenic agent, the anti-tumor and anti-metastatic effects of CTCE-9908 were markedly enhanced, suggesting potentially new effective combinatorial therapeutic strategies in the treatment of breast cancer, which include targeting the SDF-1/CXCR4 ligand/receptor pair.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Division/drug effects , Mammary Neoplasms, Experimental/pathology , Neoplasm Metastasis/prevention & control , Peptides/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Taxoids/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Blotting, Western , Disease Models, Animal , Docetaxel , Immunohistochemistry , Male , Mice , Mice, Transgenic
17.
Blood ; 115(19): 3970-9, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20203268

ABSTRACT

The transcription factor growth factor independence 1 (Gfi1) and the growth factor granulocyte colony-stimulating factor (G-CSF) are individually essential for neutrophil differentiation from myeloid progenitors. Here, we provide evidence that the functions of Gfi1 and G-CSF are linked in the regulation of granulopoiesis. We report that Gfi1 promotes the expression of Ras guanine nucleotide releasing protein 1 (RasGRP1), an exchange factor that activates Ras, and that RasGRP1 is required for G-CSF signaling through the Ras/mitogen-activated protein/extracellular signal-regulated kinase (MEK/Erk) pathway. Gfi1-null mice have reduced levels of RasGRP1 mRNA and protein in thymus, spleen, and bone marrow, and Gfi1 transduction in myeloid cells promotes RasGRP1 expression. When stimulated with G-CSF, Gfi1-null myeloid cells are selectively defective at activating Erk1/2, but not signal transducer and activator of transcription 1 (STAT1) or STAT3, and fail to differentiate into neutrophils. Expression of RasGRP1 in Gfi1-deficient cells rescues Erk1/2 activation by G-CSF and allows neutrophil maturation by G-CSF. These results uncover a previously unknown function of Gfi1 as a regulator of RasGRP1 and link Gfi1 transcriptional control to G-CSF signaling and regulation of granulopoiesis.


Subject(s)
DNA-Binding Proteins/physiology , Granulocyte Colony-Stimulating Factor/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Signal Transduction , Transcription Factors/physiology , Animals , Blotting, Western , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Flow Cytometry , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , Hematopoietic Stem Cells/metabolism , Immunoenzyme Techniques , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/metabolism
18.
Am J Pathol ; 175(1): 66-73, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19497995

ABSTRACT

The chemokine stromal cell-derived factor-1 (SDF-1) may function to attract CXCR4-expressing cancer cells to metastatic organs. We have previously demonstrated that low plasma SDF-1, a host-derived marker, increases distant metastatic risk in breast cancer. We therefore hypothesized that tumors overexpressing the SDF-1 receptor CXCR4 have an enhanced ability to metastasize in patients with low plasma SDF-1 levels. In this study, we determined the prognostic significance of activated CXCR4, or phosphorylated CXCR4 (p-CXCR4), and CXCR7, another receptor for SDF-1. Immunohistochemistry was performed on a tissue microarray built using 237 samples from the same cohort of patients for which we measured plasma SDF-1 levels. We found that the prognostic value of p-CXCR4 expression (hazard ratio or HR, 3.95; P = 0.004) was superior to total CXCR4 expression (HR, 3.20; P = 0.03). The rate of breast cancer-specific mortality was much higher in patients with both high p-CXCR4 expression and low plasma SDF-1 levels (HR, 5.96; P < 0.001) than either low plasma SDF-1 (HR, 3.59; P = 0.01) or high p-CXCR4 expression (HR, 3.83; P = 0.005) alone. The added prognostic value of low plasma SDF-1 was only effective in patients with high p-CXCR4 expression, and as such, provides clinical validation for modulation of the metastatic potential of tumor cells by an inherent host-derived metastatic risk factor.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chemokine CXCL12/blood , Receptors, CXCR4/biosynthesis , Blotting, Western , Breast Neoplasms/genetics , Female , Humans , Immunohistochemistry , Ligands , Neoplasm Invasiveness/genetics , Phosphorylation , Prognosis , Receptors, CXCR/biosynthesis , Receptors, CXCR/genetics , Receptors, CXCR4/genetics , Risk Factors , Tissue Array Analysis
19.
Blood ; 114(8): 1707-16, 2009 Aug 20.
Article in English | MEDLINE | ID: mdl-19411631

ABSTRACT

EphrinB transmembrane ligands and their cognate EphB receptor tyrosine kinases regulate vascular development through bidirectional cell-to-cell signaling, but little is known about the role of EphrinB during postnatal vascular remodeling. We report that EphrinB is a critical mediator of postnatal pericyte-to-endothelial cell assembly into vascular structures. This function is dependent upon extracellular matrix-supported cell-to-cell contact, engagement of EphrinB by EphB receptors expressed on another cell, and Src-dependent phosphorylation of the intracytoplasmic domain of EphrinB. Phosphorylated EphrinB marks angiogenic blood vessels in the developing and hypoxic retina, the wounded skin, and tumor tissue, and is detected at contact points between endothelial cells and pericytes. Furthermore, inhibition ofEphrinB activity prevents proper assembly of pericytes and endothelial cells into vascular structures. These results reveal a role for EphrinB signaling in orchestrating pericyte/endothelial cell assembly, and suggest that therapeutic targeting of EphrinB may prove useful for disrupting angiogenesis when it contributes to disease.


Subject(s)
Blood Vessels/growth & development , Endothelial Cells/physiology , Ephrins/physiology , Neovascularization, Physiologic/physiology , Pericytes/physiology , Animals , Animals, Newborn , Blood Vessels/metabolism , Bone Marrow Cells/physiology , Cell Adhesion/genetics , Cells, Cultured , Endothelial Cells/metabolism , Ephrin-B2/antagonists & inhibitors , Ephrin-B2/genetics , Ephrin-B2/physiology , Ephrins/genetics , Ephrins/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neovascularization, Physiologic/genetics , Pericytes/metabolism , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Receptors, Eph Family/physiology , Signal Transduction/physiology
20.
Anticancer Res ; 28(2A): 763-9, 2008.
Article in English | MEDLINE | ID: mdl-18507018

ABSTRACT

BACKGROUND: Increased expression of Eph receptors and their ephrin ligands has been implicated in promoting angiogenesis and tumour progression in several malignancies. Here the expression of mRNA for ephrin-B and EphB receptors in rhabdomyosarcoma (RMS) cell lines and primary tumours was investigated. MATERIALS AND METHODS: Expression of mRNA for ephrin-B and EphB receptors in RMS cell lines and primary tumours was measured by real-time RT-PCR and compared with the expression in normal striated muscle. RESULTS: A dysregulation of both ligands and receptors was found in all cell lines. In embryonal tumours, overexpression of ephrin-B1 correlated with overexpression of EphB1 (r = 0.97, p < 0.01) and EphB3 (r = 0.94, p < 0.05); overexpression of ephrin-B2 correlated with overexpression of EphB1 (r = 0.94, p < 0.05), EphB2 (r = 0.88, p < 0.01) and EphB4 (r = 0.76, p < 0.01). In alveolar tumours, no similar correlations were found. A correlation between EphB2 and EphB4 receptors was demonstrated in both tumour types, being positive in embryonal cases (r = 0.81, p < 0.01) and negative in alveolar (r = -1.00, p < 0.01). CONCLUSION: A global up-regulation of ephrin-B and EphB receptors in RMS tumours was found. The correlation between EphB2 and EphB4 receptors suggests a possible role for ephrin-B and EphB receptors in RMS development.


Subject(s)
Ephrins/genetics , Receptors, Eph Family/genetics , Rhabdomyosarcoma/genetics , Up-Regulation , Cell Line, Tumor , Ephrin-B1/metabolism , Ephrin-B2 , Humans , Ligands , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL