Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Clin Cancer Res ; 25(15): 4832-4845, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31010839

ABSTRACT

PURPOSE: Lung metastasis is an important cause of breast cancer-related deaths, in which SDF-1/CXCR4 signaling pathway plays a critical role. Single transmembrane protein LRP6 is viewed as an oncogene via activating the Wnt/ß-catenin signaling pathway. Our work aims to investigate the relationship between SDF-1/CXCR4 and LRP6 in breast cancer lung metastasis. EXPERIMENTAL DESIGN: We examined the expressions and functions of SDF-1/CXCR4 and LRP6 as well as their relationship in breast cancer in vitro and in vivo. RESULTS: LRP6 ectodomain (LRP6N) directly bound to CXCR4 and competitively prevented SDF-1 binding to CXCR4. LRP6N prevented SDF-1/CXCR4-induced metastasis to lung and prolonged survival in mice bearing breast tumors, whereas LRP6 knockdown activated SDF-1/CXCR4 signal transduction and promoted lung metastasis and tumor death. Furthermore, patients with breast cancer with high CXCR4 expression had poor prognosis, which was exacerbated by low LRP6 expression but improved by high LRP6 expression. Interestingly, a secreted LRP6N was found in the serum of mice and humans, which was downregulated by the onset of cancer metastasis in both mice bearing breast cancer as well as in patients with breast cancer. CONCLUSIONS: LRP6N might be a promising diagnostic marker for the early detection of breast cancer metastasis as well as an inhibitor of SDF-1/CXCR4-induced breast cancer metastasis. LRP6N also provides an interesting link between Wnt signaling and SDF-1/CXCR4 signaling, the two key pathways involved in cancer development.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/prevention & control , Chemokine CXCL12/adverse effects , Chemokine CXCL12/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Lung Neoplasms/prevention & control , Receptors, CXCR4/metabolism , Adolescent , Adult , Aged , Animals , Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , Receptors, CXCR4/administration & dosage , Survival Rate , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , Young Adult
2.
Theranostics ; 7(18): 4517-4536, 2017.
Article in English | MEDLINE | ID: mdl-29158842

ABSTRACT

PURPOSE: Gold standard beam radiation for glioblastoma (GBM) treatment is challenged by resistance phenomena occurring in cellular populations well prepared to survive or to repair damage caused by radiation. Among signals that have been linked with radio-resistance, the SDF1/CXCR4 axis, associated with cancer stem-like cell, may be an opportune target. To avoid the problem of systemic toxicity and blood-brain barrier crossing, the relevance and efficacy of an original system of local brain internal radiation therapy combining a radiopharmaceutical with an immuno-nanoparticle was investigated. EXPERIMENT DESIGN: The nanocarrier combined lipophilic thiobenzoate complexes of rhenium-188 loaded in the core of a lipid nanocapsule (LNC188Re) with a function-blocking antibody, 12G5 directed at the CXCR4, on its surface. The efficiency of 12G5-LNC188Re was investigated in an orthotopic and xenogenic GBM model of CXCR4-positive U87MG cells implanted in the striatum of Scid mice. RESULTS: We demonstrated that 12G5-LNC188Re single infusion treatment by convection-enhanced delivery resulted in a major clinical improvement in median survival that was accompanied by locoregional effects on tumor development including hypovascularization and stimulation of the recruitment of bone marrow derived CD11b- or CD68-positive cells as confirmed by immunohistochemistry analysis. Interestingly, thorough analysis by spectral imaging in a chimeric U87MG GBM model containing CXCR4-positive/red fluorescent protein (RFP)-positive- and CXCR4-negative/RFP-negative-GBM cells revealed greater confinement of DiD-labeled 12G5-LNCs than control IgG2a-LNCs in RFP compartments. Main conclusion: These findings on locoregional impact and targeting of disseminated cancer cells in tumor margins suggest that intracerebral active targeting of nanocarriers loaded with radiopharmaceuticals may have considerable benefits in clinical applications.


Subject(s)
Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Nanoparticles/administration & dosage , Radioisotopes/administration & dosage , Radiopharmaceuticals/administration & dosage , Receptors, CXCR4/administration & dosage , Rhenium/administration & dosage , Animals , Blood-Brain Barrier/metabolism , Brain/radiation effects , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Lipids/administration & dosage , Mice , Nanocapsules/administration & dosage , Neoplastic Stem Cells/radiation effects , Xenograft Model Antitumor Assays/methods
3.
Nanotechnology ; 28(1): 015102, 2017 Jan 06.
Article in English | MEDLINE | ID: mdl-27893441

ABSTRACT

Bacterial inclusion bodies are non-toxic, mechanically stable and functional protein amyloids within the nanoscale size range that are able to naturally penetrate into mammalian cells, where they deliver the embedded protein in a functional form. The potential use of inclusion bodies in protein delivery or protein replacement therapies is strongly impaired by the absence of specificity in cell binding and penetration, thus preventing targeting. To address this issue, we have here explored whether the genetic fusion of two tumor-homing peptides, the CXCR4 ligands R9 and T22, to an inclusion body-forming green fluorescent protein (GFP), would keep the interaction potential and the functionality of the fused peptides and then confer CXCR4 specificity in cell binding and further uptake of the materials. The fusion proteins have been well produced in Escherichia coli in their full-length form, keeping the potential for fluorescence emission of the partner GFP. By using specific inhibitors of CXCR4 binding, we have demonstrated that the engineered protein particles are able to penetrate CXCR4+ cells, in a receptor-mediated way, without toxicity or visible cytopathic effects, proving the availability of the peptide ligands on the surface of inclusion bodies. Since no further modification is required upon their purification, the biological production of genetically targeted inclusion bodies opens a plethora of cost-effective possibilities in the tissue-specific intracellular transfer of functional proteins through the use of structurally and functionally tailored soft materials.


Subject(s)
Amyloid/administration & dosage , Amyloid/chemistry , Inclusion Bodies/chemistry , Nanostructures/administration & dosage , Nanostructures/chemistry , Neoplasms/drug therapy , Amyloid/metabolism , Cell Line, Tumor , Drug Delivery Systems/methods , Escherichia coli/metabolism , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Inclusion Bodies/metabolism , Peptides/administration & dosage , Peptides/chemistry , Peptides/metabolism , Receptors, CXCR4/administration & dosage , Receptors, CXCR4/chemistry , Receptors, CXCR4/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
4.
Belo Horizonte; CCATES; 2016. tab.
Non-conventional in Portuguese | BRISA/RedTESA | ID: biblio-876499

ABSTRACT

CONTEXTO: O transplante de células estaminais hematopoiéticas é um tratamento eletivo, que utiliza a célula progenitora do próprio paciente ou de um doador compatível, para reestabelecimento da hematopoiese normal, após supressão medular com altas doses de quimioterapia. Pode ser classificado como alogênico, singênico ou autólogo, de acordo com o doador, a partir de células obtidas de três fontes: medula óssea, sangue periférico e sangue de cordão umbilical e placentário. TECNOLOGIA: Plerixafor. PERGUNTA: Existe evidência para o uso de plerixafor em pacientes com indicação de transplante autólogo de células estaminais hematopoiéticas? EVIDENCIAS: Foram encontradas duas revisões sistemáticas que avaliaram o medicamento para mobilização de células hematopoiéticas em pacientes com MM e LNH. Os resultados indicam que a adição de plerixafor ao tratamento com G-CSF aumenta de forma significativa a coleta de células hematopoiéticas em pacientes com linfoma em um menor prazo de tempo, mas que esse resultado é menor quando se avalia pacientes com mieloma. Entretanto, existem outras opções para o aumento das células hematopoiéticas para coleta, como o aumento da dose do G-CSF ou a associação do G-CSF com a quimioterapia, que também se mostraram eficazes. Os resultados de segurança e sobrevida não foram conclusivos. As revisões sistemáticas apresentam como limitações o fato de que poucos estudos avaliaram o uso de plerixafor, sendo que a maior parte dos estudos incluídos contou com financiamento do fabricante do produto. CONCLUSÕES: Plerixafor aumenta a mobilização de células estaminais hematopoiéticas em pacientes com linfoma e mieloma, o que também foi observado com aumento de dose do G-CSF ou a associação de G-CSF com quimioterapia para posterior transplante autólogo.


Subject(s)
Humans , Neoplastic Stem Cells/pathology , Receptors, CXCR4/administration & dosage , Transplantation, Autologous , Cost-Benefit Analysis/economics , Technology Assessment, Biomedical , Treatment Outcome
5.
Cytotherapy ; 16(9): 1280-93, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25108653

ABSTRACT

BACKGROUND AIMS: Inadequate engraftment of hematopoietic stem cells (HSCs) after in utero HSC transplantation (IUHSCT) remains a major obstacle for the prenatal correction of numerous hereditary disorders. HSCs express CXCR4 receptors that allow homing and engraftment in response to stromal-derived factor 1 (SDF-1) ligand present in the bone marrow stromal niche. Plerixafor, a mobilization drug, works through the interruption of the CXCR4-SDF-1 axis. METHODS: We used the fetal sheep large-animal model to test our hypotheses that (i) by administering plerixafor in utero before performing IUHSCT to release fetal HSCs and thus vacating recipient HSC niches, (ii) by using human mesenchymal stromal/stem cells (MSCs) to immunomodulate and humanize the fetal BM niches and (iii) by increasing the CXCR4(+) fraction of CD34(+) HSCs, we could improve engraftment. Human cord blood-derived CD34(+) cells and human bone marrow-derived MSCs were used for these studies. RESULTS: When MSCs were transplanted 1 week before CD34(+) cells with plerixafor treatment, we observed 2.80% donor hematopoietic engraftment. Combination of this regimen with additional CD34(+) cells at the time of MSC infusion increased engraftment levels to 8.77%. Next, increasing the fraction of CXCR4(+) cells in the CD34(+) population albeit transplanting at a late gestation age was not beneficial. Our results show engraftment of both lymphoid and myeloid lineages. CONCLUSIONS: Prior MSC and HSC cotransplantation followed by manipulation of the CXCR4-SDF-1 axis in IUHSCT provides an innovative conceptual approach for conferring competitive advantage to donor HSCs. Our novel approach could provide a clinically relevant approach for enhancing engraftment early in the fetus.


Subject(s)
Genetic Diseases, Inborn/therapy , Hematopoietic Stem Cell Transplantation , Heterocyclic Compounds/administration & dosage , Mesenchymal Stem Cells/physiology , Receptors, CXCR4/administration & dosage , Animals , Antigens, CD34/metabolism , Benzylamines , Cell Differentiation , Cell Lineage , Cells, Cultured , Chemokine CXCL12/metabolism , Clinical Protocols , Cyclams , Disease Models, Animal , Female , Fetus , Graft Survival , Humans , Immunomodulation , Pregnancy , Sheep
6.
Blood ; 124(2): 277-86, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24735964

ABSTRACT

Megakaryocyte (MK) development in the bone marrow progresses spatially from the endosteal niche, which promotes MK progenitor proliferation, to the sinusoidal vascular niche, the site of terminal maturation and thrombopoiesis. The chemokine stromal cell-derived factor-1 (SDF-1), signaling through CXCR4, is implicated in the maturational chemotaxis of MKs toward sinusoidal vessels. Here, we demonstrate that both IV administration of SDF-1 and stabilization of endogenous SDF-1 acutely increase MK-vasculature association and thrombopoiesis with no change in MK number. In the setting of radiation injury, we find dynamic fluctuations in marrow SDF-1 distribution that spatially and temporally correlate with variations in MK niche occupancy. Stabilization of altered SDF-1 gradients directly affects MK location. Importantly, these SDF-1-mediated changes have functional consequences for platelet production, as the movement of MKs away from the vasculature decreases circulating platelets, while MK association with the vasculature increases circulating platelets. Finally, we demonstrate that manipulation of SDF-1 gradients can improve radiation-induced thrombocytopenia in a manner additive with earlier TPO treatment. Taken together, our data support the concept that SDF-1 regulates the spatial distribution of MKs in the marrow and consequently circulating platelet numbers. This knowledge of the microenvironmental regulation of the MK lineage could lead to improved therapeutic strategies for thrombocytopenia.


Subject(s)
Cell Movement , Chemokine CXCL12/physiology , Megakaryocytes/cytology , Megakaryocytes/physiology , Radiation Injuries, Experimental , Stem Cell Niche/genetics , Thrombopoiesis/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Bone Marrow Cells/radiation effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/radiation effects , Cell Movement/drug effects , Cell Movement/genetics , Cell Movement/radiation effects , Cells, Cultured , Chemokine CXCL12/administration & dosage , Female , Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/drug effects , Megakaryocyte Progenitor Cells/physiology , Megakaryocyte Progenitor Cells/radiation effects , Megakaryocytes/drug effects , Megakaryocytes/radiation effects , Mice , Mice, Inbred C57BL , Radiation Injuries, Experimental/genetics , Radiation Injuries, Experimental/pathology , Receptors, CXCR4/administration & dosage , Receptors, CXCR4/metabolism , Stem Cell Niche/drug effects , Stem Cell Niche/radiation effects , Thrombopoiesis/drug effects , Thrombopoiesis/radiation effects
7.
Clin Exp Metastasis ; 31(3): 339-49, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24390633

ABSTRACT

Current combined surgical and neo-adjuvant chemotherapy of primary metastatic osteosarcoma (OS) is ineffective, reflected by a 5-year survival rate of affected patients of less than 20 %. Studies in experimental OS metastasis models pointed to the CXCR4/CXCL12 homing axis as a novel target for OS metastasis-suppressive treatment. The present study investigated for the first time the CXCR4-blocking principle in a spontaneously metastasizing human 143B OS cell line-derived orthotopic xenograft mouse model. The highly metastatic 143B cells, unlike the parental non-metastatic HOS cells, express functional CXCR4 receptors at the cell surface, as revealed in this study by RT/PCR of gene transcripts, by FACS analysis with the monoclonal anti CXCR4 antibody 12G5 (mAb 12G5) and by CXCL12 time- and dose-dependent stimulation of AKT and ERK phosphorylation. A significantly (p < 0.05) higher CXCL12 dose-dependent chemotactic response of 143B compared to HOS cells in a Boyden chamber trans-well migration assay suggested a crucial role of the CXCL12/CXCR4 homing axis in 143B cell lung metastasis. Repetitive treatment of mice with 143B cell-derived intratibial tumors given intravenous bolus injections of mAb12G5 indeed inhibited significantly (p < 0.01) the number of X-gal-stainable lung micrometastases of lacZ-transduced 143B cells. Antibody treatment had also a mild inhibitory effect on primary tumor growth associated with remarkably less osteolysis, but it did not affect the number of developing lung macrometastases. In conclusion, these results demonstrate considerable potential of high-affinity CXCR4-blocking agents for OS tumor cell homing suppressive treatment in metastasizing OS complementary to current (neo)-adjuvant chemotherapy.


Subject(s)
Antibodies/administration & dosage , Lung Neoplasms/secondary , Neoplasm Metastasis/drug therapy , Receptors, CXCR4/administration & dosage , Animals , Antibodies/immunology , Cell Line, Tumor , Disease Models, Animal , Humans , Immunotherapy , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Osteosarcoma/drug therapy , Osteosarcoma/immunology , Osteosarcoma/pathology , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/immunology , Signal Transduction , Xenograft Model Antitumor Assays
8.
Cytotherapy ; 15(7): 840-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23623275

ABSTRACT

BACKGROUND AIMS: The interaction between stromal cell-derived factor (SDF)-1 and its receptor CXCR4 is one of the mechanisms by which mesenchymal stromal cells (MSCs) are recruited to sites of injury. SDF-1 is upregulated in damaged tissues, but because the surface expression of CXCR4 on cultured MSCs is low, we investigated whether the delivery of CXCR4 into MSCs with the use of the cationic liposomal reagent IBAfect would increase their migration toward SDF-1. METHODS: We examined (i) the effect of MSC confluency, passage number, duration of transfection and amount of IBAfect and plasmid on transfection efficiency as determined by flow cytometric analysis of CXCR4 and (ii) whether IBAfect-mediated CXCR4 transfection affected the viability, proliferation and differentiation of MSCs as well as their response toward an SDF-1 gradient in a trans-Matrigel migration assay. RESULTS: We found that transfection efficiency of up to 40% was achieved after 24-h transfection of 50% confluent MSCs (at passage 4) with an IBAfect:plasmid ratio of 3.6 µL:0.6 µg, and CXCR4 transcript expression in transfected MSCs was 10(5)-fold higher than in non-transfected cells. Transfected MSCs retained their ability to differentiate to osteocytes and chondrocytes but had lower proliferation. Importantly, overexpression of surface CXCR4 with the use of IBAfect significantly increased (>3-fold) the number of cells migrating toward an SDF-1 gradient relative to cells migrating to media alone, compared with non-transfected cells (1.3-fold). CONCLUSIONS: Our results suggest that IBAfect-mediated delivery of CXCR4 into MSCs is a highly efficient technique that may be useful for enhancing the recruitment of systemically infused MSCs for tissue repair.


Subject(s)
Chemokine CXCL12/metabolism , Fetal Blood/cytology , Mesenchymal Stem Cells/cytology , Receptors, CXCR4/metabolism , Cell Differentiation/genetics , Cell Movement/genetics , Cells, Cultured , Chemokine CXCL12/genetics , Chondrocytes/cytology , Fetal Blood/metabolism , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Liposomes/administration & dosage , Liposomes/chemistry , Mesenchymal Stem Cells/metabolism , Osteocytes/cytology , Receptors, CXCR4/administration & dosage , Receptors, CXCR4/genetics , Transfection , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
SELECTION OF CITATIONS
SEARCH DETAIL