Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 328
Filter
1.
J Mol Histol ; 52(6): 1155-1164, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34642827

ABSTRACT

Cell-based therapeutics bring great hope in areas of unmet medical needs. Mesenchymal stem cells (MSCs) have been suggested to facilitate neovascularization mainly by paracrine action. Endothelial progenitor cells (EPCs) can migrate to ischemic sites and participate in angiogenesis. The combination cell therapy that includes MSCs and EPCs has a favorable effect on ischemic limbs. However, the mechanism of combination cell therapy remains unclear. Herein, we investigate whether stromal cell-derived factor (SDF)-1 secreted by MSCs contributes to EPC migration to ischemic sites via CXCR4/Phosphoinositide 3-Kinases (PI3K)/protein kinase B (termed as AKT) signaling pathway. First, by a "dual-administration" approach, intramuscular MSC injections were supplemented with intravenous Qdot® 525 labeled-EPC injections in the mouse model of hind limb ischemia. Then, the mechanism of MSC effect on EPC migration was detected by the transwell system, tube-like structure formation assays, western blot assays in vitro. Results showed that the combination delivery of MSCs and EPCs enhanced the incorporation of EPCs into the vasculature and increased the capillary density in mouse ischemic hind limb. The numbers of CXCR4-positive EPCs increased after incubation with MSC-conditioned medium (CM). MSCs contributed to EPC migration and tube-like structure formation, both of which were suppressed by AMD3100 and wortmannin. Phospho-AKT induced by MSC-CM was attenuated when EPCs were pretreated with AMD3100 and wortmannin. In conclusion, we confirmed that MSCs contributes to EPC migration, which is mediated via CXCR4/PI3K/AKT signaling pathway.


Subject(s)
Chemokine CXCL12/biosynthesis , Endothelial Progenitor Cells/metabolism , Mesenchymal Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR4/metabolism , Signal Transduction , Animals , Cell Communication , Cell Movement , Cells, Cultured , Disease Models, Animal , Extremities/blood supply , Extremities/pathology , Immunophenotyping , Ischemia/etiology , Ischemia/metabolism , Ischemia/pathology , Ischemia/therapy , Mesenchymal Stem Cell Transplantation/methods , Mice , Neovascularization, Physiologic/genetics , Phosphorylation
2.
Transfus Apher Sci ; 60(5): 103240, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34404617

ABSTRACT

BACKGROUND AND OBJECTIVES: Stem cell transplantation is a growing treatment strategy for most malignant and non- malignant hematological diseases. Plerixafor and granulocyte colony stimulating factor (G-CSF) are usually used in mobilization regimens to increase the CD34+ cell count in the harvest. Heparin is a sulphated glycosaminoglycated polymer with 12-15 kDa mass. Heparin inhibits the CXCR4/SDF1 axis, as does plerixafor. In this study, our aim was to investigate the effect of using heparin on stem cell mobilization and harvesting. MATERIALS AND METHODS: We administered 5000 units of unfractioned heparin intravenously in 150 mL (mL) of isotonic sodium chloride solution, 15 min before the stem cell harvesting procedure to 141 patients who underwent bone marrow transplantation between the years of 2018 and 2019 at our Stem Cell Transplantation Unit. Thirty patients were included as a control group, and they were not given heparin. The study population included patients with multiple myeloma and lymphoma equally in each group. RESULTS: In all patients hematopoeitic stem cells were successfully harvested in a single cycle of apheresis. In multiple myeloma patients who received heparin, the mean collected CD34+ cell number was 8 × 106/kg, and the mean CD34+ cell number yield was 12,555/µl. In the control group, the mean collected CD34+ cell number was 4,2 × 106/kg, and mean CD34+ cell number in yield was 492/µl. In lymphoma patients who received heparin, the mean collected CD34+ cell number was 6,8 × 106/kg, and the mean CD34+ cell number was 1421/µl. In the control group the mean collected CD34+ cell number was 4,3 × 106/kg, and the mean CD34+ cell number was 358/µl. The effect of heparin on the collected stem cell number in both myeloma and lymphoma patients was statistically significant (p < 0.01). CONCLUSIONS: Our results have shown that heparin increases harvested stem cell numbers significantly. Heparin may be a promising agent for stem cell harvesting.


Subject(s)
Benzylamines/administration & dosage , Cyclams/administration & dosage , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Mobilization/instrumentation , Hematopoietic Stem Cell Mobilization/methods , Heparin/therapeutic use , Stem Cells/cytology , Adult , Aged , Antigens, CD34/biosynthesis , Bone Marrow Transplantation , Chemokine CXCL12/biosynthesis , Diffusion of Innovation , Female , Humans , Lymphoma/therapy , Male , Middle Aged , Multiple Myeloma/therapy , Peripheral Blood Stem Cell Transplantation/methods , Receptors, CXCR4/biosynthesis , Retrospective Studies , Young Adult
3.
Mol Neurobiol ; 58(11): 5421-5436, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33983546

ABSTRACT

Maintaining an intact pool of neural progenitor cells (NPCs) is crucial for generating new and functionally active neurons. Methamphetamine (METH) can exacerbate the HIV-induced deficit of adult neurogenesis; however, potential mechanisms of this influence are still poorly understood. In the present study, we present evidence that chronic exposure to METH combined with brain infection by EcoHIV results in enhanced proliferation of NPCs in the subventricular zone (SVZ) in mice. This effect was long-lasting as it was preserved ex vivo in NPCs isolated from the exposed mice over several passages in the absence of additional treatments. Increased proliferation in response to METH plus HIV was associated with dysregulation of cyclin B1 and cyclin D. Transcriptomic studies indicated that 27 out of the top 30 differentially expressed genes in response to METH plus EcoHIV were targets of the forkhead box O transcriptional factor (FOXO) and primarily FOXO3. Additional ex vivo studies and in vitro experiments using human NPCs exposed to METH and infected with HIV revealed upregulation of the CXCL12-CXCR4 axis, leading to activation of downstream pAkt and pErk, the pathways that can phosphorylate FOXO3 and force its exports from the nuclei into the cytoplasm. Indeed, nuclear expulsion of FOXO3 was demonstrated both in mice exposed to METH and infected with EcoHIV and in cell cultures of human NPCs. These results provide novel information that exposure to METH combined with HIV infection can induce aberrant proliferation of SVZ-derived NPCs and identifies CXCL12-CXCR4-Akt-1-mediated phosphorylation of FOXO3 as the mechanism responsible for this effect.


Subject(s)
Forkhead Box Protein O3/physiology , HIV-1/physiology , Methamphetamine/toxicity , Neural Stem Cells/drug effects , AIDS Dementia Complex/complications , AIDS Dementia Complex/virology , Animals , Brain/pathology , Brain/virology , Cell Cycle , Cell Division , Cells, Cultured , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/genetics , Chromones/pharmacology , Disease Models, Animal , Gene Expression Regulation, Viral , HIV Infections/complications , HIV Infections/pathology , Humans , Lateral Ventricles/pathology , Male , Methamphetamine/pharmacology , Mice , Mice, Inbred C57BL , Morpholines/pharmacology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neural Stem Cells/virology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, CXCR4/biosynthesis , Receptors, CXCR4/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Substance-Related Disorders/complications
4.
Sci Rep ; 11(1): 775, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436863

ABSTRACT

Since colorectal cancer (CRC) remains one of the most common malignancies, a tremendous amount of studies keep taking place in this field. Over the past 25 years, a notable part of the scientific community has focused on the association between the immune system and colorectal cancer. A variety of studies have shown that high densities of infiltrating CD8+ T-cells are associated with improved disease-free and overall survival in CRC. Stromal cell-derived factor-1 (SDF-1) is a protein that regulates leukocyte trafficking and is variably expressed in several healthy and malignant tissues. There is strong evidence that SDF-1 has a negative prognostic impact on a variety of solid tumors. However, the existing data do not provide sufficient evidence that the expression of SDF-1 has an influence on CRC. Knowing nowadays, that the microenvironment plays a crucial role in the development of cancer, we hypothesized that the expression of SDF-1 in CRC could influence the prognostic significance of CD8+ T-cells, as an indicator of the essential role of the immune microenvironment in cancer development. Therefore, we explored the combined prognostic significance of CD8+ T-cell density and SDF-1 expression in a large CRC collective. We analyzed a tissue microarray of 613 patient specimens of primary CRCs by immunohistochemistry (IHC) for the CD8 + T-cells density and the expression of SDF-1 by tumor cells and tumor-infiltrating immune cells. Besides, we analyzed the expression of SDF-1 at the RNA level in The Cancer Genome Atlas cohort. We found that the combined high CD8+ T-cell infiltration and expression of SDF-1 shows a favorable 5-year overall survival rate (66%; 95% CI 48-79%) compared to tumors showing a high expression of CD8+ T-cell only (55%; 95% CI 45-64%; p = 0.0004). After stratifying the patients in nodal negative and positive groups, we found that the prognostic significance of CD8+ T-cell density in nodal positive colorectal cancer depends on SDF-1 expression. Univariate and multivariate Hazard Cox regression survival analysis considering the combination of both markers revealed that the combined high expression of SDF-1 and CD8+ T-cell density was an independent, favorable, prognostic marker for overall survival (HR = 0.34, 95% CI 0.17-0.66; p = 0.002 and HR = 0.45, 95% CI 0.23-0.89; p = 0.021, respectively). In our cohort there was a very weak correlation between SDF-1 and CD8+ T-cells (rs = 0.13, p = 0.002) and in the trascriptomic expression of these two immune markers display a weak correlation (rs = 0.28, p < 0.001) which was significantly more pronounced in stage III cancers (rs = 0.40, p < 0.001). The combination of high CD8+ T-cell density and expression of SDF-1 represents an independent, favorable, prognostic condition in CRC, mostly in patients with stage III disease.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokine CXCL12/biosynthesis , Colorectal Neoplasms/immunology , Tumor Microenvironment/immunology , Adult , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/pathology , Chemokine CXCL12/immunology , Cohort Studies , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Databases, Genetic , Female , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Survival Rate
5.
J Leukoc Biol ; 110(4): 711-722, 2021 10.
Article in English | MEDLINE | ID: mdl-33438259

ABSTRACT

High mobility group (HMG)A proteins are nonhistone chromatin proteins that bind to the minor groove of DNA, interact with transcriptional machinery, and facilitate DNA-directed nuclear processes. HMGA1 has been shown to regulate genes involved with systemic inflammatory processes. We hypothesized that HMGA1 is important in the function of mesenchymal stromal cells (MSCs), which are known to modulate inflammatory responses due to sepsis. To study this process, we harvested MSCs from transgenic (Tg) mice expressing a dominant-negative (dn) form of HMGA1 in mesenchymal cells. MSCs harvested from Tg mice contained the dnHMGA1 transgene, and transgene expression did not change endogenous HMGA1 levels. Immunophenotyping of the cells, along with trilineage differentiation revealed no striking differences between Tg and wild-type (WT) MSCs. However, Tg MSCs growth was decreased compared with WT MSCs, although Tg MSCs were more resistant to oxidative stress-induced death and expressed less IL-6. Tg MSCs administered after the onset of Escherichia coli-induced sepsis maintained their ability to improve survival when given in a single dose, in contrast with WT MSCs. This survival benefit of Tg MSCs was associated with less tissue cell death, and also a reduction in tissue neutrophil infiltration and expression of neutrophil chemokines. Finally, Tg MSCs promoted bacterial clearance and enhanced neutrophil phagocytosis, in part through their increased expression of stromal cell-derived factor-1 compared with WT MSCs. Taken together, these data demonstrate that expression of dnHMGA1 in MSCs provides a functional advantage of the cells when administered during bacterial sepsis.


Subject(s)
Genes, Dominant , HMGA1a Protein/genetics , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Sepsis/pathology , Sepsis/therapy , Transgenes , Adipocytes/cytology , Animals , Cell Death , Cell Proliferation , Cell Survival , Chemokine CXCL12/biosynthesis , Escherichia coli/physiology , HMGA1a Protein/metabolism , Inflammation/pathology , Interleukin-6/biosynthesis , Male , Mice, Inbred C57BL , Mice, Transgenic , Neutrophil Infiltration , Neutrophils/metabolism , Oxidative Stress , Phagocytosis , Sepsis/microbiology
6.
Int J Hematol ; 113(3): 348-361, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33398631

ABSTRACT

Histidine decarboxylase (HDC), a histamine synthase, is expressed in various hematopoietic cells and is induced by hematopoietic cytokines such as granulocyte colony-stimulating factor (G-CSF). We previously showed that nitrogen-containing bisphosphonate (NBP)-treatment induces extramedullary hematopoiesis via G-CSF stimulation. However, the function of HDC in NBP-induced medullary and extramedullary hematopoiesis remains unclear. Here, we investigated changes in hematopoiesis in wild-type and HDC-deficient (HDC-KO) mice. NBP treatment did not induce anemia in wild-type or HDC-KO mice, but did produce a gradual increase in serum G-CSF levels in wild-type mice. NBP treatment also enhanced Hdc mRNA expression and erythropoiesis in the spleen and reduced erythropoiesis in bone marrow and the number of vascular adhesion molecule 1 (VCAM-1)-positive macrophages in wild-type mice, as well as increased the levels of hematopoietic progenitor cells and proliferating cells in the spleen and enhanced expression of bone morphogenetic protein 4 (Bmp4), CXC chemokine ligand 12 (Cxcl12), and hypoxia inducible factor 1 (Hif1) in the spleen. However, such changes were not observed in HDC-KO mice. These results suggest that histamine may affect hematopoietic microenvironments of the bone marrow and spleen by changing hematopoiesis-related factors in NBP-induced extramedullary hematopoiesis.


Subject(s)
Alendronate/antagonists & inhibitors , Bone Marrow/drug effects , Cellular Microenvironment/drug effects , Hematopoiesis, Extramedullary/drug effects , Histidine Decarboxylase/deficiency , Spleen/drug effects , Alendronate/pharmacology , Alendronate/toxicity , Anemia/chemically induced , Animals , Bone Marrow/metabolism , Bone Morphogenetic Protein 4/biosynthesis , Bone Morphogenetic Protein 4/genetics , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/genetics , Enzyme Induction/drug effects , Erythroid Cells/pathology , Flow Cytometry , Granulocyte Colony-Stimulating Factor/blood , Histamine/biosynthesis , Histidine Decarboxylase/biosynthesis , Histidine Decarboxylase/genetics , Histidine Decarboxylase/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Spleen/metabolism
7.
Biotechnol Bioeng ; 118(2): 725-736, 2021 02.
Article in English | MEDLINE | ID: mdl-33064302

ABSTRACT

Enhancing angiogenesis is the prime target of current biomaterial-based wound healing strategies. However, these approaches largely overlook the angiogenic role of the cells of the nervous system. Therefore, we explored the role of a collagen-chondroitin sulfate scaffold functionalized with a proangiogenic gene stromal-derived factor-1α (SDF-1α)-an SDF-1α gene-activated scaffold on the functional regulation of human Schwann cells (SCs). A preliminary 2D study was conducted by delivering plasmids encoding for the SDF-1α gene into a monolayer of SCs using polyethyleneimine-based nanoparticles. The delivery of the SDF-1α gene into the SCs enhanced the production of proangiogenic vascular endothelial growth factor (VEGF). Subsequently, we investigated the impact of SDF-1α gene-activated scaffold (3D) on the SCs for 2 weeks, using a gene-free scaffold as control. The transfection of the SCs within the gene-activated scaffold resulted in transient overexpression of SDF-1α transcripts and triggered the production of bioactive VEGF that enhanced endothelial angiogenesis. The overexpression of SDF-1α also caused transient activation of the transcription factor c-Jun and supported the differentiation of SCs towards a repair phenotype. This was characterized by elevated expression of neurotrophin receptor p75NGFR. During this developmental stage, the SCs also performed an extensive remodelling of the basement matrix (fibronectin, collagen IV, and laminin) to enrich their environment with the pro-neurogenic matrix protein laminin, revealing an enhanced pro-neurogenic behavior. Together, this study shows that SDF-1α gene-activated scaffold is a highly bioinstructive scaffold capable of enhancing proangiogenic regenerative response in human SCs for improved wound healing.


Subject(s)
Cell Differentiation , Chemokine CXCL12 , Collagen/chemistry , Schwann Cells/metabolism , Tissue Scaffolds/chemistry , Wound Healing , Cells, Cultured , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/genetics , Endothelial Cells/metabolism , Gene Transfer Techniques , Humans , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/biosynthesis
8.
Mol Med Rep ; 23(1)2021 01.
Article in English | MEDLINE | ID: mdl-33215217

ABSTRACT

Stromal cell­derived factor­1 (SDF­1) is a chemokine involved in neuronal differentiation, as well as proliferation and migration. In the present study, the effects of recombinant SDF­1 on neurite outgrowth for nerve regeneration and engineering were evaluated in PC­12 cells. The effects of purified SDF­1 protein on cell toxicity, proliferation and migration were also assessed. SDF­1 significantly augmented cell proliferation in a dose­dependent manner, with low cytotoxicity in PC­12 cells. Cell migration also increased in the presence of SDF­1. SDF­1 significantly increased neurite number and length, compared with the control (untreated cells). Neurofilament mRNA levels, which are involved in neuronal differentiation, were also significantly upregulated in the presence of SDF­1. These results suggested that SDF­1 might prove useful for tissue engineering through induction of neuronal differentiation.


Subject(s)
Chemokine CXCL12/biosynthesis , Chemokine CXCL12/physiology , Neurites/metabolism , Neuronal Outgrowth/physiology , Animals , Cell Movement/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Chemokine CXCL12/isolation & purification , Intermediate Filaments/genetics , Microscopy, Confocal , Microscopy, Electron, Scanning , PC12 Cells , RNA, Messenger/metabolism , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Wound Healing/physiology
9.
Front Endocrinol (Lausanne) ; 11: 566761, 2020.
Article in English | MEDLINE | ID: mdl-33362712

ABSTRACT

Background: Nonfunctioning pituitary neuroendocrine tumor (NF-PitNET) is difficult to resect. Except for surgery, there is no effective treatment for NF-PitNET. MicroRNA-134 (miR-134) has been reported to inhibit proliferation and invasion ability of tumor cells. Herein, the mechanism underlying the effect of miR-134 on alleviating NF-PitNET tumor cells growth is explored. Methods: Mouse pituitary αT3-1 cells were transfected with miR-134 mimics and inhibitor, followed by treatment with stromal cell-derived factor-1α (SDF-1α) in vitro. MiR-134 expression level: we used quantitative real-time PCR (qRT-PCR) to detect the expression of miR-134. Cell behavior level: cell viability and invasion ability were assessed using a cell counting kit-8 (CCK8) assay and Transwell invasion assay respectively. Cytomolecular level: tumor cell proliferation was evaluated by Ki-67 staining; propidium iodide (PI) staining analyzed the effect of miR-134 on cell cycle arrest; western blot analysis and immunofluorescence staining evaluated tumor migration and invasive ability. Additionally, we collected 27 NF-PitNET tumor specimens and related clinical data. The specimens were subjected to qRT-PCR to obtain the relative miR-134 expression level of each specimen; linear regression analysis was used to analyze the miR-134 expression level in tumor specimens and the age of the NF-PitNET population, gender, tumor invasion, prognosis, and other indicators. Results: In vitro experiment, miR-134 was observed to significantly inhibit αT3-1 cells proliferation characterized by inhibited cell viability and expressions of vascular endothelial growth factor A (VEGFA) and cell cycle transition from G1 to S phase (P < 0.01). VEGFA was verified as a target of miR-134. Additionally, miR-134-induced inhibition of αT3-1 cell proliferation and invasion was attenuated by SDF-1α and VEGFA overexpression (P < 0.01). In primary NF-PitNET tumor analysis, miR-134 expression level was negatively correlated with tumor invasion (P = 0.003). Conclusion: The regulation of the SDF-1α/miR-134/VEGFA axis represents a novel mechanism in the pathogenesis of NF-PitNETs and may serve as a potential therapeutic target for the treatment of NF-PitNETs.


Subject(s)
Adenoma/metabolism , Chemokine CXCL12/biosynthesis , MicroRNAs/biosynthesis , Neuroendocrine Tumors/metabolism , Pituitary Neoplasms/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Adenoma/genetics , Adenoma/pathology , Adult , Animals , Cell Line, Tumor , Chemokine CXCL12/genetics , Female , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Male , Mice , MicroRNAs/genetics , Middle Aged , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Pituitary Gland/cytology , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Vascular Endothelial Growth Factor A/genetics
10.
Front Immunol ; 11: 2176, 2020.
Article in English | MEDLINE | ID: mdl-33013914

ABSTRACT

Chemokine receptor cxcr4 and its ligand cxcl12 have evolved two paralogs in the teleost lineage. In this study, we have identified four duplicated cxcr4 and cxcl12 genes from hexaploid gibel carp, Carassius gibelio, respectively. Cgcxcr4bs and Cgcxcl12as were dynamically and differentially expressed in immune-related tissues, and significantly up-regulated in head kidney and spleen after crucian carp herpesvirus (CaHV) infection. Blocking Cxcr4/Cxcl12 axis by injecting AMD3100 brought more severe bleeding symptom and lower survival rate in CaHV-infected fish. AMD3100 treatment also suppressed the up-regulation of key antiviral genes in head kidney and spleen, and resulted in more acute replication of CaHV in vivo. Consistently, the similar suppression of up-regulated expression of key antiviral genes were also observed in CAB cells treated by AMD3100 after poly(I:C) stimulation. Finally, MAPK3 and JAK/STAT were identified as the possible pathways that CgCxcr4s and CgCxcl12s participate in to promote the antiviral response in vitro.


Subject(s)
Carps/genetics , Chemokine CXCL12/genetics , Fish Diseases/genetics , Herpesviridae Infections/veterinary , Herpesviridae/physiology , Receptors, CXCR4/genetics , Amino Acid Sequence , Animals , Antiviral Agents/pharmacology , Base Sequence , Benzylamines/pharmacology , Carps/immunology , Carps/virology , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/immunology , Conserved Sequence , Cyclams/pharmacology , DNA, Complementary/genetics , Fish Diseases/immunology , Fish Diseases/virology , Gene Duplication , Gene Expression Regulation , Head Kidney/immunology , Head Kidney/metabolism , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Organ Specificity , Phylogeny , Poly I-C/pharmacology , Polyploidy , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/biosynthesis , Receptors, CXCR4/immunology , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction/immunology , Spleen/immunology , Spleen/metabolism , Virus Replication
11.
J Cell Mol Med ; 24(18): 10589-10603, 2020 09.
Article in English | MEDLINE | ID: mdl-32761803

ABSTRACT

Low-energy shock wave (LESW) has been recognized as a promising non-invasive intervention to prevent the organs or tissues against ischaemia reperfusion injury (IRI), whereas its effect on kidney injury is rarely explored. To investigate the protective role of pretreatment with LESW on renal IRI in rats, animals were randomly divided into Sham, LESW, IRI and LESW + IRI groups. At 4, 12, 24 hours and 3 and 7 days after reperfusion, serum samples and renal tissues were harvested for performing the analysis of renal function, histopathology, immunohistochemistry, flow cytometry and Western blot, as well as enzyme-linked immunosorbent assay. Moreover, circulating endothelial progenitor cells (EPCs) were isolated, labelled with fluorescent dye and injected by tail vein. The fluorescent signals of EPCs were detected using fluorescence microscope and in vivo imaging system to track the distribution of injected circulating EPCs. Results showed that pretreatment with LESW could significantly reduce kidney injury biomarkers, tubular damage, and cell apoptosis, and promote cell proliferation and vascularization in IRI kidneys. The renoprotective role of LESW pretreatment would be attributed to the remarkably increased EPCs in the treated kidneys, part of which were recruited from circulation through SDF-1/CXCR7 pathway. In conclusion, pretreatment with LESW could increase the recruitment of circulating EPCs to attenuate and repair renal IRI.


Subject(s)
Endothelial Progenitor Cells/physiology , Extracorporeal Shockwave Therapy , Kidney/blood supply , Reperfusion Injury/prevention & control , Animals , Apoptosis , Cell Movement , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/genetics , Chemokine CXCL12/physiology , Extracorporeal Shockwave Therapy/methods , Fluorescent Dyes/pharmacokinetics , In Situ Nick-End Labeling , Kidney/pathology , Kidney/physiology , Male , Microscopy, Fluorescence , Microvessels/pathology , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR/biosynthesis , Receptors, CXCR/genetics , Receptors, CXCR/physiology , Regeneration , Reperfusion Injury/blood , Reperfusion Injury/pathology , Signal Transduction , Time Factors
12.
Prostate ; 80(13): 1145-1156, 2020 09.
Article in English | MEDLINE | ID: mdl-32659025

ABSTRACT

BACKGROUND: Epithelial stem cells (ESCs) demonstrate a capacity to maintain normal tissues homeostasis and ESCs with a deregulated behavior can contribute to cancer development. The ability to reprogram normal tissue epithelial cells into prostate or mammary stem-like cells holds great promise to help understand cell of origin and lineage plasticity in prostate and breast cancers in addition to understanding normal gland development. We previously showed that an intracellular chemokine, CXCL12γ induced cancer stem cells and neuroendocrine characteristics in both prostate and breast adenocarcinoma cell lines. However, its role in normal prostate or mammary epithelial cell fate and development remains unknown. Therefore, we sought to elucidate the functional role of CXCL12γ in the regulation of ESCs and tissue development. METHODS: Prostate epithelial cells (PNT2) or mammary epithelial cells (MCF10A) with overexpressed CXCL12γ was characterized by quantitative real-time polymerase chain reaction, Western blots, and immunofluorescence for lineage marker expression, and fluorescence activated cell sorting analyses and sphere formation assays to examine stem cell surface phenotype and function. Xenotransplantation animal models were used to evaluate gland or acini formation in vivo. RESULTS: Overexpression of CXCL12γ promotes the reprogramming of cells with a differentiated luminal phenotype to a nonluminal phenotype in both prostate (PNT2) and mammary (MCF10A) epithelial cells. The CXCL12γ-mediated nonluminal type cells results in an increase of epithelial stem-like phenotype including the subpopulation of EPCAMLo /CD49fHi /CD24Lo /CD44Hi cells capable of sphere formation. Critically, overexpression of CXCL12γ promotes the generation of robust gland-like structures from both prostate and mammary epithelial cells in in vivo xenograft animal models. CONCLUSIONS: CXCL12γ supports the reprogramming of epithelial cells into nonluminal cell-derived stem cells, which facilitates gland development.


Subject(s)
Chemokine CXCL12/biosynthesis , Mammary Glands, Human/growth & development , Prostate/growth & development , Animals , Cellular Reprogramming/physiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Heterografts , Humans , Male , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mice , Prostate/cytology , Prostate/metabolism , Protein Isoforms
13.
J Endovasc Ther ; 27(4): 669-675, 2020 08.
Article in English | MEDLINE | ID: mdl-32419594

ABSTRACT

Purpose: To present the 6-month results of the Stromal Cell-Derived Factor-1 Plasmid Treatment for Patients with Peripheral Artery Disease (STOP-PAD) trial. The trial was an attempt to alter the course of chronic limb-threatening ischemia (CLTI) with a biological agent vs placebo after successful arterial revascularization at or below the knee. Materials and Methods: The multicenter, randomized, double-blinded, placebo-controlled, phase 2B STOP-PAD trial (ClinicalTrials.gov identifier NCT02544204) randomized 109 patients (mean age 71 years; 68 men) with Rutherford category 5 or 6 CLTI and evidence of persistent impaired forefoot perfusion following recent successful revascularization to 8- (n=34) or 16-mg (n=36) intramuscular injections of a non-viral DNA plasmid-based treatment vs placebo (n=34). The primary efficacy outcome was the 6-month wound healing score evaluated by an independent wound core laboratory; the primary safety endpoint was major adverse limb events (MALE), a composite of major amputation plus clinically-driven target lesion revascularization at 6 months. Results: Only one-third of the patients had complete wound healing at 6 months in the placebo (31%), 8-mg injection (33%), and 16-mg injection (33%) groups. In addition, the observed increase in the toe-brachial index from baseline to 6 months was statistically significant in each group; however, this did not result in lower rates of MALE at 6 months (24% in the placebo, 29% in the 8-mg injection, and 11% in the 16-mg injection groups). During the 6-month period, 6 patients (6%) died, and 24 patients (23%) had an amputation [only 4 (4%) major]. Conclusion: Combining revascularization and biological therapy failed to improve outcomes in CLTI at 6 months. STOP-PAD has provided insights for future trials to evaluate biological therapy.


Subject(s)
Chemokine CXCL12/biosynthesis , Genetic Therapy , Ischemia/therapy , Neovascularization, Physiologic , Peripheral Arterial Disease/therapy , Plasmids , Aged , Amputation, Surgical , Chemokine CXCL12/genetics , Chronic Disease , Double-Blind Method , Female , Genetic Therapy/adverse effects , Humans , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Limb Salvage , Male , Peripheral Arterial Disease/genetics , Peripheral Arterial Disease/metabolism , Peripheral Arterial Disease/physiopathology , Recovery of Function , Regional Blood Flow , Time Factors , Treatment Outcome , United States , Vascular Surgical Procedures , Wound Healing
14.
Diabetes ; 69(7): 1562-1572, 2020 07.
Article in English | MEDLINE | ID: mdl-32345753

ABSTRACT

Mobilization of hematopoietic stem/progenitor cells (HSPC) from the bone marrow (BM) is impaired in diabetes. Excess oncostatin M (OSM) produced by M1 macrophages in the diabetic BM signals through p66Shc to induce Cxcl12 in stromal cells and retain HSPC. BM adipocytes are another source of CXCL12 that blunts mobilization. We tested a strategy of pharmacologic macrophage reprogramming to rescue HSPC mobilization. In vitro, PPAR-γ activation with pioglitazone switched macrophages from M1 to M2, reduced Osm expression, and prevented transcellular induction of Cxcl12 In diabetic mice, pioglitazone treatment downregulated Osm, p66Shc, and Cxcl12 in the hematopoietic BM, restored the effects of granulocyte-colony stimulation factor (G-CSF), and partially rescued HSPC mobilization, but it increased BM adipocytes. Osm deletion recapitulated the effects of pioglitazone on adipogenesis, which was p66Shc independent, and double knockout of Osm and p66Shc completely rescued HSPC mobilization. In the absence of OSM, BM adipocytes produced less CXCL12, being arguably devoid of HSPC-retaining activity, whereas pioglitazone failed to downregulate Cxcl12 in BM adipocytes. In patients with diabetes on pioglitazone therapy, HSPC mobilization after G-CSF was partially rescued. In summary, pioglitazone reprogrammed BM macrophages and suppressed OSM signaling, but sustained Cxcl12 expression by BM adipocytes could limit full recovery of HSPC mobilization.


Subject(s)
Bone Marrow Cells/drug effects , Diabetes Mellitus, Experimental/drug therapy , Hematopoietic Stem Cell Mobilization , Macrophages/drug effects , PPAR gamma/agonists , Pioglitazone/pharmacology , Adipogenesis , Animals , Bone Marrow Cells/physiology , Cellular Reprogramming , Chemokine CXCL12/biosynthesis , Female , Humans , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Oncostatin M/antagonists & inhibitors , Src Homology 2 Domain-Containing, Transforming Protein 1/physiology
15.
J Pain ; 21(9-10): 1060-1074, 2020.
Article in English | MEDLINE | ID: mdl-32006698

ABSTRACT

Complex regional pain syndrome (CRPS) results in chronic and excruciating pain in patients. Conventional therapies lack effectiveness, rendering it one of the most difficult to treat neurological conditions.. Electroacupuncture (EA) is an effective alternative therapy for pain relief. Here, we investigated whether EA exerts analgesic effect on a rat model of CRPS type-I (CRPS-I) and related mechanisms. The rat chronic postischemic pain (CPIP) model was established to mimic CRPS-I. 100Hz EA exerted robust and persistent antiallodynic effect on CPIP model compared with 2 Hz EA or sham EA. EA markedly suppressed the overexpression of CXCL12/CXCR4 in spinal cord dorsal horn (SCDH) of CPIP model, leading to substantial decrease in neuronal and glial cell activities in SCDH. Pharmacological blocking CXCR4 mimicked EA-induced antiallodynic effect and related cellular events in SCDH, whereas exogenous CXCL12 abolished EA's effect. CXCR4 signaling resulted in ERK activation in SCDH, contributing to mechanical allodynia of CPIP model rats, whereas EA markedly reduced ERK activation. Therefore, we demonstrated that EA interferes with CXCL12/CXCR4 signaling in SCDH and downstream ERK pathway to exert robust antiallodynic effect on an animal model of CRPS-I. Our work suggests that EA may be a potential therapeutic option for CRPS-I in clinic. PERSPECTIVE: Our work identified that EA exerts robust antiallodynic effect on an animal model of CRPS-I, via mechanisms involving inhibition of CXCL12/CXCR4 signaling. EA further attenuates downstream neuronal and glial cell activation and ERK pathway in SCDH. This work suggests that EA may be a potential therapeutic option for CRPS-I management in clinic.


Subject(s)
Chemokine CXCL12/antagonists & inhibitors , Complex Regional Pain Syndromes/therapy , Electroacupuncture/methods , Hyperalgesia/therapy , Receptors, CXCR4/antagonists & inhibitors , Spinal Cord/metabolism , Animals , Chemokine CXCL12/biosynthesis , Complex Regional Pain Syndromes/metabolism , Disease Models, Animal , Hyperalgesia/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptors, CXCR4/biosynthesis , Signal Transduction/physiology
16.
Br J Haematol ; 188(2): 283-294, 2020 01.
Article in English | MEDLINE | ID: mdl-31423579

ABSTRACT

Monocytes polarize into pro-inflammatory macrophage-1 (M1) or alternative macrophage-2 (M2) states with distinct phenotypes and physiological functions. M2 cells promote tumour growth and metastasis whereas M1 macrophages show anti-tumour effects. We found that M2 cells were increased whereas M1 cells were decreased in bone marrow (BM) from multiple myeloma (MM) patients with progressive disease (PD) compared to those in complete remission (CR). Gene expression of Tribbles homolog 1 (TRIB1) protein kinase, an inducer of M2 polarization, was increased in BM from MM patients with PD compared to those in CR. Ruxolitinib (RUX) is an inhibitor of the Janus kinase family of protein tyrosine kinases (JAKs) and is effective for treating patients with myeloproliferative disorders. RUX markedly reduces both M2 polarization and TRIB1 gene expression in MM both in vitro and in vivo in human MM xenografts in severe combined immunodeficient mice. RUX also downregulates the expression of CXCL12, CXCR4, MUC1, and CD44 in MM cells and monocytes co-cultured with MM tumour cells; overexpression of these genes is associated with resistance of MM cells to the immunomodulatory agent lenalidomide. These results provide the rationale for evaluation of JAK inhibitors, including MM BM in combination with lenalidomide, for the treatment of MM patients.


Subject(s)
Chemokines, CXC/biosynthesis , Intracellular Signaling Peptides and Proteins/biosynthesis , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Janus Kinases/metabolism , Lenalidomide/pharmacology , Mucin-1/biosynthesis , Multiple Myeloma/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Case-Control Studies , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/metabolism , Chemokines, CXC/metabolism , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, SCID , Monocytes/drug effects , Monocytes/metabolism , Mucin-1/metabolism , Multiple Myeloma/blood , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Receptors, CXCR4/biosynthesis , Receptors, CXCR4/metabolism , Signal Transduction , THP-1 Cells
17.
Inflammation ; 43(2): 664-672, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31814061

ABSTRACT

The pathogenesis of keloids has not been elucidated, and the disease is thought to be caused by abnormal secretion of proinflammatory mediators and irregular responses to other inflammatory signals mediated by keloid fibroblasts (KFs). In this study, we investigated whether a local increase in interleukin IL-17 in keloid tissues stimulates the production of stromal cell-derived factor-1 (SDF-1) in KFs causing further recruitment of IL-17-producing T helper 17 (Th17) cells, which subsequently creates a positive feedback loop. Histological assessment was performed and the change in the expression of IL-17, IL-1ß, IL-6, and TNF-α which of fibrosis and inflammation associated markers was examined. In addition, fibroblasts were treated with IL-17 in the presence or absence of STAT3 inhibitor STA-21; SDF-1 levels and fibrosis genes were measured. Our results showed that fibrotic reaction and expression of proinflammatory cytokines including IL-17 were most prominent in the growing margin (perilesional area) of keloid tissue and Th17 cells significantly infiltrated the perilesional area. In addition, IL-17 upregulated the expression of SDF-1, collagen, and α-SMA in KFs. Finally, STA-21 decreased SDF-1α expression and the expression of fibrosis genes in KFs even after IL-17 stimulation. Our study demonstrated that a local increase in IL-17 in keloid tissues stimulates the production of SDF-1 in KFs causing further recruitment of IL-17-producing T helper 17 (Th17) cells, which subsequently creates a positive feedback loop. These findings suggest that STAT3 inhibition can be used to treat keloid scars by reversing the vicious cycle between Th17 cells and KFs.


Subject(s)
Chemokine CXCL12/biosynthesis , Fibroblasts/metabolism , Interleukin-17/pharmacology , Keloid/metabolism , STAT3 Transcription Factor/biosynthesis , Skin/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chemokine CXCL12/agonists , Fibroblasts/drug effects , Humans , Keloid/pathology , STAT3 Transcription Factor/agonists , Signal Transduction/drug effects , Signal Transduction/physiology , Skin/drug effects
18.
J Neuroimmune Pharmacol ; 15(3): 554-563, 2020 09.
Article in English | MEDLINE | ID: mdl-31823250

ABSTRACT

Microglia are implicated in the neuropathogenesis of HIV. Tetraspanin 2 (Tspan2) is closely related to CD9 and CD81 proteins, and are expressed on microglia cells. They have been implicated in cell fusion and adhesion and in the immune response, and neuroinflammation. Developing therapeutics that target microglia remains a challenge as these therapeutics must cross the Blood-Brain Barrier (BBB). Our goal was to use microglia derived exosomes as a vehicle to deliver siRNA across the BBB to target human telomerase reverse transcriptase immortalized human microglial cells (HTHU) latently infected by HIV (HTHU-HIV) and to evaluate if the knockdown of Tspan2 gene expression in changes the activation state of microglia cells, thereby modulating the neuroinflammatory response. A blood brain barrier (BBB) model that closely mimics and accurately reflects the characteristics and functional properties of the in vivo BBB was used to examine HTHU microglia exosome effects on BBB permeability, and their ability to migrate across the and delivery small interfering RNA (siRNA) to cells on the CNS side of the BBB model. Exosomes were loaded with Texas-Red control siRNA (20 pmol) or Cy5-Tspan2 siRNA and then placed in the apical side of the BBB model, 24 h after incubation, HTHU-HIV cells microglial cells on the lower chamber were either imaged for siRNA uptake or analyzed for gene expression induced modifications. HTHU exosomes transmigrate from the apical side of the BBB to deliver Texas-Red control siRNA or Cy5-Tspan2 siRNA to HTHU-HIV microglia cells on the CNS side of the BBB model. A dose dependent (5-40 pmol) increase in Cy5-Tspan2 uptake with a corresponding decrease in gene expression for Tspan2 occurred in HTHU-HIV microglia. A decrease in Tspan2 gene expression as a consequence of knockdown with Tspan2 siRNA at both 20 and 40 pmol concentrations resulted in a significant decrease in C-X-C motif chemokine 12 (CXCL12) and C-X-C chemokine receptor type 4 (CXCR4) gene expression in HTHU-HIV microglia. Furthermore, a decrease in the gene expression levels of the Interleukins, IL-13 and IL-10 and an increase in the gene expression levels for the Fc gamma receptor 2A(FCGR2A) and TNF-α occurred in HTHU-HIV microglial cells These data demonstrate that HTHU exosomes cross the BBB and are efficient delivery vehicles to the CNS. Moreover, modifying the expression levels of Tspan2, has downstream consequences that includes alterations in cytokines and microglia biomarkers. Graphical Abstract Microglia-derived exosomes loaded with Tspan2 siRNA transmigrate across the BBB and knockdown Tspan2 gene expression in human microglial cells latently infected by HIV. This knockdown increases CXCL12, CXCR4, FCGR2A and TNF-α while decreasing IL-13 and IL-10 gene expression in HTHU-HIV microglial cells. Modulating Tspan2 modulates microglia cytokines and phenotype biomarkers.


Subject(s)
Blood-Brain Barrier/metabolism , Exosomes/metabolism , Immunity, Cellular , Microglia/immunology , Microglia/metabolism , Nerve Tissue Proteins/metabolism , RNA, Small Interfering/genetics , Tetraspanins/metabolism , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/genetics , Cytokines/biosynthesis , Fluorescent Dyes/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Nerve Tissue Proteins/genetics , Primary Cell Culture , Receptors, CXCR4/biosynthesis , Receptors, CXCR4/genetics , Tetraspanins/genetics , Xanthenes/metabolism
19.
Brain Behav Immun ; 84: 72-79, 2020 02.
Article in English | MEDLINE | ID: mdl-31751616

ABSTRACT

Conditioned place preference (CPP) is a learned behavior, in which animals learn to associate environmental contexts with rewarding effects. The formation of CPP is an integrated outcome of multiple learning processes. Although multiple anatomical substrates underlying this contextual learning have been proposed, it remains unknown whether a specific molecular signaling pathway within CA1 mediates context learning associated with morphine conditioning. Here, we showed that repeated context learning associated with morphine conditioning significantly increased CXCL12 levels in hippocampal CA1 neurons, and the inhibition of CXCL12 expression ameliorated the CPP behavior following context exposure with morphine conditioning. Additionally, repeated context exposure with morphine conditioning increased the phosphorylation of STAT3 and the acetylation of histone H4 in CXCL12-expressing neurons in CA1. Immunoprecipitation and chromatin immunoprecipitation assays demonstrated that repeated context exposure with morphine conditioning increased the binding of STAT3 to the CXCL12 gene promoter and the interaction between STAT3 and p300, which contributed to the enhanced transcription of CXCL12 by increasing the acetylation of histone H4 in the CXCL12 gene promoter. The inhibition of STAT3 by intrathecal injection of S3I-201 suppressed the acetylation of histone H4. These data demonstrated the epigenetic upregulation of CXCL12 following repeated context exposure with morphine conditioning, which potentially contributed to the spatial memory consolidation associated with conditioned place preference induced by morphine conditioning.


Subject(s)
Chemokine CXCL12/genetics , Conditioning, Psychological , Epigenesis, Genetic , Hippocampus/drug effects , Hippocampus/metabolism , Morphine/pharmacology , Spatial Memory/drug effects , Up-Regulation/drug effects , Animals , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/metabolism , Male , Narcotics/pharmacology , Rats , Rats, Sprague-Dawley
20.
Inflamm Res ; 69(2): 167-178, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31865399

ABSTRACT

OBJECTIVE: To elucidate the regulation, function of the chemokine CXC-motif ligand 12 (CXCL12) and its receptors (CXCR) 4 and 7 in prostate cancer tumor microenvironment. MATERIAL: In-silico-analysis of expression in prostate cancer tissues. In-vitro comparison, testing of regulation in human prostate cancer cells LNCaP, DU145, and PC3. TREATMENT: Dihydrotestosterone (DHT) treatments (0-10 nM) were for 0-48 h. The inflammatory agent Flagellin treatment (20 ng/ml) was for 2 h. Migration assays were performed for 24 h using 10 ng/ml CXCL12. METHODS: Real-time PCR, western analysis, and migration assays were used to determine mRNA, protein, and functional changes, respectively. RESULTS: Malignant prostate cancer tissues exhibit higher CXCR4/7 mRNA ratio, and higher CXCR7 mRNA levels were detected in the androgen-responsive LNCaP cells. Putative androgen-responsive elements were identified in CXCR4, 7 gene, and exposure to DHT, flagellin increased CXCR4 mRNA but decreased CXCR7 mRNA levels in LNCaP cells. Androgen receptor siRNA significantly attenuated the effects of DHT on CXCR4, 7 mRNA in LNCaP cells. However, DHT and flagellin only decrease CXCR7 protein and additively increased migration of LNCaP cells towards CXCL12. CONCLUSIONS: Down regulation of CXCR7 protein by DHT and flagellin increased migration, supporting CXCR7 as decoy receptor counteracting CXCL12/CXCR4-mediated migration in prostate cancer cells.


Subject(s)
Androgens/metabolism , Inflammation/metabolism , Prostatic Neoplasms/genetics , Receptors, CXCR4/genetics , Receptors, CXCR/genetics , Cell Line, Tumor , Cell Movement , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/genetics , Computer Simulation , Dihydrotestosterone/pharmacology , Flagellin/pharmacology , Humans , Male , Prostatic Neoplasms/pathology , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Receptors, CXCR/biosynthesis , Receptors, CXCR4/biosynthesis , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...