Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cells ; 11(3)2022 01 26.
Article in English | MEDLINE | ID: mdl-35159233

ABSTRACT

Altered hypoxia-inducible factor-alpha (HIF-α) activity may have significant consequences in the hippocampus, which mediates declarative memory, has limited vascularization, and is vulnerable to hypoxic insults. Previous studies have reported that neurovascular coupling is reduced in aged brains and that diseases which cause hypoxia increase with age, which may render the hippocampus susceptible to acute hypoxia. Most studies have investigated the actions of HIF-α in aging cortical structures, but few have focused on the role of HIF-α within aged hippocampus. This study tests the hypothesis that aging is associated with impaired hippocampal HIF-α activity. Dorsal hippocampal sections from mice aged 3, 9, 18, and 24 months were probed for the presence of HIF-α isoforms or their associated gene products using immunohistochemistry and fluorescent in situ hybridization (fISH). A subset of each age was exposed to acute hypoxia (8% oxygen) for 3 h to investigate changes in the responsiveness of HIF-α to hypoxia. Basal mean intensity of fluorescently labeled HIF-1α protein increases with age in the hippocampus, whereas HIF-2α intensity only increases in the 24-month group. Acute hypoxic elevation of HIF-1α is lost with aging and is reversed in the 24-month group. fISH reveals that glycolytic genes induced by HIF-1α (lactose dehydrogenase-a, phosphoglycerate kinase 1, and pyruvate dehydrogenase kinase 1) are lower in aged hippocampus than in 3-month hippocampus, and mRNA for monocarboxylate transporter 1, a lactose transporter, increases. These results indicate that lactate, used in neurotransmission, may be limited in aged hippocampus, concurrent with impaired HIF-α response to hypoxic events. Therefore, impaired HIF-α may contribute to age-associated cognitive decline during hypoxic events.


Subject(s)
Hypoxia , Lactose , Animals , Hippocampus/metabolism , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , In Situ Hybridization, Fluorescence , Mice , RNA, Messenger/metabolism
2.
Br J Anaesth ; 113(4): 652-60, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24880828

ABSTRACT

BACKGROUND: Preoperative anaemia is associated with increased morbidity in patients undergoing major surgery. Whether erythrocytes are the only bone-marrow-derived cell lineage that associates with increased surgical complications is unknown. This prospective observational trial studied the mobilization of endothelial progenitor cells (EPCs) in response to exercise in association with postoperative complications. METHODS: After IRB approval, 60 subjects undergoing major thoracic surgery were exercised to exhaustion (peak V̇(O2)). Peripheral blood collected before and after peak exercise was quantified for EPC lineages by fluorescence-activated cell sorter analysis. Complication analysis was based on the Clavien-Dindo classification. RESULTS: Exhaustive exercise increased EPC [CD45-133+34+ cells=150 (0.00-5230) to 220 (0.00-1270) cells µl(-1); median change (range)=20 (-4,180-860) cells µl(-1); P=0.03] but not mature endothelial cell (EC) subpopulations. Pre-exercise levels [odds ratio (OR)=0.86, 95% confidence interval (CI): 0.37-2.00, P=0.72), change after exercise as a continuous variable (OR=0.95, 95% CI: 0.41-2.22, P=0.91) and a positive response after exercise (change >0 cells µl(-1); OR=0.41, 95% CI: 0.13-1.28, P=0.12) were not statistically significantly associated with the incidence of postoperative complications. Post-hoc receiver operating characteristic curve analyses revealed that subjects with a CD45-133+34+ increase ≥60 cells µl(-1) in response to exercise suffered fewer postoperative complications [86% sensitivity, 48% specificity and AUC=0.67 (95% CI: 0.52-0.81)]. CONCLUSIONS: Preoperative exercise induces EPC into the peripheral circulation. Subjects with a poor EPC response had a pre-existing propensity for postoperative complications. This warrants further research into the role of bone marrow function as a critical component to endothelial repair mechanisms. CLINICAL TRIAL REGISTRATION: IRB 2003-0434 (University of Texas M.D. Anderson Cancer Center, Houston, TX, USA).


Subject(s)
Endothelial Cells/physiology , Exercise Therapy/methods , Hematopoietic Stem Cell Mobilization , Postoperative Complications/prevention & control , Preoperative Period , Adult , Aged , Blood Gas Analysis , Bone Marrow/physiology , Endpoint Determination , Ethnicity , Exercise Test , Exercise Tolerance/physiology , Female , Flow Cytometry , Hemodynamics/physiology , Humans , Male , Middle Aged , Postoperative Complications/epidemiology , Prospective Studies , ROC Curve , Risk Assessment , Risk Factors , Stress, Physiological , Thoracic Surgical Procedures , Treatment Outcome
3.
Clin Cancer Res ; 17(14): 4872-81, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21632852

ABSTRACT

PURPOSE: VEGF and infiltrating myeloid cells are known regulators of tumor angiogenesis and vascular permeability in glioblastoma. We investigated potential blood-based markers associated with radiographic changes to aflibercept, which binds VEGF and placental growth factor (PlGF) in patients with recurrent glioblastoma. EXPERIMENTAL DESIGN: In this single-arm phase II trial, aflibercept was given intravenously every two weeks until disease progression. Plasma and peripheral blood mononuclear cells were collected at baseline and 24 hours, 14 days, and 28 days posttreatment. Plasma cytokines and angiogenic factors were quantified by using ELISA and multiplex bead assays, and myeloid cells were assessed by flow cytometry in a subset of patients. RESULTS: Circulating levels of VEGF significantly decreased 24 hours after treatment with aflibercept, coincident with radiographic response observed by MRI. PlGF initially decreased 24 hours posttreatment but increased significantly by days 14 and 28. Lower baseline levels of PlGF, elevated baseline levels of CTACK/CCL27, MCP3/CCL7, MIF, and IP-10/CXCL10, and a decrease in VEGFR1(+) monocytes from baseline to 24 hours were all associated with improved response. Tumor progression was associated with increases in circulating matrix metalloproteinase 9. CONCLUSIONS: These data suggest that decreases in VEGF posttreatment are associated with radiographic response to aflibercept. Elevated baseline chemokines of monocyte lineage in responding patients supports a role for myeloid cells and chemokines as potential biomarkers and regulators of glioma angiogenesis.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Glioblastoma/drug therapy , Recombinant Fusion Proteins/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiogenesis Inducing Agents/blood , Cytokines/blood , Disease Progression , Glioblastoma/diagnostic imaging , Glioblastoma/metabolism , Humans , Membrane Proteins/blood , Molecular Targeted Therapy , Monocytes/metabolism , Radiography , Receptors, Vascular Endothelial Growth Factor , Recurrence , Vascular Endothelial Growth Factor A/urine , Vascular Endothelial Growth Factor Receptor-1/metabolism
4.
Cancer ; 116(7): 1751-6, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20120033

ABSTRACT

BACKGROUND: : Tumor angiogenesis has been associated with a poor prognosis in patients with metastatic melanoma (MM). Microtubule stabilizers and cyclooxygenase 2 (COX-2) inhibitors, alone and in combination, have produced inhibitory effects on endothelial cells and tumor angiogenesis. Angiogenesis, which is the growth of new blood vessels, is necessary for tumor growth and progression. Thus, the authors tested the safety and efficacy of a low dose of paclitaxel and celecoxib in patients with MM. METHODS: : Patients received paclitaxel 10 mg/m(2) for 96 hours weekly as a continuous intravenous infusion and oral celecoxib 400 mg twice daily. Systemic tumor response was assessed at 6-week intervals. Tumor measurements at the end of Cycle 1 were used as the baseline for assessment of tumor progression. Patients with unacceptable toxicity or disease progression after Cycle 2 relative to the end of Cycle 1 were taken off study. RESULTS: : Twenty patients were enrolled. Twelve of 20 patients (60%) had received > or =2 previous systemic therapies. Three patients did not receive treatment because of rapid disease progression. Treatment-related grade 3/4 toxicities were limited to catheter-related complications. One patient achieved a partial response, and 3 of 20 patients (15%) had stable disease for >6 months. The median time to progression was 57 days (95% confidence interval, 43-151 days), and the median overall survival was 212 days (95% confidence interval, 147-811 days). CONCLUSIONS: : Low-dose, continuous intravenous infusion paclitaxel and oral celecoxib produced disease stabilization in a significant proportion of heavily pretreated patients with MM. These findings support a role for metronomic therapy in patients with this disease. Cancer 2010. (c) 2010 American Cancer Society.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/drug therapy , Paclitaxel/administration & dosage , Pyrazoles/administration & dosage , Skin Neoplasms/drug therapy , Sulfonamides/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Celecoxib , Cytokines/blood , Drug Administration Schedule , Female , Humans , Male , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Pilot Projects , Skin Neoplasms/pathology
5.
Clin Cancer Res ; 15(10): 3484-94, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19447865

ABSTRACT

PURPOSE: The epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKI) gefitinib and erlotinib benefit some non-small cell lung cancer (NSCLC) patients, but most do not respond (primary resistance) and those who initially respond eventually progress (acquired resistance). EGFR TKI resistance is not completely understood and has been associated with certain EGFR and K-RAS mutations and MET amplification. EXPERIMENTAL DESIGN: We hypothesized that dual inhibition of the vascular endothelial growth factor (VEGF) and EGFR pathways may overcome primary and acquired resistance. We investigated the VEGF receptor/EGFR TKI vandetanib, and the combination of bevacizumab and erlotinib in vivo using xenograft models of EGFR TKI sensitivity, primary resistance, and three models of acquired resistance, including models with mutated K-RAS and secondary EGFR T790M mutation. RESULTS: Vandetanib, gefitinib, and erlotinib had similar profiles of in vitro activity and caused sustained tumor regressions in vivo in the sensitive HCC827 model. In all four resistant models, vandetanib and bevacizumab/erlotinib were significantly more effective than erlotinib or gefitinib alone. Erlotinib resistance was associated with a rise in both host and tumor-derived VEGF but not EGFR secondary mutations in the KRAS mutant-bearing A549 xenografts. Dual inhibition reduced tumor endothelial proliferation compared with VEGF or EGFR blockade alone, suggesting that the enhanced activity of dual inhibition is due at least in part to antiendothelial effects. CONCLUSION: These studies suggest that erlotinib resistance may be associated with a rise in both tumor cell and host stromal VEGF and that combined blockade of the VEGFR and EGFR pathways can abrogate primary or acquired resistance to EGFR TKIs. This approach merits further evaluation in NSCLC patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Bevacizumab , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Gefitinib , Humans , Lung Neoplasms/metabolism , Mice , Mice, Nude , Mutation , NIH 3T3 Cells , Piperidines/administration & dosage , Piperidines/pharmacology , Quinazolines/administration & dosage , Quinazolines/pharmacology , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection , Tumor Burden/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
6.
Cancer ; 115(10 Suppl): 2346-54, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19402074

ABSTRACT

In recent years, there has been significant progress in the clinical development and application of antiangiogenic therapies in renal cell carcinomas, particularly inhibitors of the vascular endothelial growth factor (VEGF) pathway. Despite this progress, no validated methods are currently available for identifying which patients are most likely to respond to treatment or experience toxic effects, selecting the optimal dose, or determining whether the intended molecular target has been effectively inhibited. However, recent studies have suggested that some of the biomarkers currently under investigation in clear cell renal cell carcinoma for VEGF pathway inhibitors are promising. These biomarkers include circulating proangiogenic factors and receptors; markers of hypoxia and endothelial damage; and cellular populations in peripheral blood, such as circulating endothelial cells. Further preclinical and translational validation studies are still needed to determine their practical utility in the clinical setting.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Renal Cell/blood , Kidney Neoplasms/blood , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Endothelial Cells/pathology , Humans , Stem Cells/pathology
7.
Clin Cancer Res ; 15(1): 267-73, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19118054

ABSTRACT

PURPOSE: Antigenic overlap among circulating endothelial cells (CEC) and progenitors (CEP), platelets, and other blood cells led to the need to develop a reliable standardized method for CEC and CEP quantification. These cells are emerging as promising preclinical/clinical tools to define optimal biological doses of antiangiogenic therapies and to help stratify patients in clinical trials. EXPERIMENTAL DESIGN: We report the experimental validation of a novel flow cytometry method that precisely dissects CEC/CEP from platelets and other cell populations and provides information about CEC/CEP viability. RESULTS: Sorted DNA/Syto16(+)CD45(-)CD31(+)CD146(+) CECs, investigated by electron microscopy, were found to be bona fide endothelial cells by the presence of Weibel-Palade bodies. More than 75% of the circulating mRNAs of the endothelial-specific gene, VE-cadherin, found in the blood were present in the sorted population. CECs were 140 +/- 171/mL in healthy subjects (n = 37) and 951 +/- 1,876/mL in cancer patients (n = 78; P < 0.0001). The fraction of apoptotic/necrotic CECs was 77 +/- 14% in healthy subjects and 43 +/- 23% in cancer patients (P < 0.0001). CEPs were 181 +/- 167/mL in healthy donors and 429 +/- 507/mL in patients (P = 0.00019). Coefficients of variation were 4 +/- 4% (intrareader), 17 +/- 4% (interreader), and 17 +/- 7% (variability over 0-72 h), respectively. Parallel samples were frozen by a standardized protocol. After thawing, coefficients of variation were 12 +/- 8% (intrareader), 16 +/- 10% (interreader), and 26 +/- 16% (variability over 0-14 days of frozen storage), respectively. CONCLUSIONS: This procedure enumerates a truly endothelial cell population with limited intrareader and interreader variability. It appears possible to freeze samples for large-scale CEC enumeration during clinical trials. This approach could be enlarged to investigate other angiogenic cell populations as well.


Subject(s)
Endothelial Cells , Flow Cytometry/methods , Neoplasms/blood , Cell Count/methods , Cell Survival , Endothelial Cells/chemistry , Humans , Microscopy, Electron , Reproducibility of Results , Stem Cells , Weibel-Palade Bodies
8.
Clin Lab ; 53(5-6): 285-95, 2007.
Article in English | MEDLINE | ID: mdl-17605403

ABSTRACT

An increase in the number of circulating endothelial cells (CECs) and of bone marrow derived endothelial progenitors (CEPs) in the peripheral blood (PB) is normally associated with vascular injury, repair, and neovascularization. These cells rarely exist in the PB of healthy individuals. Therefore, when they are present in the PB of individuals, their phenotypes and quantity in the PB may serve as surrogate diagnostic or prognostic parameters of vascular injury and/or as an indication of tumor growth. An elevated level of CEPs may suggest an ongoing repair of ischemic vascular injuries and/or angiogenesis. Recently, more advanced techniques for CEC isolation and CEP enumeration have become available. In particular, immunobeads isolation and fluorescence-activated cell sorting (FACS) techniques have been employed with success in evaluation of vascular dysfunctions. Therefore, CECs and CEPs may serve as potential surrogate markers for monitoring various vascular diseases, which could help to determine pathological process and clinical treatment. In this article, we will present an overview of CECs and CEPs by discussing their origins, reviewing methodologies adapted to the measurement of rare events, describing pathological situations associated with CECs/CEPs, and correlating them with a broad spectrum of disease processes.


Subject(s)
Biomarkers/blood , Endothelial Cells , Hematopoietic Stem Cells , Immunomagnetic Separation/methods , Vascular Diseases/blood , Vascular Diseases/diagnosis , Flow Cytometry/methods , Humans
10.
J Surg Res ; 126(2): 193-8, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15919419

ABSTRACT

BACKGROUND: The isolation, differentiation, and expansion of endothelial progenitor cells (EPCs) from peripheral blood have potential applicability in areas of therapeutic neovascularization, vascular repair, and tissue engineering. The purpose of the current study was to elucidate a simple method of isolation and differentiation of EPCs by defining the endothelial morphology, surface marker expression, and proliferative capacity of EPC outgrowth from canine peripheral blood mononuclear cells (PBMCs). MATERIALS AND METHODS: PBMCs were isolated from fresh canine blood and cultured in fibronectin-coated plates in which EPCs were identified from cell morphology and outgrowth characteristics. Cell surface markers were determined with flow cytometry analysis to identify differentiation of cultured and subcultured colonies. A hematologic counter with phase contrast microscopy was used to study cell growth curves of EPCs as compared with mature human coronary artery endothelial cells. RESULTS: During the first week of canine PBMC culture, cells were morphologically round and varied in size, but in the course of the second and third week of culture, the cells, respectively, became spindle-shaped and displayed an endothelium-like cobblestone morphology with outgrowth. CD34 was significantly decreased at 21 days as compared with 7 days culture (36.04% to 21.37%), whereas vWF (from 77.26% to 96.37%) and eNOS (from 0% to 14.97%) were significantly increased. VEGFR-2 was slightly increased, and P1H12 (CD146) was unchanged. Subcultured canine EPCs displayed a higher proliferation rate as compared to mature human coronary artery endothelial cells in the same culture conditions. CONCLUSIONS: These data demonstrate that canine EPCs can be isolated and cultured from the canine PBMC fraction. These outgrowth cells displayed characteristics of endothelial morphology with endothelial cell-specific surface markers. Furthermore, it was revealed that canine EPCs have a greater growth potential as compared to mature endothelial cells. This study suggests that PBMCs could be used as a source of EPCs for potential applications in tissue engineering and vascular therapy.


Subject(s)
Endothelial Cells/cytology , Monocytes/cytology , Stem Cells/cytology , Animals , Antigens, CD/metabolism , Biomarkers/metabolism , CD146 Antigen , Cell Count , Cell Differentiation , Cell Membrane/metabolism , Cell Proliferation , Cells, Cultured , Dogs , Endothelial Cells/metabolism , Monocytes/metabolism , Neural Cell Adhesion Molecules/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Stem Cells/metabolism , Time Factors , Vascular Endothelial Growth Factor Receptor-2/metabolism , von Willebrand Factor/metabolism
11.
Artif Organs ; 28(7): 668-75, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15209861

ABSTRACT

The objective of this study was to determine the effect of basic fibroblast growth factor (bFGF) coating on endothelial cell seeding and proliferation on a decellularized heparin coated vascular graft and to determine the retention of seeded cells on the graft under flow conditions. Disks of heparin coated decellularized grafts were incubated for 24 h as controls or with bFGF. Human microvascular endothelial cells (HMECs) or canine peripheral blood endothelial progenitor cells (CEPC) were seeded onto the disks and incubated for 96 h or 48 h, respectively. HMECs were also seeded onto the luminal surfaces of two heparin-coated decellularized grafts for 3 h. One graft was placed in a perfusion culture system and cultured for an additional 6 h with flow and pressure. After culturing, there were 4.7 +/- 1.4 cells/mm(2) HMECs on control grafts and 11.4 +/- 1.4 cells/mm(2) in bFGF treated grafts (P < 0.05). Likewise, with CEPCs, there were 14.8 +/- 4.8 cells/mm(2) in control grafts and 33.3 +/- 7.3 cells/mm(2) in bFGF treated grafts. After only 3 h of cell attachment, 60% of HMECs were retained in the intact graft exposed flow relative to the static control graft, which is an acceptable level. These data demonstrate that bFGF coating on the heparin bound decellularized grafts significantly increases both HMEC and dog EPC proliferation and that seeded cells are stable under perfusion conditions.


Subject(s)
Blood Vessel Prosthesis , Coated Materials, Biocompatible , Endothelial Cells/cytology , Fibroblast Growth Factor 2 , Animals , Carotid Arteries/cytology , Cells, Cultured , Female , Heparin , Male , Swine , Tissue Engineering
12.
J Surg Res ; 118(1): 38-44, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15093715

ABSTRACT

BACKGROUND: Heterogeneity of endothelial cells may affect angiogenesis, vascular healing, and cardiovascular disease formation. The objective of this study was to identify and characterize populations of human coronary artery endothelial cells (HCAECs), their gene expression levels, and response to TNF-alpha stimulation. MATERIALS AND METHODS: Commercial HCAECs were cultured with EGM-2 complete medium. Cell population was determined by flow cytometry analysis based on size-scatter distribution. Gene expression for each population with or without TNF-alpha (1 ng/ml) stimulation for 16 h was also studied by flow cytometry analysis with proper gating and fluorescence labeling of antibodies. RESULTS: Two distinguished populations, HCAEC-I (small) and HCAEC-II (large), were identified through all passages (from 2 to 10) during cultures. Both HCAEC-I and HCAEC-II showed more than 90% positive staining for both von Willebrand factor and CD31 and were negative for CD34 and CD4 (less than 2%). HCAEC-I had substantially higher expression of cadherin-5 (71.91% versus 51.07%) than HCAEC-II. HCAEC-I also expressed much higher levels of vascular endothelial growth factor receptor-2 (VEGF-R2) (17.35% versus 0.77%), endothelial nitric oxide synthase (eNOS) (44.64% versus 2.54%), VCAM-1 (6.08% versus 1.18%), E-selectin (18.68% versus 1.42%), CXCR4 (61.05% versus 7.98%), and CCR5 (48.66% versus 1.97%) than HCAEC-II. HCAEC-II, however, had substantially higher expression of P1H12 (93.07% versus 58.73%) and ICAM-1 (94.37% versus 58.62%) than HCAEC-I. Following TNF-alpha stimulation, both populations substantially increased the expression of ICAM-1, VCAM-1, and E-selectin. HCAEC-I, however, had much higher expressions of VEGFR-2, eNOS, CXCR4, and CCR-5 than HACEC-II after TNF-alpha stimulation. CONCLUSIONS: These data demonstrate that commercial HCAECs have two distinguished populations with different gene expression patterns and different responses following TNF-alpha stimulation. This study indicates that the heterogeneity of HCAECs may have biologic or pathologic significances in coronary arteries.


Subject(s)
Coronary Vessels/cytology , Endothelium, Vascular/cytology , Arteries , Biomarkers/analysis , Cells, Cultured , Coronary Vessels/metabolism , E-Selectin/metabolism , Endothelium, Vascular/metabolism , Flow Cytometry , Gene Expression , Humans , Intercellular Adhesion Molecule-1/metabolism , Microscopy, Phase-Contrast , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...