Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Melanoma Res ; 21(5): 405-16, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21900792

ABSTRACT

'Cancer stem cells' (CSCs) are tumor cells with stem cell properties hypothesized to be responsible for tumorigenesis, metastatis, and resistance to treatment, and have been identified in different tumors including cutaneous melanoma, using stem cell markers such as CD133. This study explored expression of CD133 and other putative stem cell markers in uveal melanoma. Eight uveal melanoma cell lines were subjected to flow-cytometric (fluorescence-activated cell sorting) analysis of CD133 and other stem cell markers. Eight paraffin-embedded tumors were analyzed by immunohistochemistry for CD133, Pax6, Musashi, nestin, Sox2, ABCB5, and CD68 expressions. Ocular, uveal melanoma, and hematopoietic stem cell distributions of C-terminal and N-terminal CD133 mRNA splice variants were compared by reverse-transcription PCR. Fluorescence-activated cell sorting analysis revealed a population of CD133-positive/nestin-positive cells in cell lines Mel270, OMM 2.3, and OMM2.5. All cell lines studied were positive for nestin, CXCR-4, CD44, and c-kit. Immunohistochemistry identified cells positive for CD133, Pax6, Musashi, nestin, Sox2, ABCB5, and CD68 predominantly at the invading tumor front. C-terminal primers interacting with CD133 splice variant s2 detected a novel variant lacking exon 27. Differential expression of CD133 splice variants was found in iris, ciliary body, retina, and retinal pigment epithelium/choroid as well as in uveal melanoma cell lines. mRNA for nestin, Sox2, and Musashi was present in all studied cell lines. Uveal melanoma such as cutaneous melanoma may therefore contain CSCs. Further experiments are needed to isolate stem cell marker-positive cells, to evaluate their functional properties and to explore therapeutical approaches to these putative CSCs in uveal melanoma.


Subject(s)
Antigens, CD/biosynthesis , Glycoproteins/biosynthesis , Melanoma/metabolism , Neoplastic Stem Cells/metabolism , Uveal Neoplasms/metabolism , AC133 Antigen , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Female , Glycoproteins/genetics , Humans , Immunohistochemistry , Intermediate Filament Proteins/biosynthesis , Male , Melanoma/genetics , Melanoma/pathology , Mice , Middle Aged , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/biosynthesis , Nestin , Peptides/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology
2.
Int J Artif Organs ; 34(7): 529-45, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21786252

ABSTRACT

Worldwide, cardiovascular disease results in an estimated 14.3 million deaths per year, giving rise to an increased demand for alternative and advanced treatment. Current approaches include medical management, cardiac transplantation, device therapy, and, most recently, stem cell therapy. Research into cell-based therapies has shown this option to be a promising alternative to the conventional methods. In contrast to early trials, modern approaches now attempt to isolate specific stem cells, as well as increase their numbers by means of amplifying in a culture environment. The method of delivery has also been improved to minimize the risk of micro-infarcts and embolization, which were often observed after the use of coronary catheterization. The latest approach entails direct, surgical, trans-epicardial injection of the stem cell mixture, as well as the use of tissue-engineered meshes consisting of embedded progenitor cells.


Subject(s)
Cardiac Surgical Procedures , Heart Failure/surgery , Myocardium/pathology , Myocytes, Cardiac/transplantation , Stem Cell Transplantation , Tissue Engineering , Animals , Cell Differentiation , Cell Proliferation , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Myocytes, Cardiac/pathology , Regeneration , Tissue Scaffolds , Treatment Outcome
3.
Tissue Eng Part C Methods ; 16(5): 1069-81, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20073989

ABSTRACT

CD133+ cells are hemangioblasts that have capacity to generate into both hematopoietic and endothelial cells (ECs). Hypoxia/normoxia has shown to be the regulator of the balance between stemness and differentiation. In this study we performed Agilent's whole human genome oligo microarray analysis and examined the differentiation potential of the bone-marrow-derived CD133+ cells after hypoxic/normoxic preconditioning of CD133+ cells. Results showed that there was no significant increase in erythroid colony forming unit (CFU-E) and CFU-granulocyte, erythrocyte, monocyte, and megakaryocyte formation with cells treated under hypoxia/normoxia. However, a significant increment of EC forming unit at 24 h (143.2 +/- 8.0%) compared to 0 h (100 +/- 11.4%) was observed in CFU-EC analysis. Reverse transcription-polymerase chain reaction and immunostaining analysis showed that the differentiated cells diminished hematopoietic stem cell surface markers and acquired the gene markers and functional phenotype of ECs. The transcriptome profile revealed a cluster of 232 downregulated and 498 upregulated genes in cells treated for 24 h under hypoxia. The upregulated genes include angiogenic genes, angiogenic growth factor genes, angiogenic cytokine and chemokine genes, as well as angiogenic-positive regulatory genes, including FGFBP1, PDGFB, CCL15, CXCL12, CXCL6, IL-6, PTN, EREG, ERBB2, EDG5, FGF3, FHF2, GDF15, JUN, L1CAM, NRG1, NGFR, and PDGFB. On the other hand, angiogenesis inhibitors and related genes, including IL12A, MLLT7, STAB1, and TIMP2, are downregulated. Taken together, hypoxic/normoxic preconditioning may lead to the differentiation of CD133+ cells toward endothelial lineage, which may improve the current clinical trial studies.


Subject(s)
Antigens, CD/immunology , Bone Marrow Cells/cytology , Cell Differentiation , Cell Hypoxia , Endothelium/cytology , Glycoproteins/immunology , Oxygen/metabolism , Peptides/immunology , AC133 Antigen , Base Sequence , Bone Marrow Cells/immunology , Cell Lineage , Cell Separation , DNA Primers , Flow Cytometry , Humans , Oligonucleotide Array Sequence Analysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
4.
Transfusion ; 50(4): 843-55, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20003054

ABSTRACT

BACKGROUND: Dendritic cells (DCs) are applied worldwide in several clinical studies of immune therapy of malignancies, autoimmune diseases, and transplantations. Most legislative bodies are demanding high standards for cultivation and transduction of cells. Closed-cell cultivating systems like cell culture bags would simplify and greatly improve the ability to reach these cultivation standards. We investigated if a new polyolefin cell culture bag enables maturation and adenoviral modification of human DCs in a closed system and compare the results with standard polystyrene flasks. STUDY DESIGN AND METHODS: Mononuclear cells were isolated from HLA-A*0201-positive blood donors by leukapheresis. A commercially available separation system (CliniMACS, Miltenyi Biotec) was used to isolate monocytes by positive selection using CD14-specific immunomagnetic beads. The essentially homogenous starting cell population was cultivated in the presence of granulocyte-macrophage-colony-stimulating factor and interleukin-4 in a closed-bag system in parallel to the standard flask cultivation system. Genetic modification was performed on Day 4. After induction of maturation on Day 5, mature DCs could be harvested and cryopreserved on Day 7. During the cultivation period comparative quality control was performed using flow cytometry, gene expression profiling, and functional assays. RESULTS: Both flasks and bags generated mature genetically modified DCs in similar yields. Surface membrane markers, expression profiles, and functional testing results were comparable. The use of a closed-bag system facilitated clinical applicability of genetically modified DCs. CONCLUSIONS: The polyolefin bag-based culture system yields DCs qualitatively and quantitatively comparable to the standard flask preparation. All steps including cryopreservation can be performed in a closed system facilitating standardized, safe, and reproducible preparation of therapeutic cells.


Subject(s)
Cell Culture Techniques/methods , Dendritic Cells/physiology , Adult , Antigens, CD/analysis , Antigens, CD/genetics , Antigens, Surface/analysis , Cell Count , Cell Culture Techniques/instrumentation , Cell Division/physiology , Cellular Senescence/physiology , Child , Dendritic Cells/cytology , Dendritic Cells/ultrastructure , Female , Gene Transfer Techniques , Humans , Leukapheresis/methods , Leukocytes/cytology , Leukocytes/physiology , Lipopolysaccharide Receptors/analysis , Lipopolysaccharide Receptors/genetics , Male , Microscopy, Electron, Scanning , Middle Aged , Oligonucleotide Array Sequence Analysis
5.
BMC Immunol ; 8: 13, 2007 Aug 06.
Article in English | MEDLINE | ID: mdl-17683591

ABSTRACT

BACKGROUND: Siglec-7, a sialic acid binding inhibitory receptor expressed by NK cells is masked in vivo by a so far unknown ligand. It shows a strong binding prevalence for alpha-2,8-linked disialic acids in vitro. RESULTS: Here we describe the expression of PSA-NCAM (alpha-2,8-linked polysialic acid modified NCAM) on functional adult peripheral blood natural killer cells and examine its possible role in masking Siglec-7. Unmasking of Siglec-7 using Clostridium perfringens neuraminidase massively reduces NK cell cytotoxicity. By contrast a specific removal of PSA using Endo-NF does not lead to a reduction of NK cell cytotoxicity. CONCLUSION: The results presented here therefore indicate that PSA-NCAM is not involved in masking Siglec-7.


Subject(s)
CD56 Antigen/immunology , CD56 Antigen/metabolism , Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/immunology , Sialic Acids/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cell Line , Cell Separation , Humans , Killer Cells, Natural/enzymology , Lectins/metabolism , Neuraminidase/metabolism , Sialyltransferases/metabolism
6.
Stem Cells Dev ; 16(3): 447-60, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17610375

ABSTRACT

Neural stem cells (NSCs) are potential sources for cell therapy of neurodegenerative diseases and for drug screening. Despite their potential benefits, ethical and practical considerations limit the application of NSCs derived from human embryonic stem cells (ES) or adult brain tissue. Thus, alternative sources are required to satisfy the criteria of ready accessibility, rapid expansion in chemically defined media and reliable induction to a neuronal fate. We isolated somatic stem cells from the human periodontium that were collected during minimally invasive periodontal access flap surgery as part of guided tissue regeneration therapy. These cells could be propagated as neurospheres in serum-free medium, which underscores their cranial neural crest cell origin. Culture in the presence of epidermal growth factor (EGF) and fibroblast growth factor-2 (FGF-2) under serum-free conditions resulted in large numbers of nestin-positive/Sox-2-positive NSCs. These periodontium-derived (pd) NSCs are highly proliferative and migrate in response to chemokines that have been described as inducing NSC migration. We used immunocytochemical techniques and RT-PCR analysis to assess neural differentiation after treatment of the expanded cells with a novel induction medium. Adherence to substrate, growth factor deprivation, and retinoic acid treatment led to the acquisition of neuronal morphology and stable expression of markers of neuronal differentiation by more than 90% of the cells. Thus, our novel method might provide nearly limitless numbers of neuronal precursors from a readily accessible autologous adult human source, which could be used as a platform for further experimental studies and has potential therapeutic implications.


Subject(s)
Cell Differentiation/physiology , Neurons/physiology , Periodontium , Stem Cells , Adolescent , Adult , Biomarkers/metabolism , Calcium/metabolism , Cell Lineage , Cell Proliferation , Cells, Cultured , Flow Cytometry , Guided Tissue Regeneration, Periodontal , Humans , Middle Aged , Neurons/cytology , Periodontium/cytology , Periodontium/surgery , Phenotype , Stem Cells/cytology , Stem Cells/physiology
7.
Stem Cells ; 25(8): 2118-27, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17478584

ABSTRACT

Engraftment of mesenchymal stem cells (MSCs) derived from adult bone marrow has been proposed as a potential therapeutic approach for postinfarction left ventricular dysfunction. However, limited cell viability after transplantation into the myocardium has restricted its regenerative capacity. In this study, we genetically modified MSCs with an antiapoptotic Bcl-2 gene and evaluated cell survival, engraftment, revascularization, and functional improvement in a rat left anterior descending ligation model via intracardiac injection. Rat MSCs were manipulated to overexpress the Bcl-2 gene. In vitro, the antiapoptotic and paracrine effects were assessed under hypoxic conditions. In vivo, the Bcl-2 gene-modified MSCs (Bcl-2-MSCs) were injected after myocardial infarction. The surviving cells were tracked after transplantation. Capillary density was quantified after 3 weeks. The left ventricular function was evaluated by pressure-volume loops. The Bcl-2 gene protected MSCs against apoptosis. In vitro, Bcl-2 overexpression reduced MSC apoptosis by 32% and enhanced vascular endothelial growth factor secretion by more than 60% under hypoxic conditions. Transplantation with Bcl-2-MSCs increased 2.2-fold, 1.9-fold, and 1.2-fold of the cellular survival at 4 days, 3 weeks, and 6 weeks, respectively, compared with the vector-MSC group. Capillary density in the infarct border zone was 15% higher in Bcl-2-MSC transplanted animals than in vector-MSC treated animals. Furthermore, Bcl-2-MSC transplanted animals had 17% smaller infarct size than vector-MSC treated animals and exhibited functional recovery remarkably. Our current findings support the premise that transplantation of antiapoptotic gene-modified MSCs may have values for mediating substantial functional recovery after acute myocardial infarction.


Subject(s)
Apoptosis , Genes, bcl-2 , Heart/physiology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Animals , Apoptosis/genetics , Cell Differentiation , Cell Hypoxia/genetics , Cells, Cultured , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Myocardial Infarction/therapy , Organisms, Genetically Modified , Rats , Rats, Inbred Lew , Regeneration , Transfection , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
8.
Cardiovasc Res ; 71(1): 158-69, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16730684

ABSTRACT

OBJECTIVE: The regenerative potential of endothelial and hematopoietic progenitor cells in the heart may vary according to their origin. This study was designed to compare the functional effects of CD133+ cells from human cord blood and bone marrow in a mouse model of myocardial injury. METHODS: 5 x 10(5) CD133+ cells from bone marrow (BM(CD133)) or cord blood (UCB(CD133)) were injected in the necrosis border zone of NOD/SCID (non-obese diabetic/severe combined immunodeficiency) mice with left ventricular cryoinjury (CI+). Transplanted cells were tracked by immunostaining for hNuclear antigen and by PCR for hDNA. Echocardiography was used to measure contractility. Scar size, capillary density, and cardiomyocyte apoptosis were evaluated by histology. In addition, the myogenic and endothelial differentiation capacity of BM(CD133) and UCB(CD133) was compared in vitro. RESULTS: DNA was detected 4 weeks after cell injection by PCR, but hNuc+ cells were found by immunostaining only after 48 h. Capillary density in both BM(CD133) and UCB(CD133) cell-treated CI+ mice was higher than in control CI+ mice, but not different between BM(CD133) and UCB(CD133) cell-treated hearts. There were no differences in scar size and myocardial mass among BM(CD133), UCB(CD133) and control CI+ mice, but cardiomyocyte apoptosis was reduced by both BM(CD133) and UCB(CD133) cells. The post-injury deterioration of shortening fraction (46.2+/-1% in sham-operated mice and 41.3+/-0.8% in control CI+ mice) was prevented by BM(CD133) cells (45.4+/-0.9%), but not by UCB(CD133) cells (40.8+/-0.7%). On the other hand, both BM(CD133) and UCB(CD133) cells abolished post-injury mortality. In vitro, neither cultivated BM(CD133) or UCB(CD133) cells developed into myocytes, but both readily differentiated towards an endothelial cell phenotype. CONCLUSIONS: While both cord blood and marrow CD133+ cells have some beneficial effects on post-injury angiogenesis and survival, only marrow cells appear to improve myocardial contractility.


Subject(s)
Bone Marrow Transplantation/methods , Cardiomyopathies/surgery , Cord Blood Stem Cell Transplantation/methods , Myocardium/pathology , AC133 Antigen , Animals , Antigens, CD/immunology , Apoptosis , Capillaries , Cardiomyopathies/immunology , Cardiomyopathies/physiopathology , Cell Differentiation , Cells, Cultured , Cold Temperature/adverse effects , Echocardiography , Endothelial Cells/cytology , Female , Flow Cytometry , Glycoproteins/immunology , Humans , Immunohistochemistry/methods , Injections , Mice , Mice, Inbred NOD , Mice, SCID , Models, Animal , Myocardium/immunology , Myocytes, Cardiac/cytology , Necrosis , Neovascularization, Physiologic , Peptides/immunology , Transplantation, Heterologous , Treatment Outcome
9.
J Am Soc Echocardiogr ; 19(5): 563-8, 2006 May.
Article in English | MEDLINE | ID: mdl-16644442

ABSTRACT

OBJECTIVES: Intramyocardial transplantation of bone marrow-derived cells is currently under clinical evaluation as a therapy of heart failure. A major limitation of all clinical studies dealing with myocardial cell engraftment is the inability to track the fate of the transplanted cells. We present a clinically applicable technique using transesophageal echocardiography (TEE) of CliniMACS nanoparticle labeled transplanted CD133+ cells in ischemic hearts. METHODS AND RESULTS: CD133+ cells were isolated from human bone marrow by CliniMACS magnetic bead selection. Positive (5 x 10(6) cells) cells were transplanted into porcine ischemic myocardium (n = 6). Control animals (n = 5) received medium injection. TEE was performed to demonstrate the distribution of the cells during and 8 weeks after the procedure. Accumulations of labeled CD133+ cells of adjacent injections in the myocardium were discriminatively identified by TEE. CONCLUSIONS: TEE is an elegant method for real-time documentation of successful transplantation and cell colony localization of magnetically labeled CD133+ cells in the ischemic myocardium. Our method should be of special interest for TEE-guided transcatheter injection of cells.


Subject(s)
Echocardiography, Transesophageal/methods , Echocardiography/methods , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/surgery , Peripheral Blood Stem Cell Transplantation/methods , Stem Cells/diagnostic imaging , Animals , Cell Count/methods , Cells, Cultured , Contrast Media , Feasibility Studies , Humans , Image Enhancement/methods , Immunomagnetic Separation/methods , Myocardium , Nanostructures/analysis , Swine
10.
Stem Cells ; 23(4): 463-70, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15790766

ABSTRACT

The liver has a large capacity for regeneration after resection. However, below a critical level of future liver remnant volume (FLRV), partial hepatectomy is accompanied by a significant increase of postoperative liver failure. There is accumulating evidence for the contribution of bone marrow stem cells (BMSCs) to participate in liver regeneration. Here we report on three patients subjected to intraportal administration of autologous CD133(+) BMSCs subsequent to portal venous embolization of right liver segments, used to expand left lateral hepatic segments as FLRV. Computerized tomography scan volumetry revealed 2.5-fold increased mean proliferation rates of left lateral segments compared with a group of three consecutive patients treated without application of BMSCs. This early experience with portovenous application of CD133(+) BMSCs could suggest that this novel therapeutic approach bears the potential of enhancing and accelerating hepatic regeneration in a clinical setting.


Subject(s)
Antigens, CD/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Glycoproteins/metabolism , Liver Neoplasms/therapy , Liver Regeneration , Peptides/metabolism , Portal Vein , AC133 Antigen , Aged , Bone Marrow Cells/cytology , Bone Marrow Transplantation/adverse effects , Embolization, Therapeutic , Female , Hepatectomy , Humans , Liver Neoplasms/pathology , Liver Neoplasms/surgery , Male , Middle Aged , Transplantation, Autologous
11.
Methods Mol Med ; 109: 55-70, 2005.
Article in English | MEDLINE | ID: mdl-15585913

ABSTRACT

Dendritic cells (DC) can either be generated from progenitors such as stem cells or CD14+ monocytes, or isolated directly from the blood. Blood-derived DC are present as at least two distinct populations-myeloid and plasmacytoid DC. Here we describe methods for the clinical-grade isolation of blood DC and DC precursors using the CliniMACS. We describe the isolation of ultra-pure monocytes in order to generate large numbers of monocyte-derived DC, and also new methods for the direct isolation of blood DC. Isolation of blood DC in large numbers means that natural DC with different properties can be investigated for their clinical function for the first time.


Subject(s)
Cell Separation/methods , Dendritic Cells/cytology , Dendritic Cells/immunology , Immunomagnetic Separation/methods , Cell Differentiation , Flow Cytometry , Humans , Lipopolysaccharide Receptors/immunology , Monocytes/cytology , Monocytes/immunology
12.
J Immunol Methods ; 275(1-2): 57-68, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12667670

ABSTRACT

BACKGROUND: Dendritic cells (DCs) are the most potent antigen-presenting cells. They can be generated in vitro from CD14+ cells, and also from CD34+ progenitor cells. Although T cell proliferation using [3H] thymidine incorporation assay has been used widely to check DC function, this technique only provides limited information about the T cell proliferation. Here, we describe a novel method for quantitative analysis of T cell proliferation using flow cytometry. MATERIALS AND METHODS: DCs were generated from CD14+ cells from six healthy blood donors. Monocytes were isolated using positive selection with magnetic cell sorting (MACS) and then cultured with IL-4, GM-CSF, IL-1beta, IL-6, TNF-alpha and PGE(2) to yield fully mature DCs. Allogeneic naive T lymphocytes with known mismatches in HLA classes I and II were cocultured with DCs. Naive T cells without DC stimulation served as negative controls. T cells were harvested on days 0, 3, 5, 7, 9, 11 and analysed by flow cytometry. CD3-ECD and CD4-fluorescein isothiocyanate (FITC) or CD8-FITC antibodies were used to distinguish T cell subsets, whereas T cell activation was measured by assessment of HLA-DR, CD45RO, CD25 and CD71 expression. For T cell quantification, fluorescent microparticles were used. Dead cells were excluded with 7-AAD. The bromdeoxyridine (BrdU)-incorporation ELISA procedure was also performed in order to compare with the T cell proliferation assay with regard to absolute cell counts and CD71 expression. RESULTS: The initial T cell concentration on day 1 was 203.9+/-39.7 (173-265) CD3+/CD4+ cells/micro l and 184.5+/-41.6 (148-260) CD3+/CD8+ cells/micro l. The maximal T cell proliferation was recorded on day 7 with a five- to tenfold T cell expansion which resulted in 1994.9+/-383 (1446-2404) CD3+/CD4+ cells/micro l and 944+/-303.7 (560-1483) CD3+/CD8+ cells/micro l. Furthermore, activation markers of both cell lineages were upregulated and reached maxima on days 7 (CD71) and 9 (CD25, HLA-DR). T cell count/micro l as well as CD71 expression both correlated significantly with BrdU incorporation. CONCLUSION: Flow cytometric analysis permits simple, precise and rapid quantification of T cell proliferation in a mixed lymphocyte reaction with DCs. Activation, proliferation and cell viability can be simultaneously determined. CD71 is particularly well suited as an activation marker for the simultaneous measurement of T cell proliferation. Thus, specific T cell subsets involved in antigen-specific proliferation can be evaluated in detail.


Subject(s)
Dendritic Cells/immunology , Flow Cytometry/methods , Lymphocyte Activation , Lymphocyte Culture Test, Mixed/methods , T-Lymphocytes/immunology , Bromodeoxyuridine/metabolism , CD3 Complex/metabolism , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Humans , In Vitro Techniques , Lipopolysaccharide Receptors/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...