Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Gut ; 72(10): 1887-1903, 2023 10.
Article in English | MEDLINE | ID: mdl-37399271

ABSTRACT

OBJECTIVE: Colorectal tumours are often densely infiltrated by immune cells that have a role in surveillance and modulation of tumour progression but are burdened by immunosuppressive signals, which might vary from primary to metastatic stages. Here, we deployed a multidimensional approach to unravel the T-cell functional landscape in primary colorectal cancers (CRC) and liver metastases, and genome editing tools to develop CRC-specific engineered T cells. DESIGN: We paired high-dimensional flow cytometry, RNA sequencing and immunohistochemistry to describe the functional phenotype of T cells from healthy and neoplastic tissue of patients with primary and metastatic CRC and we applied lentiviral vectors (LV) and CRISPR/Cas9 genome editing technologies to develop CRC-specific cellular products. RESULTS: We found that T cells are mainly localised at the front edge and that tumor-infiltrating T cells co-express multiple inhibitory receptors, which largely differ from primary to metastatic sites. Our data highlighted CD39 as the major driver of exhaustion in both primary and metastatic colorectal tumours. We thus simultaneously redirected T-cell specificity employing a novel T-cell receptor targeting HER-2 and disrupted the endogenous TCR genes (TCR editing (TCRED)) and the CD39 encoding gene (ENTPD1), thus generating TCREDENTPD1KOHER-2-redirected lymphocytes. We showed that the absence of CD39 confers to HER-2-specific T cells a functional advantage in eliminating HER-2+ patient-derived organoids in vitro and in vivo. CONCLUSION: HER-2-specific CD39 disrupted engineered T cells are promising advanced medicinal products for primary and metastatic CRC.


Subject(s)
Antigens, CD , Apyrase , Colorectal Neoplasms , Liver Neoplasms , T-Lymphocytes , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Receptors, Antigen, T-Cell , Apyrase/genetics , Antigens, CD/genetics , Cell Engineering
3.
Stem Cell Reports ; 13(5): 832-846, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31587993

ABSTRACT

Neuroferritinopathy (NF) is a movement disorder caused by alterations in the L-ferritin gene that generate cytosolic free iron. NF is a unique pathophysiological model for determining the direct consequences of cell iron dysregulation. We established lines of induced pluripotent stem cells from fibroblasts from two NF patients and one isogenic control obtained by CRISPR/Cas9 technology. NF fibroblasts, neural progenitors, and neurons exhibited the presence of increased cytosolic iron, which was also detectable as: ferritin aggregates, alterations in the iron parameters, oxidative damage, and the onset of a senescence phenotype, particularly severe in the neurons. In this spontaneous senescence model, NF cells had impaired survival and died by ferroptosis. Thus, non-ferritin-bound iron is sufficient per se to cause both cell senescence and ferroptotic cell death in human fibroblasts and neurons. These results provide strong evidence supporting the primary role of iron in neuronal aging and degeneration.


Subject(s)
Ferroptosis , Iron Metabolism Disorders/pathology , Iron/metabolism , Neuroaxonal Dystrophies/pathology , Neurons/pathology , Cells, Cultured , Cellular Senescence , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Iron Metabolism Disorders/metabolism , Middle Aged , Neuroaxonal Dystrophies/metabolism , Neurons/metabolism
4.
Front Immunol ; 10: 1391, 2019.
Article in English | MEDLINE | ID: mdl-31275324

ABSTRACT

Pemphigus vulgaris (PV) is an autoimmune bullous disease of the skin and mucous membranes characterized by the presence of circulating and tissue-bound autoantibodies against keratinocyte cell surface antigens, specifically desmoglein (Dsg) 1 and 3. The pathogenic role of anti-Dsg antibodies is well-established, while the mechanism of blister formation is only partly defined. We have applied a previously developed method for the efficient immortalization of IgG+ memory B cells to identify novel target antigens in PV. A human monoclonal antibody reactive with a hitherto unreported non-Dsg antigen was isolated. Immunoprecipitation and immunoblotting studies with keratinocyte extracts indicated α-catenin as the putative antigen, then confirmed by immunoblotting on the recombinant protein. Four of ten PV sera reacted with recombinant α-catenin. Although the isolated human monoclonal antibody was per se unable to dissociate keratinocyte monolayers and also to synergize with a pathogenic antibody in vitro, further studies are warranted to assess its possible in vivo contribution in the multifactorial pathogenesis and heterogeneous manifestations of PV disease.


Subject(s)
Autoantigens/immunology , Autoimmunity , Disease Susceptibility/immunology , Pemphigus/immunology , Pemphigus/pathology , Autoantibodies/immunology , Autoantigens/metabolism , Biomarkers , Desmogleins/immunology , Desmogleins/metabolism , Epithelium/immunology , Epithelium/metabolism , Epithelium/pathology , Humans , Pemphigus/metabolism , alpha Catenin/immunology , alpha Catenin/metabolism
5.
Front Immunol ; 9: 3171, 2018.
Article in English | MEDLINE | ID: mdl-30719025

ABSTRACT

Immunomodulatory drugs (IMiDs) are effective therapeutics for multiple myeloma (MM), where in different clinical settings they exert their function both directly on MM cells and indirectly by modulating immune cell subsets, although with not completely defined mechanisms. Here we studied the role of IMiDs in the context of autologous hematopoietic stem cell transplantation on the T cell subset distribution in the bone marrow of newly diagnosed MM patients. We found that after transplantation pro-tumor Th17-Th1 and Th22 cells and their related cytokines were lower in patients treated with IMiDs during induction chemotherapy compared to untreated patients. Of note, lower levels of IL-17, IL-22, and related IL-6, TNF-α, IL-1ß, and IL-23 in the bone marrow sera correlated with treatment with IMiDs and favorable clinical outcome. Collectively, our results suggest a novel anti-inflammatory role for IMiDs in MM.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Hematopoietic Stem Cell Transplantation , Immunomodulation/drug effects , Lymphocyte Count , Multiple Myeloma/immunology , Multiple Myeloma/therapy , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Adult , Aged , Antineoplastic Agents, Immunological/pharmacology , Biomarkers , Combined Modality Therapy , Cytokines/metabolism , Female , Hematopoietic Stem Cell Transplantation/methods , Humans , Immunophenotyping , Male , Middle Aged , Multiple Myeloma/diagnosis , T-Lymphocyte Subsets/metabolism , Treatment Outcome
6.
Oncoimmunology ; 5(4): e1093278, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27141378

ABSTRACT

We recently reported that in multiple myeloma increased Th22 cell frequencies correlate with poor prognosis. Here we show that within the same patients' cohort Th17 cells associate with bone disease and not with prognosis. Thus, we propose that Th22 and Th17 cells play non-redundant roles in multiple myeloma and constitute independent therapeutic targets.

7.
Methods Mol Biol ; 1393: 37-51, 2016.
Article in English | MEDLINE | ID: mdl-27033214

ABSTRACT

CD4(+) T cells comprise a significant portion of tumor-infiltrating lymphocytes. Different subsets of CD4(+) T cells exist and they exert different effector functions in tumor immunity depending on the cytokines produced going from antitumor to pro-tumor. Methods that use small aliquots of cells to identify ex vivo the frequency and functional orientation of tumor-specific CD4(+) T cells in the blood and visualization of the presence of different CD4(+) T cell subsets and their localization at the tumor site are valuable tools to determine their clinical impact in neoplastic diseases.


Subject(s)
CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Cell Separation , Cells, Cultured , GATA3 Transcription Factor/metabolism , Humans , T-Box Domain Proteins/metabolism
8.
Oncoimmunology ; 4(5): e1005460, 2015 May.
Article in English | MEDLINE | ID: mdl-26155400

ABSTRACT

There is increased production of plasmacytoid dendritic cells (pDCs) in the bone marrow (BM) of multiple myeloma (MM) patients and these favor Th22 cell differentiation. Here, we found that the frequency of interleukin (IL)-22+IL-17-IL-13+ T cells is significantly increased in peripheral blood (PB) and BM of stage III and relapsed/refractory MM patients compared with healthy donors and patients with asymptomatic or stage I/II disease. Th22 cells cloned from the BM of MM patients were CCR6+CXCR4+CCR4+CCR10- and produced IL-22 and IL-13 but not IL-17. Furthermore, polyfunctional Th22-Th2 and Th22-Th1 clones were identified based on the co-expression of additional chemokine receptors and cytokines (CRTh2 or CXCR3 and IL-5 or interferon gamma [IFNγ], respectively). A fraction of MM cell lines and primary tumors aberrantly expressed the IL-22RA1 and IL-22 induced STAT-3 phosphorylation, cell growth, and resistance to drug-induced cell death in MM cells. IL-13 treatment of normal BM mesenchymal stromal cells (MSCs) induced STAT-6 phosphorylation, adhesion molecule upregulation, and increased IL-6 production and significantly favored MM cell growth compared with untreated BM MSCs. Collectively, our data show that increased frequency of IL-22+IL-17-IL-13+ T cells correlates with poor prognosis in MM through IL-22 and IL-13 protumor activity and suggest that interference with IL-22 and IL-13 signaling pathways could be exploited for therapeutic intervention.

9.
J Clin Invest ; 122(10): 3781-90, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22996451

ABSTRACT

Pemphigus vulgaris (PV) is an autoimmune blistering disease of skin and mucous membranes caused by autoantibodies to the desmoglein (DSG) family proteins DSG3 and DSG1, leading to loss of keratinocyte cell adhesion. To learn more about pathogenic PV autoantibodies, we isolated 15 IgG antibodies specific for DSG3 from 2 PV patients. Three antibodies disrupted keratinocyte monolayers in vitro, and 2 were pathogenic in a passive transfer model in neonatal mice. The epitopes recognized by the pathogenic antibodies were mapped to the DSG3 extracellular 1 (EC1) and EC2 subdomains, regions involved in cis-adhesive interactions. Using a site-specific serological assay, we found that the cis-adhesive interface on EC1 recognized by the pathogenic antibody PVA224 is the primary target of the autoantibodies present in the serum of PV patients. The autoantibodies isolated used different heavy- and light-chain variable region genes and carried high levels of somatic mutations in complementary-determining regions, consistent with antigenic selection. Remarkably, binding to DSG3 was lost when somatic mutations were reverted to the germline sequence. These findings identify the cis-adhesive interface of DSG3 as the immunodominant region targeted by pathogenic antibodies in PV and indicate that autoreactivity relies on somatic mutations generated in the response to an antigen unrelated to DSG3.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Desmoglein 3/immunology , Immunoglobulin G/immunology , Pemphigus/immunology , Somatic Hypermutation, Immunoglobulin , Amino Acid Sequence , Animals , Animals, Newborn , Antigen-Antibody Reactions , Autoantigens/chemistry , Cells, Cultured , Complementarity Determining Regions/immunology , Desmoglein 3/chemistry , Epitopes/chemistry , Epitopes/immunology , Humans , Immunization, Passive , Immunoglobulin Variable Region/genetics , Keratinocytes/immunology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Protein Conformation , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid
10.
PLoS One ; 7(8): e42340, 2012.
Article in English | MEDLINE | ID: mdl-22879946

ABSTRACT

BACKGROUND: Knowledge of antigen-specific CD4(+) T cells frequencies is pivotal to the choice of the antigen to be used in anti-viral and anti-tumor vaccination procedures and for monitoring of immune responses. Methods that employ small cell numbers from patient samples, are easy to perform and do not require complex techniques/instrumentations and therefore standardization are desirable. METHODOLOGY/PRINCIPAL FINDINGS: Purified blood CD4(+) T cells from healthy donors were cultured with autologous antigen presenting cells in several replicate wells in equal numbers in the absence (un-stimulated wells) or in the presence of synthetic peptides corresponding to viral antigens promiscuous HLA-DR epitopes (antigen-stimulated wells). At day 7 of culture low dose IL-2 was added and at day 14 IFN-γ and IL-5 release in the supernatant was measured. A statistical analysis approach, based on Poisson distribution, was then implemented to calculate the frequency of viral-specific CD4(+) T cells. We first determined a patient-specific exceptionality threshold of cytokine release in the un-stimulated wells and then, based on this threshold, we counted the inactive/active wells within the antigen-stimulated wells. This number, along with the number of cells per well, allowed the point and interval estimates of frequencies. A ready-to-use Excel worksheet template with automatic calculations for frequencies estimate was developed and is provided as a supplemental file (Table S9). CONCLUSIONS/SIGNIFICANCE: We report a simple experimental procedure combining short term in vitro cell culture with statistical analysis to calculate the frequency of antigen-specific CD4(+) T cells. The detailed experimental procedure along with the Excel applicative are a valuable tool for monitoring immune responses in the clinical practice.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Epitopes/immunology , CD4 Lymphocyte Count , Cell Proliferation , Cells, Cultured , Epstein-Barr Virus Nuclear Antigens/immunology , Humans , Immunologic Memory/immunology , Interferon-gamma/metabolism , Interleukin-5/biosynthesis , Lymphocyte Activation/immunology , Poisson Distribution , Reproducibility of Results
11.
J Virol Methods ; 163(2): 195-204, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19778556

ABSTRACT

Modified vaccinia virus Ankara (MVA) is employed as a human vaccine vector for the high expression of heterologous genes and the lack of replication in mammalian cells. This study demonstrates that cells infected by recombinant viruses can be obtained by fluorescence-activated cell sorting. Recombinant viruses are generated by a swapping event between a red fluorescent protein gene in the acceptor virus and a plasmid cassette coding for both a green fluorescent marker and a transgene. To prevent the carry-over of parental virus, due to superinfection of the cells harbouring recombinant viruses, the sorting is performed on cells infected at low m.o.i. in the presence of a reversible inhibitor of viral particle release. Terminal dilution cloning is then used to isolate both green and marker-free recombinant viruses, which can be identified by whole-plate fluoroimaging. The differential visualization of all the viral types involved allows a stepwise monitoring of all recombinations and leads to a straightforward and efficient flow cytometry-based cell sorting purification protocol. As an example of the efficacy of this sorting procedure, the construction of rMVA's coding for the rat nuclear protein HMGB1 and H5N1 influenza A virus hemagglutinin is reported. The entire recombinant MVA production process is carried out in serum-free media employing primary chicken embryo fibroblasts (CEF), which are certified for the preparation of human vaccines. This rMVA production method is faster, simpler and more reliable than any other available procedure for obtaining safe vaccine stocks for human use.


Subject(s)
Genes, Viral , Recombination, Genetic , Smallpox Vaccine , Vaccinia virus/growth & development , Vaccinia virus/isolation & purification , Animals , Cell Culture Techniques/methods , Cells, Cultured , Chick Embryo , Chickens , Culture Media, Serum-Free , Fibroblasts/virology , Flow Cytometry/methods , Green Fluorescent Proteins/genetics , HMGB1 Protein/genetics , Influenza A Virus, H5N1 Subtype/genetics , Luminescent Proteins/genetics , Rats , Staining and Labeling/methods , Vaccinia virus/genetics , Red Fluorescent Protein
12.
J Virol Methods ; 156(1-2): 37-43, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19038289

ABSTRACT

Modified Vaccinia Virus Ankara (MVA) is employed widely as an experimental and human vaccine vector for its lack of replication in mammalian cells and high expression of heterologous genes. Recombinant MVA technology can be improved greatly by combining transient host-range selection (based on the restoration in MVA of the deleted vaccinia gene K1L) with the differential expression of fluorescent proteins. Recombinant virus results from swapping a red protein gene (in the acceptor virus) with a cassette of the transfer plasmid comprising the transgene and the green marker K1Lgfp (a chimeric gene comprising K1L and EGFP). Recombinant selection is performed in the selective host RK13. Finally, in the non-selective host BHK-21, a single crossover between identical flanking regions excises the marker gene. The three types of viruses involved (red parental, green intermediate and colourless final recombinant) are visualized differentially by fluorescence microscopy or fluoro-imaging of terminal dilution microcultures, leading to a straightforward and efficient purification protocol. This method (Red-to-Green gene swapping) reduces greatly the time needed to obtain marker-free recombinant MVA and increases the reliability of the construction process.


Subject(s)
Defective Viruses/genetics , Genetic Engineering/methods , Vaccinia virus/genetics , Animals , Cell Line , Cricetinae , DNA, Recombinant/genetics , DNA, Viral/genetics , Fluorescent Dyes/metabolism , Genes, Reporter , Genes, Viral , Genetic Vectors , Microscopy, Fluorescence , Plasmids , Rabbits , Species Specificity , Transfection , Transgenes
13.
J Immunol ; 174(9): 5602-11, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15843559

ABSTRACT

Interaction of secretory IgE with FcepsilonRI is the prerequisite for allergen-driven cellular responses, fundamental events in immediate and chronic allergic manifestations. Previous studies reported the binding of soluble FcepsilonRIalpha to membrane IgE exposed on B cells. In this study, the functional interaction between human membrane IgE and human FcepsilonRI is presented. Four different IgE versions were expressed in mouse B cell lines, namely: a truncation at the Cepsilon2-Cepsilon3 junction of membrane IgE isoform long, membrane IgE isoform long (without Igalpha/Igbeta BCR accessory proteins), and both epsilonBCRs (containing membrane IgE isoforms short and long). All membrane IgE versions activated a rat basophilic leukemia cell line transfected with human FcepsilonRI, as detected by measuring the release of both preformed and newly synthesized mediators. The interaction led also to Ca(2+) responses in the basophil cell line, while membrane IgE-FcepsilonRI complexes were detected by immunoprecipitation. FcepsilonRI activation by membrane IgE occurs in an Ag-independent manner. Noteworthily, human peripheral blood basophils and monocytes also were activated upon contact with cells bearing membrane IgE. In humans, the presence of FcepsilonRI in several cellular entities suggests a possible membrane IgE-FcepsilonRI-driven cell-cell dialogue, with likely implications for IgE homeostasis in physiology and pathology.


Subject(s)
Antigens/physiology , Immunoglobulin E/metabolism , Receptors, Antigen, B-Cell/metabolism , Receptors, IgE/metabolism , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Basophils/immunology , Basophils/metabolism , Binding Sites, Antibody , Binding, Competitive/immunology , CHO Cells , Calcium/metabolism , Cell Communication/immunology , Cell Count , Cell Line, Tumor , Cricetinae , Humans , Immunoglobulin E/physiology , Mice , Monocytes/immunology , Monocytes/metabolism , Multiprotein Complexes/metabolism , Protein Binding/immunology , Protein Isoforms/biosynthesis , Protein Isoforms/metabolism , Rats , Receptors, Antigen, B-Cell/physiology , Receptors, IgE/antagonists & inhibitors , Receptors, IgE/biosynthesis , SRS-A/analogs & derivatives , SRS-A/metabolism , Solubility , Time Factors
14.
J Virol ; 79(5): 2807-13, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15708999

ABSTRACT

Human herpesvirus 6 (HHV-6) is a potentially immunosuppressive CD4(+)-T-lymphotropic betaherpesvirus that causes severe human thymocyte depletion in heterochimeric SCID-hu thy/liv mice and has been implicated as a potential cofactor in the progression of AIDS. However, the mechanisms of HHV-6-mediated immunosuppression have not yet been fully elucidated. We investigated the phenotypic and functional alterations induced by HHV-6 on peripheral blood-derived human dendritic cells (DC). The infection of DC with HHV-6 A or B was nonproductive, as revealed by calibrated real-time PCR measuring the accumulation of viral genome equivalents over time. Nevertheless, preexposure to HHV-6 markedly impaired the maturation of DC driven by gamma interferon and lipopolysaccharide, as shown by the reduced surface expression of major histocompatibility complex class I molecules, HLA-DR, CD40, and CD80. Moreover, HHV-6, but not the closely related betaherpesvirus HHV-7, dramatically suppressed the secretion of interleukin-12 (IL-12) p70 by DC, while the production of other cytokines that influence DC maturation, i.e., IL-10 and tumor necrosis factor alpha, was not significantly modified. Likewise, the secretion of the CC chemokines macrophage inflammatory protein 1beta and RANTES was unaltered. Functionally, a pretreatment with HHV-6 impaired the ability of DC to stimulate allogeneic T-cell proliferation. Altogether, these data identify interference with the functional maturation of DC as a potential mechanism of HHV-6-mediated immunosuppression.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , Herpesvirus 6, Human/immunology , Herpesvirus 6, Human/pathogenicity , Interleukin-12/biosynthesis , Protein Subunits/biosynthesis , Cell Differentiation , Chemokines/biosynthesis , Cytokines/biosynthesis , Dendritic Cells/cytology , Herpesvirus 6, Human/physiology , Herpesvirus 7, Human/immunology , Herpesvirus 7, Human/pathogenicity , Humans , Immune Tolerance , In Vitro Techniques , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Lymphocyte Activation , Recombinant Proteins , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Virus Replication
15.
Blood ; 102(8): 2877-84, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-12829600

ABSTRACT

Human herpesvirus 6 (HHV-6) is a potentially immunosuppressive agent that has been suggested to act as a cofactor in the progression of HIV disease. Exposure of human macrophages to HHV-6A or HHV-6B profoundly impaired their ability to produce interleukin 12 (IL-12) upon stimulation with interferon-gamma (IFN-gamma) and lipopolysaccharide (LPS). By contrast, the production of tumor necrosis factor-alpha (TNF-alpha); regulated on activation, normal T-cell expressed and secreted (RANTES); and macrophage inflammatory protein 1 beta (MIP-1 beta) was not negatively affected. To exclude the involvement of IL-12-suppressive cytokines, such as IL-10 and TNF-alpha, the viral stocks were fractionated by ultra-centrifugation. The bulk of the suppressive activity was recovered within the virion-rich pelleted fraction that was virtually devoid of such cytokines. IL-12 suppression was independent of viral replication, and the effect was not abrogated upon ultraviolet-light inactivation of the viral inoculum. The mechanism of HHV-6-mediated IL-12 suppression was investigated by RNase protection assays, which demonstrated unaltered levels of IL-12 p35 mRNA and only a modest reduction in p40 mRNA, which was insufficient to account for the near-complete loss of both extracellular and intracellular IL-12 protein. Moreover, both the IFN-gamma and the LPS signaling pathways were intact in HHV-6-treated cells. These data suggest that HHV-6 can dramatically affect the generation of effective cellular immune responses, providing a novel potential mechanism of HHV-6-mediated immunosuppression.


Subject(s)
Herpesvirus 6, Human/metabolism , Interleukin-12/antagonists & inhibitors , Antigens, CD/biosynthesis , Blotting, Western , Chemokine CCL4 , Chemokine CCL5/metabolism , Chemokines/metabolism , Cytokines/metabolism , Flow Cytometry , HIV/metabolism , Humans , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-12/metabolism , Lipopolysaccharides/metabolism , Macrophage Inflammatory Proteins/metabolism , Macrophages/metabolism , Membrane Cofactor Protein , Membrane Glycoproteins/biosynthesis , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Ribonucleases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ultracentrifugation , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...