Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Front Neurol ; 10: 992, 2019.
Article in English | MEDLINE | ID: mdl-31611837

ABSTRACT

Cardiac involvement is one of the most important manifestations of the multisystemic phenotype of patients affected by myotonic dystrophy (DM) and represents the second cause of premature death. Molecular mechanisms responsible for DM cardiac defects are still unclear; however, missplicing of the cardiac isoform of troponin T (TNNT2) and of the cardiac sodium channel (SCN5A) genes might contribute to the reduced myocardial function and conduction abnormalities seen in DM patients. Since, in DM skeletal muscle, the TNNT2 gene shows the same aberrant splicing pattern observed in cardiac muscle, the principal aim of this work was to verify if the TNNT2 aberrant fetal isoform expression could be secondary to myopathic changes or could reflect the DM cardiac phenotype. Analysis of alternative splicing of TNNT2 and of several genes involved in DM pathology has been performed on muscle biopsies from patients affected by DM type 1 (DM1) or type 2 (DM2) with or without cardiac involvement. Our analysis shows that missplicing of muscle-specific genes is higher in DM1 and DM2 than in regenerating control muscles, indicating that these missplicing could be effectively important in DM skeletal muscle pathology. When considering the TNNT2 gene, missplicing appears to be more evident in DM1 than in DM2 muscles since, in DM2, the TNNT2 fetal isoform appears to be less expressed than the adult isoform. This evidence does not seem to be related to less severe muscle histopathological alterations that appear to be similar in DM1 and DM2 muscles. These results seem to indicate that the more severe TNNT2 missplicing observed in DM1 could not be related only to myopathic changes but could reflect the more severe general phenotype compared to DM2, including cardiac problems that appear to be more severe and frequent in DM1 than in DM2 patients. Moreover, TNNT2 missplicing significantly correlates with the QRS cardiac parameter in DM1 but not in DM2 patients, indicating that this splicing event has good potential to function as a biomarker of DM1 severity and it should be considered in pharmacological clinical trials to monitor the possible effects of different therapeutic approaches on skeletal muscle tissues.

2.
PLoS One ; 14(3): e0214254, 2019.
Article in English | MEDLINE | ID: mdl-30901379

ABSTRACT

Myotonic dystrophy type 1 (DM1) and type 2 (DM2) are autosomal dominant multisystemic disorders linked to two different genetic loci and characterized by several features including myotonia, muscle atrophy and insulin resistance. The aberrant alternative splicing of insulin receptor (IR) gene and post-receptor signalling abnormalities have been associated with insulin resistance, however the precise molecular defects that cause metabolic dysfunctions are still unknown. Thus, the aims of this study were to investigate in DM skeletal muscle biopsies if beyond INSR missplicing, altered IR protein expression could play a role in insulin resistance and to verify if the lack of insulin pathway activation could contribute to skeletal muscle wasting. Our analysis showed that DM skeletal muscle exhibits a lower expression of the insulin receptor in type 1 fibers which can contribute to the defective activation of the insulin pathway. Moreover, the aberrant insulin signalling activation leads to a lower activation of mTOR and to an increase in MuRF1 and Atrogin-1/MAFbx expression, possible explaining DM skeletal muscle fiber atrophy. Taken together our data indicate that the defective insulin signalling activation can contribute to skeletal muscle features in DM patients and are probably linked to an aberrant specific-fiber type expression of the insulin receptor.


Subject(s)
Antigens, CD/analysis , Insulin Resistance , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Myotonic Dystrophy/pathology , Receptor, Insulin/analysis , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult
3.
Sci Rep ; 8(1): 11058, 2018 07 23.
Article in English | MEDLINE | ID: mdl-30038349

ABSTRACT

A patient with an early severe myotonia diagnosed for Myotonic Dystrophy type 2 (DM2) was found bearing the combined effects of DM2 mutation and Nav1.4 S906T substitution. To investigate the mechanism underlying his atypical phenotype,whole-cell patch-clamp in voltage- and current-clamp mode was performed in myoblasts and myotubes obtained from his muscle biopsy. Results characterizing the properties of the sodium current and of the action potentials have been compared to those obtained in muscle cells derived from his mother, also affected by DM2, but without the S906T polymorphism. A faster inactivation kinetics and a +5 mV shift in the availability curve were found in the sodium current recorded in patient's myoblasts compared to his mother. 27% of his myotubes displayed spontaneous activity. Patient's myotubes showing a stable resting membrane potential had a lower rheobase current respect to the mother's while the overshoot and the maximum slope of the depolarizing phase of action potential were higher. These findings suggest that SCN4A polymorphisms may be responsible for a higher excitability of DM2 patients sarcolemma, supporting the severe myotonic phenotype observed. We suggest SCN4A as a modifier factor and that its screening should be performed in DM2 patients with uncommon clinical features.


Subject(s)
Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , NAV1.4 Voltage-Gated Sodium Channel/metabolism , Action Potentials/physiology , Adult , Chloride Channels/genetics , Chloride Channels/metabolism , Female , Humans , Male , Membrane Potentials/genetics , Membrane Potentials/physiology , Middle Aged , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Mutation/genetics , Myoblasts/cytology , Myoblasts/metabolism , NAV1.4 Voltage-Gated Sodium Channel/genetics , Sarcolemma/genetics , Sarcolemma/metabolism
4.
Clin Chim Acta ; 484: 111-116, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29803895

ABSTRACT

BACKGROUND: Myotonic dystrophy type 1 (DM1) is caused by an expanded CTG repeat in the non-coding 3' UTR of the DMPK gene. PCR and Southern Blot Analysis (SBA) of long-range PCR represent the routine molecular testing most widely used for DM1 diagnosis. However, in these conventional methods artifacts such as allele dropout (ADO) represent a risk of misdiagnosis for DM1. Subjects, who show a single product by conventional methods, require a complementary technique such as triplet repeat primed PCR (TP-PCR). OBJECT: To estimate and minimize the incidence of allele dropout event in our diagnostic molecular laboratory by the use of new kit TP-PCR-based. METHODS: We retrospectively studied 190 DMPK alleles, on blood samples from to ninety-five subjects, previously genotyped by traditional methods to validate a new assay. The pedigree of a DM1 family was used to expand our analysis. RESULTS: TP-PCR assay correctly identified all 95/95 (100%) subjects and these results were in agreement with the other molecular laboratory. By conventional methods the amplification failure due to allele dropout in our cohort was in 12/190 (6.3%) DMPK alleles analyzed. When these 12 alleles were detected and solved by new assay, we found that the 2.6% was caused by primer sequence-dependent and the remaining 3.6% by polymerase-hindering secondary structures. CONCLUSIONS: Allele dropout could be considered as a potentially important problem in DM1 diagnosis that may lead to the attribution of a wrong genotype with long-term consequences for both proband and family.


Subject(s)
Alleles , Molecular Diagnostic Techniques , Multiplex Polymerase Chain Reaction , Myotonin-Protein Kinase/genetics , Nucleic Acid Amplification Techniques , Pathology, Molecular , Female , Humans , Male , Middle Aged
5.
PLoS One ; 12(9): e0184987, 2017.
Article in English | MEDLINE | ID: mdl-28915272

ABSTRACT

Myotonic dystrophy type 1 (DM1) and type 2 (DM2) are autosomal dominant multisystemic disorders caused by expansion of microsatellite repeats. In both forms, the mutant transcripts accumulate in nuclear foci altering the function of alternative splicing regulators which are necessary for the physiological mRNA processing. Missplicing of insulin receptor (IR) gene (INSR) has been associated with insulin resistance, however, it cannot be excluded that post-receptor signalling abnormalities could also contribute to this feature in DM. We have analysed the insulin pathway in skeletal muscle biopsies and in myotube cultures from DM patients to assess whether downstream metabolism might be dysregulated and to better characterize the mechanism inducing insulin resistance. DM skeletal muscle exhibits alterations of basal phosphorylation levels of Akt/PKB, p70S6K, GSK3ß and ERK1/2, suggesting that these changes might be accompanied by a lack of further insulin stimulation. Alterations of insulin pathway have been confirmed on control and DM myotubes expressing fetal INSR isoform (INSR-A). The results indicate that insulin action appears to be lower in DM than in control myotubes in terms of protein activation and glucose uptake. Our data indicate that post-receptor signalling abnormalities might contribute to DM insulin resistance regardless the alteration of INSR splicing.


Subject(s)
Alternative Splicing , Antigens, CD , Gene Expression Regulation , Insulin Resistance/genetics , Myotonic Dystrophy , Receptor, Insulin , Signal Transduction , Adult , Aged , Aged, 80 and over , Antigens, CD/biosynthesis , Antigens, CD/genetics , Female , Humans , Male , Middle Aged , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Receptor, Insulin/biosynthesis , Receptor, Insulin/genetics
6.
Am J Pathol ; 183(2): 413-21, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23731727

ABSTRACT

Despite the evidence that tumor necrosis factor (TNF) inhibitors block TNF and the downstream inflammatory cascade, their primary mechanism of action in inhibiting the self-sustaining pathogenic cycle in psoriasis is not completely understood. This study has the aim to identify early critical events for the resolution of inflammation in skin lesions using anti-TNF therapy. We used a translational approach that correlates gene expression fold change in lesional skin with the Psoriasis Area and Severity Index score decrease induced by TNF blockade after 4 weeks of treatment. Data were validated by immunofluorescence microscopy on skin biopsy specimens. We found that the anti-TNF-modulated genes that mostly associated with the clinical amelioration were Ccr7, its ligand, Ccl19, and dendritic cell maturation genes. Decreased expression of T-cell activation genes and Vegf also associated with the clinical response. More important, the down-regulation of Ccr7 observed at 4 weeks significantly correlated with the clinical remission occurring at later time points. Immunofluorescence microscopy on skin biopsy specimens showed that reduction of CCR7(+) cells and chemokine ligand (CCL) 19 was paralleled by disaggregation of the dermal lymphoid-like tissue. These data show that an early critical event for the clinical remission of psoriasis in response to TNF inhibitors is the inhibition of the CCR7/CCL19 axis and support its role in psoriasis pathogenesis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Chemokine CCL19/antagonists & inhibitors , Psoriasis/drug therapy , Receptors, CCR7/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adalimumab , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , CD4 Antigens/metabolism , Cellular Senescence/drug effects , Chemokine CCL19/genetics , Etanercept , Female , Humans , Immunoglobulin G/therapeutic use , Infliximab , Langerhans Cells/drug effects , Lymphocyte Activation/genetics , Male , Middle Aged , Psoriasis/genetics , Receptors, CCR7/genetics , Receptors, Tumor Necrosis Factor/therapeutic use , Remission Induction , Skin/metabolism , T-Lymphocytes/drug effects
7.
Inflamm Bowel Dis ; 19(4): 729-39, 2013.
Article in English | MEDLINE | ID: mdl-23448791

ABSTRACT

BACKGROUND: Antibodies against tumor necrosis factor represent an effective therapy for patients with inflammatory bowel disease. Despite their successful results, the exact mechanism by which infliximab suppresses intestinal inflammation is still a matter of debate. In this study, we used a translational approach to identify the key mechanisms associated with resolution of mucosal inflammation induced by infliximab. METHODS: A total of 16 patients with active inflammatory bowel disease (9 with Crohn's disease and 7 with ulcerative colitis) and 16 controls were enrolled in the study. Patients received infliximab infusions at 0, 2, and 6 weeks. At enrollment and at week 6, patients underwent flexible sigmoidoscopy, and biopsies were taken from the sigmoid colon. RNA was extracted, and mucosal expression of 96 immune-related genes was evaluated by qRT-PCR and confirmed by immunofluorescence microscopy on tissue. Correlation between infliximab-induced gene expression modulation and endoscopic response to therapy was calculated. Lamina propria mononuclear cell apoptosis induced by infliximab was evaluated on tissue sections by the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. RESULTS: We found that infliximab-induced downregulation of macrophage and Th17 pathway genes was significantly associated with both endoscopic response to the therapy and achievement of mucosal healing. Importantly, the observed reduction of lamina propria CD68 macrophages was associated with an increased rate of macrophage apoptosis. CONCLUSIONS: The 2 mechanisms associated with infliximab-induced resolution of intestinal inflammation are the reduction of lamina propria infiltrating CD68 macrophages and the downregulation of interleukin 17A. Moreover, the data suggest that infliximab-induced macrophage apoptosis may represent a key mechanism for the therapeutic success of anti-tumor necrosis factor antibodies.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Colitis, Ulcerative/drug therapy , Crohn Disease/drug therapy , Interleukin-17/metabolism , Intestinal Mucosa/pathology , Macrophages/pathology , Wound Healing , Adult , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Apoptosis/drug effects , Case-Control Studies , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Colitis, Ulcerative/immunology , Colitis, Ulcerative/metabolism , Crohn Disease/immunology , Crohn Disease/metabolism , Down-Regulation , Endoscopy , Female , Gastrointestinal Agents/adverse effects , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Infliximab , Interleukin-17/genetics , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Microscopy, Fluorescence , Middle Aged , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/antagonists & inhibitors
8.
Haematologica ; 97(7): 980-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22271892

ABSTRACT

BACKGROUND: During late differentiation, erythroid cells undergo profound changes involving actin filament remodeling. One of the proteins controlling actin dynamics is gelsolin, a calcium-activated actin filament severing and capping protein. Gelsolin-null (Gsn(-/-)) mice generated in a C57BL/6 background are viable and fertile.1 DESIGN AND METHODS: We analyzed the functional roles of gelsolin in erythropoiesis by: (i) evaluating gelsolin expression in murine fetal liver cells at different stages of erythroid differentiation (using reverse transcription polymerase chain reaction analysis and immunohistochemistry), and (ii) characterizing embryonic and adult erythropoiesis in Gsn(-/-) BALB/c mice (morphology and erythroid cultures). RESULTS: In the context of a BALB/c background, the Gsn(-/-) mutation causes embryonic death. Gsn(-/-) embryos show defective erythroid maturation with persistence of circulating nucleated cells. The few Gsn(-/-) mice reaching adulthood fail to recover from phenylhydrazine-induced acute anemia, revealing an impaired response to stress erythropoiesis. In in vitro differentiation assays, E13.5 fetal liver Gsn(-/-) cells failed to undergo terminal maturation, a defect partially rescued by Cytochalasin D, and mimicked by administration of Jasplakinolide to the wild-type control samples. CONCLUSIONS: In BALB/c mice, gelsolin deficiency alters the equilibrium between erythrocyte actin polymerization and depolymerization, causing impaired terminal maturation. We suggest a non-redundant role for gelsolin in terminal erythroid differentiation, possibly contributing to the Gsn(-/-) mice lethality observed in mid-gestation.


Subject(s)
Embryonic Stem Cells/pathology , Erythrocytes/pathology , Erythropoiesis/genetics , Gelsolin/genetics , Liver/pathology , Actins/antagonists & inhibitors , Actins/metabolism , Anemia/chemically induced , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Cytochalasin D/pharmacology , Depsipeptides/pharmacology , Embryo, Mammalian , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Erythrocytes/drug effects , Erythrocytes/metabolism , Fetus , Gelsolin/deficiency , Gene Expression Regulation, Developmental/drug effects , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Phenylhydrazines/toxicity
9.
Clin Immunol ; 139(2): 164-76, 2011 May.
Article in English | MEDLINE | ID: mdl-21334981

ABSTRACT

The impact of anti-TNF therapy on systemic immune responses in patients has not been clearly defined. Here, we examined Th1/Th2/Th17 cytokine expression, activation and proliferation of peripheral T cells from patients with psoriasis and inflammatory bowel disease before and during anti-TNF therapy. In parallel, we calculated the correlation with the clinical response and we monitored cytokine expression in biopsies from inflamed tissues. We evidenced a dual role of TNF-blockade. In peripheral blood, it increased the expression of cytokines such as IL-17, IL-10, and IFN-γ, and enhanced the expression of activation markers and the proliferative response of CD4 T cells to TCR stimulation. By contrast, in biopsies from target tissues, TNF-blockade diminished the expression of Th17/Th1 cytokine and early inflammatory genes. Importantly, the enhanced T cell responses to TCR-stimulation did not impair the clinical response to the therapy and, in responder patients, occurred with the concomitant down-regulation of inflammatory genes in the target tissues.


Subject(s)
Blood/immunology , Lymphocyte Activation/drug effects , Psoriasis/drug therapy , Receptors, Antigen, T-Cell/metabolism , Skin/immunology , T-Lymphocytes/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adalimumab , Adolescent , Adult , Aged , Aged, 80 and over , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antigens, CD/genetics , Antigens, CD/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Bacterial Toxins/immunology , Blood/drug effects , Blood/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cytokines/blood , Cytokines/genetics , Cytokines/metabolism , Enterotoxins/immunology , Etanercept , Female , Gene Expression/drug effects , Gene Expression/immunology , Humans , Immunoglobulin G/pharmacology , Immunoglobulin G/therapeutic use , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Infliximab , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/immunology , Male , Middle Aged , Psoriasis/immunology , Psoriasis/metabolism , Receptors, Tumor Necrosis Factor/therapeutic use , Skin/drug effects
10.
Blood ; 117(13): 3669-79, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21263153

ABSTRACT

Sox6 belongs to the Sry (sex-determining region Y)-related high-mobility-group-box family of transcription factors, which control cell-fate specification of many cell types. Here, we explored the role of Sox6 in human erythropoiesis by its overexpression both in the erythroleukemic K562 cell line and in primary erythroid cultures from human cord blood CD34+ cells. Sox6 induced significant erythroid differentiation in both models. K562 cells underwent hemoglobinization and, despite their leukemic origin, died within 9 days after transduction; primary erythroid cultures accelerated their kinetics of erythroid maturation and increased the number of cells that reached the final enucleation step. Searching for direct Sox6 targets, we found SOCS3 (suppressor of cytokine signaling-3), a known mediator of cytokine response. Sox6 was bound in vitro and in vivo to an evolutionarily conserved regulatory SOCS3 element, which induced transcriptional activation. SOCS3 overexpression in K562 cells and in primary erythroid cells recapitulated the growth inhibition induced by Sox6, which demonstrates that SOCS3 is a relevant Sox6 effector.


Subject(s)
Erythroid Precursor Cells/physiology , Erythropoiesis/genetics , SOXD Transcription Factors/physiology , Animals , Antigens, CD34/metabolism , Cell Differentiation/genetics , Cell Growth Processes/genetics , Cells, Cultured , Colony-Forming Units Assay , Erythroid Precursor Cells/metabolism , Erythropoiesis/physiology , Gene Expression Regulation/physiology , Humans , K562 Cells , Mice , Models, Biological , SOXD Transcription Factors/genetics , SOXD Transcription Factors/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Suppressor of Cytokine Signaling Proteins/physiology , Transfection
11.
Mol Cell Biol ; 26(10): 3942-54, 2006 May.
Article in English | MEDLINE | ID: mdl-16648487

ABSTRACT

We observed that binding sites for the ubiquitously expressed transcription factor CP2 were present in regulatory regions of multiple erythroid genes. In these regions, the CP2 binding site was adjacent to a site for the erythroid factor GATA-1. Using three such regulatory regions (from genes encoding the transcription factors GATA-1, EKLF, and p45 NF-E2), we demonstrated the functional importance of the adjacent CP2/GATA-1 sites. In particular, CP2 binds to the GATA-1 HS2 enhancer, generating a ternary complex with GATA-1 and DNA. Mutations in the CP2 consensus greatly impaired HS2 activity in transient transfection assays with K562 cells. Similar results were obtained by transfection of EKLF and p45 NF-E2 mutant constructs. Chromatin immunoprecipitation with K562 cells showed that CP2 binds in vivo to all three regulatory elements and that both GATA-1 and CP2 were present on the same GATA-1 and EKLF regulatory elements. Adjacent CP2/GATA-1 sites may represent a novel module for erythroid expression of a number of genes. Additionally, coimmunoprecipitation and glutathione S-transferase pull-down experiments demonstrated a physical interaction between GATA-1 and CP2. This may contribute to the functional cooperation between these factors and provide an explanation for the important role of ubiquitous CP2 in the regulation of erythroid genes.


Subject(s)
GATA1 Transcription Factor/metabolism , Gene Expression Regulation , Promoter Regions, Genetic , Repressor Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Motifs , Amino Acid Sequence , Base Sequence , Binding Sites , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Enhancer Elements, Genetic , GATA1 Transcription Factor/chemistry , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/isolation & purification , Genes, Regulator , Genes, Reporter , Glutathione Transferase/metabolism , Humans , K562 Cells , Kruppel-Like Transcription Factors/chemistry , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Luciferases/metabolism , Molecular Sequence Data , Mutation , NF-E2 Transcription Factor, p45 Subunit/chemistry , NF-E2 Transcription Factor, p45 Subunit/genetics , NF-E2 Transcription Factor, p45 Subunit/metabolism , Plasmids/genetics , Protein Binding , Recombinant Fusion Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/isolation & purification , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL