Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Blood Adv ; 7(5): 697-711, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36477543

ABSTRACT

Emerging gene therapy clinical trials test the correction of hemophilia A (HA) by replacing factor VIII (FVIII) in autologous hematopoietic stem cells (HSCs). Although it is known that platelets, monocyte/macrophages, and mesenchymal stromal cells can secrete transgenic FVIII, a systematic examination of blood lineages as extrahepatic sources of FVIII, to our knowledge, has not yet been performed. In this study, we sought to provide a comprehensive map of native and lentivirus-based transgenic FVIII production from HSC stage to mature blood cells, through a flow cytometry analysis. In addition, we generated a model of transient HA in zebrafish based on antisense RNA, to assess the corrective potential of the FVIII-transduced HSCs. We discovered that FVIII production begins at the CD34+ progenitor stage after cytokine stimulation in culture. Among all mature white blood cells, monocytes are the largest producers of native FVIII and can maintain protein overexpression during differentiation from HSCs when transduced by a FVIII lentiviral vector. Moreover, the addition of the HSC self-renewal agonist UM171 to CD34+ cells during transduction expanded a subpopulation of CD14+/CD31+ monocytes with excellent ability to carry the FVIII transgene, allowing the correction of HA phenotype in zebrafish. Finally, the HA zebrafish model showed that f8 RNA is predominantly localized in the hematopoietic system at the larval stage, which indicates a potential contributory role of FVIII in hematopoiesis that warrants further investigation. We believe that this study may be of broad interest to hematologists and researchers striving to advance knowledge and permanent treatments for patients with HA.


Subject(s)
Hemophilia A , Hemostatics , Animals , Factor VIII/genetics , Hematopoietic Stem Cells/metabolism , Hemophilia A/therapy , Monocytes/metabolism , Zebrafish/metabolism , Humans
2.
Sci Rep ; 11(1): 10364, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990655

ABSTRACT

Rosiglitazone is an effective insulin-sensitizer, however associated with bone loss mainly due to increased bone resorption and bone marrow adiposity. We investigated the effect of the co-administration of fish oil rich in omega-3 fatty acids (FAs) on rosiglitazone-induced bone loss in C57BL/6 mice and the mechanisms underlying potential preventive effect. Mice fed the iso-caloric diet supplemented with fish oil exhibited significantly higher levels of bone density in different regions compared to the other groups. In the same cohort of mice, reduced activity of COX-2, enhanced activity of alkaline phosphatase, lower levels of cathepsin k, PPAR-γ, and pro-inflammatory cytokines, and a higher level of anti-inflammatory cytokines were observed. Moreover, fish oil restored rosiglitazone-induced down-regulation of osteoblast differentiation and up-regulation of adipocyte differentiation in C3H10T1/2 cells and inhibited the up-regulation of osteoclast differentiation of RANKL-treated RAW264.7 cells. We finally tested our hypothesis on human Mesenchymal Stromal Cells differentiated to osteocytes and adipocytes confirming the beneficial effect of docosahexaenoic acid (DHA) omega-3 FA during treatment with rosiglitazone, through the down-regulation of adipogenic genes, such as adipsin and FABP4 along the PPARγ/FABP4 axis, and reducing the capability of osteocytes to switch toward adipogenesis. Fish oil may prevent rosiglitazone-induced bone loss by inhibiting inflammation, osteoclastogenesis, and adipogenesis and by enhancing osteogenesis in the bone microenvironment.


Subject(s)
Bone Diseases, Metabolic/prevention & control , Dietary Supplements , Fatty Acids, Omega-3/administration & dosage , Rosiglitazone/adverse effects , Adipogenesis/drug effects , Aging/physiology , Animals , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/physiopathology , Cell Differentiation/drug effects , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Female , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/physiology , Osteoclasts/drug effects , Osteoclasts/physiology , Osteogenesis/drug effects , Primary Cell Culture , RAW 264.7 Cells
3.
Front Immunol ; 11: 589381, 2020.
Article in English | MEDLINE | ID: mdl-33584653

ABSTRACT

The progress in the isolation and characterization of tumor antigen (TA)-specific T lymphocytes and in the genetic modification of immune cells allowed the clinical development of adoptive cell therapy (ACT). Several clinical studies highlighted the striking clinical activity of T cells engineered to express either Chimeric Antigen (CAR) or T Cell (TCR) Receptors to target molecularly defined antigens expressed on tumor cells. The breakthrough of immunotherapy is represented by the approval of CAR-T cells specific for advanced or refractory CD19+ B cell malignancies by both the Food and Drug Administration (FDA) and the European Medicinal Agency (EMA). Moreover, advances in the manufacturing and gene editing of engineered immune cells contributed to the selection of drug products with desired phenotype, refined specificity and decreased toxicity. An important step toward the optimization of CAR-T cell therapy is the development of "off-the shelf" T cell products that allow to reduce the complexity and the costs of the manufacturing and to render these drugs available for a broad number of cancer patients. The Engineered Immune Cells in Cancer Immunotherapy (EICCI) workshop hosted in Doha, Qatar, renowned experts, from both academia and industry, to present and discuss the progress on both pre-clinical and clinical development of genetically modified immune cells, including advances in the "off-the-shelf" manufacturing. These experts have addressed also organizational needs and hurdles for the clinical grade production and application of these biological drugs.


Subject(s)
Immunotherapy , Neoplasms/therapy , Animals , Genetic Engineering , Humans , Immunotherapy, Adoptive , Qatar , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology
4.
Mol Ther Methods Clin Dev ; 17: 1-12, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-31886317

ABSTRACT

Detection of factor VIII (FVIII) in cells by flow cytometry is controversial, and no monoclonal fluorescent antibody is commercially available. In this study, we optimized such an assay and successfully used it as a platform to study the functional properties of phosphoglycerate kinase (PGK)-FVIII lentiviral vector-transduced cells by directly visualizing FVIII in cells after different gene transfer conditions. We could measure cellular stress parameters after transduction by correlating gene expression and protein accumulation data. Flow cytometry performed on transduced cell lines showed that increasing MOI rates resulted in increased protein levels, plateauing after an MOI of 30. We speculated that, at higher MOI, FVIII production could be impaired by a limiting factor required for proper folding. To test this hypothesis, we interfered with the unfolded protein response by blocking proteasomal degradation and measured the accumulation of intracellular misfolded protein. Interestingly, at higher MOIs the cells displayed signs of toxicity with reactive oxygen species accumulation. This suggests the need for identifying a safe window of transduction dose to avoid consequent cell toxicity. Herein, we show that our flow cytometry platform for intracytoplasmic FVIII protein detection is a reliable method for optimizing gene therapy protocols in hemophilia A by shedding light on the functional status of cells after gene transfer.

5.
Probiotics Antimicrob Proteins ; 11(2): 478-492, 2019 06.
Article in English | MEDLINE | ID: mdl-29512027

ABSTRACT

Safety and efficacy was investigated for two candidate probiotic B. flexus MCC2427 and B. licheniformis MCC2512 via in vivo studies on albino Wistar rats. In acute toxicity assay, rats were fed with single dose of 1010 cfu mL-1 of probiotics. The follow-up studies for next 14 days did not reveal any toxicity-related criteria indicating the non-toxicity nature of probiotics. In 90-day repeated dosage studies, the cultures were administered in three doses (106, 107, 108 cfu mL-1). Results showed no overt toxic effect and no drastic treatment-related changes pertaining to histopathology of vital organs. DNA fingerprinting indicated the lack of bacterial translocation. Superoxide dismutase and catalase activity indicated their antioxidant potential. Reduced serum cholesterol with improved HDL-cholesterol specified the cholesterol-reducing ability of the cultures, which was also apparent with increased excretion of cholic acid in feces. Both probiotic cultures positively altered the gut microbial environment, retained lactic acid bacterial effect, and simultaneously reduced pathogenic strains. A sensitive and rapid tool was developed using strain-specific qPCR primers, which facilitated appropriate estimation of test culture in feces. The data strongly advocate the safety of tested probiotics at levels used in the study.


Subject(s)
Bacillus , Gastrointestinal Microbiome/drug effects , Liver/drug effects , Probiotics/pharmacology , Animals , Bacterial Load , Feces/chemistry , Feces/microbiology , Lipids/blood , Liver/physiology , Male , Rats , Rats, Wistar , Species Specificity , Superoxide Dismutase/metabolism
6.
J Biosci ; 42(4): 603-611, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29229878

ABSTRACT

In continuation of our studies on the bioaccessibility of phenolic compounds from food grains as influenced by domestic processing, we examined the uptake of phenolics from native/sprouted finger millet (Eleucine coracana) and green gram (Vigna radiata) and native/heat-processed onion (Allium cepa) in human Caco-2 cells. Absorption of pure phenolic compounds, as well as the uptake of phenolic compounds from finger millet, green gram, and onion, was investigated in Caco-2 monolayer model. Transport of individual phenolic compounds from apical compartment to the basolateral compartment across Caco-2 monolayer was also investigated. Sprouting enhanced the uptake of syringic acid from both these grains. Open-pan boiling reduced the uptake of quercetin from the onion. Among pure phenolic compounds, syringic acid was maximally absorbed, while the flavonoid isovitexin was least absorbed. Apparent permeability coefficient P(app) of phenolic compounds from their standard solutions was 2.02 x 10-6cm/s to 8.94 x 10-6cm/s. Sprouting of grains enhanced the uptake of syringic acid by the Caco-2 cells. Open-pan boiling drastically reduced the uptake of quercetin from the onion. The permeability of phenolic acids across Caco-2 monolayer was higher than those of flavonoids.


Subject(s)
Apigenin/metabolism , Flavonoids/metabolism , Gallic Acid/analogs & derivatives , Phenols/metabolism , Quercetin/metabolism , Apigenin/isolation & purification , Biological Transport , Caco-2 Cells , Eleusine/chemistry , Flavonoids/isolation & purification , Gallic Acid/isolation & purification , Gallic Acid/metabolism , Humans , Kinetics , Onions/chemistry , Phenols/isolation & purification , Quercetin/isolation & purification , Vigna/chemistry
7.
J Transl Med ; 13: 119, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25890290

ABSTRACT

OBJECTIVES: In Qataris, a population characterized by a small size and a high rate of consanguinity, between two-thirds to three-quarters of adults are overweight or obese. We investigated the relevance of 23 obesity-related loci in the Qatari population. METHODS: Eight-hundred-four individuals assessed to be third generation Qataris were included in the study and assigned to 3 groups according to their body mass index (BMI): 190 lean (BMI < 25 kg/m(2)); 131 overweight (25 kg/m(2) ≤ BMI < 30 kg/m(2)) and 483 obese (BMI ≥ 30 kg/m(2)). Genomic DNA was isolated from peripheral blood and genotyped by TaqMan. RESULTS: Two loci significantly associated with obesity in Qataris: the TFAP2B variation (rs987237) (A allele versus G allele: chi-square = 10.3; P = 0.0013) and GNPDA2 variation (rs10938397) (A allele versus G allele: chi-square = 6.15; P = 0.013). The TFAP2B GG genotype negatively associated with obesity (OR = 0.21; P = 0.0031). Conversely, the GNDPA2 GG homozygous genotype associated with higher risk of obesity in subjects of age < 32 years (P = 0.0358). CONCLUSION: We showed a different genetic profile associated with obesity in the Qatari population compared to Western populations. Studying the genetic background of Qataris is of primary importance as the etiology of a given disease might be population-specific.


Subject(s)
Arabs/genetics , Consanguinity , Genetic Loci , Genetic Predisposition to Disease , Obesity/genetics , Adult , Body Mass Index , Female , Humans , Logistic Models , Male , Middle Aged , Phenotype , Polymorphism, Single Nucleotide/genetics , Principal Component Analysis , Qatar , Racial Groups/genetics , Thinness/genetics
8.
J Transl Med ; 11: 121, 2013 May 13.
Article in English | MEDLINE | ID: mdl-23668334

ABSTRACT

BACKGROUND: Large databases focused on genetic susceptibility to prostate cancer have been accumulated from population studies of different ancestries, including Europeans and African-Americans. Arab populations, however, have been only rarely studied. METHODS: Using Affymetrix Genome-Wide Human SNP Array 6, we conducted a genome-wide association study (GWAS) in which 534,781 single nucleotide polymorphisms (SNPs) were genotyped in 221 Tunisians (90 prostate cancer patients and 131 age-matched healthy controls). TaqMan SNP Genotyping Assays on 11 prostate cancer associated SNPs were performed in a distinct cohort of 337 individuals from Arab ancestry living in Qatar and Saudi Arabia (155 prostate cancer patients and 182 age-matched controls). In-silico expression quantitative trait locus (eQTL) analysis along with mRNA quantification of nearby genes was performed to identify loci potentially cis-regulated by the identified SNPs. RESULTS: Three chromosomal regions, encompassing 14 SNPs, are significantly associated with prostate cancer risk in the Tunisian population (P = 1 × 10-4 to P = 1 × 10-5). In addition to SNPs located on chromosome 17q21, previously found associated with prostate cancer in Western populations, two novel chromosomal regions are revealed on chromosome 9p24 and 22q13. eQTL analysis and mRNA quantification indicate that the prostate cancer associated SNPs of chromosome 17 could enhance the expression of STAT5B gene. CONCLUSION: Our findings, identifying novel GWAS prostate cancer susceptibility loci, indicate that prostate cancer genetic risk factors could be ethnic specific.


Subject(s)
Arabs/genetics , Genome-Wide Association Study , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics , Aged , Aged, 80 and over , Alleles , Case-Control Studies , Chromosome Mapping/methods , Cohort Studies , Genetic Predisposition to Disease , Genome , Genotype , Humans , Male , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/ethnology , Qatar , Quantitative Trait Loci , RNA, Messenger/metabolism , Reproducibility of Results , Risk Factors , Saudi Arabia , Tunisia
9.
Cancer Res ; 73(9): 2840-9, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23447579

ABSTRACT

Although the linkage between germline mutations of BRCA1 and hereditary breast/ovarian cancers is well established, recent evidence suggests that altered expression of wild-type BRCA1 might contribute to the sporadic forms of breast cancer. The breast cancer gene trinucleotide-repeat-containing 9 (TNRC9; TOX3) has been associated with disease susceptibility but its function is undetermined. Here, we report that TNRC9 is often amplified and overexpressed in breast cancer, particularly in advanced breast cancer. Gene amplification was associated with reduced disease-free and metastasis-free survival rates. Ectopic expression of TNRC9 increased breast cancer cell proliferation, migration, and survival after exposure to apoptotic stimuli. These phenotypes were associated with tumor progression in a mouse model of breast cancer. Gene expression profiling, protein analysis, and in silico assays of large datasets of breast and ovarian cancer samples suggested that TNRC9 and BRCA1 expression were inversely correlated. Notably, we found that TNRC9 bound to both the BRCA1 promoter and the cAMP-responsive element-binding protein (CREB) complex, a regulator of BRCA1 transcription. In support of this connection, expression of TNRC9 downregulated expression of BRCA1 by altering the methylation status of its promoter. Our studies unveil a function for TNRC9 in breast cancer that highlights a new paradigm in BRCA1 regulation.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Genes, BRCA1 , Receptors, Progesterone/metabolism , Adult , Animals , Apoptosis Regulatory Proteins , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Cyclic AMP Response Element-Binding Protein/metabolism , DNA Methylation , Disease Progression , Disease-Free Survival , Female , Gene Expression Profiling , HEK293 Cells , High Mobility Group Proteins , Humans , Mice , Middle Aged , Models, Genetic , Neoplasm Invasiveness , Neoplasm Metastasis , Phenotype , Trans-Activators
SELECTION OF CITATIONS
SEARCH DETAIL
...