Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Acta Haematol ; 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38471491

ABSTRACT

Introduction Pre exposure prophylaxis with monoclonal antibodies (mAbs) were developed in addition to COVID19 vaccine for immunocompromised and those with insufficient immune response, among them patients with CLL. Omicron variant and its sublineages evolved mutations that escape mAbs neutralizing effect, yet the extent of which was not studied. Methods We evaluated anti-spike titters and neutralization activity of COVID-19 wild type (WT) , Delta , Omicron, BA2, BA4 and BA5 before and after tixagevimab-cilgavimab (TGM/CGM) dose of 150/150mg or 300/300mg in patients with CLL. Results 70 patients were tested 2 weeks before and 4 weeks after receiving TGM/CGM mAbs. After TGM/CGM anti-spike ab level increased 170 folds from 13.6 BAU/ml (IQR, 0.4-288) to 2328 BAU/ml (IQR, 1681-3500). Neutralization activity increased in all variants, and was 176 folds higher in WT and 55 folds higher in Delta compared to 10 folds higher in Omicron and its sublineages (BA2 x11, BA4 x4 , BA5 x18). Over follow-up period of 3 months, 20 patients (29%) with CLL acquired COVID-19 infection, all recovered uneventfully. In a multivariate analysis anti-spike antibody titer was found a significant predictor for post TGM/CGM COVID19 infection. Conclusion Efficacy of preexposure prophylaxis with TGM/CGM in patients with CLL is significantly reduced in era of Omicron and its sublineages BA2, BA4 and BA5.

2.
J Orthop Surg Res ; 14(1): 198, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31262323

ABSTRACT

BACKGROUND: Adipose tissue-derived mesenchymal stem cells (AT-MSCs) are one of the most potent adult stem cells, capable of differentiating into bone, cartilage, adipose, muscle, and others. An innovative autologous AT-MSC-derived cell-based product (BonoFill-II) for bone tissue regeneration was developed to be suited as a bone graft for segmental bone defects. METHODS: BonoFill-II was transplanted into 8 sheep with 3.2-cm full cortex segmental defect formed in the tibia. Bone regeneration was followed by X-ray radiographs for 12 weeks. At experiment termination, the healed tibia bones were analyzed by computed tomography, histology, and mechanical tests. RESULTS: Our results indicate that one dose of BonoFill-II injectable formula led to an extensive bone growth within the transplantation site and to a complete closure of the critical gap in the sheep's tibia in a relatively short time (8-12 weeks), with no inflammation and no other signs of graft rejection. This new and innovative product opens new prospects for the treatment of long bone defects. CONCLUSIONS: Injection of BonoFill-II (an innovative autologous cell therapy product for bone tissue regeneration) into a critical size segmental defect model (3.2 cm), generated in the sheep tibia, achieved full bridging of the gap in an extremely short period (8-12 weeks).


Subject(s)
Bone Regeneration/physiology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Tibia/diagnostic imaging , Tibia/physiology , Transplantation, Autologous/methods , Animals , Cell- and Tissue-Based Therapy/methods , Female , Humans , Random Allocation , Sheep , Tibia/injuries
3.
Cell Metab ; 21(3): 392-402, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25738455

ABSTRACT

Loss of pluripotency is a gradual event whose initiating factors are largely unknown. Here we report the earliest metabolic changes induced during the first hours of differentiation. High-resolution NMR identified 44 metabolites and a distinct metabolic transition occurring during early differentiation. Metabolic and transcriptional analyses showed that pluripotent cells produced acetyl-CoA through glycolysis and rapidly lost this function during differentiation. Importantly, modulation of glycolysis blocked histone deacetylation and differentiation in human and mouse embryonic stem cells. Acetate, a precursor of acetyl-CoA, delayed differentiation and blocked early histone deacetylation in a dose-dependent manner. Inhibitors upstream of acetyl-CoA caused differentiation of pluripotent cells, while those downstream delayed differentiation. Our results show a metabolic switch causing a loss of histone acetylation and pluripotent state during the first hours of differentiation. Our data highlight the important role metabolism plays in pluripotency and suggest that a glycolytic switch controlling histone acetylation can release stem cells from pluripotency.


Subject(s)
Acetyl Coenzyme A/metabolism , Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/physiology , Glycolysis/physiology , Histones/metabolism , Acetyl Coenzyme A/genetics , Acetylation , Animals , Cell Differentiation/genetics , Cell Line , Glycolysis/genetics , Histones/genetics , Humans , Mice , Transcription, Genetic/genetics , Transcription, Genetic/physiology
4.
Hum Mol Genet ; 23(14): 3629-40, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24549038

ABSTRACT

Human telomeric regions are packaged as constitutive heterochromatin, characterized by extensive subtelomeric DNA methylation and specific histone modifications. ICF (immunodeficiency, centromeric instability, facial anomalies) type I patients carry mutations in DNA methyltransferase 3B (DNMT3B) that methylates de novo repetitive sequences during early embryonic development. ICF type I patient fibroblasts display hypomethylated subtelomeres, abnormally short telomeres and premature senescence. In order to study the molecular mechanism by which the failure to de novo methylate subtelomeres results in accelerated telomere shortening, we generated induced pluripotent stem cells (iPSCs) from 3 ICF type I patients. Telomeres were elongated in ICF-iPSCs during reprogramming, and the senescence phenotype was abolished despite sustained subtelomeric hypomethylation and high TERRA levels. Fibroblast-like cells (FLs) isolated from differentiated ICF-iPSCs maintained abnormally high TERRA levels, and telomeres in these cells shortened at an accelerated rate, leading to early senescence, thus recapitulating the telomeric phenotype of the parental fibroblasts. These findings demonstrate that the abnormal telomere phenotype associated with subtelomeric hypomethylation is overridden in cells expressing telomerase, therefore excluding telomerase inhibition by TERRA as a central mechanism responsible for telomere shortening in ICF syndrome. The data in the current study lend support to the use of ICF-iPSCs for modeling of phenotypic and molecular defects in ICF syndrome and for unraveling the mechanism whereby subtelomeric hypomethylation is linked to accelerated telomeric loss in this syndrome.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Face/abnormalities , Immunologic Deficiency Syndromes/genetics , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Telomere Shortening , Telomere/metabolism , Cells, Cultured , Cellular Reprogramming , Cellular Senescence , Chromosome Aberrations , Chromosomes, Human , DNA Methylation , DNA-Binding Proteins/metabolism , Face/physiopathology , Female , Fibroblasts/physiology , Humans , Immunologic Deficiency Syndromes/physiopathology , Induced Pluripotent Stem Cells/cytology , Male , Primary Immunodeficiency Diseases , DNA Methyltransferase 3B
5.
Stem Cells Dev ; 23(5): 443-56, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24192045

ABSTRACT

Adenosine-to-inosine (A-to-I) RNA editing is a post-transcriptional, site-specific modification process that is catalyzed by Adenosine Deaminase Acting on RNA (ADAR) gene family members. Since ADARs act on double-stranded RNA, most A-to-I editing occurs within repetitive elements, particularly Alu elements, as the result of the inherent property of these sequences to fold and form double strands. ADAR1-mediated A-to-I RNA editing was recently implicated in the regulation of human embryonic stem cells (hESCs). Spontaneous and neuronal differentiation of hESC was shown to result in a decrease in A-to-I editing levels. Knockdown of ADAR1 in hESCs results in an elevation of the expression of differentiation-related genes. In addition, we found that hESCs over-expressing ADAR1 could not be generated. The current study shows that the editing levels of induced pluripotent stem cells (iPSCs) change throughout reprogramming, from a source cell level to a level similar to that of hESCs. Up- or down-regulation of the ADAR1 level in human foreskin fibroblast (HFF) cells before induction of reprogramming results in varied reprogramming efficiencies. Furthermore, HFF-iPSC early clones derived from source cells in which the ADAR1 level was down-regulated lose their iPSC properties shortly after iPSC colony formation and instead exhibit characteristics of cancer cells. Taken together, our results imply a role for ADAR1 in the regulation of pluripotency induction as well as in the maintenance of early iPSC properties.


Subject(s)
Adenosine Deaminase/biosynthesis , Cell Differentiation/genetics , Embryonic Stem Cells , Induced Pluripotent Stem Cells , Adenosine Deaminase/genetics , Fibroblasts , Gene Expression Regulation , Gene Knockdown Techniques , Humans , RNA-Binding Proteins
6.
Eur J Hum Genet ; 20(12): 1248-55, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22713809

ABSTRACT

Pluripotency and proliferative capacity of human embryonic stem cells (hESCs) make them a promising source for basic and applied research as well as in therapeutic medicine. The introduction of human induced pluripotent cells (hiPSCs) holds great promise for patient-tailored regenerative medicine therapies. However, for hESCs and hiPSCs to be applied for therapeutic purposes, long-term genomic stability in culture must be maintained. Until recently, G-banding analysis was considered as the default approach for detecting chromosomal abnormalities in stem cells. Our goal in this study was to apply fluorescence in-situ hybridization (FISH) and comparative genomic hybridization (CGH) for the screening of pluripotent stem cells, which will enable us identifying chromosomal abnormalities in stem cells genome with a better resolution. We studied three hESC lines and two hiPSC lines over long-term culture. Aneuploidy rates were evaluated at different passages, using FISH probes (12,13,16,17,18,21,X,Y). Genomic integrity was shown to be maintained at early passages of hESCs and hiPSCs but, at late passages, we observed low rates mosaiciam in hESCs, which implies a direct correlation between number of passages and increased aneuploidy rate. In addition, CGH analysis revealed a recurrent genomic instability, involving the gain of chromosome 1q. This finding was detected in two unrelated cell lines of different origin and implies that gains of chromosome 1q may endow a clonal advantage in culture. These findings, which could only partially be detected by conventional cytogenetic methods, emphasize the importance of using molecular cytogenetic methods for tracking genomic instability in stem cells.


Subject(s)
Aneuploidy , Chromosome Duplication/genetics , Chromosomes, Human, Pair 1/genetics , Mosaicism , Pluripotent Stem Cells , Cell Line , Comparative Genomic Hybridization , Genomic Instability/genetics , Humans , In Situ Hybridization, Fluorescence
7.
Tissue Eng Part A ; 18(21-22): 2290-302, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22731654

ABSTRACT

Human mesenchymal stem cells (hMSCs) can be derived from various adult and fetal tissues. However, the quality of tissues for the isolation of adult and fetal hMSCs is donor dependent with a nonreproducible yield. In addition, tissue engineering and cell therapy require large-scale production of a pure population of lineage-restricted stem cells that can be easily induced to differentiate into a specific cell type. Therefore, human embryonic stem cells (hESCs) can provide an alternative, plentiful source for generation of reproducible hMSCs. We have developed efficient differentiation protocols for derivation of hMSCs from hESCs, including coculture with murine OP9 stromal cells and feeder layer-free system. Our protocols have resulted in the generation of up to 49% of hMSCs, which expressed CD105, CD90, CD29, and CD44. The hMSCs exhibited high adipogenic, chondrocytic, and osteogenic differentiation in vitro. The latter correlated with osteocalcin secretion and vascular endothelial growth factor (VEGF) production by the differentiating hMSCs. hMSC-derived osteoblasts further differentiated and formed ectopic bone in vivo, and induced the formation of blood vessels in Matrigel implants. Our protocol enables generation of a purified population of hESC-derived MSCs, with the potential of differentiating into several mesodermal lineages, and particularly into vasculogenesis-inducing osteoblasts, which can contribute to the development of bone repair protocols.


Subject(s)
Bone and Bones/blood supply , Choristoma/pathology , Embryonic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic , Tissue Engineering/methods , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Bone and Bones/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Separation , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Coculture Techniques , Collagen/pharmacology , Drug Combinations , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Humans , Kinetics , Laminin/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, SCID , Neovascularization, Physiologic/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteocalcin/metabolism , Osteogenesis/drug effects , Proteoglycans/pharmacology , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism
8.
Nat Protoc ; 6(5): 572-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21527915

ABSTRACT

Human pluripotent (embryonic or induced) stem cells (hPSCs) have many potential applications, not only for research purposes but also for clinical and industrial uses. While culturing these cells as undifferentiated lines, an adherent cell culture based on supportive layers or matrices is most often used. However, the use of hPSCs for industrial or clinical applications requires a scalable, reproducible and controlled process. Here we present a suspension culture system for undifferentiated hPSCs, based on a serum-free medium supplemented with interleukins and basic fibroblast growth factor, suitable for the mass production of these cells. The described system supports a suspension culture of hPSC lines, in both static and dynamic cultures. Results showed that hPSCs cultured with the described dynamic method maintained all hPSC features after 20 passages, including stable karyotype and pluripotency, and increased in cell numbers by 25-fold in 10 d. Thus, the described suspension method is suitable for large-scale culture of undifferentiated hPSCs.


Subject(s)
Cell Culture Techniques/methods , Pluripotent Stem Cells/cytology , Humans , Teratoma/pathology
9.
Stem Cell Rev Rep ; 6(2): 248-59, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20431964

ABSTRACT

Alongside their contribution to research, human embryonic stem cells (hESC) may also prove valuable for cell-based therapies. Traditionally, these cells have been grown in adhesion culture either with or without feeder cells, allowing for their continuous growth as undifferentiated cells. However, to be applicable in therapy and industry they must be produced in a scalable and controlled process. Here we present for the first time a suspension culture system for undifferentiated hESC and induced pluripotent stem cells (iPSC), based on medium supplemented with the IL6RIL6 chimera (interleukin-6 receptor fused to interleukin-6), and basic fibroblast growth factor. Four hESC lines cultured in this system maintained all ESC features after 20 passages, including stable karyotype and pluripotency. Similar results were obtained when hESC were replaced with iPSC from two different cell lines. We demonstrate that the IL6RIL6 chimera supports the self-renewal and expansion of undifferentiated hESC and iPSC in suspension, and thus present another efficient system for large-scale propagation of undifferentiated pluripotent cells for clinical and translational applications.


Subject(s)
Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Apoptosis/drug effects , Blotting, Western , Cell Differentiation/physiology , Flow Cytometry , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Karyotyping , Polymerase Chain Reaction , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology
10.
Stem Cells Dev ; 17(6): 1227-41, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19006458

ABSTRACT

The promise of human embryonic stem cells (hESCs) to provide an unlimited supply of cells for cell therapy depends on the availability of a controllable bioprocess for their expansion and differentiation. We describe here a robust and well-defined scale up platform for human embryoid body (EB) formation, propagation, and differentiation. The efficacy of the dynamic process as compared to the static cultivation in Petri dishes was analyzed. Our optimized conditions include specific bioreactor and impeller type, seeding and propagation parameters, and scale up. Quantitative analyses of viable cell concentrations, apoptosis percentages, and EB yield revealed 6.7-fold enhancement in the generation of hESC-derived cells after 10 cultivation days. Other metabolic indices such as glucose consumption, lactic acid production and pH all pointed to efficient cell expansion in the dynamic cultures. The hydrodynamic conditions during seeding and cultivation were found to be crucial for the EB formation and propagation. The EBs' prearrangement in the static system and EB cultivation in the Glass Ball Impeller spinner flask resulted in high EB yield, a round homogenous shape, and the fastest growth rate. The appearance of representative genes of the three germ layers as well as primitive neuronal tube organization and blood vessel formation indicated that the initial developmental events in the human EBs are not interfered by the dynamic system. Furthermore, well developed endothelial networks and contracting EBs with functional cardiac muscle were also obtained after two cultivation weeks. Collectively, our study defines the technological platform for the controlled large-scale generation of hESC-derived cells for clinical and industrial applications.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Embryonic Stem Cells/cytology , Animals , Apoptosis , Cell Culture Techniques/instrumentation , Cell Line , Embryonic Stem Cells/metabolism , Humans , Mice , Time Factors
11.
Prenat Diagn ; 23(10): 788-90, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14558020

ABSTRACT

Ethmocephaly is a rare anomaly associated with partial failure of cleavage of the prosencephalon. Morphologically, it is closely related to cyclopia. We present an extremely rare case of ethmocephaly diagnosed in utero and caused by an unbalanced de novo translocation 18;21.


Subject(s)
Chromosomes, Human, Pair 18 , Chromosomes, Human, Pair 21 , Fetal Diseases/diagnostic imaging , Holoprosencephaly/diagnostic imaging , Translocation, Genetic , Ultrasonography, Prenatal , Diagnosis, Differential , Female , Fetal Diseases/embryology , Fetal Diseases/genetics , Holoprosencephaly/embryology , Holoprosencephaly/genetics , Humans , Pregnancy , Pregnancy Trimester, Third
SELECTION OF CITATIONS
SEARCH DETAIL
...