Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Cyst Fibros ; 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37980178

ABSTRACT

BACKGROUND: Population genetic carrier screening (PGCS) for cystic fibrosis (CF) has been offered to couples in Israel since 1999 and was included in a fully subsidized national program in 2008. We evaluated the impact of PGCS on CF incidence, genetic and clinical features. METHODS: This was a retrospective national study. Demographic and clinical characteristics of children with CF born in Israel between 2008 and 2018 were obtained from the national CF registry and from patients' medical records. Data on CF births, preimplantation genetic testing (PGT), pregnancy termination and de-identified data from the PGCS program were collected. RESULTS: CF births per 100,000 live births decreased from 8.29 in 2008 to 0.54 in 2018 (IRR = 0.84, p < 0.001). The CF pregnancy termination rate did not change (IRR = 1, p=  0.9) while the CF-related PGT rate increased markedly (IRR = 1.33, p < 0.001). One hundred and two children were born with CF between 2008 and 2018 with a median age at diagnosis of 4.8 months, range 0-111 months. Unlike the generally high uptake nationally, 65/102 had not performed PGCS. Even if all had utilized PGCS, only 51 would have been detected by the existing genetic screening panel. Clinically, 34 % of children were pancreatic sufficient compared to 23 % before 2008 (p = 0.04). CONCLUSIONS: Since institution of a nationwide PGCS program, the birth of children with CF decreased markedly. Residual function variants and pancreatic sufficiency were more common. A broader genetic screening panel and increased PGCS utilization may further decrease the birth of children with CF.

2.
Dig Liver Dis ; 55(7): 880-887, 2023 07.
Article in English | MEDLINE | ID: mdl-36740502

ABSTRACT

BACKGROUND: Autosomal recessive conditions are common in consanguineous populations. Since consanguinity is common in the Israeli Arab population, we evaluated the rate of MUTYH polyposis (MAP) among polyposis patients in this population and studied Pathogenic Variants (PVs) spectrum. METHODS: We reviewed health records of all Arab and Druze polyposis patients referred for counseling during 2013-2020 who fulfilled the Israeli Genetic Society criteria for MUTYH/APC testing, in a tertiary center in Northern Israel and four additional gastro-genetic clinics in Israel. RESULTS: The Northern cohort included 37 patients from 30 unrelated families; 8(26.6%) carried bi-allelic MUTYH PVs. The major variant p.Glu452del was detected in 6/8 Druze and Muslim families who shared the same haplotype. Other PVs detected in both cohorts included p.Tyr56Ter, p.His57Arg, c.849+3A>C, p.Ala357fs, and p.Tyr151Cys. Among bi-allelic carriers, 88% reported consanguinity, and 100% had positive family history for polyposis or colorectal cancer (CRC). Generally, the age of CRC was 10 years younger than reported in the general MAP population. CONCLUSIONS: MAP accounted for 27% of polyposis cases in the Arab population of Northern Israel. PVs spectrum is unique, with high frequency of the founder variant p.Glu452del. Our results may inform the genetic testing strategy in the Israeli Arab population.


Subject(s)
Colorectal Neoplasms , Genetic Predisposition to Disease , Humans , Child , Israel/epidemiology , Prevalence , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Health Disparate Minority and Vulnerable Populations , Mutation
3.
Stem Cell Rev Rep ; 8(3): 792-802, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22592939

ABSTRACT

New sources of beta cells are needed in order to develop cell therapies for patients with diabetes. An alternative to forced expansion of post-mitotic beta cells is the induction of differentiation of stem-cell derived progenitor cells that have a natural self-expansion capacity into insulin-producing cells. In order to learn more about these progenitor cells at different stages along the differentiation process in which they become progressively more committed to the final beta cell fate, we took the approach of identifying, isolating and characterizing stage specific progenitor cells. We generated human embryonic stem cell (HESC) clones harboring BAC GFP reporter constructs of SOX17, a definitive endoderm marker, and PDX1, a pancreatic marker, and identified subpopulations of GFP expressing cells. Using this approach, we isolated a highly enriched population of pancreatic progenitor cells from hESCs and examined their gene expression with an emphasis on the expression of stage-specific cell surface markers. We were able to identify novel molecules that are involved in the pancreatic differentiation process, as well as stage-specific cell markers that may serve to define (alone or in combination with other markers) a specific pancreatic progenitor cell. These findings may help in optimizing conditions for ultimately generating and isolating beta cells for transplantation therapy.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/physiology , Membrane Proteins/metabolism , Pancreas/cytology , Arabidopsis Proteins/biosynthesis , Arabidopsis Proteins/genetics , Biomarkers/metabolism , Carbon-Nitrogen Lyases , Cell Separation , Cells, Cultured , Chromosomes, Artificial, Bacterial/genetics , Diabetes Mellitus, Type 1/therapy , Embryonic Stem Cells/metabolism , Flow Cytometry , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Insulin-Secreting Cells/transplantation , Nitrogenous Group Transferases/biosynthesis , Nitrogenous Group Transferases/genetics , Oligonucleotide Array Sequence Analysis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , SOXF Transcription Factors/biosynthesis , SOXF Transcription Factors/genetics , Transcriptome , Transgenes
4.
Stem Cell Rev Rep ; 8(1): 137-49, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21732092

ABSTRACT

The regulatory pathways responsible for maintaining human embryonic stem cells (hESCs) in an undifferentiated state have yet to be elucidated. Since these pathways are thought to be governed by complex protein cues, deciphering the changes that occur in the proteomes of the ESCs during differentiation is important for understanding the expansion and differentiation processes involved. In this study, we present the first quantitative comparison of the hESC protein profile in the undifferentiated and early differentiated states. We used iTRAQ (isobaric tags for relative and absolute quantification) labeling combined with two dimensional capillary chromatography coupled with tandem mass spectrometry (µLC-MS/MS) to achieve comparative proteomics of hESCs at the undifferentiated stage, and at 6, 48, and 72 h after initiation of differentiation. In addition, two dimensional electrophoresis (2-DE) was performed on differentiating hESCs at eleven points of time during the first 72 h of differentiation. The results indicate that during the first 48 h of hESC differentiation, many processes are initiated and are later reversed, including chromatin remodeling, heterochromatin spreading, a decrease in transcription and translation, a decrease in glycolytic proteins and cytoskeleton remodeling, and a decrease in focal and cell adhesion. Only 72 h after differentiation induction did the expression of the homeobox prox1 protein increase, indicating the beginning of developmental processes.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/physiology , Proteome/metabolism , Cell Shape , Cells, Cultured , Electrophoresis, Gel, Two-Dimensional , Embryonic Stem Cells/metabolism , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Humans , Karyotype , Proteome/genetics , Proteome/isolation & purification , Proteomics , Tandem Mass Spectrometry
5.
Cell Reprogram ; 12(6): 665-78, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20964482

ABSTRACT

Induced pluripotent stem cells (iPSCs) represent an ideal cell source for future cell therapy and regenerative medicine. However, most iPSC lines described to date have been isolated from skin fibroblasts or other cell types that require harvesting by surgical intervention. Because it is desirable to avoid such intervention, an alternative cell source that can be readily and noninvasively isolated from patients and efficiently reprogrammed, is required. Here we describe a detailed and reproducible method to derive iPSCs from plucked human hair follicle keratinocytes (HFKTs). HFKTs were isolated from single plucked hair, then expanded and reprogrammed by a single polycistronic excisable lentiviral vector. The reprogrammed HFKTs were found to be very sensitive to human embryonic stem cell (hESC) growth conditions, generating a built-in selection with easily obtainable and very stable iPSCs. All emerging colonies were true iPSCs, with characteristics typical of human embryonic stem cells, differentiated into derivatives of all three germ layers in vitro and in vivo. Spontenaeouly differentiating functional cardiomyocytes (CMs) were successfully derived and characterized from these HFKT-iPSCs. The contracting CMs exhibited well-coordinated intracellular Ca²+ transients and contractions that were readily responsive to ß-adrenergic stimulation with isoproterenol. The introduction of Cre-recombinase to HFKT-iPSC clones was able to successfully excise the integrated vector and generate transgene-free HFKT-iPSC clone that could be better differentiated into contracting CMs, thereby revealing the desired cells for modeling human diseases. Thus, HFKTs are easily obtainable, and highly reprogrammed human cell source for all iPSC applications.


Subject(s)
Cell Differentiation/physiology , Hair Follicle/chemistry , Heart/embryology , Keratinocytes/physiology , Lentivirus/metabolism , Myocardium/cytology , Cell Culture Techniques/methods , Cells, Cultured , Humans , Keratinocytes/cytology , Lentivirus/genetics , Patch-Clamp Techniques , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology
6.
Dev Growth Differ ; 47(5): 295-306, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16026538

ABSTRACT

During early embryogenesis, the cardiovascular system is the first system to be established and is initiated by a process involving the hypoblastic cells of the primitive endoderm. Human embryonic stem (hES) cells provide a model to investigate the early developmental stages of this system. When removed from their feeder layer, hESC create embryoid bodies (EB) which, when plated, develop areas of beating cells in 21.5% of the EB. These spontaneously contracting cells were demonstrated using histology, immunostaining and reverse transcription-polymerase chain reaction (RT-PCR), to possess morphological and molecular characteristics consistent with cardiomyocytic phenotypes. In addition, the expression pattern of specific cardiomyocytic genes in human EB (hEB) was demonstrated and analyzed for the first time. GATA-4 is the first gene to be expressed in 6-day-old EB. Alpha cardiac actin and atrial natriuretic factor are expressed in older hEB at 10 and 20 days, respectively. Light chain ventricular myosin (MLC-2V) was expressed only in EB with beating areas and its expression increased with time. Alpha heavy chain myosin (alpha-MHC) expression declined in the pulsating hEB with time, in contrast to events in EB derived from mice. We conclude that human embryonic stem cells can provide a useful tool for research on embryogenesis in general and cardiovascular development in particular.


Subject(s)
Myocytes, Cardiac/metabolism , Stem Cells/cytology , Biomarkers , Cardiac Myosins/genetics , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , GATA4 Transcription Factor , Gene Expression Regulation, Developmental , Heart/embryology , Humans , Immunohistochemistry , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myosin Heavy Chains/genetics , Myosin Light Chains/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Transcription Factors/genetics
7.
Stem Cells ; 22(3): 265-74, 2004.
Article in English | MEDLINE | ID: mdl-15153604

ABSTRACT

Type I diabetes mellitus is caused by an autoimmune destruction of the insulin-producing beta cells. The major obstacle in using transplantation for curing the disease is the limited source of insulin-producing cells. The isolation of human embryonic stem (hES) cells introduced a new prospect for obtaining a sufficient number of beta cells for transplantation. We present here a method for forming immature islet-like clusters of insulin-producing cells derived from hES cells. The protocol consisted of several steps. Embryoid bodies were first cultured and plated in insulin-transferrin-selenium-fibronectin medium, followed by medium supplemented with N2, B27, and basic fibroblast growth factor (bFGF). Next, the glucose concentration in the medium was lowered, bFGF was withdrawn, and nicotinamide was added. Dissociating the cells and growing them in suspension resulted in the formation of clusters which exhibited higher insulin secretion and had longer durability than cells grown as monolayers. Reverse transcription-polymerase chain reaction detected an enhanced expression of pancreatic genes in the differentiated cells. Immunofluorescence and in situ hybridization analyses revealed a high percentage of insulin-expressing cells in the clusters. In addition to insulin, most cells also coexpressed glucagon or somatostatin, indicating a similarity to immature pancreatic cells. Further improvement of this insulin-producing cell protocol may lead to the formation of an unlimited source of cells suitable for transplantation.


Subject(s)
Cell Differentiation/physiology , Insulin/metabolism , Islets of Langerhans/cytology , Stem Cells/cytology , Apoptosis/physiology , Cell Culture Techniques , Cell Differentiation/drug effects , Cells, Cultured , Diabetes Mellitus, Type 1/therapy , Embryo, Mammalian/cytology , Glucagon/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Insulin Secretion , Islets of Langerhans/metabolism , Niacinamide/pharmacology , Somatostatin/metabolism , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...