Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Ann Hematol ; 101(6): 1227-1237, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35380239

ABSTRACT

Therapy-related myeloid neoplasm (t-MN) is a threatening complication of autologous stem cell transplantation (ASCT). Detecting clonal hematopoiesis (CH) mutations in cryopreserved cells before ASCT has been associated with a higher risk of t-MN, but the evolution of molecular abnormalities from pre-ASCT to t-MN, within the same patient, remains to be elucidated. We evaluated the mutational profile of 19 lymphoma/myeloma patients, at both pre-ASCT and t-MN diagnosis, using a targeted NGS approach; 26 non-developing t-MN control patients were also studied pre-ASCT. At ASCT, we found a higher frequency of CH in patients developing t-MN (58%) than in those who did not (23%) (P = 0.029); mutations in epigenetic (DNMT3A, TET2, and ASXL1) and DNA repair genes (PPM1D, RAD21, TP53, and STAG2) were the most represented. At t-MN, CH increased to 82% of patients. Cumulative mutational burden and variant allele frequency (VAF) also increased at t-MN. CH clones detected at ASCT were found at t-MN in eight out of 16 patients, mainly with stable VAF. Among the new driver mutations appeared at t-MN, TP53 increased from one to 13 mutations, in nine patients; being associated with complex karyotype. Mutations in transcription factor (RUNX1, CEBPA) and intracellular signaling genes (FLT3, RAS genes) also increased from three to 17 mutations in eight patients, presenting with a normal karyotype. Overall, we found that preexisting CH at ASCT rarely causes t-MN directly, but may rather facilitate the appearance of new mutations, especially those involving TP53, RUNX1, and RAS, that can drive the evolution to t-MN of at least two distinct types.


Subject(s)
Hematopoietic Stem Cell Transplantation , Myeloproliferative Disorders , Neoplasms, Second Primary , Clonal Hematopoiesis/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Mutation , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/therapy , Neoplasms, Second Primary/genetics , Transplantation, Autologous/adverse effects
3.
J Allergy Clin Immunol ; 132(3): 656-664.e17, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23830146

ABSTRACT

BACKGROUND: Combined immunodeficiency with multiple intestinal atresias (CID-MIA) is a rare hereditary disease characterized by intestinal obstructions and profound immune defects. OBJECTIVE: We sought to determine the underlying genetic causes of CID-MIA by analyzing the exomic sequences of 5 patients and their healthy direct relatives from 5 unrelated families. METHODS: We performed whole-exome sequencing on 5 patients with CID-MIA and 10 healthy direct family members belonging to 5 unrelated families with CID-MIA. We also performed targeted Sanger sequencing for the candidate gene tetratricopeptide repeat domain 7A (TTC7A) on 3 additional patients with CID-MIA. RESULTS: Through analysis and comparison of the exomic sequence of the subjects from these 5 families, we identified biallelic damaging mutations in the TTC7A gene, for a total of 7 distinct mutations. Targeted TTC7A gene sequencing in 3 additional unrelated patients with CID-MIA revealed biallelic deleterious mutations in 2 of them, as well as an aberrant splice product in the third patient. Staining of normal thymus showed that the TTC7A protein is expressed in thymic epithelial cells, as well as in thymocytes. Moreover, severe lymphoid depletion was observed in the thymus and peripheral lymphoid tissues from 2 patients with CID-MIA. CONCLUSIONS: We identified deleterious mutations of the TTC7A gene in 8 unrelated patients with CID-MIA and demonstrated that the TTC7A protein is expressed in the thymus. Our results strongly suggest that TTC7A gene defects cause CID-MIA.


Subject(s)
Immunologic Deficiency Syndromes/genetics , Intestinal Atresia/genetics , Intestines/abnormalities , Proteins/genetics , Animals , Child, Preschool , Exome/genetics , Female , Humans , Infant , Infant, Newborn , Male , Mice , Mutation , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Thymus Gland/metabolism , Tissue Array Analysis
5.
Hepatology ; 55(1): 153-60, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21898485

ABSTRACT

UNLABELLED: The cross-talk of cluster of differentiation (CD)40/CD40 ligand (CD40L) plays a key role in CD4(+) T-cell priming, B-cell terminal maturation, and immunoglobulin (Ig) class-switch recombination. Genetic defects in the CD40L lead to a disorder characterized by elevated concentrations of serum IgM and immunodeficiency. Patients with primary biliary cirrhosis (PBC) characteristically show circulating antimitochondrial antibodies (AMAs), liver-infiltrating autoreactive T lymphocytes against mitochondrial antigens, and high levels of IgM. We hypothesized that CD40L may play a key role in the pathogenesis of the elevated serum IgM and analyzed genetic and epigenetic modifications of the gene coding for CD40L in CD4(+) and CD8(+) T cells isolated from circulating mononuclear cells from PBC patients and healthy controls. We herein demonstrate significantly lower levels of DNA methylation of the CD40L promoter in CD4(+) T cells from PBC patients, as compared with controls, and this decreased methylation was inversely correlated with levels of serum IgM in PBC patients. CONCLUSION: The findings of an absence of genetic modifications of the CD40L gene, in concert with decreased DNA methylation of the CD40L promoter in PBC patients, suggests that environmental factors, rather than genetics, must play a major role in the pathogenesis of elevated serum IgM in PBC.


Subject(s)
CD40 Ligand/genetics , DNA Methylation/physiology , Immunoglobulin M/blood , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/immunology , Adult , Aged , Aged, 80 and over , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/physiology , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/physiology , Female , Genotype , HapMap Project , Humans , Immunoglobulin M/immunology , Liver Cirrhosis, Biliary/blood , Middle Aged , Promoter Regions, Genetic/physiology , Receptor Cross-Talk/immunology
6.
Am J Med Genet A ; 161A(10): 2614-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23950054

ABSTRACT

Wave-shaped ribs were detected at prenatal ultrasound in a 20(+1) week female fetus. At birth, skeletal radiographs showed marked hypomineralization and suggested hypophosphatasia. However, elevated blood calcium and alkaline phosphatase excluded hypophosphatasia and raised the possibility of Jansen metaphyseal dysplasia. Molecular analysis of the PTH/PTHrP receptor gene (PTH1R) showed heterozygosity for a previously undescribed transversion variant (c.1373T>A), which predicts p.Ile458Lys. In vitro evaluation of wild type and mutant PTH/PTHrP receptors supported the pathogenic role of the p.Ile458Lys substitution, and confirmed the diagnosis of Jansen metaphyseal dysplasia. This disorder may present prenatally with wavy ribs and in the newborn with hypomineralization, and may therefore be confused with hypophosphatasia. The mottled metaphyseal lesions typically associated with this disease appear only in childhood.


Subject(s)
Mutation, Missense , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/genetics , Receptor, Parathyroid Hormone, Type 1/genetics , Adult , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Female , Humans , Infant, Newborn , Pregnancy , Radiography , Ultrasonography, Prenatal
7.
Brain Sci ; 13(8)2023 Aug 08.
Article in English | MEDLINE | ID: mdl-37626534

ABSTRACT

Very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency is a rare autosomal recessive long-chain fatty acid oxidation disorder caused by mutations in the ACADVL gene. The myopathic form presents with exercise intolerance, exercise-related rhabdomyolysis, and muscle pain, usually starting during adolescence or adulthood. We report on a 17-year-old boy who has presented with exercise-induced muscle pain and fatigue since childhood. In recent clinical history, episodes of exercise-related severe hyperCKemia and myoglobinuria were reported. Electromyography was normal, and a muscle biopsy showed only "moth-eaten" fibers, and a mild increase in lipid storage in muscle fibers. NGS analysis displayed the already known heterozygote c.1769G>A variant and the unreported heterozygote c.523G>C change in ACADVL both having disease-causing predictions. Plasma acylcarnitine profiles revealed high long-chain acylcarnitine species levels, especially C14:1. Clinical, histopathological, biochemical, and genetic tests supported the diagnosis of VLCAD deficiency. Our report of a novel pathogenic missense variant in ACADVL expands the allelic heterogeneity of the disease. Since dietary treatment is the only therapy available for treating VLCAD deficiency and it is more useful the earlier it is started, prompt diagnosis is essential in order to minimize muscle damage and slow the disease progression.

8.
Blood ; 116(26): 5867-74, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-20702779

ABSTRACT

CD40/CD40 ligand (CD40L) cross-talk plays a key role in B-cell terminal maturation in the germinal centers. Genetic defects affecting CD40 cause a rare form of hyper-immunoglobulin M (IgM) syndrome, a disorder characterized by low or absent serum IgG and IgA, associated with recurrent infections. We previously reported on a few patients with homozygous CD40 mutations resulting in lack or severe reduction of CD40 cell surface expression. Here we characterize the 3 CD40 mutants due to missense mutations or small in-frame deletions, and show that the mutated proteins are synthesized but retained in the endoplasmic reticulum (ER), likely due to protein misfolding. Interestingly, the intracellular behavior and fate differ significantly among the mutants: progressive accumulation of the P2 mutant causes endoplasmic reticulum stress and the activation of an unfolded protein response; the mutant P4 is rather efficiently disposed by the ER-associated degradation pathway, while the P5 mutant partially negotiates transport to the plasma membrane, and is competent for CD40L binding. Interestingly, this latter mutant activates downstream signaling elements when overexpressed in transfected cells. These results give new important insights into the molecular pathogenesis of HIGM disease, and suggest that CD40 deficiency can also be regarded as an ER-storage disease.


Subject(s)
CD40 Antigens/genetics , CD40 Antigens/metabolism , Frameshift Mutation/genetics , Hyper-IgM Immunodeficiency Syndrome/genetics , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense/genetics , Amino Acid Sequence , B-Lymphocytes/metabolism , Blotting, Western , CD40 Antigens/chemistry , Cell Membrane/metabolism , Cells, Cultured , Child , Child, Preschool , Endoplasmic Reticulum/metabolism , Female , Flow Cytometry , Glycosylation , Humans , Hyper-IgM Immunodeficiency Syndrome/immunology , Infant , Kidney/cytology , Kidney/metabolism , Male , Molecular Sequence Data , Mutant Proteins/chemistry , Pedigree , Protein Conformation , Protein Folding , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
10.
Stem Cell Res ; 54: 102430, 2021 07.
Article in English | MEDLINE | ID: mdl-34182252

ABSTRACT

We have developed Joubert syndrome (JS)-derived induced pluripotent stem cell (iPSC) lines from dermal fibroblasts biopsied from a female patient harbouring novel compound heterozygous mutations in CC2D2A gene. The newly established iPSC lines provide tremendous promises for development of JS-derived neuronal cell lines to uncover the molecular and cellular mechanisms underlying the pathogenesis of JS and to develop therapeutic interventions for treatment of JS.


Subject(s)
Abnormalities, Multiple , Eye Abnormalities , Induced Pluripotent Stem Cells , Kidney Diseases, Cystic , Cell Differentiation , Cerebellum/abnormalities , Eye Abnormalities/genetics , Female , Fibroblasts , Humans , Mutation , Retina/abnormalities
13.
Stem Cell Res ; 42: 101660, 2020 01.
Article in English | MEDLINE | ID: mdl-31794943

ABSTRACT

Autosomal recessive osteopetrosis (ARO) is a rare inherited disorder leading to increased bone density with impairment in bone resorption. Among the genes responsible for ARO, the TCIRG1 gene, coding for the a3 subunit of the osteoclast proton pump, is mutated in more than 50% of the cases, increasing the importance of TCIRG1-iPSCs as disease model. We generated 3 iPSC clones derived from Peripheral Blood Mononuclear Cells (PBMCs) of a patient carrying the heterozygous mutations p.Y512X and c.2236 + 1G > A. A Sendai virus-based vector was used and the iPSCs were characterized for genetic identity to parental cells, genomic integrity, pluripotency, and differentiation ability.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Osteopetrosis/genetics , Vacuolar Proton-Translocating ATPases/genetics , Humans , Infant , Male , Mutation
14.
Stem Cell Res ; 49: 102007, 2020 12.
Article in English | MEDLINE | ID: mdl-33010677

ABSTRACT

We have generated new disease-specific induced pluripotent stem cell (iPSC) lines from skin fibroblasts obtained from a female patient with Joubert syndrome (JS) caused by compound heterozygous mutations in C5orf42 gene. The generated iPSCs offer an unprecedented opportunity to obtain iPSC-derived neurons to investigate the pathogenesis of JS in vitro and to develop therapeutic strategies.


Subject(s)
Abnormalities, Multiple , Eye Abnormalities , Induced Pluripotent Stem Cells , Kidney Diseases, Cystic , Cell Differentiation , Cerebellum/abnormalities , Eye Abnormalities/genetics , Female , Humans , Mutation , Retina/abnormalities
15.
Clin Pharmacol Ther ; 108(2): 358-367, 2020 08.
Article in English | MEDLINE | ID: mdl-32243572

ABSTRACT

Ataxia telangiectasia (AT) and Aicardi-Goutières syndrome (AGS) are inherited disorders of immunity with prevalent neurological phenotype. Available treatments are only partially effective, and the prognosis is poor. Induced pluripotent stem cells (iPSCs) are obtained by reprogramming patient somatic cells, preserving the donor individual genetic heritage and creating patient-specific disease models, useful to investigate pathogenesis and drug effects and to develop precision therapies. The aim is to investigate the cytotoxicity of a panel of immunomodulators using iPSCs of patients with AT or different forms of AGS (AGS1, AGS2, and AGS7). iPSCs were obtained by reprogramming AT and AGS patients' cells and, as a control, the BJ normal human fibroblast line, using Sendai virus. Cytotoxic effects of two drugs proposed to treat respectively AT and AGS (dexamethasone and mepacrine) were tested by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay after 72 hours' exposure. Data were obtained also for other immunomodulatory drugs (thioguanine, mercaptopurine, thalidomide, and lenalidomide). Relative expression of genes involved in the tested drug pathways was analyzed. AGS7-derived iPSCs displayed altered viability when treated with a low dose of mepacrine and higher expression of cyclic guanosine monophosphate-adenosine monophosphate synthase, which is the main target for mepacrine action. AGS7-derived iPSCs were also more sensitive to thioguanine, while AGS2 and AT iPSCs were less sensitive to this medication than the BJ-iPSC. All iPSCs were equally sensitive to mercaptopurine and resistant to dexamethasone, thalidomide, and lenalidomide. This work establishes an innovative in vitro model that is useful to investigate the mechanisms of drugs potentially effective in AT and AGS.


Subject(s)
Ataxia Telangiectasia/drug therapy , Autoimmune Diseases of the Nervous System/drug therapy , Immunologic Factors/pharmacology , Induced Pluripotent Stem Cells/drug effects , Nervous System Malformations/drug therapy , Precision Medicine , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/immunology , Ataxia Telangiectasia/metabolism , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/metabolism , Biomarkers/metabolism , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Clinical Decision-Making , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Drug Resistance , Genetic Predisposition to Disease , Humans , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/metabolism , Lenalidomide/pharmacology , Mercaptopurine/pharmacology , Nervous System Malformations/genetics , Nervous System Malformations/immunology , Nervous System Malformations/metabolism , Phenotype , Predictive Value of Tests , Quinacrine/pharmacology , Thalidomide/pharmacology , Thioguanine/pharmacology
16.
Dermatol Ther (Heidelb) ; 10(1): 213-220, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31691923

ABSTRACT

We report a rare case of a newborn male affected by incontinentia pigmenti, Klinefelter syndrome, and aplasia cutis congenita, who developed severe cutaneous, neurological, and ophthalmological manifestations. Genetic analysis showed the presence of the common mutation of NEMO (exon 4-10 deletion), Klinefelter syndrome karyotype (47 XXY), and random X inactivation. This is in accordance with the severity of involvement of the affected tissues (skin, central nervous system, and retina). Indeed, the patient developed typical skin lesions all over the body, except the head. Equally, multiple lesions diffusely involving both the cortical grey matter and subcortical white matter of the cerebellum and cerebral hemispheres were observed. Discussing current knowledge about the etiopathogenesis of skin and brain lesions in incontinentia pigmenti, our case seems to support the proapoptotic origin of central nervous system involvement. Possibly, incontinentia pigmenti patients suffer an impaired protection against apoptosis at the level of cerebral endothelial cells of small vessels, leading to vascular damage and subsequent ischemic brain lesions.

17.
J Clin Med ; 9(10)2020 Oct 17.
Article in English | MEDLINE | ID: mdl-33080915

ABSTRACT

Activated phosphoinositide 3-kinase delta syndrome 1 (APDS-1) is a recently described inborn error of immunity caused by monoallelic gain-of-function mutations in the PIK3CD gene. We reviewed for the first time medical records and laboratory data of eight Italian APDS-1 patients. Recurrent sinopulmonary infections were the most common clinical feature at onset of disease. Seven patients presented lymphoproliferative disease, at onset or during follow-up, one of which resembled hemophagocytic lymphohistiocytosis (HLH). Genetic analysis of the PIK3CD gene revealed three novel mutations: functional testing confirmed their activating nature. In the remaining patients, the previously reported variants p.E1021K (n = 4) and p.E525A (n = 1) were identified. Six patients were started on immunoglobulin replacement treatment (IgRT). One patient successfully underwent hematopoietic stem cell transplantation (HSCT), with good chimerism and no GVHD at 21 months post-HSCT. APDS-1 is a combined immune deficiency with a wide variety of clinical manifestations and a complex immunological presentation. Besides IgRT, specific therapies targeting the PI3Kδ pathway will most likely become a valid aid for the amelioration of patients' clinical management and their quality of life.

18.
Stem Cell Res ; 35: 101393, 2019 03.
Article in English | MEDLINE | ID: mdl-30711802

ABSTRACT

The Cri du Chat Syndrome (CdCS) is a genetic disease resulting from variable size deletion occurring on the short arm of chromosome 5. The main clinical features are a high-pitched monochromatic cry, microcephaly, severe psychomotor and mental retardation with characteristics of autism spectrum disorders such as hand flapping, obsessive attachments to objects, twirling objects, repetitive movements, and rocking. We reprogrammed to pluripotency peripheral blood mononuclear cells derived from a patient carrying large deletion on the short arm of chromosome 5, using a commercially available non-integrating expression system. The iPSCs expressed pluripotency markers and differentiated in the three embryonic germ layers.


Subject(s)
Cellular Reprogramming Techniques , Cri-du-Chat Syndrome , Induced Pluripotent Stem Cells , Leukocytes, Mononuclear , Adult , Cri-du-Chat Syndrome/genetics , Cri-du-Chat Syndrome/metabolism , Cri-du-Chat Syndrome/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Male
19.
Stem Cell Res ; 41: 101623, 2019 12.
Article in English | MEDLINE | ID: mdl-31698194

ABSTRACT

Aicardi-Goutières syndrome (AGS) is an early-onset monogenic encephalopathy characterized by intracranial calcification, leukodystrophy and cerebrospinal fluid lymphocytosis. To date, seven genes have been related to AGS. Among these, IFIH1 encodes for MDA5, a cytosolic double-stranded RNA receptor, and is responsible for AGS type 7. We generated three isogenic iPSC clones, using a Sendai virus-based vector, starting from fibroblasts of a patient carrying a dominant mutation in IFIH1. All lines were characterized for genomic integrity, genetic uniqueness, pluripotency, and differentiation capability. Our clones might offer a good model to investigate AGS7 pathophysiological mechanism and to discover new biomarkers for this condition treatment.


Subject(s)
Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/pathology , Cell Culture Techniques/methods , Cell Line/pathology , Fibroblasts/pathology , Interferon-Induced Helicase, IFIH1/genetics , Mutation/genetics , Nervous System Malformations/genetics , Nervous System Malformations/pathology , Adolescent , Base Sequence , Humans , Induced Pluripotent Stem Cells , Male , Reproducibility of Results
20.
Stem Cell Res ; 41: 101620, 2019 12.
Article in English | MEDLINE | ID: mdl-31678772

ABSTRACT

We report the generation of three isogenic iPSC clones (UNIBSi007-A, UNIBSi007-B, and UNIBSi007-C) obtained from fibroblasts of a patient with Aicardi Goutières Syndrome (AGS) carrying a homozygous mutation in RNaseH2B. Cells were transduced using a Sendai virus based system, delivering the human OCT4, SOX2, c-MYC and KLF4 transcription factors. The resulting transgene-free iPSC lines retained the disease-causing DNA mutation, showed normal karyotype, expressed pluripotent markers and could differentiate in vitro toward cells of the three embryonic germ layers.


Subject(s)
Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/pathology , Cell Culture Techniques/methods , Cell Line/pathology , Fibroblasts/pathology , Induced Pluripotent Stem Cells/pathology , Mutation/genetics , Nervous System Malformations/genetics , Nervous System Malformations/pathology , Ribonuclease H/genetics , Base Sequence , Child , Female , Humans , Kruppel-Like Factor 4 , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL