Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Haematologica ; 108(8): 2130-2145, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36794499

ABSTRACT

Sustained ANKRD26 expression associated with germline ANKRD26 mutations causes thrombocytopenia 2 (THC2), an inherited platelet disorder associated with a predisposition to leukemia. Some patients also present with erythrocytosis and/or leukocytosis. Using multiple human-relevant in vitro models (cell lines, primary patients' cells and patient-derived induced pluripotent stem cells) we demonstrate for the first time that ANKRD26 is expressed during the early steps of erythroid, megakaryocyte and granulocyte differentiation, and is necessary for progenitor cell proliferation. As differentiation progresses, ANKRD26 expression is progressively silenced, to complete the cellular maturation of the three myeloid lineages. In primary cells, abnormal ANKRD26 expression in committed progenitors directly affects the proliferation/differentiation balance for the three cell types. We show that ANKRD26 interacts with and crucially modulates the activity of MPL, EPOR and G-CSFR, three homodimeric type I cytokine receptors that regulate blood cell production. Higher than normal levels of ANKRD26 prevent the receptor internalization that leads to increased signaling and cytokine hypersensitivity. These findings afford evidence how ANKRD26 overexpression or the absence of its silencing during differentiation is responsible for myeloid blood cell abnormalities in patients with THC2.


Subject(s)
Leukemia , Receptors, Cytokine , Humans , Cytokines , Hematopoiesis , Leukemia/pathology , Cell Differentiation , Intercellular Signaling Peptides and Proteins
2.
Mov Disord ; 35(3): 401-408, 2020 03.
Article in English | MEDLINE | ID: mdl-31724242

ABSTRACT

BACKGROUND: Although Huntington's disease (HD) is caused by a single dominant gene, it is clear that there are genetic modifiers that may influence the age of onset and disease progression. OBJECTIVES: We sought to investigate whether new inflammation-related genetic variants may contribute to the onset and progression of HD. METHODS: We first used postmortem brain material from patients at different stages of HD to look at the protein expression of toll-like receptor 4 (TLR4) and triggering receptor expressed on myeloid cells 2 (TREM2). We then genotyped the TREM2 R47H gene variant and 3 TLR4 single nucleotide polymorphisms in a large cohort of HD patients from the European Huntington's Disease Network REGISTRY. RESULTS: We found an increase in the number of cells expressing TREM2 and TLR4 in postmortem brain samples from patients dying with HD. We also found that the TREM2 R47H gene variant was associated with changes in cognitive decline in the large cohort of HD patients, whereas 2 of 3 TLR4 single nucleotide polymorphisms assessed were associated with changes in motor progression in this same group. CONCLUSIONS: These findings identify TREM2 and TLR4 as potential genetic modifiers for HD and suggest that inflammation influences disease progression in this condition. © 2019 International Parkinson and Movement Disorder Society.


Subject(s)
Alzheimer Disease , Huntington Disease , Brain , Humans , Huntington Disease/genetics , Membrane Glycoproteins/genetics , Myeloid Cells , Receptors, Immunologic/genetics , Toll-Like Receptor 4/genetics
3.
Br J Cancer ; 121(9): 768-775, 2019 10.
Article in English | MEDLINE | ID: mdl-31558803

ABSTRACT

BACKGROUND: Current approaches aimed at inducing immunogenic cell death (ICD) to incite an immune response against cancer neoantigens are based on the use of chemotherapeutics and other agents. Results are hampered by issues of efficacy, combinatorial approaches, dosing and toxicity. Here, we adopted a strategy based on the use of an immunomolecule that overcomes pharmachemical limitations. METHODS: Cytofluorometry, electron microscopy, RT-PCR, western blotting, apotome immunofluorescence, MLR and xenografts. RESULTS: We report that an ICD process can be activated without the use of pharmacological compounds. We show that in Kras-mut/TP53-mut colorectal cancer cells the 15 kDa ßGBP cytokine, a T cell effector with onco-suppressor properties and a potential role in cancer immunosurveillance, induces key canonical events required for ICD induction. We document ER stress, autophagy that extends from cancer cells to the corresponding xenograft tumours, CRT cell surface shifting, ATP release and evidence of dendritic cell activation, a process required for priming cytotoxic T cells into a specific anticancer immunogenic response. CONCLUSIONS: Our findings provide experimental evidence for a rationale to explore a strategy based on the use of an immunomolecule that as a single agent couples oncosuppression with the activation of procedures necessary for the induction of long term response to cancer.


Subject(s)
Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/immunology , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Apoptosis/immunology , Autophagic Cell Death/drug effects , Autophagic Cell Death/immunology , Calreticulin/immunology , Calreticulin/metabolism , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Death/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Female , Galectins/pharmacology , Heterografts , Humans , Immunologic Surveillance , Mice , Mice, Nude , Proto-Oncogene Proteins p21(ras)/metabolism , Unfolded Protein Response/drug effects
4.
J Cell Physiol ; 233(7): 5396-5404, 2018 07.
Article in English | MEDLINE | ID: mdl-29215726

ABSTRACT

SEL1L (suppressor/enhancer of Lin-12-like) is a highly conserved gene associated with the endoplasmic reticulum-associated degradation (ERAD) pathway and involved in mediating the balance between stem cells self-renewal and differentiation of neural progenitors. It has been recently shown that SEL1L KO mice are embryonic lethal and display altered organogenesis. To better characterize the function of SEL1L in the early stages of embryonic development, we turned to the zebrafish model (Danio rerio). After exploring sel1l expression by RT-PCR and in situ hybridization, we employed a morpholino-mediated down-regulation approach. Results showed extensive impairments in the vasculature, which supports the mice knock-out findings.


Subject(s)
Embryonic Development/genetics , Endoplasmic Reticulum-Associated Degradation/genetics , Endoplasmic Reticulum/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Blood Vessels/growth & development , Blood Vessels/metabolism , Endoplasmic Reticulum/metabolism , Endothelium/cytology , Gene Expression Regulation, Developmental/genetics , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/genetics
5.
J Cell Physiol ; 233(4): 3152-3163, 2018 04.
Article in English | MEDLINE | ID: mdl-28816361

ABSTRACT

mSEL-1L is a highly conserved ER-resident type I protein, involved in the degradation of misfolded peptides through the ubiquitin-proteasome system (UPS), a pathway known to control the plasticity of the vascular smooth muscle cells (VSMC) phenotype and survival. In this article, we demonstrate that mSEL-1L deficiency interferes with the murine embryonic vascular network, showing particular irregularities in the intracranic and intersomitic neurovascular units and in the cerebral capillary microcirculation. During murine embryogenesis, mSEL-1L is expressed in cerebral areas known to harbor progenitor neural cells, while in the adult brain the protein is specifically restricted to the stem cell niches, co-localizing with Sox2 and Nestin. Null mice are characterized by important defects in the development of telenchephalic regions, revealing conspicuous aberration in neural stem cell lineage commitment. Moreover, mSEL-1L depletion in vitro and in vivo appears to affect the harmonic differentiation of the NSCs, by negatively influencing the corticogenesis processes. Overall, the data presented suggests that the drastic phenotypic characteristics exhibited in mSEL-1L null mice can, in part, be explained by the negative influence it plays on Notch1 signaling pathway.


Subject(s)
Cell Lineage , Neovascularization, Physiologic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Proteins/metabolism , Animals , Brain/growth & development , Brain/metabolism , Cell Proliferation , Cell Self Renewal , Genome , Intracellular Signaling Peptides and Proteins , Mice, Knockout , Receptors, Notch/metabolism , Transcriptome/genetics
6.
Mol Cell Neurosci ; 82: 46-57, 2017 07.
Article in English | MEDLINE | ID: mdl-28476540

ABSTRACT

DNA methylation (DNAm) changes are of increasing relevance to neurodegenerative disorders, including Huntington's disease (HD). We performed genome-wide screening of possible DNAm changes occurring during striatal differentiation in human induced pluripotent stem cells derived from a HD patient (HD-hiPSCs) as cellular model. We identified 240 differentially methylated regions (DMRs) at promoters in fully differentiated HD-hiPSCs. Subsequently, we focused on the methylation differences in a subcluster of genes related to Jumonji Domain Containing 3 (JMJD3), a demethylase that epigenetically regulates neuronal differentiation and activates neuronal progenitor associated genes, which are indispensable for neuronal fate acquisition. Noticeably among these genes, WD repeat-containing protein 5 (WDR5) promoter was found hypermethylated in HD-hiPSCs, resulting in a significant down-modulation in its expression and of the encoded protein. A similar WDR5 expression decrease was seen in a small series of HD-hiPSC lines characterized by different CAG length. The decrease in WDR5 expression was particularly evident in HD-hiPSCs compared to hESCs and control-hiPSCs from healthy subjects. WDR5 is a core component of the MLL/SET1 chromatin remodeling complexes essential for H3K4me3, previously reported to play an important role in stem cells self-renewal and differentiation. These results suggest the existence of epigenetic mechanisms in HD and the identification of genes, which are able to modulate HD phenotype, is important both for biomarker discovery and therapeutic interventions.


Subject(s)
Cell Differentiation/genetics , Epigenesis, Genetic/genetics , Histone-Lysine N-Methyltransferase/genetics , Huntington Disease/metabolism , Induced Pluripotent Stem Cells/cytology , Cell Line , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , Humans , Huntington Disease/genetics , Intracellular Signaling Peptides and Proteins , Neurons/metabolism
7.
J Biol Chem ; 289(5): 2826-38, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24311781

ABSTRACT

Valproic acid (VPA), an histone deacetylase inhibitor, is emerging as a promising therapeutic agent for the treatments of gliomas by virtue of its ability to reactivate the expression of epigenetically silenced genes. VPA induces the unfolded protein response (UPR), an adaptive pathway displaying a dichotomic yin yang characteristic; it initially contributes in safeguarding the malignant cell survival, whereas long-lasting activation favors a proapoptotic response. By triggering UPR, VPA might tip the balance between cellular adaptation and programmed cell death via the deregulation of protein homeostasis and induction of proteotoxicity. Here we aimed to investigate the impact of proteostasis on glioma stem cells (GSC) using VPA treatment combined with subversion of SEL1L, a crucial protein involved in homeostatic pathways, cancer aggressiveness, and stem cell state maintenance. We investigated the global expression of GSC lines untreated and treated with VPA, SEL1L interference, and GSC line response to VPA treatment by analyzing cell viability via MTT assay, neurosphere formation, and endoplasmic reticulum stress/UPR-responsive proteins. Moreover, SEL1L immunohistochemistry was performed on primary glial tumors. The results show that (i) VPA affects GSC lines viability and anchorage-dependent growth by inducing differentiative programs and cell cycle progression, (ii) SEL1L down-modulation synergy enhances VPA cytotoxic effects by influencing GSCs proliferation and self-renewal properties, and (iii) SEL1L expression is indicative of glioma proliferation rate, malignancy, and endoplasmic reticulum stress statuses. Targeting the proteostasis network in association to VPA treatment may provide an alternative approach to deplete GSC and improve glioma treatments.


Subject(s)
Brain Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Glioma/pathology , Proteins/metabolism , Unfolded Protein Response/drug effects , Valproic Acid/toxicity , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Down-Regulation/drug effects , Down-Regulation/physiology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/toxicity , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Glioma/drug therapy , Glioma/metabolism , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Proteins/genetics , Unfolded Protein Response/physiology
8.
J Cell Physiol ; 230(8): 1770-80, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25502508

ABSTRACT

MicroRNAs (miRNAs) are important regulators of several cellular processes. During hematopoiesis, specific expression signatures have been reported in different blood cell lineages and stages of hematopoietic stem cell (HSC) differentiation. Here we explored the expression of miRNAs in umbilical cord blood stem (HSC) and progenitor cells (HPC) and compared it to unilineage granulocyte and granulo-monocyte differentiation as well as to primary blasts from patients with acute myeloid leukemia (AML). CD34 + CD38- ad CD34 + CD38 + cells were profiled using a global array consisting of about 2000 miRNAs. An approach combining bioinformatic prediction of miRNA targets with mRNA expression profiling was used to search for putative biologically enriched functions and networks. At least 15 miRNAs to be differentially expressed between HSC and HPC cell population, a cluster of 7 miRNAs are located in the q32 region of human chromosome 14 (miR-377-3p, -136-5p, 376a-3p, 495-3p, 654-3p, 376c-3p and 381-3p) whose expression decreased during the early stages of normal myelopoiesis but were markedly increased in a small set of AML. Interestingly, miR-4739 and -4516, two novel microRNA whose function and targets are presently unknown, showed specific and peculiar expression profile during the hematopoietic stem cells differentiation into unilineages and resulted strongly upregulated in almost all AML subsets. miR-181, -126-5p, -29b-3p and -22-3p resulted dis-regulated in specific leukemias phenotypes. This study provides the first evidence of a miRNA signature in human cord blood stem and progenitor cells with a potential role in hematopoietic stemness properties and possibly in leukemogenesis of specific AML subtypes.


Subject(s)
Cell Differentiation/genetics , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Leukemia, Myeloid, Acute/genetics , MicroRNAs/genetics , Transcriptome/genetics , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Computational Biology , Humans , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction
9.
Toxicol Appl Pharmacol ; 280(3): 421-33, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25173957

ABSTRACT

Cell differentiation and response to hormonal signals were studied in a 3D environment on an in-house generated mouse fibroblast cell line expressing a reporter gene under the control of estrogen responsive sequences (EREs). 3D cell culture conditions were obtained in a Rotary Cell Culture System; (RCCS™), a microgravity based bioreactor that promotes the aggregation of cells into multicellular spheroids (MCS). In this bioreactor the cells maintained a better differentiated phenotype and more closely resembled in vivo tissue. The RCCS™ cultured fibroblasts showed higher expression of genes regulating cell assembly, differentiation and hormonal functions. Microarray analysis showed that genes related to cell cycle, proliferation, cytoskeleton, migration, adhesion and motility were all down-regulated in 3D as compared to 2D conditions, as well as oncogene expression and inflammatory cytokines. Controlled remodeling of ECM, which is an essential aspect of cell organization, homeostasis and tissue was affected by the culture method as assessed by immunolocalization of ß-tubulin. Markers of cell organization, homeostasis and tissue repair, metalloproteinase 2 (MMP2) and its physiological inhibitor (TIMP4) changed expression in association with the relative formation of cell aggregates. The fibroblasts cultured in the RCCS™ maintain a better responsiveness to estrogens, measured as expression of ERα and regulation of an ERE-dependent reporter and of the endogenous target genes CBP, Rarb, MMP1 and Dbp. Our data highlight the interest of this 3D culture model for its potential application in the field of cell response to hormonal signals and the pharmaco-toxicological analyses of chemicals and natural molecules endowed of estrogenic potential.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/physiology , Estrogens/physiology , Extracellular Matrix/physiology , Fibroblasts/physiology , Animals , Gene Expression Profiling , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , RNA/chemistry , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction
10.
J Clin Invest ; 134(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39007270

ABSTRACT

Copy number variation (CNV) at 7q11.23 causes Williams-Beuren syndrome (WBS) and 7q microduplication syndrome (7Dup), neurodevelopmental disorders (NDDs) featuring intellectual disability accompanied by symmetrically opposite neurocognitive features. Although significant progress has been made in understanding the molecular mechanisms underlying 7q11.23-related pathophysiology, the propagation of CNV dosage across gene expression layers and their interplay remains elusive. Here we uncovered 7q11.23 dosage-dependent symmetrically opposite dynamics in neuronal differentiation and intrinsic excitability. By integrating transcriptomics, translatomics, and proteomics of patient-derived and isogenic induced neurons, we found that genes related to neuronal transmission follow 7q11.23 dosage and are transcriptionally controlled, while translational factors and ribosomal genes are posttranscriptionally buffered. Consistently, we found phosphorylated RPS6 (p-RPS6) downregulated in WBS and upregulated in 7Dup. Surprisingly, p-4EBP was changed in the opposite direction, reflecting dosage-specific changes in total 4EBP levels. This highlights different dosage-sensitive dyregulations of the mTOR pathway as well as distinct roles of p-RPS6 and p-4EBP during neurogenesis. Our work demonstrates the importance of multiscale disease modeling across molecular and functional layers, uncovers the pathophysiological relevance of ribosomal biogenesis in a paradigmatic pair of NDDs, and uncouples the roles of p-RPS6 and p-4EBP as mechanistically actionable relays in NDDs.


Subject(s)
Chromosomes, Human, Pair 7 , DNA Copy Number Variations , Neurons , Humans , Neurons/metabolism , Neurons/pathology , Chromosomes, Human, Pair 7/genetics , Ribosomes/metabolism , Ribosomes/genetics , Neurogenesis/genetics , Williams Syndrome/genetics , Williams Syndrome/metabolism , Williams Syndrome/pathology , Williams Syndrome/physiopathology , Ribosomal Protein S6/metabolism , Ribosomal Protein S6/genetics , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Male , Cell Differentiation , Female
11.
J Biol Chem ; 286(21): 18708-19, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21454627

ABSTRACT

Murine SEL-1L (mSEL-1L) is a key component of the endoplasmic reticulum-associated degradation pathway. It is essential during development as revealed by the multi-organ dysfunction and in uterus lethality occurring in homozygous mSEL-1L-deficient mice. Here we show that mSEL-1L is highly expressed in pluripotent embryonic stem cells and multipotent neural stem cells (NSCs) but silenced in all mature neural derivatives (i.e. astrocytes, oligodendrocytes, and neurons) by mmu-miR-183. NSCs derived from homozygous mSEL-1L-deficient embryos (mSEL-1L(-/-) NSCs) fail to proliferate in vitro, show a drastic reduction of the Notch effector HES-5, and reveal a significant down-modulation of the early neural progenitor markers PAX-6 and OLIG-2, when compared with the wild type (mSEL-1L(+/+) NSCs) counterpart. Furthermore, these cells are almost completely deprived of the neural marker Nestin, display a significant decrease of SOX-2 expression, and rapidly undergo premature astrocytic commitment and apoptosis. The data suggest severe self-renewal defects occurring in these cells probably mediated by misregulation of the Notch signaling. The results reported here denote mSEL-1L as a primitive marker with a possible involvement in the regulation of neural progenitor stemness maintenance and lineage determination.


Subject(s)
Antigens, Differentiation/metabolism , Apoptosis/physiology , Cell Lineage/physiology , Multipotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Proteins/metabolism , Animals , Antigens, Differentiation/genetics , Astrocytes/cytology , Astrocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Eye Proteins/genetics , Eye Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , Multipotent Stem Cells/cytology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neural Stem Cells/cytology , Oligodendrocyte Transcription Factor 2 , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Proteins/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism
12.
J Cell Physiol ; 227(3): 1226-34, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21618531

ABSTRACT

The health of cells is preserved by the levels and correct folding states of the proteome, which is generated and maintained by the proteostasis network, an integrated biological system consisting of several cytoprotective and degradative pathways. Indeed, the health conditions of the proteostasis network is a fundamental prerequisite to life as the inability to cope with the mismanagement of protein folding arising from genetic, epigenetic, and micro-environment stress appears to trigger a whole spectrum of unrelated diseases. Here we describe the potential functional role of the proteostasis network in tumor biology and in conformational diseases debating on how the signaling branches of this biological system may be manipulated to develop more efficacious and selective therapeutic strategies. We discuss the dual strategy of these processes in modulating the folding activity of molecular chaperones in order to counteract the antithetic proteostasis deficiencies occurring in cancer and loss/gain of function diseases. Finally, we provide perspectives on how to improve the outcome of these disorders by taking advantage of proteostasis modeling.


Subject(s)
Drug Delivery Systems/methods , Molecular Chaperones/metabolism , Molecular Targeted Therapy/methods , Neoplasms/metabolism , Neoplasms/therapy , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/therapy , Humans , Neoplasms/pathology , Proteostasis Deficiencies/pathology
13.
J Cell Physiol ; 227(1): 14-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21412777

ABSTRACT

The use of human stem cells in biomedical research projects is increasing steadily and the number of cells that are being derived develops at a remarkable pace. However, stem cells around the world are vastly different in their provenance, programming, and potentials. Furthermore, knowledge on the actual number of cell types, their derivation, availability, and characteristics is rather sparse. Usually, "colleague-supply" avenues constantly furnish cells to laboratories around the world without ensuring their correct identity, characterization, and quality. These parameters are critical if the cells will be eventually used in toxicology studies and drug discovery. Here, we outline some basic principles in establishing a stem cell-specific bank.


Subject(s)
Stem Cells , Tissue Banks/trends , Humans , Tissue Banks/organization & administration
14.
Neurobiol Dis ; 46(1): 41-51, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22405424

ABSTRACT

Neuronal disorders, like Huntington's disease (HD), are difficult to study, due to limited cell accessibility, late onset manifestations, and low availability of material. The establishment of an in vitro model that recapitulates features of the disease may help understanding the cellular and molecular events that trigger disease manifestations. Here, we describe the generation and characterization of a series of induced pluripotent stem (iPS) cells derived from patients with HD, including two rare homozygous genotypes and one heterozygous genotype. We used lentiviral technology to transfer key genes for inducing reprogramming. To confirm pluripotency and differentiation of iPS cells, we used PCR amplification and immunocytochemistry to measure the expression of marker genes in embryoid bodies and neurons. We also analyzed teratomas that formed in iPS cell-injected mice. We found that the length of the pathological CAG repeat did not increase during reprogramming, after long term growth in vitro, and after differentiation into neurons. In addition, we observed no differences between normal and mutant genotypes in reprogramming, growth rate, caspase activation or neuronal differentiation. However, we observed a significant increase in lysosomal activity in HD-iPS cells compared to control iPS cells, both during self-renewal and in iPS-derived neurons. In conclusion, we have established stable HD-iPS cell lines that can be used for investigating disease mechanisms that underlie HD. The CAG stability and lysosomal activity represent novel observations in HD-iPS cells. In the future, these cells may provide the basis for a powerful platform for drug screening and target identification in HD.


Subject(s)
Cell Culture Techniques/methods , Huntington Disease/genetics , Huntington Disease/metabolism , Lysosomes/genetics , Nerve Tissue Proteins/genetics , Pluripotent Stem Cells/metabolism , Animals , Cell Line , Fibroblasts/cytology , Fibroblasts/physiology , Heterozygote , Homozygote , Humans , Huntingtin Protein , Huntington Disease/pathology , Lysosomes/metabolism , Mice , Mice, SCID , Mutation , Nerve Tissue Proteins/metabolism , Phenotype , Teratoma/genetics , Teratoma/metabolism , Transcriptional Activation/physiology
15.
Dig Dis Sci ; 57(4): 905-12, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22350780

ABSTRACT

BACKGROUND: SEL1L gene product is implicated in the endoplasmic reticulum (ER)-associated protein degradation and Unfolded Protein Response pathways. This gene and associated miRNAs have been indicated as predictive and prognostic markers of pancreatic cancer. AIM: Explore the role of SEL1L in colorectal cancer (CRC) progression. METHODS: SEL1L expression was analysed immunohistochemically in 153 adenomas and 71 CRCs from African American and North Italian patients. The distribution of stained cells was determined by computing median and inter quartile range. The receiver operating characteristics plot was used as discriminate power of SEL1L expression, CRC diagnosis and the effects on patient survival. RESULTS: SEL1L was low in normal mucosa and confined to few scattered cells at the base crypt of the villi and in the foveolar glandular compartment. The highest levels were in Paneth cells within the lysosomes. The enterocytic progenitor cells and mature enterocytes showed less cytoplasmic staining. In CRCs, SEL1L expression significantly correlated with the progression from adenoma to carcinoma (P = 0.0001) being stronger in well-to-moderately differentiated cancers. No correlation was found with other clinicopathological characteristics or ethnicity. CONCLUSIONS: SEL1L expression is a potential CRC tissue biomarker since its expression is significantly higher in adenoma cells with respect to normal mucosa. The levels of expression decrease sensibly in undifferentiated CRC cancers. Interestingly, Paneth cells contain high levels of SEL1L protein that could indicate pre-neoplastic mucosa undergoing neoplastic transformation. Since SEL1L's major function lies within ER stress and active ERAD response, it may identify CRCs with differentiated secretory phenotype and acute cellular stress.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic , Colorectal Neoplasms/diagnosis , Proteins/metabolism , Adenocarcinoma/diagnosis , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenoma/diagnosis , Adenoma/metabolism , Adenoma/pathology , Blotting, Western , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease Progression , Female , Humans , Immunohistochemistry , Intestinal Mucosa/metabolism , Male , Middle Aged , Paneth Cells/metabolism , Paneth Cells/pathology , Survival Rate , Tissue Array Analysis , Unfolded Protein Response
16.
J Biol Chem ; 285(18): 13694-703, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20197277

ABSTRACT

Stress in the endoplasmic reticulum (ER) plays an important causal role in the pathogenesis of several chronic diseases such as Alzheimer, Parkinson, and diabetes mellitus. Insight into the genetic determinants responsible for ER homeostasis will greatly facilitate the development of therapeutic strategies for the treatment of these debilitating diseases. Suppressor enhancer Lin12 1 like (SEL1L) is an ER membrane protein and was thought to be involved in the quality control of secreted proteins. Here we show that the mice homozygous mutant for SEL1L were embryonic lethal. Electron microscopy studies revealed a severely dilated ER in the fetal liver of mutant embryos, indicative of alteration in ER homeostasis. Consistent with this, several ER stress responsive genes were significantly up-regulated in the mutant embryos. Mouse embryonic fibroblast cells deficient in SEL1L exhibited activated unfolded protein response at the basal state, impaired ER-associated protein degradation, and reduced protein secretion. Furthermore, markedly increased apoptosis was observed in the forebrain and dorsal root ganglions of mutant embryos. Taken together, our results demonstrate an essential role for SEL1L in protein quality control during mouse embryonic development.


Subject(s)
Embryo, Mammalian/metabolism , Embryonic Development , Endoplasmic Reticulum/metabolism , Gene Expression Regulation, Developmental , Proteins/metabolism , Unfolded Protein Response , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Embryo Loss/genetics , Embryo Loss/metabolism , Embryo Loss/pathology , Embryo, Mammalian/ultrastructure , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Intracellular Signaling Peptides and Proteins , Mice , Mice, Mutant Strains , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Proteins/genetics
17.
BioTech (Basel) ; 10(3)2021 Jul 02.
Article in English | MEDLINE | ID: mdl-35822767

ABSTRACT

BACKGROUND: The emergence of "multi-omics" and "multi-parametric" types of analysis based on a high number of biospecimens enforces the use of a great number of high-quality "Biological Materials and Associated Data" (BMaD). To meet the demands of biomedical research, several Biological Resource Centers (BRCs) or Biobanks world-wide have implemented a specific Quality Management System (QMS) certified ISO 9001:2015 or accredited by CAP9 ISO 20387:2018. For the first time, ISO, with the support of several Biobanking experts, issued the ISO 20387:2018 which is the first ISO norm specific for Biobanks. The fundamental difference with present certification/accreditation standards is that the ISO 20387:2018 focuses not only on the operational aspects of the Biobank, but also on the "competence of the Biobank to carry our specific Biobanking tasks". METHODS: The accreditation process for ISO 20387:2018 required the definition of: (1) objectives, goals and organizational structure of the Biobank, including procedures for governance, confidentiality and impartiality policies; (2) standard operating procedures (SOPs) of all activities performed, including acquisition, analysis, collection, data management, distribution, preparation, preservation, testing facility and equipment maintenance, calibration, and monitoring; (3) procedures for control of documents and records, the identification of risks and opportunities, improvements, corrective actions, nonconforming records and evaluation of external providers (4) an internal audit and management reviews, verification of QMS performance, monitoring of quality objectives and personnel qualification and competency in carrying out specific Biobanking tasks. RESULTS: The accreditation process is performed by an independent authorized organization which certifies that all processes are performed according to the QMS, and that the infrastructure is engineered and managed according to the GDP and/or GMP guidelines. CONCLUSION: Accreditation is given by an accreditation body, which recognizes formally that the Biobank is "competent to carry out specific Biobanking tasks".

18.
J Histochem Cytochem ; 69(7): 451-460, 2021 07.
Article in English | MEDLINE | ID: mdl-34142588

ABSTRACT

"Multi-Omics" technologies have contributed greatly to the understanding of various diseases by enabling researchers to accurately and rapidly investigate the molecular circuitry that connects cellular systems. The tissue-engineered, three-dimensional (3D), in vitro disease model "organoid" integrates the "omics" results in a model system, elucidating the complex links between genotype and phenotype. These 3D structures have been used to model cancer, infectious disease, toxicity, and neurological disorders. Here, we describe the advantage of using the tissue microarray (TMA) technology to analyze human-induced pluripotent stem cell-derived cerebral organoids. Compared with the conventional processing of individual samples, sectioning and staining of TMA slides are faster and can be automated, decreasing labor and reagent costs. The TMA technology faithfully captures cell morphology variations and detects specific biomarkers. The use of this technology can scale up organoid research results in at least two ways: (1) in the number of specimens that can be analyzed simultaneously and (2) in the number of consecutive sections that can be produced for analysis with different probes and antibodies.


Subject(s)
Brain/cytology , Organoids/cytology , Tissue Array Analysis , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/cytology , Neurons/cytology
19.
Sci Rep ; 11(1): 13982, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34234171

ABSTRACT

Samples used in biomedical research are often collected over years, in some cases from subjects that may have died and thus cannot be retrieved in any way. The value of these samples is priceless. Sample misidentification or mix-up are unfortunately common problems in biomedical research and can eventually result in the publication of incorrect data. Here we have compared the Fluidigm SNPtrace and the Agena iPLEX Sample ID panels for the authentication of human genomic DNA samples. We have tested 14 pure samples and simulated their cross-contamination at different percentages (2%, 5%, 10%, 25% and 50%). For both panels, we report call rate, allele intensity/probability score, performance in distinguishing pure samples and contaminated samples at different percentages, and sex typing. We show that both panels are reliable and efficient methods for sample authentication and we highlight their advantages and disadvantages. We believe that the data provided here is useful for sample authentication especially in biorepositories and core facility settings.


Subject(s)
Biological Specimen Banks/standards , Biomedical Research/standards , Biometric Identification , Biomedical Research/methods , Biometric Identification/methods , DNA Contamination , Female , Humans , Male , Microsatellite Repeats , Polymorphism, Single Nucleotide
20.
Elife ; 102021 06 01.
Article in English | MEDLINE | ID: mdl-34059198

ABSTRACT

Thrombocytopenic disorders have been treated with the Thrombopoietin-receptor agonist Eltrombopag. Patients with the same apparent form of thrombocytopenia may respond differently to the treatment. We describe a miniaturized bone marrow tissue model that provides a screening bioreactor for personalized, pre-treatment response prediction to Eltrombopag for individual patients. Using silk fibroin, a 3D bone marrow niche was developed that reproduces platelet biogenesis. Hematopoietic progenitors were isolated from a small amount of peripheral blood of patients with mutations in ANKRD26 and MYH9 genes, who had previously received Eltrombopag. The ex vivo response was strongly correlated with the in vivo platelet response. Induced Pluripotent Stem Cells (iPSCs) from one patient with mutated MYH9 differentiated into functional megakaryocytes that responded to Eltrombopag. Combining patient-derived cells and iPSCs with the 3D bone marrow model technology allows having a reproducible system for studying drug mechanisms and for individualized, pre-treatment selection of effective therapy in Inherited Thrombocytopenias.


Platelets are tiny cell fragments essential for blood to clot. They are created and released into the bloodstream by megakaryocytes, giant cells that live in the bone marrow. In certain genetic diseases, such as Inherited Thrombocytopenia, the bone marrow fails to produce enough platelets: this leaves patients extremely susceptible to bruising, bleeding, and poor clotting after an injury or surgery. Certain patients with Inherited Thrombocytopenia respond well to treatments designed to boost platelet production, but others do not. Why these differences exist could be investigated by designing new test systems that recreate the form and function of bone marrow in the laboratory. However, it is challenging to build the complex and poorly understood bone marrow environment outside of the body. Here, Di Buduo et al. have developed an artificial three-dimensional miniature organ bioreactor system that recreates the key features of bone marrow. In this system, megakaryocytes were grown from patient blood samples, and hooked up to a tissue scaffold made of silk. The cells were able to grow as if they were in their normal environment, and they could shed platelets into an artificial bloodstream. After treating megakaryocytes with drugs to stimulate platelet production, Di Buduo et al. found that the number of platelets recovered from the bioreactor could accurately predict which patients would respond to these drugs in the clinic. This new test system enables researchers to predict how a patient will respond to treatment, and to tailor therapy options to each individual. This technology could also be used to test new drugs for Inherited Thrombocytopenias and other blood-related diseases; if scaled-up, it could also, one day, generate large quantities of lab-grown blood cells for transfusion.


Subject(s)
Benzoates/pharmacology , Blood Platelets/drug effects , Hematopoietic Stem Cells/drug effects , Hydrazines/pharmacology , Induced Pluripotent Stem Cells/drug effects , Megakaryocytes/drug effects , Pyrazoles/pharmacology , Receptors, Thrombopoietin/agonists , Stem Cell Niche , Thrombocytopenia/drug therapy , Thrombopoiesis/drug effects , Adult , Aged , Bioreactors , Blood Platelets/metabolism , Cell Culture Techniques , Cells, Cultured , Female , Fibroins/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Male , Megakaryocytes/metabolism , Middle Aged , Miniaturization , Mutation , Myosin Heavy Chains/genetics , Receptors, Thrombopoietin/metabolism , Thrombocytopenia/blood , Thrombocytopenia/genetics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL