Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Genes Dev ; 37(5-6): 243-257, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36810209

ABSTRACT

Transfer RNAs (tRNAs) are small adaptor RNAs essential for mRNA translation. Alterations in the cellular tRNA population can directly affect mRNA decoding rates and translational efficiency during cancer development and progression. To evaluate changes in the composition of the tRNA pool, multiple sequencing approaches have been developed to overcome reverse transcription blocks caused by the stable structures of these molecules and their numerous base modifications. However, it remains unclear whether current sequencing protocols faithfully capture tRNAs existing in cells or tissues. This is specifically challenging for clinical tissue samples that often present variable RNA qualities. For this reason, we developed ALL-tRNAseq, which combines the highly processive MarathonRT and RNA demethylation for the robust assessment of tRNA expression, together with a randomized adapter ligation strategy prior to reverse transcription to assess tRNA fragmentation levels in both cell lines and tissues. Incorporation of tRNA fragments not only informed on sample integrity but also significantly improved tRNA profiling of tissue samples. Our data showed that our profiling strategy effectively improves classification of oncogenic signatures in glioblastoma and diffuse large B-cell lymphoma tissues, particularly for samples presenting higher levels of RNA fragmentation, further highlighting the utility of ALL-tRNAseq for translational research.


Subject(s)
Protein Biosynthesis , RNA, Transfer , RNA, Transfer/genetics , RNA, Transfer/metabolism , RNA, Messenger/metabolism , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, RNA/methods
2.
Ann Neurol ; 86(5): 780-792, 2019 11.
Article in English | MEDLINE | ID: mdl-31433864

ABSTRACT

OBJECTIVE: Astrocytes have gained attention as important players in neurological disease. In line with their heterogeneous character, defects in specific astrocyte subtypes have been identified. Leukodystrophy vanishing white matter (VWM) shows selective vulnerability in white matter astrocytes, but the underlying mechanisms remain unclear. Induced pluripotent stem cell technology is being extensively explored in studies of pathophysiology and regenerative medicine. However, models for distinct astrocyte subtypes for VWM are lacking, thereby hampering identification of disease-specific pathways. METHODS: Here, we characterize human and mouse pluripotent stem cell-derived gray and white matter astrocyte subtypes to generate an in vitro VWM model. We examined morphology and functionality, and used coculture methods, high-content microscopy, and RNA sequencing to study VWM cultures. RESULTS: We found intrinsic vulnerability in specific astrocyte subpopulations in VWM. When comparing VWM and control cultures, white matter-like astrocytes inhibited oligodendrocyte maturation, and showed affected pathways in both human and mouse cultures, involving the immune system and extracellular matrix. Interestingly, human white matter-like astrocytes presented additional, human-specific disease mechanisms, such as neuronal and mitochondrial functioning. INTERPRETATION: Astrocyte subtype cultures revealed disease-specific pathways in VWM. Cross-validation of human- and mouse-derived protocols identified human-specific disease aspects. This study provides new insights into VWM disease mechanisms, which helps the development of in vivo regenerative applications, and we further present strategies to study astrocyte subtype vulnerability in neurological disease. ANN NEUROL 2019;86:780-792.


Subject(s)
Astrocytes/pathology , Culture Techniques , Induced Pluripotent Stem Cells , Leukoencephalopathies/pathology , Animals , Humans , Mice
3.
Bio Protoc ; 9(17): e3350, 2019 Sep 05.
Article in English | MEDLINE | ID: mdl-33654852

ABSTRACT

Crosstalk between neurons and oligodendrocytes is important for proper brain functioning. Multiple co-culture methods have been developed to study oligodendrocyte maturation, myelination or the effect of oligodendrocytes on neurons. However, most of these methods contain cells derived from animal models. In the current protocol, we co-culture human neurons with human oligodendrocytes. Neurons and oligodendrocyte precursor cells (OPCs) were differentiated separately from pluripotent stem cells according to previously published protocols. To study neuron-glia cross-talk, neurons and OPCs were plated in co-culture mode in optimized conditions for additional 28 days, and prepared for OPC maturation and neuronal morphology analysis. To our knowledge, this is one of the first neuron-OPC protocols containing all human cells. Specific neuronal abnormalities not observed in mono-cultures of Tuberous Sclerosis Complex (TSC) neurons, became apparent when TSC neurons were co-cultured with TSC OPCs. These results show that this co-culture system can be used to study human neuron-OPC interactive mechanisms involved in health and disease.

4.
Stem Cell Reports ; 12(1): 42-56, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30581017

ABSTRACT

Tuberous sclerosis complex (TSC) is a rare neurodevelopmental disorder resulting from autosomal dominant mutations in the TSC1 or TSC2 genes, leading to a hyperactivated mammalian target of rapamycin (mTOR) pathway, and gray and white matter defects in the brain. To study the involvement of neuron-glia interactions in TSC phenotypes, we generated TSC patient induced pluripotent stem cell (iPSC)-derived cortical neuronal and oligodendrocyte (OL) cultures. TSC neuron mono-cultures showed increased network activity, as measured by calcium transients and action potential firing, and increased dendritic branching. However, in co-cultures with OLs, neuronal defects became more apparent, showing cellular hypertrophy and increased axonal density. In addition, TSC neuron-OL co-cultures showed increased OL cell proliferation and decreased OL maturation. Pharmacological intervention with the mTOR regulator rapamycin suppressed these defects. Our patient iPSC-based model, therefore, shows a complex cellular TSC phenotype arising from the interaction of neuronal and glial cells and provides a platform for TSC disease modeling and drug development.


Subject(s)
Neurons/physiology , Oligodendroglia/physiology , Tuberous Sclerosis/pathology , Action Potentials , Cell Proliferation , Cells, Cultured , Coculture Techniques , Humans , Induced Pluripotent Stem Cells/cytology , Neuronal Outgrowth , Neurons/cytology , Oligodendroglia/cytology , Phenotype
5.
Stem Cell Res ; 32: 25-34, 2018 10.
Article in English | MEDLINE | ID: mdl-30172094

ABSTRACT

The neural tube consists of neural progenitors (NPs) that acquire different characteristics during gestation due to patterning factors. However, the influence of such patterning factors on human pluripotent stem cells (hPSCs) during in vitro neural differentiation is often unclear. This study compared neural induction protocols involving in vitro patterning with single SMAD inhibition (SSI), retinoic acid (RA) administration and dual SMAD inhibition (DSI). While the derived NP cells expressed known NP markers, they differed in their NP expression profile and differentiation potential. Cortical neuronal cells generated from 1) SSI NPs exhibited less mature neuronal phenotypes, 2) RA NPs exhibited an increased GABAergic phenotype, and 3) DSI NPs exhibited greater expression of glutamatergic lineage markers. Further, although all NPs generated astrocytes, astrocytes derived from the RA-induced NPs had the highest GFAP expression. Differences between NP populations included differential expression of regional identity markers HOXB4, LBX1, OTX1 and GSX2, which persisted into mature neural cell stages. This study suggests that patterning factors regulate how potential NPs may differentiate into specific neuronal and glial cell types in vitro. This challenges the utility of generic neural induction procedures, while highlighting the importance of carefully selecting specific NP protocols.


Subject(s)
Cell Differentiation/physiology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Astrocytes/cytology , Astrocytes/metabolism , Cell Line , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Neurons/cytology , Neurons/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Vet Clin Pathol ; 37(1): 112-4, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18366553

ABSTRACT

A 14-year-old Italian Griffon and an 11-year-old mixed breed dog were presented to our clinic with monolateral testicular enlargement. In both dogs, a firm, nodular, and nonpainful mass was palpated, and ultrasonographic examination of testicular parenchyma showed a large and irregular nodular area with hyperechogenic features. Fine-needle aspirates of the masses were highly cellular and consisted of populations of large elongated vacuolated cells in sheets and palisades, with finely granular chromatin and prominent nucleoli, consistent with neoplastic Sertoli cells. A variable number of structures also were observed that consisted of a central round area of amorphous, deeply eosinophilic, hyaline material surrounded by a peripheral, rosette-like arrangement of single or multiple rows of Sertoli cells. These structures were suggestive of Call-Exner bodies. Histologic sections of the tumors obtained following castration confirmed the diagnosis of Sertoli cell neoplasia and the presence of Call-Exner bodies. The Call-Exner bodies were intensely positive with PAS, toluidine blue, and Ziehl-Neelsen stains, moderately positive with alcian blue, and negative with Congo red and Luxol fast blue. Call-Exner bodies, thought to represent an attempt by neoplastic cells to form basement membrane, are seen most frequently in granulosa cell tumors, but are occasionally reported in testicular tumors that contain epithelial elements of sex-cord origin. To our knowledge, this is the first description of Call-Exner bodies in cytologic specimens from dogs, and only the fifth report of their presence in canine testicular neoplasms.


Subject(s)
Dog Diseases/pathology , Sertoli Cell Tumor/veterinary , Testicular Neoplasms/veterinary , Animals , Dogs , Male , Sertoli Cell Tumor/pathology , Testicular Neoplasms/pathology
7.
J Mol Cell Cardiol ; 42(4): 746-59, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17300799

ABSTRACT

Human amniotic fluid-derived stem (AFS) cells, similarly to embryonic stem cells, could possess privileged immunological characteristics suitable for a successful transplantation even in a discordant xenograft system. We investigated whether AFS cells could be fruitfully used in a rat model of myocardial infarction. c-kit immunomagnetic-sorted AFS cells were characterized by flow cytometric analysis and cytospins as well as reverse-transcription polymerase chain reaction, Western blotting and immunocytochemistry for cardiovascular differentiation markers. In vitro, AFS cell phenotypic conversion was assayed by cardiovascular-specific induction media or co-cultured with rat neonatal cardiomyocytes. AFS cells showed mRNAs and/or protein for endothelial (angiopoietin, CD146) and smooth muscle (smoothelin) cells, and cardiomyocyte (Nkx2.5, MLC-2v, GATA-4, beta-MyHC) markers. Acquisition of a cardiomyocyte-like phenotype in rare AFS cells could be seen only in co-cultures with rat neonatal cells. In vivo, AFS cells xenotransplantated in a rat model of myocardial infarction, with or without cyclosporine treatment, or in intact heart from immuno-competent or immuno-deficient animals were acutely rejected due to the different recruitment of recipient CD4(+), CD8(+) T and B lymphocytes, NK cells and macrophages. This reaction is most likely to be linked to expression of B7 co-stimulatory molecules CD80 and CD86 as well as macrophage marker CD68 on AFS cells. Xenotransplanted AFS cells gave also rise in some animals to cell masses in the subendocardium and myocardium suggestive of a process of chondro-osteogenic differentiation. Despite AFS cells in vitro can differentiate to some extent to cells of cardiovascular lineages, their in vivo use in xenotransplantation for cell therapy of myocardial infarction is hampered by their peculiar immunogenic properties and phenotypic instability.


Subject(s)
Amniotic Fluid/cytology , Cell Differentiation , Myocardial Ischemia/therapy , Myocytes, Cardiac/cytology , Stem Cell Transplantation , Stem Cells/cytology , Adult , Animals , Biomarkers/metabolism , Cell Survival , Cells, Cultured , Coculture Techniques , Female , Humans , Immunosuppression Therapy , Myocardial Ischemia/immunology , Myocardial Ischemia/metabolism , Myocytes, Cardiac/immunology , Rats , Rats, Sprague-Dawley , Transplantation, Heterologous
8.
J Urol ; 177(1): 369-76, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17162093

ABSTRACT

PURPOSE: Wound healing of the cryo-injured bladder can bring about organ remodeling because of incomplete reconstitution of depleted smooth muscle cells. Stem cell transplantation could be beneficial to improve smooth muscle cell regeneration and/or modulate the remodeling process. The repair of bladder injury using adult-type stem cells would be useful for adult urological patients but unsuited for neonatal patients, in whom major benefits are likely to derive from fetal-type stem cells. MATERIALS AND METHODS: The smooth muscle cell differentiation potential of fetal-type vs adult-type stem cells was evaluated by injecting green fluorescent protein labeled mesenchymal stem cells from rat amniotic fluid or bone marrow, respectively, in cryo-injured rat bladder walls. RESULTS: At 30 days after transplantation only a few fetal-type or adult-type mesenchymal stem cells gave rise to enteric or vascular smooth muscle cells, whereas most mesenchymal stem cells appeared incapable of specific differentiation. In vitro co-culture experiments of smooth muscle cells with fetal-type or adult-type mesenchymal stem cells selectively labeled with distinct fluorochromes showed the presence of hybrid cells, suggesting that some mesenchymal stem cells can undergo cell fusion. Surprisingly the major effect of rat bone marrow or amniotic fluid mesenchymal stem cell transplantation seemed to be preventing cryo-injury induced hypertrophy of surviving smooth muscle cells. CONCLUSIONS: In this model stem cell transplantation has a limited effect on smooth muscle cell regeneration. Instead it can regulate post-injury bladder remodeling, possibly via a paracrine mechanism.


Subject(s)
Amniotic Fluid/cytology , Bone Marrow Cells , Cell Differentiation , Mesenchymal Stem Cells/cytology , Muscle, Smooth/cytology , Muscle, Smooth/pathology , Stem Cell Transplantation , Urinary Bladder/cytology , Animals , Female , Hypertrophy/prevention & control , Rats , Rats, Nude
9.
J Anat ; 209(3): 271-87, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16928198

ABSTRACT

The existence of specific differentiation markers for arterial smooth muscle (SM) cells is still a matter of debate. A clone named MM1 was isolated from a library of monoclonal antibodies to adult porcine aorta, which in vivo binds to arterial but not venous SM cells, except for the pulmonary vein. MM1 immunoreactivity in Western blotting involved bands in the range of M(r) 33-226 kDa, in both arterial and venous SM tissues. However, immunoprecipitation experiments revealed that MM1 bound to a 100-kDa polypeptide that was present only in the arterial SM extract. By mass spectrometry analysis of tryptic digests from MM1-positive 130- and 120-kDa polypeptides of aorta SM extract, the antigen recognized by the antibody was identified as a decorin precursor. Using a crude decorin preparation from this tissue MM1 reacted strongly with the 33-kDa polypeptide and this pattern did not change after chondroitinase ABC treatment. In vitro, decorin immunoreactivity was found in secreted grainy material produced by confluent arterial SM cells, although lesser amounts were also seen in venous SM cells. Western blotting of extracts from these cultures showed the presence of the 33-kDa band but not of the high-molecular-weight components, except for the 100-kDa monomer. The 100/33-kDa combination was more abundant in arterial SM cells than in the venous counterpart. In the early phase of neointima formation, induced by endothelial injury of the carotid artery or vein-to-artery transposition, the decorin precursor was not expressed, but it was up-regulated in the SM cells of the media underlying the neointima in both models. Collectively, these data suggest a different processing/utilization of the 100-kDa monomer of proteoglycan decorin in arterial and venous SM cells, which is abolished after vein injury.


Subject(s)
Antibodies, Monoclonal/immunology , Extracellular Matrix Proteins/immunology , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle/immunology , Proteoglycans/immunology , Animals , Animals, Newborn , Antibody Specificity , Antigen-Antibody Reactions , Aorta , Blood Vessel Prosthesis Implantation , Blotting, Western , Cells, Cultured , Coronary Vessels , Decorin , Female , Humans , Hybridomas , Immunoprecipitation , Jugular Veins , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Pulmonary Veins , Swine
10.
Eur J Cardiothorac Surg ; 28(5): 677-84, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16188450

ABSTRACT

OBJECTIVE: Transplantation of stem cells in the acute ischemic myocardium (AMI) may play a role in the recovery of cardiac function. Here, we investigated the ability of amniotic fluid-derived mesenchymal cells (AFC) for phenotypic conversion to vascular cells and cardiomyocytes (CM) when autotransplanted in a porcine model of AMI. METHODS: Single AFC preparations were taken from 12 fetuses 3 days before normal delivery. AFC were expanded in vitro and stored separately until animals of the original litter weighed 22-25 kg. A new model of AMI, i.e. 45-min circumflex coronary occlusion followed by wall dissection, was used to assess AFC differentiation potential. CMFDA-labeled AFC were autogenically transplanted in the ischemic area 1 week after AMI induction. Thirty days later, pigs were sacrificed and the phenotypic profile of transplanted AFC was assessed and compared to the corresponding pre-injection pattern. RESULTS: AFC showed in vitro to be of mesenchymal type also expressing markers of 'embryonic stem' cells (SSEA4 and Oct-4), as well as endothelial (von Willebrand factor, VE-cadherin) and smooth muscle (SM alpha-actin, SM22) cells. Thirty days after transplantation, in the survived AFC (5+/-1%) 'embryonic stem' cell markers disappeared and mesenchymal cell markers were down regulated with the exception of smooth muscle and endothelial antigens. No evidence for expression of cardiac troponin I was found. CONCLUSIONS: In the conditions used in this study, AFC were able to transdifferentiate to cells of vascular cell lineages but not to CM. Thus, porcine AFC may require further ex vivo re-programming to be suitable for therapeutic use in AMI.


Subject(s)
Amniotic Fluid/cytology , Cell Differentiation , Mesenchymal Stem Cell Transplantation/methods , Myocardial Ischemia/therapy , Animals , Biomarkers/metabolism , Cells, Cultured , Disease Models, Animal , Immunophenotyping , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocytes, Cardiac/cytology , Swine
11.
J Endovasc Ther ; 11(2): 196-210, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15056026

ABSTRACT

PURPOSE: To investigate the role of smooth muscle cell (SMC) response and platelet activation in peripheral venous catheterization using a model of catheter injury associated with thrombocytopenic treatment. METHODS: Silicon elastic catheters were inserted into New Zealand White rabbit external jugular veins from 24 hours to 60 days. Immunocytochemical procedures with antibodies to differentiation markers specific for SMCs, myofibroblasts, and endothelial cells were used to ascertain the phenotypic features of injured venous SMCs and the tissue sleeve formed around the catheter. Thrombocytopenia was induced in rabbits by busulfan treatment and the effect on catheter injury development examined after 15 days. The putative direct effect of this drug on the venous SMC proliferation, migration, and differentiation was assayed in vitro for 48 hours. RESULTS: Catheter injury is characterized by the progressive formation of (1) a neointima, containing differentiating SMCs, which are derived from the media and adventitial layer, and (2) by the organizing thrombus formed around the catheter, which contains myofibroblasts. In busulfan-treated thrombocytopenic animals, there was no evidence for either neointimal development or thrombus formation. A direct role of this drug in the unresponsiveness of vascular wall can be excluded by the unchanged proliferation and migration pattern of cultured venous SMCs treated with busulfan compared to control cultures. CONCLUSIONS: In our model, accumulation of differentiated SMCs in the neointima and myofibroblast appearance in the thrombus are linked, although distinct, events regulated by platelet activation, which is able to furnish the appropriate microenvironment for vascular SMC recruitment from the media/adventitial layer.


Subject(s)
Catheterization, Peripheral/adverse effects , Endothelium, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Platelet Activation , Animals , Blotting, Western , Busulfan/pharmacology , Cells, Cultured , Endothelium, Vascular/injuries , Immunohistochemistry , Immunosuppressive Agents/pharmacology , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Rabbits , Thrombocytopenia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...