Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Acta Neuropathol Commun ; 11(1): 50, 2023 03 25.
Article in English | MEDLINE | ID: mdl-36966348

ABSTRACT

Gangliogliomas are brain tumors composed of neuron-like and macroglia-like components that occur in children and young adults. Gangliogliomas are often characterized by a rare population of immature astrocyte-appearing cells expressing CD34, a marker expressed in the neuroectoderm (neural precursor cells) during embryogenesis. New insights are needed to refine tumor classification and to identify therapeutic approaches. We evaluated five gangliogliomas with single nucleus RNA-seq, cellular indexing of transcriptomes and epitopes by sequencing, and/or spatially-resolved RNA-seq. We uncovered a population of CD34+ neoplastic cells with mixed neuroectodermal, immature astrocyte, and neuronal markers. Gene regulatory network interrogation in these neuroectoderm-like cells revealed control of transcriptional programming by TCF7L2/MEIS1-PAX6 and SOX2, similar to that found during neuroectodermal/neural development. Developmental trajectory analyses place neuroectoderm-like tumor cells as precursor cells that give rise to neuron-like and macroglia-like neoplastic cells. Spatially-resolved transcriptomics revealed a neuroectoderm-like tumor cell niche with relative lack of vascular and immune cells. We used these high resolution results to deconvolute clinically-annotated transcriptomic data, confirming that CD34+ cell-associated gene programs associate with gangliogliomas compared to other glial brain tumors. Together, these deep transcriptomic approaches characterized a ganglioglioma cellular hierarchy-confirming CD34+ neuroectoderm-like tumor precursor cells, controlling transcription programs, cell signaling, and associated immune cell states. These findings may guide tumor classification, diagnosis, prognostication, and therapeutic investigations.


Subject(s)
Brain Neoplasms , Ganglioglioma , Neural Stem Cells , Child , Humans , Ganglioglioma/pathology , Transcriptome , Neural Plate/pathology , Neural Stem Cells/pathology , Brain Neoplasms/pathology
2.
Hum Mol Genet ; 29(2): 305-319, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31813957

ABSTRACT

Kabuki syndrome is an autosomal dominant developmental disorder with high similarities to CHARGE syndrome. It is characterized by a typical facial gestalt in combination with short stature, intellectual disability, skeletal findings and additional features like cardiac and urogenital malformations, cleft palate, hearing loss and ophthalmological anomalies. The major cause of Kabuki syndrome are mutations in KMT2D, a gene encoding a histone H3 lysine 4 (H3K4) methyltransferase belonging to the group of chromatin modifiers. Here we provide evidence that Kabuki syndrome is a neurocrestopathy, by showing that Kmt2d loss-of-function inhibits specific steps of neural crest (NC) development. Using the Xenopus model system, we find that Kmt2d loss-of-function recapitulates major features of Kabuki syndrome including severe craniofacial malformations. A detailed marker analysis revealed defects in NC formation as well as migration. Transplantation experiments confirm that Kmt2d function is required in NC cells. Furthermore, analyzing in vivo and in vitro NC migration behavior demonstrates that Kmt2d is necessary for cell dispersion but not protrusion formation of migrating NC cells. Importantly, Kmt2d knockdown correlates with a decrease in H3K4 monomethylation and H3K27 acetylation supporting a role of Kmt2d in the transcriptional activation of target genes. Consistently, using a candidate approach, we find that Kmt2d loss-of-function inhibits Xenopus Sema3F expression, and overexpression of Sema3F can partially rescue Kmt2d loss-of-function defects. Taken together, our data reveal novel functions of Kmt2d in multiple steps of NC development and support the hypothesis that major features of Kabuki syndrome are caused by defects in NC development.


Subject(s)
Abnormalities, Multiple/enzymology , Face/abnormalities , Hematologic Diseases/enzymology , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Neural Crest/metabolism , Vestibular Diseases/enzymology , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Acetylation , Animals , Cell Movement/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Face/pathology , Hematologic Diseases/genetics , Hematologic Diseases/metabolism , Hematologic Diseases/pathology , Histones/metabolism , Loss of Function Mutation , Methylation , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neural Crest/enzymology , Neural Crest/pathology , Neural Plate/growth & development , Neural Plate/metabolism , Neural Plate/pathology , Semaphorins/genetics , Semaphorins/metabolism , Vestibular Diseases/genetics , Vestibular Diseases/metabolism , Vestibular Diseases/pathology , Xenopus/embryology , Xenopus/genetics , Xenopus/metabolism , Xenopus Proteins/physiology
3.
J Comp Pathol ; 172: 107-109, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31690407

ABSTRACT

A juvenile female Moroccan uromastyx (Uromastyx acanthinurus nigriventris) that died unexpectedly was necropsied. Necropsy examination revealed minimal intracoelomic fat, small numbers of intestinal nematodes and intraocular masses within the vitreous chamber of both eyes. One of the intraocular masses was focally contiguous with the optic nerve and composed of neuroparenchyma with rare glial cells, consistent with a diagnosis of neural heterotopia. This condition is considered a neuroectodermal malformation, readily recognized in human medicine but rarely reported in animals. To the authors' knowledge, this is the first case of intraocular neural heterotopia reported in a reptile.


Subject(s)
Choristoma/veterinary , Lizards , Nervous System Malformations/veterinary , Optic Nerve/pathology , Animals , Eye/pathology , Female , Neural Plate/growth & development , Neural Plate/pathology
4.
J Cell Physiol ; 234(11): 19511-19522, 2019 11.
Article in English | MEDLINE | ID: mdl-30950033

ABSTRACT

Mitochondrial disorders (MDs) arise as a result of a respiratory chain dysfunction. While some MDs can affect a single organ, many involve several organs, the brain being the most affected, followed by heart and/or muscle. Many of these diseases are associated with heteroplasmic mutations in the mitochondrial DNA (mtDNA). The proportion of mutated mtDNA must exceed a critical threshold to produce disease. Therefore, understanding how embryonic development determines the heteroplasmy level in each tissue could explain the organ susceptibility and the clinical heterogeneity observed in these patients. In this report, the dynamics of heteroplasmy and the influence in cardiac commitment of the mutational load of the m.13513G>A mutation has been analyzed. This mutation has been reported as a frequent cause of Leigh syndrome (LS) and is commonly associated with cardiac problems. In this report, induced pluripotent stem cell (iPSc) technology has been used to delve into the molecular mechanisms underlying cardiac disease in LS. When mutation m.13513G>A is above a threshold, iPSc-derived cardiomyocytes (iPSc-CMs) could not be obtained due to an inefficient epithelial-mesenchymal transition. Surprisingly, these cells are redirected toward neuroectodermal lineages that would give rise to the brain. However, when mutation is below that threshold, dysfunctional CM are generated in a mutant-load dependent way. We suggest that distribution of the m.13513G>A mutation during cardiac differentiation is not at random. We propose a possible explanation of why neuropathology is a frequent feature of MD, but cardiac involvement is not always present.


Subject(s)
DNA, Mitochondrial/genetics , Electron Transport/genetics , Heart Diseases/genetics , Leigh Disease/genetics , Mitochondrial Diseases/genetics , Cell Differentiation/genetics , Electron Transport Complex I/genetics , Embryonic Development/genetics , Epithelial-Mesenchymal Transition/genetics , Heart Diseases/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Leigh Disease/pathology , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/genetics , Mutation , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Neural Plate/growth & development , Neural Plate/pathology , Phenotype
5.
Development ; 145(9)2018 05 08.
Article in English | MEDLINE | ID: mdl-29636380

ABSTRACT

The last stage of neural tube (NT) formation involves closure of the caudal neural plate (NP), an embryonic structure formed by neuromesodermal progenitors and newly differentiated cells that becomes incorporated into the NT. Here, we show in mouse that, as cell specification progresses, neuromesodermal progenitors and their progeny undergo significant changes in shape prior to their incorporation into the NT. The caudo-rostral progression towards differentiation is coupled to a gradual reliance on a unique combination of complex mechanisms that drive tissue folding, involving pulses of apical actomyosin contraction and planar polarised cell rearrangements, all of which are regulated by the Wnt-PCP pathway. Indeed, when this pathway is disrupted, either chemically or genetically, the polarisation and morphology of cells within the entire caudal NP is disturbed, producing delays in NT closure. The most severe disruptions of this pathway prevent caudal NT closure and result in spina bifida. In addition, a decrease in Vangl2 gene dosage also appears to promote more rapid progression towards a neural fate, but not the specification of more neural cells.


Subject(s)
Cell Differentiation , Neural Plate/embryology , Neural Stem Cells/metabolism , Neural Tube/embryology , Wnt Signaling Pathway , Animals , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Plate/pathology , Neural Stem Cells/pathology , Neural Tube/pathology , Spinal Dysraphism/epidemiology , Spinal Dysraphism/genetics , Spinal Dysraphism/pathology
6.
PLoS One ; 11(12): e0167573, 2016.
Article in English | MEDLINE | ID: mdl-27907123

ABSTRACT

Charcot-Marie-Tooth disease type 2A (CMT2A), the most common axonal form of hereditary sensory motor neuropathy, is caused by mutations of mitofusin-2 (MFN2). Mitofusin-2 is a GTPase required for fusion of mitochondrial outer membranes, repair of damaged mitochondria, efficient mitochondrial energetics, regulation of mitochondrial-endoplasmic reticulum calcium coupling and axonal transport of mitochondria. We knocked T105M MFN2 preceded by a loxP-flanked STOP sequence into the mouse Rosa26 locus to permit cell type-specific expression of this pathogenic allele. Crossing these mice with nestin-Cre transgenic mice elicited T105M MFN2 expression in neuroectoderm, and resulted in diminished numbers of mitochondria in peripheral nerve axons, an alteration in skeletal muscle fiber type distribution, and a gait abnormality.


Subject(s)
Alleles , Charcot-Marie-Tooth Disease/genetics , GTP Phosphohydrolases/genetics , Hemizygote , Muscle Fibers, Skeletal/metabolism , Peripheral Nerves/metabolism , Amino Acid Substitution , Animals , Axonal Transport , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Disease Models, Animal , Embryo, Mammalian , GTP Phosphohydrolases/deficiency , Gait , Gene Expression Regulation , Hindlimb/pathology , Humans , Integrases/genetics , Integrases/metabolism , Mice , Mice, Transgenic , Mitochondria/metabolism , Mitochondria/pathology , Muscle Fibers, Skeletal/pathology , Nestin/genetics , Nestin/metabolism , Neural Plate/metabolism , Neural Plate/pathology , Peripheral Nerves/pathology , Phenotype , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
7.
Dev Biol ; 415(2): 371-382, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26874011

ABSTRACT

Mandibulofacial dysostosis (MFD) is a human developmental disorder characterized by defects of the facial bones. It is the second most frequent craniofacial malformation after cleft lip and palate. Nager syndrome combines many features of MFD with a variety of limb defects. Mutations in SF3B4 (splicing factor 3b, subunit 4) gene, which encodes a component of the pre-mRNA spliceosomal complex, were recently identified as a cause of Nager syndrome, accounting for 60% of affected individuals. Nothing is known about the cellular pathogenesis underlying Nager type MFD. Here we describe the first animal model for Nager syndrome, generated by knocking down Sf3b4 function in Xenopus laevis embryos, using morpholino antisense oligonucleotides. Our results indicate that Sf3b4-depleted embryos show reduced expression of the neural crest genes sox10, snail2 and twist at the neural plate border, associated with a broadening of the neural plate. This phenotype can be rescued by injection of wild-type human SF3B4 mRNA but not by mRNAs carrying mutations that cause Nager syndrome. At the tailbud stage, morphant embryos had decreased sox10 and tfap2a expression in the pharyngeal arches, indicative of a reduced number of neural crest cells. Later in development, Sf3b4-depleted tadpoles exhibited hypoplasia of neural crest-derived craniofacial cartilages, phenocopying aspects of the craniofacial skeletal defects seen in Nager syndrome patients. With this animal model we are now poised to gain important insights into the etiology and pathogenesis of Nager type MFD, and to identify the molecular targets of Sf3b4.


Subject(s)
Disease Models, Animal , Gene Expression Regulation, Developmental/genetics , Mandibulofacial Dysostosis/genetics , Maxillofacial Development/genetics , RNA Splicing Factors/genetics , Xenopus Proteins/deficiency , Xenopus laevis/genetics , Amino Acid Sequence , Animals , Branchial Region/embryology , Branchial Region/metabolism , Branchial Region/pathology , Cartilage/growth & development , Cartilage/metabolism , Cartilage/pathology , Codon, Nonsense , Frameshift Mutation , Gene Knockdown Techniques , Genetic Complementation Test , Humans , Mandibulofacial Dysostosis/embryology , Mandibulofacial Dysostosis/physiopathology , Molecular Sequence Data , Morpholinos/pharmacology , Neural Crest/cytology , Neural Crest/embryology , Neural Crest/metabolism , Neural Plate/embryology , Neural Plate/pathology , Phenotype , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Skull/abnormalities , Skull/embryology , Skull/growth & development , Xenopus Proteins/biosynthesis , Xenopus Proteins/genetics , Xenopus Proteins/physiology , Xenopus laevis/embryology , Xenopus laevis/growth & development
8.
Sci Rep ; 5: 16917, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26593875

ABSTRACT

Failure to close the neural tube results in birth defects, with severity ranging from spina bifida to lethal anencephaly. Few genetic risk factors for neural tube defects are known in humans, highlighting the critical role of environmental risk factors, such as maternal diabetes. Yet, it is not well understood how altered maternal metabolism interferes with embryonic development, and with neurulation in particular. We present evidence from two independent mouse models of diabetic pregnancy that identifies impaired migration of nascent mesodermal cells in the primitive streak as the morphogenetic basis underlying the pathogenesis of neural tube defects. We conclude that perturbed gastrulation not only explains the neurulation defects, but also provides a unifying etiology for the broad spectrum of congenital malformations in diabetic pregnancies.


Subject(s)
Diabetes, Gestational/genetics , Nerve Tissue Proteins/genetics , Neural Plate/metabolism , Neural Tube Defects/genetics , Animals , Diabetes, Gestational/metabolism , Diabetes, Gestational/pathology , Disease Models, Animal , Embryo, Mammalian , Female , Gastrulation/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , High-Throughput Nucleotide Sequencing , Humans , Laser Capture Microdissection , Mice , Mice, Inbred NOD , Nerve Tissue Proteins/metabolism , Neural Plate/embryology , Neural Plate/pathology , Neural Tube Defects/embryology , Neural Tube Defects/metabolism , Neural Tube Defects/pathology , Pregnancy
9.
Mol Brain ; 7: 67, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-25223405

ABSTRACT

BACKGROUND: Alcohol is detrimental to early development. Fetal alcohol spectrum disorders (FASD) due to maternal alcohol abuse results in a series of developmental abnormalities including cranial facial dysmorphology, ocular anomalies, congenital heart defects, microcephaly and intellectual disabilities. Previous studies have been shown that ethanol exposure causes neural crest (NC) apoptosis and perturbation of neural crest migration. However, the underlying mechanism remains elusive. In this report we investigated the fetal effect of alcohol on the process of neural crest development in the Xenopus leavis. RESULTS: Pre-gastrulation exposure of 2-4% alcohol induces apoptosis in Xenopus embryo whereas 1% alcohol specifically impairs neural crest migration without observing discernible apoptosis. Additionally, 1% alcohol treatment considerably increased the phenotype of small head (43.4% ± 4.4%, total embryo n = 234), and 1.5% and 2.0% dramatically augment the deformation to 81.2% ± 6.5% (n = 205) and 91.6% ± 3.0% (n = 235), respectively (P < 0.05). Significant accumulation of Homocysteine was caused by alcohol treatment in embryos and 5-mehtyltetrahydrofolate restores neural crest migration and alleviates homocysteine accumulation, resulting in inhibition of the alcohol-induced neurocristopathies. CONCLUSIONS: Our study demonstrates that prenatal alcohol exposure causes neural crest cell migration abnormality and 5-mehtyltetrahydrofolate could be beneficial for treating FASD.


Subject(s)
Cell Movement/drug effects , Ethanol/toxicity , Neural Crest/pathology , Tetrahydrofolates/pharmacology , Animals , Apoptosis/drug effects , Cartilage/drug effects , Cartilage/embryology , Models, Animal , Neural Crest/drug effects , Neural Plate/drug effects , Neural Plate/pathology , Pigments, Biological/metabolism , Xenopus laevis/embryology
11.
Dev Med Child Neurol ; 56(12): 1207-1211, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24954233

ABSTRACT

AIM: Patients with tuberous sclerosis complex (TSC) with brain involvement usually have both tubers and subependymal nodules (SENs) and the occurrence of one lesion without the other seems to be rare. The aim of this study was to assess the specific clinical manifestations and genotype of patients with one type of lesion or the other but not both. METHOD: The magnetic resonance images of 220 patients with TSC were reviewed, and patients with either tubers or SENs, but not both, were identified. RESULTS: Out of a total of 220 patients (95 males, 125 females; mean age 22y 9mo, range 9mo-81y), six (3%) had tubers without SENs (two males; four females; mean age 34y 10mo, range 11-48y); however, no patients with SENs and without tubers were identified. No mutation was identified (NMI) in any of the six patients who underwent mutational analysis of TSC1 and TSC2. Five of the six patients had three or fewer tubers. INTERPRETATION: We found no patients with SENs but without tubers among our cohort. In all patients with tubers but without SENs, mutational studies of TSC1/TSC2 were negative, and the majority of these had three or fewer tubers. A possible mechanism for patients with NMI and an absence of SENs is a mosaicism with a first postzygotic mutation in the neuroectoderm.


Subject(s)
Brain/pathology , Mosaicism , Neural Plate/pathology , Tuberous Sclerosis/genetics , Tuberous Sclerosis/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Humans , Infant , Magnetic Resonance Imaging , Male , Middle Aged , Mutation/genetics , Retrospective Studies , Young Adult
12.
Reprod Toxicol ; 48: 88-97, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24887031

ABSTRACT

Malformations of the facial midline are a consistent feature among individuals with defects in primary cilia. Here, we provide a framework in which to consider how these primary cilia-dependent facial anomalies occur. We generated mice in which the intraflagellar transport protein Kif3a was deleted in cranial neural crest cells. The Kif3a phenotypes included isolated metopic craniosynostosis, delayed closure of the anterior fontanelles, and hydrocephalus, as well as midline facial anomalies including hypertelorism, cleft palate, and bifid nasal septum. Although all cranial neural crest cells had truncated primary cilia as a result of the conditional deletion, only those in the midline showed evidence of hyper-proliferation and ectopic Wnt responsiveness. Thus, cranial neural crest cells do not rely on primary cilia for their migration but once established in the facial prominences, midline cranial neural crest cells require Kif3a function in order to integrate and respond to Wnt signals from the surrounding epithelia.


Subject(s)
Cilia/physiology , Cleft Palate/genetics , Craniosynostoses/genetics , Hydrocephalus/genetics , Hypertelorism/genetics , Kinesins/genetics , Animals , Cranial Fontanelles/abnormalities , Mice, Transgenic , Mutation , Neural Crest/metabolism , Neural Crest/pathology , Neural Plate/metabolism , Neural Plate/pathology , Phenotype , Wnt Signaling Pathway
13.
Article in English | MEDLINE | ID: mdl-24902834

ABSTRACT

The neural tube (NT), the embryonic precursor of the vertebrate brain and spinal cord, is generated by a complex and highly dynamic morphological process. In mammals, the initially flat neural plate bends and lifts bilaterally to generate the neural folds followed by fusion of the folds at the midline during the process of neural tube closure (NTC). Failures in any step of this process can lead to neural tube defects (NTDs), a common class of birth defects that occur in approximately 1 in 1000 live births. These severe birth abnormalities include spina bifida, a failure of closure at the spinal level; craniorachischisis, a failure of NTC along the entire body axis; and exencephaly, a failure of the cranial neural folds to close which leads to degeneration of the exposed brain tissue termed anencephaly. The mouse embryo presents excellent opportunities to explore the genetic basis of NTC in mammals; however, its in utero development has also presented great challenges in generating a deeper understanding of how gene function regulates the cell and tissue behaviors that drive this highly dynamic process. Recent technological advances are now allowing researchers to address these questions through visualization of NTC dynamics in the mouse embryo in real time, thus offering new insights into the morphogenesis of mammalian NTC.


Subject(s)
Morphogenesis , Neural Crest/growth & development , Neural Plate/growth & development , Neural Tube/growth & development , Anencephaly/genetics , Anencephaly/pathology , Animals , Mice , Neural Plate/pathology , Neural Tube/pathology , Neural Tube Defects/genetics , Neural Tube Defects/pathology , Spinal Dysraphism/genetics , Spinal Dysraphism/pathology
14.
Cell Stem Cell ; 14(1): 27-39, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24268696

ABSTRACT

Although somatic cell nuclear transfer (SCNT) and induction of pluripotency (to form iPSCs) are both recognized reprogramming methods, there has been relatively little comparative analysis of the resulting pluripotent cells. Here, we examine the capacity of these two reprogramming approaches to rejuvenate telomeres using late-generation telomerase-deficient (Terc(-/-)) mice that exhibit telomere dysfunction and premature aging. We found that embryonic stem cells established from Terc(-/-) SCNT embryos (Terc(-/-) ntESCs) have greater differentiation potential and self-renewal capacity than Terc(-/-) iPSCs. Remarkably, SCNT results in extensive telomere lengthening in cloned embryos and improved telomere capping function in the established Terc(-/-) ntESCs. In addition, mitochondrial function is severely impaired in Terc(-/-) iPSCs and their differentiated derivatives but significantly improved in Terc(-/-) ntESCs. Thus, our results suggest that SCNT-mediated reprogramming mitigates telomere dysfunction and mitochondrial defects to a greater extent than iPSC-based reprogramming. Understanding the basis of this differential could help optimize reprogramming strategies.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Nuclear Transfer Techniques , RNA/physiology , Telomerase/physiology , Telomere/genetics , Adenosine Triphosphate/metabolism , Animals , Cell Proliferation , Cells, Cultured , Embryonic Stem Cells/metabolism , In Situ Hybridization, Fluorescence , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondria/pathology , Neural Plate/metabolism , Neural Plate/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
15.
Curr Med Chem ; 21(8): 1017-25, 2014.
Article in English | MEDLINE | ID: mdl-23992320

ABSTRACT

Advanced enteropancreatic (EP) neuroendocrine tumors (NETs) can be treated with several different therapies, including chemotherapy, biotherapy, and locoregional treatments. Over the last few decades, impressive progress has been made in the biotherapy field. Three main druggable molecular targets have been studied and developed in terms of therapy: somatostatin receptor (sstr), mammalian target of rapamycin (mTOR), and angiogenic factors. In particular, research has moved from the old somatostatin analogs (SSAs), such as octreotide (OCT) and lanreotide (LAN), specifically binding to the sstr-2, to the newer pasireotide (PAS), which presents a wider sstr spectrum. Over the last ten years, several molecular targeted agents (MTAs) have been studied in phase II trials, and very few of them have reached phase III. The mTOR inhibitor everolimus and the multitargeted inhibitor sunitinib have been approved for clinical use by the FDA and EMA in advanced well/moderately-differentiated (WD, MD) progressive pancreatic neuroendocrine tumors (PNETs), on the basis of the positive results of two international large randomized phase III trials vs. placebo. Bevacizumab has been studied in a large US phase III trial vs. interferon (IFN)-alfa2b, and results are pending. In this review, the biological and clinical aspects of MTAs introduced into clinical practice or which are currently in an advanced phase of clinical investigation are addressed.


Subject(s)
Antineoplastic Agents/therapeutic use , Molecular Targeted Therapy/methods , Neural Plate/pathology , Neuroendocrine Tumors/drug therapy , Pancreas/pathology , Pancreatic Neoplasms/drug therapy , Animals , Humans , Neural Plate/drug effects , Neural Plate/metabolism , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Pancreas/drug effects , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology
16.
J Oral Pathol Med ; 42(6): 450-3, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23336292

ABSTRACT

BACKGROUND: The glucose transporter type 1 (GLUT-1) protein is a useful marker for perineurial cells. Because of the possible neuroectodermal histogenesis of the granular cell tumour and congenital granular cell epulis, the aim of this study was to assess the immunoexpression of GLUT-1 protein in granular cell tumour and congenital granular cell epulis to aid in clarifying their histogenesis. METHODS: The protocol of this study was approved by the Committee of Bioethics in Research at Universidade Federal Minas Gerais. Six cases of granular cell tumour and three cases of congenital granular cell epulis were submitted to immunohistochemistry for GLUT-1 and S-100 using the streptavidin-biotin standard protocol. RESULTS: Five cases of granular cell tumour were located on the tongue and one case on the upper lip. All cases of congenital granular cell epulis were observed in the alveolar ridge of newborns. All lesions evaluated proved to be immunonegative for GLUT-1. S-100 was found to be positive in all granular cell tumours and negative in congenital granular cell epulis. CONCLUSIONS: Neither granular cell tumour nor congenital granular cell epulis is directly related to perineurial cells.


Subject(s)
Gingival Neoplasms/congenital , Glucose Transporter Type 1/analysis , Granular Cell Tumor/pathology , Adult , Female , Gingival Neoplasms/pathology , Humans , Immunohistochemistry , Infant, Newborn , Lip Neoplasms/pathology , Male , Middle Aged , Neural Plate/pathology , Peripheral Nerves/pathology , S100 Proteins/analysis , Tongue Neoplasms/pathology
17.
Hum Mol Genet ; 21(18): 4104-14, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22723015

ABSTRACT

Exposure to the antiepileptic drug valproic acid (VPA) during gestation causes neurofunctional and anatomic deficits in later life. At present, there are little human data on how early neural development is affected by chemicals. We used human embryonic stem cells, differentiating to neuroectodermal precursors, as a model to investigate the modes of action of VPA. Microarray expression profiling, qPCR of specific marker genes, immunostaining and the expression of green fluorescent protein under the control of the promoter of the canonical neural precursor cell marker HES5 were used as readouts. Exposure to VPA resulted in distorted marker gene expression, characterized by a relative increase in NANOG and OCT4 and a reduction in PAX6. A similar response pattern was observed with trichostatin A, a potent and specific histone deacetylase inhibitor (HDACi), but not with several other toxicants. Differentiation markers were disturbed by prolonged, but not by acute treatment with HDACi, and the strongest disturbance of differentiation was observed by toxicant exposure during early neural fate decision. The increased acetylation of histones observed in the presence of HDACi may explain the up-regulation of some genes. However, to understand the down-regulation of PAX6 and the overall complex transcript changes, we examined further epigenetic markers. Alterations in the methylation of lysines 4 and 27 of histone H3 were detected in the promoter region of PAX6 and OCT4. The changes in these activating and silencing histone marks provide a more general mechanistic rational for the regulation of developmentally important genes at non-cytotoxic drug concentrations.


Subject(s)
Abnormalities, Drug-Induced/genetics , Embryonic Stem Cells/metabolism , Epigenesis, Genetic/drug effects , Neural Plate/embryology , Abnormalities, Drug-Induced/pathology , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/drug effects , Cells, Cultured , Embryonic Stem Cells/physiology , Eye Proteins/genetics , Eye Proteins/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hydroxamic Acids/pharmacology , Methylation , Nanog Homeobox Protein , Neural Plate/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Neuroepithelial Cells/metabolism , Neuroepithelial Cells/physiology , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Oligonucleotide Array Sequence Analysis , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Principal Component Analysis , Promoter Regions, Genetic , Protein Processing, Post-Translational , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription, Genetic , Transcriptome , Valproic Acid/adverse effects
18.
Cytotherapy ; 12(4): 491-504, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20331410

ABSTRACT

BACKGROUND AIMS: In recent years, bone marrow (BM)-derived mesenchymal stromal cells (MSC) have become a promising source for neuroregenerative therapies. We evaluated the trophic effects of neuroectodermally converted MSC (mNSC) on neural stem cells (NSC). METHODS: We quantified the expression of growth factors by mNSC using real-time reverse transcription-polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) and studied the effects of mNSC conditioned medium and mNSC (in direct co-culture) on NSC proliferation, differentiation and survival. RESULTS: Neuroectodermal conversion of human MSC induced high expression of growth factors at both mRNA and protein levels, most prominently hepatocyte growth factor, vascular endothelial growth factor and amphiregulin (37 +/- 17, 92 +/- 44 and 12 +/- 11 ng/10(5) cells, respectively), which remained at high levels upon co-culturing with neural cells. Accordingly, mNSC conditioned medium and co-cultivation with mNSC reduced cell death of NSC (36% of control), stimulated their proliferation, attenuated glial differentiation of NSC (7 +/- 3 versus 59 +/- 6%; P < 0.01) and protected NSC against the neurotoxin 6-hydroxydopamine (with half-maximally concentrations EC(50) values of 217 +/- 207 microM in the presence of mNSC compared with 62 +/- 49 microM for NSC alone). CONCLUSIONS: mNSC promote survival and proliferation, and inhibit glial differentiation, of NSC. Protection of NSC by mNSC against 6-hydroxy-dopamine is probably mediated by the release of cytotrophic factors. Our results promote neuroectodermally converted MSC as promising candidate cells for the development of neuroregenerative and neuroprotective therapies.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells/metabolism , Neural Plate/metabolism , Neurons/metabolism , Stromal Cells/metabolism , Adolescent , Adult , Animals , Apoptosis/drug effects , Cell Communication , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Proliferation/drug effects , Coculture Techniques , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Cytoprotection/genetics , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/pathology , Mice , Middle Aged , Neural Plate/pathology , Neuroglia/metabolism , Neuroglia/pathology , Neurons/drug effects , Neurons/pathology , Oxidopamine/toxicity , Rats , Stromal Cells/drug effects , Stromal Cells/pathology
19.
Proc Natl Acad Sci U S A ; 106(13): 5324-9, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19289832

ABSTRACT

Ewing tumors (ET) are highly malignant, localized in bone or soft tissue, and are molecularly defined by ews/ets translocations. DNA microarray analysis revealed a relationship of ET to both endothelium and fetal neural crest. We identified expression of histone methyltransferase enhancer of Zeste, Drosophila, Homolog 2 (EZH2) to be increased in ET. Suppressive activity of EZH2 maintains stemness in normal and malignant cells. Here, we found EWS/FLI1 bound to the EZH2 promoter in vivo, and induced EZH2 expression in ET and mesenchymal stem cells. Down-regulation of EZH2 by RNA interference in ET suppressed oncogenic transformation by inhibiting clonogenicity in vitro. Similarly, tumor development and metastasis was suppressed in immunodeficient Rag2(-/-)gamma(C)(-/-) mice. EZH2-mediated gene silencing was shown to be dependent on histone deacetylase (HDAC) activity. Subsequent microarray analysis of EZH2 knock down, HDAC-inhibitor treatment and confirmation in independent assays revealed an undifferentiated phenotype maintained by EZH2 in ET. EZH2 regulated stemness genes such as nerve growth factor receptor (NGFR), as well as genes involved in neuroectodermal and endothelial differentiation (EMP1, EPHB2, GFAP, and GAP43). These data suggest that EZH2 might have a central role in ET pathology by shaping the oncogenicity and stem cell phenotype of this tumor.


Subject(s)
DNA-Binding Proteins/physiology , Endothelial Cells/pathology , Neural Plate/pathology , Sarcoma, Ewing/etiology , Transcription Factors/physiology , Animals , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , Histone Deacetylases , Humans , Mesenchymal Stem Cells , Mice , Neoplasm Metastasis , Oncogene Proteins, Fusion , Polycomb Repressive Complex 2 , Proto-Oncogene Protein c-fli-1 , RNA-Binding Protein EWS , Sarcoma, Ewing/pathology
20.
Diagn Cytopathol ; 36(8): 595-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18618728

ABSTRACT

Immature ovarian teratoma (IOT) is a rare and aggressive malignant neoplasm characterized by immature neural tissue. The cytomorphologic features have only rarely been described. We herein describe an additional case and review the literature regarding this entity. To the best of our knowledge, this is the first reported case with imprint cytology. A 35-year-old woman presented with a pelvic mass which was resected and sent for frozen section evaluation. Imprint smears and frozen section of the mass were diagnostic of IOT. IOT has diagnostic cytologic features which show complete concordance with histology. Differential diagnoses include other small round cell neoplasms such as ovarian neuroblastoma, small cell carcinoma of hypercalcemic type, primitive neuroectodermal tumor, Wilm's tumor, desmoplastic small round cell tumor, and Non-Hodgkin lymphoma. Distinguishing IOT from these tumors can be challenging however if diligent morphologic study and/or ancillary studies are performed accurate diagnosis is possible.


Subject(s)
Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/pathology , Teratoma/diagnosis , Teratoma/pathology , Adult , Diagnosis, Differential , Female , Humans , Neural Plate/pathology
SELECTION OF CITATIONS
SEARCH DETAIL