Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Front Cell Dev Biol ; 11: 1203503, 2023.
Article En | MEDLINE | ID: mdl-37519304

Peripheral nervous system (PNS) sensory alterations are present in several pathologies and syndromes. The use of induced pluripotent stem cell (iPSC) technology is an important strategy to produce sensory neurons in patients who are accomplished in terms of sensory symptoms. The iPSC technology relies on manipulating signaling pathways to resemble what occurs in vivo, and the iPSCs are known to carry a transcriptional memory after reprogramming, which can affect the produced cell. To this date, protocols described for sensory neuron production start using iPSCs derived from skin fibroblasts, which have the same ontogenetic origin as the central nervous system (CNS). Since it is already known that the cells somehow resemble their origin even after cell reprogramming, PNS cells should be produced from cells derived from the neural crest. This work aimed to establish a protocol to differentiate sensory neurons derived from stem cells from human exfoliated deciduous teeth (SHED) with the same embryonic origin as the PNS. SHED-derived iPSCs were produced and submitted to peripheral sensory neuron (PSN) differentiation. Our protocol used the dual-SMAD inhibition method, followed by neuronal differentiation, using artificial neurotrophic factors and molecules produced by human keratinocytes. We successfully established the first protocol for differentiating neural crest and PNS cells from SHED-derived iPSCs, enabling future studies of PNS pathologies.

2.
Int J Mol Sci ; 23(18)2022 Sep 07.
Article En | MEDLINE | ID: mdl-36142200

Viral infections have always been a serious burden to public health, increasing morbidity and mortality rates worldwide. Zika virus (ZIKV) is a flavivirus transmitted by the Aedes aegypti vector and the causative agent of severe fetal neuropathogenesis and microcephaly. The virus crosses the placenta and reaches the fetal brain, mainly causing the death of neuronal precursor cells (NPCs), glial inflammation, and subsequent tissue damage. Genetic differences, mainly related to the antiviral immune response and cell death pathways greatly influence the susceptibility to infection. These components are modulated by many factors, including microRNAs (miRNAs). MiRNAs are small noncoding RNAs that regulate post-transcriptionally the overall gene expression, including genes for the neurodevelopment and the formation of neural circuits. In this context, we investigated the pathways and target genes of miRNAs modulated in NPCs infected with ZIKV. We observed downregulation of miR-302b, miR-302c and miR-194, whereas miR-30c was upregulated in ZIKV infected human NPCs in vitro. The analysis of a public dataset of ZIKV-infected human NPCs evidenced 262 upregulated and 3 downregulated genes, of which 142 were the target of the aforementioned miRNAs. Further, we confirmed a correlation between miRNA and target genes affecting pathways related to antiviral immune response, cell death and immune cells chemotaxis, all of which could contribute to the establishment of microcephaly and brain lesions. Here, we suggest that miRNAs target gene expression in infected NPCs, directly contributing to the pathogenesis of fetal microcephaly.


MicroRNAs , Microcephaly , Nervous System Malformations , Zika Virus Infection , Zika Virus , Animals , Antiviral Agents , Cell Death/genetics , Chemotaxis , Female , Humans , Immunity , MicroRNAs/genetics , Microcephaly/genetics , Mosquito Vectors , Pregnancy , Zika Virus/physiology
3.
Neuroimmunomodulation ; 28(4): 229-232, 2021.
Article En | MEDLINE | ID: mdl-34082423

INTRODUCTION: The aim of this case was to investigate the association of the Zika virus infection in utero with the autism spectrum disorder (ASD) as clinical outcome that presented no congenital anomalies. METHODS: ASD was diagnosed in the second year of life by different child neurologists and confirmed by DSM-5 and ASQ. After that, an extensive clinical, epidemiological, and genetic evaluations were performed, with main known ASD causes ruled out. RESULTS: An extensive laboratorial search was done, with normal findings. SNP array identified no pathogenic variants. Normal neuroimaging and EEG findings were also obtained. ZIKV (Zika virus) IgG was positive, while IgM was negative. Other congenital infections were negative. The exome sequencing did not reveal any pathogenic variant in genes related to ASD. CONCLUSION: Accordingly, this report firstly associates ZIKV exposure to ASD.


Autism Spectrum Disorder , Pregnancy Complications, Infectious , Zika Virus Infection , Zika Virus , Autism Spectrum Disorder/complications , Autism Spectrum Disorder/genetics , Child , Female , Humans , Pregnancy , Zika Virus/genetics , Zika Virus Infection/complications
4.
Adv Neurobiol ; 25: 207-218, 2020.
Article En | MEDLINE | ID: mdl-32578148

Neurodevelopmental disorders (ND) are characterized by an impairment of the nervous system during its development, with a wide variety of phenotypes based on genetic or environmental cues. There are currently several disorders grouped under ND including intellectual disabilities (ID), attention-deficit hyperactivity disorder (ADHD), and autism spectrum disorders (ASD). Although NDs can have multiple culprits with varied diagnostics, several NDs present an inflammatory component. Taking advantage of induced pluripotent stem cells (iPSC), several disorders were modeled in a dish complementing in vivo data from rodent models or clinical data. Monogenic syndromes displaying ND are more feasible to be modeled using iPSCs also due to the ability to recruit patients and clinical data available. Some of these genetic disorders are Fragile X Syndrome (FXS), Rett Syndrome (RTT), and Down Syndrome (DS). Environmental NDs can be caused by maternal immune activation (MIA), such as the infection with Zika virus during pregnancy known to cause neural damage to the fetus. Our goal in this chapter is to review the advances of using stem cell research in NDs, focusing on the role of neuroinflammation on ASD and environmental NDs studies.


Autism Spectrum Disorder , Fragile X Syndrome , Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Zika Virus Infection , Zika Virus , Fragile X Syndrome/genetics , Humans , Inflammation
5.
Anat Rec (Hoboken) ; 303(7): 1812-1820, 2020 07.
Article En | MEDLINE | ID: mdl-31520456

Central nervous system (CNS) trauma is often related to tissue loss, leading to partial or complete disruption of spinal cord function due to neuronal death. Although generally irreversible, traditional therapeutic efforts, such as physical therapy exercises, are generally recommended, but with a poor or reduced improvement of the microenvironment, which in turn stimulates neuroplasticity and neuroregeneration. Mesenchymal stem cells (MSCs) have paracrine, immunomodulatory, and anti-inflammatory effects. Here we use stem cells to see if they can promote not only physical but also the functional regeneration of neuronal tissue in dogs with CNS traumas. Two dogs, one with chronic spinal cord injury and one with subacute spinal cord injury, underwent infusion of autologous MSCs in association with physiotherapy. The two treatments in combination were able to partially or completely recover the dog's walking movement again. The treatment of MSCs in association with physical therapy improved the microenvironment, which could be evidence of a paradigm shift that the CNS is not capable of functional regeneration after aggressive traumas. Anat Rec, 2019. © 2019 American Association for Anatomy Anat Rec, 303:1812-1820, 2020. © 2019 American Association for Anatomy.


Mesenchymal Stem Cell Transplantation , Nerve Regeneration/physiology , Paraplegia/veterinary , Recovery of Function/physiology , Spinal Cord Injuries/veterinary , Animals , Dogs , Paraplegia/etiology , Paraplegia/therapy , Spinal Cord Injuries/complications , Spinal Cord Injuries/therapy , Therapeutics
6.
Front Psychiatry ; 10: 409, 2019.
Article En | MEDLINE | ID: mdl-31231258

Several efforts in basic and clinical research have been contributing to unveiling the genetics behind autism spectrum disorders (ASD). However, despite these advancements, many individuals diagnosed with ASD and related neuropsychiatric conditions have been genetically investigated without elucidative results. The enormous genetic complexity of ASD-related conditions makes it a significant challenge to achieve, with a growing number of genes (close to a thousand) involved, belonging to different molecular pathways and presenting distinct genetic variations. Next-generation sequencing (NGS) is the approach most used in genetic research related to ASD, identifying de novo mutation, which is closely related to more severe clinical phenotypes, especially when they affect constrained and loss-of-function intolerant genes. On the other hand, de novo mutation findings contribute to a small percentage of the ASD population, since most of the cases and genetic variants associated with neuropsychiatric conditions are inherited and phenotypes are results of additive polygenic models, which makes statistical efforts more difficult. As a result, NGS investigation can sound vainly or unsuccessful, and new mutations on genes already related with ASD are classified as variants of unknown significance (VUS), hampering their endorsement to a clinical phenotype. This review is focused on currently available strategies to clarify the impact of VUS and to describe the efforts to identify more pieces of evidence throughout clinical interpretation and genetic curation process.

8.
Sci Rep ; 8(1): 1218, 2018 01 19.
Article En | MEDLINE | ID: mdl-29352135

The outbreak of the Zika virus (ZIKV) has been associated with increased incidence of congenital malformations. Although recent efforts have focused on vaccine development, treatments for infected individuals are needed urgently. Sofosbuvir (SOF), an FDA-approved nucleotide analog inhibitor of the Hepatitis C (HCV) RNA-dependent RNA polymerase (RdRp) was recently shown to be protective against ZIKV both in vitro and in vivo. Here, we show that SOF protected human neural progenitor cells (NPC) and 3D neurospheres from ZIKV infection-mediated cell death and importantly restored the antiviral immune response in NPCs. In vivo, SOF treatment post-infection (p.i.) decreased viral burden in an immunodeficient mouse model. Finally, we show for the first time that acute SOF treatment of pregnant dams p.i. was well-tolerated and prevented vertical transmission of the virus to the fetus. Taken together, our data confirmed SOF-mediated sparing of human neural cell types from ZIKV-mediated cell death in vitro and reduced viral burden in vivo in animal models of chronic infection and vertical transmission, strengthening the growing body of evidence for SOF anti-ZIKV activity.

9.
Front Pediatr ; 6: 394, 2018.
Article En | MEDLINE | ID: mdl-30619789

Recent reports show an increase in the incidence of Autism Spectrum Disorders (ASD) to 1 in every 59 children up to 8 years old in 11 states in North America. Induced pluripotent stem cell (iPSC) technology offers a groundbreaking platform for the study of polygenic neurodevelopmental disorders in live cells. Robust inflammation states and immune system dysfunctions are associated with ASD and several cell types participate on triggering and sustaining these processes. In this review, we will examine the contribution of neuroinflammation to the development of autistic features and discuss potential therapeutic approaches. We will review the available tools, emphasizing stem cell modeling as a technology to investigate the various molecular pathways and different cell types involved in the process of neuroinflammation in ASD.

10.
Hum Mol Genet ; 27(1): 41-52, 2018 01 01.
Article En | MEDLINE | ID: mdl-29048558

Although Zika virus (ZIKV) infection is often asymptomatic, in some cases, it can lead to birth defects in newborns or serious neurologic complications in adults. However, little is known about the interplay between immune and neural cells that could contribute to the ZIKV pathology. To understand the mechanisms at play during infection and the antiviral immune response, we focused on neural precursor cells (NPCs)-microglia interactions. Our data indicate that human microglia infected with the current circulating Brazilian ZIKV induces a similar pro-inflammatory response found in ZIKV-infected human tissues. Importantly, using our model, we show that microglia interact with ZIKV-infected NPCs and further spread the virus. Finally, we show that Sofosbuvir, an FDA-approved drug for Hepatitis C, blocked viral infection in NPCs and therefore the transmission of the virus from microglia to NPCs. Thus, our model provides a new tool for studying neuro-immune interactions and a platform to test new therapeutic drugs.


Zika Virus Infection/immunology , Zika Virus/pathogenicity , Cell Line , Humans , Induced Pluripotent Stem Cells/pathology , Microglia/pathology , Models, Biological , Neural Stem Cells/pathology , Sofosbuvir/pharmacology , Zika Virus/metabolism
11.
Brain Res ; 1656: 49-54, 2017 02 01.
Article En | MEDLINE | ID: mdl-26854137

Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders characterized by impaired social communication and interactions and by restricted and repetitive behaviors. Although ASD is suspected to have a heritable or sporadic genetic basis, its underlying etiology and pathogenesis are not well understood. Therefore, viable human neurons and glial cells produced using induced pluripotent stem cells (iPSC) to reprogram cells from individuals affected with ASD provide an unprecedented opportunity to elucidate the pathophysiology of these disorders, providing novel insights regarding ASD and a potential platform to develop and test therapeutic compounds. Herein, we discuss the state of art with regards to ASD modeling, including limitations of this technology, as well as potential future directions. This article is part of a Special Issue entitled SI: Exploiting human neurons.


Autism Spectrum Disorder/physiopathology , Neurons/physiology , Animals , Autism Spectrum Disorder/drug therapy , Humans
12.
Nature ; 534(7606): 267-71, 2016 06 09.
Article En | MEDLINE | ID: mdl-27279226

Zika virus (ZIKV) is an arbovirus belonging to the genus Flavivirus (family Flaviviridae) and was first described in 1947 in Uganda following blood analyses of sentinel Rhesus monkeys. Until the twentieth century, the African and Asian lineages of the virus did not cause meaningful infections in humans. However, in 2007, vectored by Aedes aegypti mosquitoes, ZIKV caused the first noteworthy epidemic on the Yap Island in Micronesia. Patients experienced fever, skin rash, arthralgia and conjunctivitis. From 2013 to 2015, the Asian lineage of the virus caused further massive outbreaks in New Caledonia and French Polynesia. In 2013, ZIKV reached Brazil, later spreading to other countries in South and Central America. In Brazil, the virus has been linked to congenital malformations, including microcephaly and other severe neurological diseases, such as Guillain-Barré syndrome. Despite clinical evidence, direct experimental proof showing that the Brazilian ZIKV (ZIKV(BR)) strain causes birth defects remains absent. Here we demonstrate that ZIKV(BR) infects fetuses, causing intrauterine growth restriction, including signs of microcephaly, in mice. Moreover, the virus infects human cortical progenitor cells, leading to an increase in cell death. We also report that the infection of human brain organoids results in a reduction of proliferative zones and disrupted cortical layers. These results indicate that ZIKV(BR) crosses the placenta and causes microcephaly by targeting cortical progenitor cells, inducing cell death by apoptosis and autophagy, and impairing neurodevelopment. Our data reinforce the growing body of evidence linking the ZIKV(BR) outbreak to the alarming number of cases of congenital brain malformations. Our model can be used to determine the efficiency of therapeutic approaches to counteracting the harmful impact of ZIKV(BR) in human neurodevelopment.


Disease Models, Animal , Microcephaly/virology , Zika Virus/pathogenicity , Animals , Apoptosis , Autophagy , Brain/pathology , Brain/virology , Brazil/epidemiology , Cell Proliferation , Female , Fetal Growth Retardation/pathology , Fetal Growth Retardation/virology , Fetus/virology , Mice , Microcephaly/epidemiology , Microcephaly/etiology , Microcephaly/pathology , Neural Stem Cells/pathology , Neural Stem Cells/virology , Organoids/pathology , Organoids/virology , Placenta/virology , Pregnancy , Zika Virus Infection/complications , Zika Virus Infection/epidemiology , Zika Virus Infection/pathology , Zika Virus Infection/virology
13.
World J Transplant ; 5(4): 209-21, 2015 Dec 24.
Article En | MEDLINE | ID: mdl-26722648

Several diseases have been successfully modeled since the development of induced pluripotent stem cell (iPSC) technology in 2006. Since then, methods for increased reprogramming efficiency and cell culture maintenance have been optimized and many protocols for differentiating stem cell lines have been successfully developed, allowing the generation of several cellular subtypes in vitro. Gene editing technologies have also greatly advanced lately, enhancing disease-specific phenotypes by creating isogenic cell lines, allowing mutations to be corrected in affected samples or inserted in control lines. Neurological disorders have benefited the most from iPSC-disease modeling for its capability for generating disease-relevant cell types in vitro from the central nervous system, such as neurons and glial cells, otherwise only available from post-mortem samples. Patient-specific iPSC-derived neural cells can recapitulate the phenotypes of these diseases and therefore, considerably enrich our understanding of pathogenesis, disease mechanism and facilitate the development of drug screening platforms for novel therapeutic targets. Here, we review the accomplishments and the current progress in human neurological disorders by using iPSC modeling for Alzheimer's disease, Parkinson's disease, Huntington's disease, spinal muscular atrophy, amyotrophic lateral sclerosis, duchenne muscular dystrophy, schizophrenia and autism spectrum disorders, which include Timothy syndrome, Fragile X syndrome, Angelman syndrome, Prader-Willi syndrome, Phelan-McDermid, Rett syndrome as well as Nonsyndromic Autism.

14.
Pesqui. vet. bras ; 32(5): 463-469, maio 2012. ilus, graf
Article Pt | LILACS | ID: lil-626488

O objetivo principal da nossa pesquisa foi avaliar o potencial de diferenciação osteogênica de células-tronco mesenquimais (MSC) obtidas da medula óssea do cão. As MSC foram separadas pelo método Ficoll e cultivadas sob duas condições distintas: DMEM baixa glicose ou DMEM/F12, ambos contendo L-glutamina, 20% de SFB e antibióticos. Marcadores de MSC foram testados, confirmando células CD44+ e CD34- através da citometria de fluxo. Para a diferenciação osteogênica, as células foram submetidas a quatro diferentes condições: Grupo 1, as mesmas condições utilizadas para a cultura de células primárias com os meios DMEM baixa glicose suplementado; Grupo 2, as mesmas condições do Grupo 1, mais os indutores de diferenciação dexametasona, ácido ascórbico e b-glicerolfosfato; Grupo 3, células cultivadas com meios DMEM/F12 suplementado; e Grupo 4, nas mesmas condições que no Grupo 3, mais indutores de diferenciação de dexametasona, ácido ascórbico e b-glicerolfosfato. A diferenciação celular foi confirmada através da coloração com alizarin red e da imunomarcação com o anticorpo SP7/Osterix. Nós observamos através da coloração com alizarin red que o depósito de cálcio foi mais evidente nas células cultivadas em DMEM/F12. Além disso, usando a imunomarcação com o anticorpo SP/7Osterix obtivemos positividade em 1:6 células para o Meio DMEM/F12 comparada com 1:12 para o meio DMEM-baixa glicose. Com base nos nossos resultados concluímos que o meio DMEM/F12 é mais eficiente para a indução da diferenciação de células-tronco mesenquimais caninas em promotores osteogênicos. Este efeito provavelmente ocorre em decorrência da maior quantidade de glicose neste meio, bem como da presença de diversos aminoácidos.


The aim of our research was to evaluate the potential for osteogenic differentiation of mesenchimal stem cells (MSC) obtained from dog bone marrow. The MSC were separated using the Ficoll method and cultured under two different conditions: DMEM low glucose or DMEM/F12, both containing L-glutamine, 20% of FBS and antibiotics. MSC markers were tested, confirming CD44+ and CD34- cells with flow cytometry. For osteogenic differentiation, cells were submitted to four different conditions: Group 1, same conditions used for primary cell culture with DMEM supplemented media; Group 2, same conditions of Group 1 plus differentiation inductors Dexametazone, ascorbic acid and β-glicerolphosphate. Group 3, Cells cultured with supplemented DMEM/F12 media, and Group 4, same conditions as in Group 3 plus differentiation inductors Dexametazone, ascorbic acid and β-glicerolphosphate. The cellular differentiation was confirmed using alizarin red and imunostaining with SP7/Osterix antibody. We observed by alizarin staining that calcium deposit was more evident in cells cultivated in DMEM/F12.Furthermore, by SP/7Osterix antibody immunostaining we obtained 1:6 positive cells when using DMEM/F12 compared with 1:12 for low-glucose DMEM. Based on our results, we conclude that the medium DMEM/F12 is more efficient for induction of differentiation of mesenchymal stem cells in canine osteogenic progenitors. This effect is probably due to the greater amount of glucose in the medium and the presence of various amino acids.


Animals , Dogs , Dogs/genetics , Mesenchymal Stem Cells/cytology , Bone Marrow/physiology , Osteogenesis/genetics , Glucose/genetics , Culture Media/isolation & purification , Cell Culture Techniques/veterinary
15.
Cell Transplant ; 20(11-12): 1707-19, 2011.
Article En | MEDLINE | ID: mdl-21457612

Induced pluripotent stem cells (iPSCs) can be created by forcing expression of certain genes in fibroblasts or other somatic cell types, reversing them to a pluripotent state similar to that of embryonic stem cells (ESC). Here, we used human immature dental pulp stem cells (hIDPSCs) as an alternative source for creating iPSC. hIDPSCs can be easily isolated from accessible tissue of young and adult patients. hIDPSCs possess a fibroblast-like morphology, retaining characteristics of adult multipotent stem cells. Reprogramming of hIDPSCs was fast, producing primary hIDPSC-iPSC colonies even under feeder-free conditions. hIDPSCs acquired ESC-like morphology, expressed pluripotent markers, possessed stable, normal karyotypes, and demonstrated the ability to differentiated in vitro and in vivo. Our data demonstrate that hIDPSCs-iPSCs offer an advantageous cell system for future cell therapy and basic studies, particularly as a model for pediatric developmental disorders.


Dental Pulp/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Cellular Reprogramming , Child , Embryoid Bodies/cytology , Humans , Karyotyping , Mice , Mice, Nude , Teratoma/pathology
16.
Cell Transplant ; 11(6): 583-92, 2002.
Article En | MEDLINE | ID: mdl-12428748

Vascular smooth muscle cells (VSMC) are ideal for systemic gene therapy because of their proximity to blood vessels and they have demonstrated long-term exogenous gene expression in vivo. However, the procedure generally followed to seed the transduced VSMC onto arteries denuded of endothelial cells usually induces stenosis and thrombosis, with a consequent high risk for use in humans. We demonstrate here that the vascular adventitia is a suitable place to introduce transduced VSMC and to secrete therapeutic proteins into the blood stream by a simple procedure, avoiding postoperative vascular complications. Transduced VSMC, with the retroviral vectors carrying the human growth hormone gene (hGH), were seeded into the adventitia of the rat abdominal aorta by single injection of a cell suspension. Based on the hGH and anti-hGH production in serum and on histological analysis of the removed aortas, we demonstrated hGH production over the 2-month experimental period. None of the animals used in the experiment showed stenosis, thrombosis, or other vascular or visible physiological abnormalities.


Cell Transplantation/methods , Endothelium, Vascular/cytology , Gene Expression , Muscle, Smooth, Vascular/transplantation , 3T3 Cells , Animals , Aorta, Abdominal , Cells, Cultured , DNA, Complementary/genetics , Endothelium, Vascular/physiology , Genetic Therapy/methods , Genetic Vectors , Human Growth Hormone/blood , Human Growth Hormone/genetics , Humans , Kinetics , Mice , Muscle, Smooth, Vascular/cytology , Promoter Regions, Genetic , Rats , Rats, Wistar , Retroviridae , Terminal Repeat Sequences , Transfection/methods , Zinc/pharmacology
...