Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Commun Biol ; 6(1): 1134, 2023 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-37945749

RESUMO

The molecular basis of reduced autofluorescence in oral squamous cell carcinoma (OSCC) cells relative to normal cells has been speculated to be due to lower levels of free flavin adenine dinucleotide (FAD). This speculation, along with differences in the intrinsic optical properties of extracellular collagen, lies at the foundation of the design of currently-used clinical optical detection devices. Here, we report that free FAD levels may not account for differences in autofluorescence of OSCC cells, but that the differences relate to FAD as a co-factor for flavination. Autofluorescence from a 70 kDa flavoprotein, succinate dehydrogenase A (SDHA), was found to be responsible for changes in optical properties within the FAD spectral region, with lower levels of flavinated SDHA in OSCC cells. Since flavinated SDHA is required for functional complexation with succinate dehydrogenase B (SDHB), decreased SDHB levels were observed in human OSCC tissue relative to normal tissues. Accordingly, the metabolism of OSCC cells was found to be significantly altered relative to normal cells, revealing vulnerabilities for both diagnosis and targeted therapy. Optimizing non-invasive tools based on optical and metabolic signatures of cancers will enable more precise and early diagnosis leading to improved outcomes in patients.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Bucais , Humanos , Succinato Desidrogenase/genética , Succinato Desidrogenase/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Neoplasias Bucais/patologia , Complexo II de Transporte de Elétrons/metabolismo
2.
bioRxiv ; 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36909618

RESUMO

Background: Gene editing in induced pluripotent stem (iPS) cells has been hailed to enable new cell therapies for various monogenetic diseases including dystrophic epidermolysis bullosa (DEB). However, manufacturing, efficacy and safety roadblocks have limited the development of genetically corrected, autologous iPS cell-based therapies. Methods: We developed Dystrophic Epidermolysis Bullosa Cell Therapy (DEBCT), a new generation GMP-compatible (cGMP), reproducible, and scalable platform to produce autologous clinical-grade iPS cell-derived organotypic induced skin composite (iSC) grafts to treat incurable wounds of patients lacking type VII collagen (C7). DEBCT uses a combined high-efficiency reprogramming and CRISPR-based genetic correction single step to generate genome scar-free, COL7A1 corrected clonal iPS cells from primary patient fibroblasts. Validated iPS cells are converted into epidermal, dermal and melanocyte progenitors with a novel 2D organoid differentiation protocol, followed by CD49f enrichment and expansion to minimize maturation heterogeneity. iSC product characterization by single cell transcriptomics was followed by mouse xenografting for disease correcting activity at 1 month and toxicology analysis at 1-6 months. Culture-acquired mutations, potential CRISPR-off targets, and cancer-driver variants were evaluated by targeted and whole genome sequencing. Findings: iPS cell-derived iSC grafts were reproducibly generated from four recessive DEB patients with different pathogenic mutations. Organotypic iSC grafts onto immune-compromised mice developed into stable stratified skin with functional C7 restoration. Single cell transcriptomic characterization of iSCs revealed prominent holoclone stem cell signatures in keratinocytes and the recently described Gibbin-dependent signature in dermal fibroblasts. The latter correlated with enhanced graftability. Multiple orthogonal sequencing and subsequent computational approaches identified random and non-oncogenic mutations introduced by the manufacturing process. Toxicology revealed no detectable tumors after 3-6 months in DEBCT-treated mice. Interpretation: DEBCT successfully overcomes previous roadblocks and represents a robust, scalable, and safe cGMP manufacturing platform for production of a CRISPR-corrected autologous organotypic skin graft to heal DEB patient wounds.

3.
Development ; 149(20)2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-35748297

RESUMO

Oligodendrocytes, the myelinating cells of the central nervous system, possess great potential for disease modeling and cell transplantation-based therapies for leukodystrophies. However, caveats to oligodendrocyte differentiation protocols ( Ehrlich et al., 2017; Wang et al., 2013; Douvaras and Fossati, 2015) from human embryonic stem and induced pluripotent stem cells (iPSCs), which include slow and inefficient differentiation, and tumorigenic potential of contaminating undifferentiated pluripotent cells, are major bottlenecks towards their translational utility. Here, we report the rapid generation of human oligodendrocytes by direct lineage conversion of human dermal fibroblasts (HDFs). We show that the combination of the four transcription factors OLIG2, SOX10, ASCL1 and NKX2.2 is sufficient to convert HDFs to induced oligodendrocyte precursor cells (iOPCs). iOPCs resemble human primary and iPSC-derived OPCs based on morphology and transcriptomic analysis. Importantly, iOPCs can differentiate into mature myelinating oligodendrocytes in vitro and in vivo. Finally, iOPCs derived from patients with Pelizaeus Merzbacher disease, a hypomyelinating leukodystrophy caused by mutations in the proteolipid protein 1 (PLP1) gene, showed increased cell death compared with iOPCs from healthy donors. Thus, human iOPCs generated by direct lineage conversion represent an attractive new source for human cell-based disease models and potentially myelinating cell grafts.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doença de Pelizaeus-Merzbacher , Diferenciação Celular/fisiologia , Fibroblastos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Oligodendroglia/metabolismo , Doença de Pelizaeus-Merzbacher/genética , Doença de Pelizaeus-Merzbacher/metabolismo , Doença de Pelizaeus-Merzbacher/terapia
4.
J Neurosci ; 42(8): 1557-1573, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-34965974

RESUMO

Collagen VI is a key component of muscle basement membranes, and genetic variants can cause monogenic muscular dystrophies. Conversely, human genetic studies recently implicated collagen VI in central nervous system function, with variants causing the movement disorder dystonia. To elucidate the neurophysiological role of collagen VI, we generated mice with a truncation of the dystonia-related collagen α3 VI (COL6A3) C-terminal domain (CTD). These Col6a3CTT mice showed a recessive dystonia-like phenotype in both sexes. We found that COL6A3 interacts with the cannabinoid receptor 1 (CB1R) complex in a CTD-dependent manner. Col6a3CTT mice of both sexes have impaired homeostasis of excitatory input to the basal pontine nuclei (BPN), a motor control hub with dense COL6A3 expression, consistent with deficient endocannabinoid (eCB) signaling. Aberrant synaptic input in the BPN was normalized by a CB1R agonist, and motor performance in Col6a3CTT mice of both sexes was improved by CB1R agonist treatment. Our findings identify a readily therapeutically addressable synaptic mechanism for motor control.SIGNIFICANCE STATEMENT Dystonia is a movement disorder characterized by involuntary movements. We previously identified genetic variants affecting a specific domain of the COL6A3 protein as a cause of dystonia. Here, we created mice lacking the affected domain and observed an analogous movement disorder. Using a protein interaction screen, we found that the affected COL6A3 domain mediates an interaction with the cannabinoid receptor 1 (CB1R). Concordantly, our COL6A3-deficient mice showed a deficit in synaptic plasticity linked to a deficit in cannabinoid signaling. Pharmacological cannabinoid augmentation rescued the motor impairment of the mice. Thus, cannabinoid augmentation could be a promising avenue for treating dystonia, and we have identified a possible molecular mechanism mediating this.


Assuntos
Canabinoides , Colágeno Tipo VI , Distonia , Distúrbios Distônicos , Neurônios Motores , Plasticidade Neuronal , Animais , Canabinoides/metabolismo , Canabinoides/farmacologia , Colágeno Tipo VI/genética , Colágeno Tipo VI/metabolismo , Distonia/genética , Distonia/metabolismo , Distúrbios Distônicos/genética , Distúrbios Distônicos/metabolismo , Feminino , Masculino , Camundongos , Neurônios Motores/efeitos dos fármacos , Mutação , Plasticidade Neuronal/efeitos dos fármacos , Receptores de Canabinoides/genética , Receptores de Canabinoides/metabolismo
5.
Cancer Cell ; 39(3): 407-422.e13, 2021 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-33545065

RESUMO

Diffuse intrinsic pontine glioma (DIPG) is an aggressive childhood tumor of the brainstem with currently no curative treatment available. The vast majority of DIPGs carry a histone H3 mutation leading to a lysine 27-to-methionine exchange (H3K27M). We engineered human induced pluripotent stem cells (iPSCs) to carry an inducible H3.3-K27M allele in the endogenous locus and studied the effects of the mutation in different disease-relevant neural cell types. H3.3-K27M upregulated bivalent promoter-associated developmental genes, producing diverse outcomes in different cell types. While being fatal for iPSCs, H3.3-K27M increased proliferation in neural stem cells (NSCs) and to a lesser extent in oligodendrocyte progenitor cells (OPCs). Only NSCs gave rise to tumors upon induction of H3.3-K27M and TP53 inactivation in an orthotopic xenograft model recapitulating human DIPGs. In NSCs, H3.3-K27M leads to maintained expression of stemness and proliferative genes and a premature activation of OPC programs that together may cause tumor initiation.


Assuntos
Neoplasias do Tronco Encefálico/genética , Neoplasias do Tronco Encefálico/parasitologia , Glioma/genética , Glioma/patologia , Histonas/genética , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Neurais/patologia , Animais , Linhagem Celular , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
6.
J Nutr Sci Vitaminol (Tokyo) ; 66(2): 136-149, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32350175

RESUMO

The acute metabolic effect of low dosages of L-carnitine under fat-mobilizing conditions was investigated. Healthy subjects (Study 1: n=5; Study 2: n=6) were asked to fast overnight. Then, 30 min of aerobic exercise on a cycle ergometer was performed after supplementation, followed by a 3.5-h sedentary recovery phase. The following ingestion patterns were used: Study 1 (i) noningestion, (ii) 750 mg of L-carnitine (LC), and (iii) 750 mg of LC+50 g of carbohydrate (CHO); Study 2 (iv) noningestion, (v) 500 mg of LC, (vi) 30 mg of CoQ10, and (vii) 500 mg of LC+30 mg of CoQ10. The energy expenditure (EE) and nonprotein respiratory quotient (npRQ) were measured during the pre-exercise, postexercise, and recovery periods. Serum free carnitine, acetylcarnitine, total carnitine (Study 1 and 2), and ketone bodies (Study 2) were measured. The 750 mg LC treatment significantly facilitated fat oxidation during the recovery phases (p<0.05) without elevating EE. The higher fat oxidation associated with LC was completely suppressed by CHO. CoQ10 affected neither npRQ nor EE. npRQ was significantly correlated with the serum total ketone bodies (R=-0.68, p<0.001) and acetylcarnitine (R=-0.61--0.70, p<0.001). The highest correlation was found between acetylcarnitine and total ketone bodies immediately after exercise (R=0.85, p<0.001). In conclusion, LC enhanced liver fat utilization and ketogenesis in an acute manner without stimulating EE under fat-mobilizing conditions.


Assuntos
Carnitina/farmacologia , Metabolismo Energético , Exercício Físico/fisiologia , Corpos Cetônicos/sangue , Lipólise/efeitos dos fármacos , Fígado/efeitos dos fármacos , Acetilcarnitina/sangue , Tecido Adiposo/metabolismo , Adulto , Ciclismo , Carnitina/administração & dosagem , Carnitina/sangue , Carboidratos da Dieta , Método Duplo-Cego , Feminino , Humanos , Fígado/metabolismo , Masculino , Músculo Esquelético/metabolismo , Oxirredução , Projetos Piloto , Respiração , Ubiquinona/análogos & derivados , Ubiquinona/farmacologia , Adulto Jovem
7.
Nat Commun ; 10(1): 2172, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-31092829

RESUMO

Inducing mitochondrial uncoupling (mUncoupling) is an attractive therapeutic strategy for treating metabolic diseases because it leads to calorie-wasting by reducing the efficiency of oxidative phosphorylation (OXPHOS) in mitochondria. Here we report a safe mUncoupler, OPC-163493, which has unique pharmacokinetic characteristics. OPC-163493 shows a good bioavailability upon oral administration and primarily distributed to specific organs: the liver and kidneys, avoiding systemic toxicities. It exhibits insulin-independent antidiabetic effects in multiple animal models of type I and type II diabetes and antisteatotic effects in fatty liver models. These beneficial effects can be explained by the improvement of glucose metabolism and enhancement of energy expenditure by OPC-163493 in the liver. Moreover, OPC-163493 treatment lowered blood pressure, extended survival, and improved renal function in the rat model of stroke/hypertension, possibly by enhancing NO bioavailability in blood vessels and reducing mitochondrial ROS production. OPC-163493 is a liver-localized/targeted mUncoupler that ameliorates various complications of diabetes.


Assuntos
Hipoglicemiantes/farmacologia , Fígado/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Desacopladores/farmacologia , Administração Oral , Animais , Pressão Sanguínea/efeitos dos fármacos , Células CHO , Cricetulus , Diabetes Mellitus/sangue , Diabetes Mellitus/tratamento farmacológico , Modelos Animais de Doenças , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/etiologia , Fígado Gorduroso/patologia , Feminino , Células Hep G2 , Humanos , Hipertensão/tratamento farmacológico , Hipertensão/etiologia , Hipertensão/mortalidade , Hipoglicemiantes/farmacocinética , Hipoglicemiantes/uso terapêutico , Rim/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Mitocôndrias/metabolismo , Fosforilação Oxidativa/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/mortalidade , Análise de Sobrevida , Desacopladores/farmacocinética , Desacopladores/uso terapêutico
8.
Elife ; 82019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30628890

RESUMO

Long noncoding RNAs (lncRNAs) have been shown to act as important cell biological regulators including cell fate decisions but are often ignored in human genetics. Combining differential lncRNA expression during neuronal lineage induction with copy number variation morbidity maps of a cohort of children with autism spectrum disorder/intellectual disability versus healthy controls revealed focal genomic mutations affecting several lncRNA candidate loci. Here we find that a t(5:12) chromosomal translocation in a family manifesting neurodevelopmental symptoms disrupts specifically lnc-NR2F1. We further show that lnc-NR2F1 is an evolutionarily conserved lncRNA functionally enhances induced neuronal cell maturation and directly occupies and regulates transcription of neuronal genes including autism-associated genes. Thus, integrating human genetics and functional testing in neuronal lineage induction is a promising approach for discovering candidate lncRNAs involved in neurodevelopmental diseases.


Assuntos
Transtorno do Espectro Autista/genética , Diferenciação Celular/genética , Mutação , Transtornos do Neurodesenvolvimento/genética , Neurônios/metabolismo , RNA Longo não Codificante/genética , Transtorno do Espectro Autista/patologia , Criança , Cromossomos Humanos Par 12/genética , Cromossomos Humanos Par 5/genética , Variações do Número de Cópias de DNA , Feminino , Perfilação da Expressão Gênica/métodos , Humanos , Masculino , Transtornos do Neurodesenvolvimento/patologia , Neurogênese/genética , Neurônios/citologia , Linhagem , Translocação Genética/genética
9.
Proc Natl Acad Sci U S A ; 115(25): 6470-6475, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29866841

RESUMO

Human cell models for disease based on induced pluripotent stem (iPS) cells have proven to be powerful new assets for investigating disease mechanisms. New insights have been obtained studying single mutations using isogenic controls generated by gene targeting. Modeling complex, multigenetic traits using patient-derived iPS cells is much more challenging due to line-to-line variability and technical limitations of scaling to dozens or more patients. Induced neuronal (iN) cells reprogrammed directly from dermal fibroblasts or urinary epithelia could be obtained from many donors, but such donor cells are heterogeneous, show interindividual variability, and must be extensively expanded, which can introduce random mutations. Moreover, derivation of dermal fibroblasts requires invasive biopsies. Here we show that human adult peripheral blood mononuclear cells, as well as defined purified T lymphocytes, can be directly converted into fully functional iN cells, demonstrating that terminally differentiated human cells can be efficiently transdifferentiated into a distantly related lineage. T cell-derived iN cells, generated by nonintegrating gene delivery, showed stereotypical neuronal morphologies and expressed multiple pan-neuronal markers, fired action potentials, and were able to form functional synapses. These cells were stable in the absence of exogenous reprogramming factors. Small molecule addition and optimized culture systems have yielded conversion efficiencies of up to 6.2%, resulting in the generation of >50,000 iN cells from 1 mL of peripheral blood in a single step without the need for initial expansion. Thus, our method allows the generation of sufficient neurons for experimental interrogation from a defined, homogeneous, and readily accessible donor cell population.


Assuntos
Diferenciação Celular/fisiologia , Transdiferenciação Celular/fisiologia , Leucócitos Mononucleares/citologia , Neurônios/citologia , Linfócitos T/citologia , Adolescente , Adulto , Idoso , Reprogramação Celular/fisiologia , Feminino , Fibroblastos/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem
10.
Cell Rep ; 23(2): 361-375, 2018 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-29641997

RESUMO

Here, we report that MYC rescues early human cells undergoing reprogramming from a proliferation pause induced by OCT3/4, SOX2, and KLF4 (OSK). We identified ESRG as a marker of early reprogramming cells that is expressed as early as day 3 after OSK induction. On day 4, ESRG positive (+) cells converted to a TRA-1-60 (+) intermediate state. These early ESRG (+) or TRA-1-60 (+) cells showed a proliferation pause due to increased p16INK4A and p21 and decreased endogenous MYC caused by OSK. Exogenous MYC did not enhance the appearance of initial reprogramming cells but instead reactivated their proliferation and improved reprogramming efficiency. MYC increased expression of LIN41, which potently suppressed p21 post-transcriptionally. MYC suppressed p16 INK4A. These changes inactivated retinoblastoma protein (RB) and reactivated proliferation. The RB-regulated proliferation pause does not occur in immortalized fibroblasts, leading to high reprogramming efficiency even without exogenous MYC.


Assuntos
Reprogramação Celular , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteína do Retinoblastoma/metabolismo , Antígenos de Superfície/metabolismo , Linhagem Celular , Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Fosforilação , Proteoglicanas/metabolismo , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myc/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteína do Retinoblastoma/antagonistas & inibidores , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
11.
Anal Chim Acta ; 985: 1-6, 2017 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-28864179

RESUMO

A microfluidic device coupled with a microfabricated Clark-type oxygen electrode was used to measure the bactericidal activity of neutrophil-like cells differentiated from HL-60 cells. The neutrophil-like cells and Escherichia coli (E. coli) cells were cultured in the same medium, which was introduced into the flow channel of the device. Changes in the respiratory activity of E. coli were measured as changes in the consumption of dissolved oxygen. As the activity of the neutrophil-like cells increased, the rate of elimination of E. coli increased. The accompanying decrease in the number of E. coli reduced the consumption of dissolved oxygen. The changes were actually observed as changes in generated current. A distinct difference in changes in dissolved oxygen concentrations was observed between E. coli cells co-incubated with IFN-γ-activated or non-activated neutrophil-like cells. The required sample volume was less than 10 µL, and results could be obtained within 1-2 h. The device may be useful for the assessment of psychological stresses that affect the activity of neutrophils.


Assuntos
Escherichia coli , Dispositivos Lab-On-A-Chip , Neutrófilos/citologia , Eletrodos , Humanos , Neutrófilos/imunologia , Oxigênio
12.
Dent Mater J ; 36(4): 408-414, 2017 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-28302944

RESUMO

The aim of this study was to investigate properties of atelocollagen/gelatin complexes (AC/Gel) and their characteristics of sustained statin release, to assess the utility of AC/Gel. AC/Gel were prepared by changing the mixing ratio of AC (0 to 40% of AC). Analysis of spectra of fluvastatin (Flu), gelatin (Gel), and Flu with Gel complex using a Fourier transform-infrared spectrometer indicates that Flu was bound to Gel through a bond involving the carboxyl and amino groups. Evaluation of characteristics of sustained release of Flu from the AC/Gel using an ultraviolet-visible spectrophotometer showed that the release rate of Flu decreased with increasing the AC content. The histological evaluation using of Sprague-Dawley rats suggest that, unlike the pure Gel sponge, the AC/Gel was not absorbed in an early stage. Therefore, the present study showed that sustained Flu release can be controlled by using an AC/Gel, suggesting the utility of this composite material.


Assuntos
Ácidos Graxos Monoinsaturados , Gelatina , Indóis , Animais , Sistemas de Liberação de Medicamentos , Fluvastatina , Inibidores de Hidroximetilglutaril-CoA Redutases , Teste de Materiais , Ratos , Ratos Sprague-Dawley
13.
Dent Mater J ; 36(4): 429-437, 2017 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-28302946

RESUMO

This study aimed to investigate influences of lyophilization factors and gelatin concentration on pore structures of ACG sponge. ACG sponges of different freezing temperatures (-30, -80 and -196oC), freezing times (1, 2 and 24 h), gelatin concentrations (0.6%AC+0.15%G, 0.6%AC+0.6%G and 0.6%AC+2.4%G), and with 500 µM fluvastatin were fabricated. Pore structures including porosity and pore size were analyzed by scanning electron microscopy and ImageJ. The cytotoxic effects of ACG sponges were evaluated in vitro. Freezing temperature did not affect porosity while high freezing temperature (-30oC) increased pore size. The high gelatin concentration group (0.6%AC+2.4%G) had decreased porosity and pore size. Freezing time and 500 µM fluvastatin did not affect pore structures. The cytotoxicity and cell proliferation assays revealed that ACG sponges had no cytotoxic effects on human mesenchymal stromal cell growth and proliferation. These results indicate that ACG sponge may be a good biomaterial scaffold for bone regeneration.


Assuntos
Materiais Biocompatíveis , Gelatina , Engenharia Tecidual , Osso e Ossos , Liofilização , Humanos , Células-Tronco Mesenquimais , Porosidade , Alicerces Teciduais
14.
J Neurosci ; 36(45): 11544-11558, 2016 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-27911758

RESUMO

Chemogenetic manipulation of neuronal activities has been enabled by a designer receptor (designer receptor exclusively activated by designer drugs, DREADD) that is activated exclusively by clozapine-N-oxide (CNO). Here, we applied CNO as a functional reporter probe to positron emission tomography (PET) of DREADD in living brains. Mutant human M4 DREADD (hM4Di) expressed in transgenic (Tg) mouse neurons was visualized by PET with microdose [11C]CNO. Deactivation of DREADD-expressing neurons in these mice by nonradioactive CNO at a pharmacological dose could also be captured by arterial spin labeling MRI (ASL-MRI). Neural progenitors derived from hM4Di Tg-induced pluripotent stem cells were then implanted into WT mouse brains and neuronal differentiation of the grafts could be imaged by [11C]CNO-PET. Finally, ASL-MRI captured chemogenetic functional manipulation of the graft neurons. Our data provide the first demonstration of multimodal molecular/functional imaging of cells expressing a functional gene reporter in the brain, which would be translatable to humans for therapeutic gene transfers and cell replacements. SIGNIFICANCE STATEMENT: The present work provides the first successful demonstration of in vivo positron emission tomographic (PET) visualization of a chemogenetic designer receptor (designer receptor exclusively activated by designer drugs, DREADD) expressed in living brains. This technology has been applied to longitudinal PET reporter imaging of neuronal grafts differentiated from induced pluripotent stem cells. Differentiated from currently used reporter genes for neuroimaging, DREADD has also been available for functional manipulation of target cells, which could be visualized by functional magnetic resonance imaging (fMRI) in a real-time manner. Multimodal imaging with PET/fMRI enables the visualization of the differentiation of iPSC-derived neural progenitors into mature neurons and DREADD-mediated functional manipulation along the time course of the graft and is accordingly capable of fortifying the utility of stem cells in cell replacement therapies.


Assuntos
Encéfalo/citologia , Genes Reporter , Células-Tronco Pluripotentes Induzidas/citologia , Imagem Multimodal/métodos , Células-Tronco Neurais/transplante , Neurônios/citologia , Neurônios/metabolismo , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Tomografia por Emissão de Pósitrons/métodos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Transplante de Células-Tronco/métodos
15.
J Oleo Sci ; 65(12): 1003-1010, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27829610

RESUMO

The technology of physicochemical surface modification is available for enhancing the bioactivity and osseointegration capability of tetragonal zirconia polycrystal (TZP). Hydrophobicity index and electrical charge play important roles in protein adsorption. We previously studied the mechanism underlying the adsorption of bovine serum albumin (BSA) on the surfaces of dental materials and hydroxyapatite in vitro. The aim of the present study was to clarify the correlation among the adsorption of BSA to TZP and physicochemically modified TZP surfaces and the zeta potential of BSA and TZP. We used TZP that was sintered at 1350°C for 2 h in air because this kind of TZP is widely applied in the field of dentistry. Surface physicochemistry was modified with ultraviolet light (UV) and atmospheric-pressure plasma treatment. The zeta potentials were measured with ELSZ-1000 and ELSZ-2000 analyzers (Otsuka Electronics, Hirakata, Japan). All experiments were conducted in 10 mM NaCl (pH 7.0). The zeta potentials of as-received TZP and BSA were negative, but those of UV- and plasma-treated TZP were positive. The reason the zeta potentials of TZP changed positive by physicochemical modification is due to an increase in the amount of basic hydroxyl groups. The zeta potentials of UV- and plasma-treated TZP after BSA adsorption were negative. These results suggested that electrostatic interactions play an important role in BSA adsorption to TZP and modified TZP surfaces, so that this modified surface may control the adsorption of protein.


Assuntos
Polímeros/química , Soroalbumina Bovina/química , Zircônio/química , Adsorção , Animais , Bovinos , Físico-Química , Eletricidade Estática , Propriedades de Superfície
16.
Cell Stem Cell ; 19(3): 341-54, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27476965

RESUMO

Variation in the differentiation capacity of induced pluripotent stem cells (iPSCs) to specific lineages is a significant concern for their use in clinical applications and disease modeling. To identify factors that affect differentiation capacity, we performed integration analyses between hematopoietic differentiation performance and molecular signatures such as gene expression, DNA methylation, and chromatin status, using 35 human iPSC lines and four ESC lines. Our analyses revealed that hematopoietic commitment of PSCs to hematopoietic precursors correlates with IGF2 expression level, which in turn depends on signaling-dependent chromatin accessibility at mesendodermal genes. Maturation capacity for conversion of PSC-derived hematopoietic precursors to mature blood associates with the amount and pattern of DNA methylation acquired during reprogramming. Our study therefore provides insight into the molecular features that determine the differential capacities seen among human iPSC lines and, through the predictive potential of this information, highlights a way to select optimal iPSCs for clinical applications.


Assuntos
Diferenciação Celular/genética , Epigênese Genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Ativinas/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Linhagem da Célula/genética , Reprogramação Celular/genética , Cromatina/química , Metilação de DNA/genética , Células Eritroides/citologia , Células Eritroides/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Redes Reguladoras de Genes , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Fator de Crescimento Insulin-Like II/metabolismo , Megacariócitos/citologia , Megacariócitos/metabolismo , Camundongos SCID , Transdução de Sinais/genética , Transplante de Células-Tronco , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo
17.
PLoS One ; 11(3): e0151927, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26986842

RESUMO

Elucidating the regulation of glucose-stimulated insulin secretion (GSIS) in pancreatic ß cells is important for understanding and treating diabetes. The pancreatic ß cell line, MIN6, retains GSIS but gradually loses it in long-term culture. The MIN6 subclone, MIN6c4, exhibits well-regulated GSIS even after prolonged culture. We previously used DNA microarray analysis to compare gene expression in the parental MIN6 cells and MIN6c4 cells and identified several differentially regulated genes that may be involved in maintaining GSIS. Here we investigated the potential roles of six of these genes in GSIS: Tmem59l (Transmembrane protein 59 like), Scgn (Secretagogin), Gucy2c (Guanylate cyclase 2c), Slc29a4 (Solute carrier family 29, member 4), Cdhr1 (Cadherin-related family member 1), and Celsr2 (Cadherin EGF LAG seven-pass G-type receptor 2). These genes were knocked down in MIN6c4 cells using lentivirus vectors expressing gene-specific short hairpin RNAs (shRNAs), and the effects of the knockdown on insulin expression and secretion were analyzed. Suppression of Tmem59l, Scgn, and Gucy2c expression resulted in significantly decreased glucose- and/or KCl-stimulated insulin secretion from MIN6c4 cells, while the suppression of Slc29a4 expression resulted in increased insulin secretion. Tmem59l overexpression rescued the phenotype of the Tmem59l knockdown MIN6c4 cells, and immunostaining analysis indicated that the TMEM59L protein colocalized with insulin and GM130, a Golgi complex marker, in MIN6 cells. Collectively, our findings suggested that the proteins encoded by Tmem59l, Scgn, Gucy2c, and Slc29a4 play important roles in regulating GSIS. Detailed studies of these proteins and their functions are expected to provide new insights into the molecular mechanisms involved in insulin secretion.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animais , Western Blotting , Caderinas/fisiologia , Linhagem Celular , Feminino , Imunofluorescência , Técnicas de Silenciamento de Genes , Genes Reguladores/fisiologia , Glucose/fisiologia , Insulina/fisiologia , Secreção de Insulina , Células Secretoras de Insulina/fisiologia , Proteínas de Membrana/fisiologia , Proteínas de Membrana Transportadoras/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Enterotoxina , Receptores Acoplados a Guanilato Ciclase/fisiologia , Receptores de Peptídeos/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Secretagoginas/fisiologia
18.
Philos Trans R Soc Lond B Biol Sci ; 370(1680): 20140368, 2015 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-26416679

RESUMO

The predominant view of embryonic development and cell differentiation has been that rigid and even irreversible epigenetic marks are laid down along the path of cell specialization ensuring the proper silencing of unrelated lineage programmes. This model made the prediction that specialized cell types are stable and cannot be redirected into other lineages. Accordingly, early attempts to change the identity of somatic cells had little success and was limited to conversions between closely related cell types. Nuclear transplantation experiments demonstrated, however, that specialized cells even from adult mammals can be reprogrammed into a totipotent state. The discovery that a small combination of transcription factors can reprogramme cells to pluripotency without the need of oocytes further supported the view that these epigenetic barriers can be overcome much easier than assumed, but the extent of this flexibility was still unclear. When we showed that a differentiated mesodermal cell can be directly converted to a differentiated ectodermal cell without a pluripotent intermediate, it was suggested that in principle any cell type could be converted into any other cell type. Indeed, the work of several groups in recent years has provided many more examples of direct somatic lineage conversions. Today, the question is not anymore whether a specific cell type can be generated by direct reprogramming but how it can be induced.


Assuntos
Linhagem da Célula , Reprogramação Celular , Animais , Diferenciação Celular , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Neurais/citologia , Neurônios/classificação , Neurônios/citologia , Técnicas de Transferência Nuclear
19.
Proc Natl Acad Sci U S A ; 111(34): 12426-31, 2014 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-25097266

RESUMO

Pluripotency can be induced in somatic cells by overexpressing transcription factors, including POU class 5 homeobox 1 (OCT3/4), sex determining region Y-box 2 (SOX2), Krüppel-like factor 4 (KLF4), and myelocytomatosis oncogene (c-MYC). However, some induced pluripotent stem cells (iPSCs) exhibit defective differentiation and inappropriate maintenance of pluripotency features. Here we show that dynamic regulation of human endogenous retroviruses (HERVs) is important in the reprogramming process toward iPSCs, and in re-establishment of differentiation potential. During reprogramming, OCT3/4, SOX2, and KLF4 transiently hyperactivated LTR7s--the long-terminal repeats of HERV type-H (HERV-H)--to levels much higher than in embryonic stem cells by direct occupation of LTR7 sites genome-wide. Knocking down LTR7s or long intergenic non-protein coding RNA, regulator of reprogramming (lincRNA-RoR), a HERV-H-driven long noncoding RNA, early in reprogramming markedly reduced the efficiency of iPSC generation. KLF4 and LTR7 expression decreased to levels comparable with embryonic stem cells once reprogramming was complete, but failure to resuppress KLF4 and LTR7s resulted in defective differentiation. We also observed defective differentiation and LTR7 activation when iPSCs had forced expression of KLF4. However, when aberrantly expressed KLF4 or LTR7s were suppressed in defective iPSCs, normal differentiation was restored. Thus, a major mechanism by which OCT3/4, SOX2, and KLF4 promote human iPSC generation and reestablish potential for differentiation is by dynamically regulating HERV-H LTR7s.


Assuntos
Retrovirus Endógenos/genética , Retrovirus Endógenos/fisiologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/virologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Reprogramação Celular/genética , Reprogramação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Células-Tronco Embrionárias/virologia , Epigênese Genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Pluripotentes Induzidas/virologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/fisiologia , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/fisiologia , Células-Tronco Pluripotentes/fisiologia , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/genética , RNA Viral/antagonistas & inibidores , RNA Viral/genética , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/fisiologia
20.
Nat Commun ; 5: 3678, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24759836

RESUMO

During mammalian embryonic development, the primitive streak initiates the differentiation of pluripotent epiblast cells into germ layers. Pluripotency can be reacquired in committed somatic cells using a combination of a handful of transcription factors, such as OCT3/4, SOX2, KLF4 and c-MYC (hereafter referred to as OSKM), albeit with low efficiency. Here we show that during OSKM-induced reprogramming towards pluripotency in human cells, intermediate cells transiently show gene expression profiles resembling mesendoderm, which is a major component of the primitive streak. Based on these findings, we discover that forkhead box H1 (FOXH1), a transcription factor required for anterior primitive streak specification during early development, significantly enhances the reprogramming efficiency of human fibroblasts by promoting their maturation, including mesenchymal to epithelial transition and the activation of late pluripotency markers. These results demonstrate that during the reprogramming process, human somatic cells go through a transient state that resembles mesendoderm.


Assuntos
Endoderma/citologia , Mesoderma/citologia , Células-Tronco Pluripotentes/citologia , Diferenciação Celular/fisiologia , Células Cultivadas , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...