Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 40(9): e106423, 2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33644903

RESUMO

Endogenous retroviruses (ERVs) make up a large fraction of mammalian genomes and are thought to contribute to human disease, including brain disorders. In the brain, aberrant activation of ERVs is a potential trigger for an inflammatory response, but mechanistic insight into this phenomenon remains lacking. Using CRISPR/Cas9-based gene disruption of the epigenetic co-repressor protein Trim28, we found a dynamic H3K9me3-dependent regulation of ERVs in proliferating neural progenitor cells (NPCs), but not in adult neurons. In vivo deletion of Trim28 in cortical NPCs during mouse brain development resulted in viable offspring expressing high levels of ERVs in excitatory neurons in the adult brain. Neuronal ERV expression was linked to activated microglia and the presence of ERV-derived proteins in aggregate-like structures. This study demonstrates that brain development is a critical period for the silencing of ERVs and provides causal in vivo evidence demonstrating that transcriptional activation of ERV in neurons results in an inflammatory response.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encefalite/genética , Retrovirus Endógenos/genética , Deleção de Genes , Proteína 28 com Motivo Tripartido/genética , Animais , Encéfalo/imunologia , Encéfalo/virologia , Sistemas CRISPR-Cas , Células Cultivadas , Encefalite/imunologia , Encefalite/virologia , Retrovirus Endógenos/imunologia , Epigênese Genética , Regulação da Expressão Gênica , Histonas/metabolismo , Camundongos , Ativação Transcricional
2.
Blood ; 139(11): 1659-1669, 2022 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-35007327

RESUMO

Stem cell transplantation is a cornerstone in the treatment of blood malignancies. The most common method to harvest stem cells for transplantation is by leukapheresis, requiring mobilization of CD34+ hematopoietic stem and progenitor cells (HSPCs) from the bone marrow into the blood. Identifying the genetic factors that control blood CD34+ cell levels could reveal new drug targets for HSPC mobilization. Here we report the first large-scale, genome-wide association study on blood CD34+ cell levels. Across 13 167 individuals, we identify 9 significant and 2 suggestive associations, accounted for by 8 loci (PPM1H, CXCR4, ENO1-RERE, ITGA9, ARHGAP45, CEBPA, TERT, and MYC). Notably, 4 of the identified associations map to CXCR4, showing that bona fide regulators of blood CD34+ cell levels can be identified through genetic variation. Further, the most significant association maps to PPM1H, encoding a serine/threonine phosphatase never previously implicated in HSPC biology. PPM1H is expressed in HSPCs, and the allele that confers higher blood CD34+ cell levels downregulates PPM1H. Through functional fine-mapping, we find that this downregulation is caused by the variant rs772557-A, which abrogates an MYB transcription factor-binding site in PPM1H intron 1 that is active in specific HSPC subpopulations, including hematopoietic stem cells, and interacts with the promoter by chromatin looping. Furthermore, PPM1H knockdown increases the proportion of CD34+ and CD34+90+ cells in cord blood assays. Our results provide the first large-scale analysis of the genetic architecture of blood CD34+ cell levels and warrant further investigation of PPM1H as a potential inhibition target for stem cell mobilization.


Assuntos
Estudo de Associação Genômica Ampla , Células-Tronco Hematopoéticas , Antígenos CD34/metabolismo , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Humanos
3.
Blood ; 138(16): 1441-1455, 2021 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-34075404

RESUMO

Changes in gene regulation and expression govern orderly transitions from hematopoietic stem cells to terminally differentiated blood cell types. These transitions are disrupted during leukemic transformation, but knowledge of the gene regulatory changes underpinning this process is elusive. We hypothesized that identifying core gene regulatory networks in healthy hematopoietic and leukemic cells could provide insights into network alterations that perturb cell state transitions. A heptad of transcription factors (LYL1, TAL1, LMO2, FLI1, ERG, GATA2, and RUNX1) bind key hematopoietic genes in human CD34+ hematopoietic stem and progenitor cells (HSPCs) and have prognostic significance in acute myeloid leukemia (AML). These factors also form a densely interconnected circuit by binding combinatorially at their own, and each other's, regulatory elements. However, their mutual regulation during normal hematopoiesis and in AML cells, and how perturbation of their expression levels influences cell fate decisions remains unclear. In this study, we integrated bulk and single-cell data and found that the fully connected heptad circuit identified in healthy HSPCs persists, with only minor alterations in AML, and that chromatin accessibility at key heptad regulatory elements was predictive of cell identity in both healthy progenitors and leukemic cells. The heptad factors GATA2, TAL1, and ERG formed an integrated subcircuit that regulates stem cell-to-erythroid transition in both healthy and leukemic cells. Components of this triad could be manipulated to facilitate erythroid transition providing a proof of concept that such regulatory circuits can be harnessed to promote specific cell-type transitions and overcome dysregulated hematopoiesis.


Assuntos
Fator de Transcrição GATA2/genética , Regulação Leucêmica da Expressão Gênica , Leucemia Mieloide Aguda/genética , Proteína 1 de Leucemia Linfocítica Aguda de Células T/genética , Células Eritroides/metabolismo , Células Eritroides/patologia , Redes Reguladoras de Genes , Hematopoese , Humanos , Leucemia Mieloide Aguda/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Regulador Transcricional ERG/genética
4.
Haematologica ; 108(11): 3095-3109, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37199130

RESUMO

Diamond-Blackfan anemia is a rare genetic bone marrow failure disorder which is usually caused by mutations in ribosomal protein genes. In the present study, we generated a traceable RPS19-deficient cell model using CRISPR-Cas9 and homology-directed repair to investigate the therapeutic effects of a clinically applicable lentiviral vector at single-cell resolution. We developed a gentle nanostraw delivery platform to edit the RPS19 gene in primary human cord bloodderived CD34+ hematopoietic stem and progenitor cells. The edited cells showed expected impaired erythroid differentiation phenotype, and a specific erythroid progenitor with abnormal cell cycle status accompanied by enrichment of TNFα/NF-κB and p53 signaling pathways was identified by single-cell RNA sequencing analysis. The therapeutic vector could rescue the abnormal erythropoiesis by activating cell cycle-related signaling pathways and promoted red blood cell production. Overall, these results establish nanostraws as a gentle option for CRISPR-Cas9- based gene editing in sensitive primary hematopoietic stem and progenitor cells, and provide support for future clinical investigations of the lentiviral gene therapy strategy.


Assuntos
Anemia de Diamond-Blackfan , Humanos , Anemia de Diamond-Blackfan/genética , Anemia de Diamond-Blackfan/terapia , Anemia de Diamond-Blackfan/metabolismo , Proteínas Ribossômicas/genética , Diferenciação Celular , Eritropoese , Células-Tronco/metabolismo , Antígenos CD34
5.
Proc Natl Acad Sci U S A ; 117(35): 21267-21273, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32817519

RESUMO

Introduction of exogenous genetic material into primary stem cells is essential for studying biological function and for clinical applications. Traditional delivery methods for nucleic acids, such as electroporation, have advanced the field, but have negative effects on stem cell function and viability. We introduce nanostraw-assisted transfection as an alternative method for RNA delivery to human hematopoietic stem and progenitor cells (HSPCs). Nanostraws are hollow alumina nanotubes that can be used to deliver biomolecules to living cells. We use nanostraws to target human primary HSPCs and show efficient delivery of mRNA, short interfering RNAs (siRNAs), DNA oligonucleotides, and dextrans of sizes ranging from 6 kDa to 2,000 kDa. Nanostraw-treated cells were fully functional and viable, with no impairment in their proliferative or colony-forming capacity, and showed similar long-term engraftment potential in vivo as untreated cells. Additionally, we found that gene expression of the cells was not perturbed by nanostraw treatment, while conventional electroporation changed the expression of more than 2,000 genes. Our results show that nanostraw-mediated transfection is a gentle alternative to established gene delivery methods, and uniquely suited for nonperturbative treatment of sensitive primary stem cells.


Assuntos
Técnicas de Transferência de Genes , Células-Tronco Hematopoéticas , Nanoestruturas , Animais , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Microinjeções
6.
Blood ; 136(19): 2151-2161, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-32582923

RESUMO

Culture conditions in which hematopoietic stem cells (HSCs) can be expanded for clinical benefit are highly sought after. Here, we report that inhibition of the epigenetic regulator lysine-specific histone demethylase 1A (LSD1) induces a rapid expansion of human cord blood-derived CD34+ cells and promotes in vitro propagation of long-term repopulating HSCs by preventing differentiation. The phenotype and molecular characteristics of cells treated with LSD1 inhibitors were highly similar to cells treated with UM171, an agent promoting expansion of HSCs through undefined mechanisms and currently being tested in clinical trials. Strikingly, we found that LSD1, as well as other members of the LSD1-containing chromatin remodeling complex CoREST, is rapidly polyubiquitinated and degraded upon UM171 treatment. CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 depletion of the CoREST core member, RCOR1, resulted in expansion of CD34+ cells similar to LSD1 inhibition and UM171. Taken together, LSD1 and CoREST restrict HSC expansion and are principal targets of UM171, forming a mechanistic basis for the HSC-promoting activity of UM171.


Assuntos
Diferenciação Celular , Proteínas Correpressoras/metabolismo , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Histona Desmetilases/antagonistas & inibidores , Indóis/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Pirimidinas/farmacologia , Antígenos CD34/metabolismo , Proliferação de Células , Proteínas Correpressoras/genética , Sangue Fetal/efeitos dos fármacos , Sangue Fetal/metabolismo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Humanos , Proteínas do Tecido Nervoso/genética
7.
Sensors (Basel) ; 22(7)2022 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-35408204

RESUMO

This paper presents a local planning approach that is targeted for pseudo-omnidirectional vehicles: that is, vehicles that can drive sideways and rotate on the spot. This local planner-MSDU-is based on optimal control and formulates a non-linear optimization problem formulation that exploits the omni-motion capabilities of the vehicle to drive the vehicle to the goal in a smooth and efficient manner while avoiding obstacles and singularities. MSDU is designed for a real platform for mobile manipulation where one key function is the capability to drive in narrow and confined areas. The real-world evaluations show that MSDU planned paths that were smoother and more accurate than a comparable local path planner Timed Elastic Band (TEB), with a mean (translational, angular) error for MSDU of (0.0028 m, 0.0010 rad) compared to (0.0033 m, 0.0038 rad) for TEB. MSDU also generated paths that were consistently shorter than TEB, with a mean (translational, angular) distance traveled of (0.6026 m, 1.6130 rad) for MSDU compared to (0.7346 m, 3.7598 rad) for TEB.


Assuntos
Movimento (Física)
8.
Genes Chromosomes Cancer ; 60(6): 410-417, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33368842

RESUMO

High hyperdiploid acute lymphoblastic leukemia (ALL) is one of the most common malignancies in children. The main driver event of this disease is a nonrandom aneuploidy consisting of gains of whole chromosomes but without overt evidence of chromosomal instability (CIN). Here, we investigated the frequency and severity of defective sister chromatid cohesion-a phenomenon related to CIN-in primary pediatric ALL. We found that a large proportion (86%) of hyperdiploid cases displayed aberrant cohesion, frequently severe, to compare with 49% of ETV6/RUNX1-positive ALL, which mostly displayed mild defects. In hyperdiploid ALL, cohesion defects were associated with increased chromosomal copy number heterogeneity, which could indicate increased CIN. Furthermore, cohesion defects correlated with RAD21 and NCAPG mRNA expression, suggesting a link to reduced cohesin and condensin levels in hyperdiploid ALL. Knockdown of RAD21 in an ALL cell line led to sister chromatid cohesion defects, aberrant mitoses, and increased heterogeneity in chromosomal copy numbers, similar to what was seen in primary hyperdiploid ALL. In summary, our study shows that aberrant sister chromatid cohesion is frequent but heterogeneous in pediatric high hyperdiploid ALL, ranging from mild to very severe defects, and possibly due to low cohesin or condensin levels. Cases with high levels of aberrant chromosome cohesion displayed increased chromosomal copy number heterogeneity, possibly indicative of increased CIN. These abnormalities may play a role in the clonal evolution of hyperdiploid pediatric ALL.


Assuntos
Cromátides/genética , Instabilidade Cromossômica , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Criança , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Ploidias , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Repressoras/genética , Variante 6 da Proteína do Fator de Translocação ETS
9.
Blood ; 129(8): 950-958, 2017 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-27899358

RESUMO

Adhesion is a key component of hematopoietic stem cell regulation mediating homing and retention to the niche in the bone marrow. Here, using an RNA interference screen, we identify cytohesin 1 (CYTH1) as a critical mediator of adhesive properties in primary human cord blood-derived hematopoietic stem and progenitor cells (HSPCs). Knockdown of CYTH1 disrupted adhesion of HSPCs to primary human mesenchymal stroma cells. Attachment to fibronectin and ICAM1, 2 integrin ligands, was severely impaired, and CYTH1-deficient cells showed a reduced integrin ß1 activation response, suggesting that CYTH1 mediates integrin-dependent functions. Transplantation of CYTH1-knockdown cells to immunodeficient mice resulted in significantly lower long-term engraftment levels, associated with a reduced capacity of the transplanted cells to home to the bone marrow. Intravital microscopy showed that CYTH1 deficiency profoundly affects HSPC mobility and localization within the marrow space and thereby impairs proper lodgment into the niche. Thus, CYTH1 is a novel major regulator of adhesion and engraftment in human HSPCs through mechanisms that, at least in part, involve the activation of integrins.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Animais , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Movimento Celular , Fibronectinas/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Integrinas/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos , Interferência de RNA
10.
Haematologica ; 104(10): 2006-2016, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30819903

RESUMO

Dysregulation of cytokines in the bone marrow (BM) microenvironment promotes acute myeloid leukemia (AML) cell growth. Due to the complexity and low throughput of in vivo stem-cell based assays, studying the role of cytokines in the BM niche in a screening setting is challenging. Here, we developed an ex vivo cytokine screen using 11 arrayed molecular barcodes, allowing for a competitive in vivo readout of leukemia-initiating capacity. With this approach, we assessed the effect of 114 murine cytokines on MLL-AF9 AML mouse cells and identified the tumor necrosis factor ligand superfamily member 13 (TNFSF13) as a positive regulator of leukemia-initiating cells. By using Tnfsf13-/- recipient mice, we confirmed that TNFSF13 supports leukemia initiation also under physiological conditions. TNFSF13 was secreted by normal myeloid cells but not by leukemia mouse cells, suggesting that mature myeloid BM cells support leukemia cells by secreting TNFSF13. TNFSF13 supported leukemia cell proliferation in an NF-κB-dependent manner by binding TNFRSF17 and suppressed apoptosis. Moreover, TNFSF13 supported the growth and survival of several human myeloid leukemia cell lines, demonstrating that our findings translate to human disease. Taken together, using arrayed molecular barcoding, we identified a previously unrecognized role of TNFSF13 as a positive regulator of AML-initiating cells. The arrayed barcoded screening methodology is not limited to cytokines and leukemia, but can be extended to other types of ex vivo screens, where a multiplexed in vivo read-out of stem cell functionality is needed.


Assuntos
Células da Medula Óssea/metabolismo , Leucemia Mieloide Aguda/metabolismo , Neoplasias Experimentais/metabolismo , Células-Tronco Neoplásicas/metabolismo , Microambiente Tumoral , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Antígeno de Maturação de Linfócitos B/genética , Antígeno de Maturação de Linfócitos B/metabolismo , Células da Medula Óssea/patologia , Linhagem Celular Tumoral , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Knockout , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Células-Tronco Neoplásicas/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
11.
Curr Opin Hematol ; 25(4): 259-265, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29697484

RESUMO

PURPOSE OF REVIEW: Disturbance of the delicate balance between self-renewal and differentiation in haematopoietic stem cells (HSCs) can lead to both leukaemia and bone marrow failure. The regulation of this balance in HSC biology has been intensely investigated in several model systems, and lately the importance of epigenetic modifications as well as the organization and architecture of chromatin has become increasingly recognized. In this review, we will focus on the role of the chromatin organizing protein complex cohesin in regulation of normal and malignant haematopoiesis. RECENT FINDINGS: Several functional studies in both mouse and human systems have implicated cohesin as a critical regulator of self-renewal and differentiation in HSCs. Together with the discovery of recurrent mutations of cohesin genes in myeloid malignancies, this points towards a direct role of perturbed cohesin function in leukemogenesis. SUMMARY: The work reviewed here provides new insights about the role of the cohesin complex and chromatin architecture in normal and malignant HSCs, and indicates how cohesin may be specifically targeted for therapeutic benefit in the future.


Assuntos
Proteínas de Ciclo Celular , Proteínas Cromossômicas não Histona , Hematopoese , Células-Tronco Hematopoéticas , Leucemia , Mutação , Proteínas de Neoplasias , Células-Tronco Neoplásicas , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Leucemia/genética , Leucemia/metabolismo , Leucemia/patologia , Camundongos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Coesinas
12.
Haematologica ; 103(9): 1444-1450, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29880606

RESUMO

Despite extensive studies, defining culture conditions in which hematopoietic stem cells can be expanded ex vivo has been challenging. Here we show that chemical inhibition of the NF-κB signaling pathway leads to a significant improvement of hematopoietic stem cell function from ex vivo cultured human umbilical cord blood derived CD34+ cells. We found a distinct peak of activation of the NF-κB pathway shortly after cells were put in culture, and consequently inhibition of the pathway was both necessary and sufficient during the first 24 hours of culture where it reduced the levels of several pro-inflammatory cytokines. Taken together, NF-κB pathway inhibition facilitates propagation of hematopoietic stem cells in culture and may complement other strategies for hematopoietic stem cell expansion by relieving stress signals that are induced as an immediate response to culture initiation.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Animais , Antígenos CD34/metabolismo , Biomarcadores , Técnicas de Cultura de Células , Proliferação de Células , Células Cultivadas , Citocinas/metabolismo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Imunofenotipagem , Mediadores da Inflamação/metabolismo , Camundongos , Fenótipo , Transdução de Sinais/efeitos dos fármacos , Antígenos Thy-1/metabolismo
13.
Nucleic Acids Res ; 44(22): 10644-10661, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27604872

RESUMO

Aberrant stem cell-like gene regulatory networks are a feature of leukaemogenesis. The ETS-related gene (ERG), an important regulator of normal haematopoiesis, is also highly expressed in T-ALL and acute myeloid leukaemia (AML). However, the transcriptional regulation of ERG in leukaemic cells remains poorly understood. In order to discover transcriptional regulators of ERG, we employed a quantitative mass spectrometry-based method to identify factors binding the 321 bp ERG +85 stem cell enhancer region in MOLT-4 T-ALL and KG-1 AML cells. Using this approach, we identified a number of known binders of the +85 enhancer in leukaemic cells along with previously unknown binders, including ETV6 and IKZF1. We confirmed that ETV6 and IKZF1 were also bound at the +85 enhancer in both leukaemic cells and in healthy human CD34+ haematopoietic stem and progenitor cells. Knockdown experiments confirmed that ETV6 and IKZF1 are transcriptional regulators not just of ERG, but also of a number of genes regulated by a densely interconnected network of seven transcription factors. At last, we show that ETV6 and IKZF1 expression levels are positively correlated with expression of a number of heptad genes in AML and high expression of all nine genes confers poorer overall prognosis.


Assuntos
Fator de Transcrição Ikaros/fisiologia , Proteínas Proto-Oncogênicas c-ets/fisiologia , Proteínas Repressoras/fisiologia , Transcrição Gênica , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Sequência Consenso , Elementos Facilitadores Genéticos , Regulação Leucêmica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Estimativa de Kaplan-Meier , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/mortalidade , Prognóstico , Modelos de Riscos Proporcionais , Ligação Proteica , Proteoma , Proteômica , Regulador Transcricional ERG/fisiologia , Variante 6 da Proteína do Fator de Translocação ETS
14.
J Neurophysiol ; 117(2): 818-835, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27903637

RESUMO

Multivariate pattern analysis of functional magnetic resonance imaging (fMRI) data is widely used, yet the spatial scales and origin of neurovascular signals underlying such analyses remain unclear. We compared decoding performance for stimulus orientation and eye of origin from fMRI measurements in human visual cortex with predictions based on the columnar organization of each feature and estimated the spatial scales of patterns driving decoding. Both orientation and eye of origin could be decoded significantly above chance in early visual areas (V1-V3). Contrary to predictions based on a columnar origin of response biases, decoding performance for eye of origin in V2 and V3 was not significantly lower than that in V1, nor did decoding performance for orientation and eye of origin differ significantly. Instead, response biases for both features showed large-scale organization, evident as a radial bias for orientation, and a nasotemporal bias for eye preference. To determine whether these patterns could drive classification, we quantified the effect on classification performance of binning voxels according to visual field position. Consistent with large-scale biases driving classification, binning by polar angle yielded significantly better decoding performance for orientation than random binning in V1-V3. Similarly, binning by hemifield significantly improved decoding performance for eye of origin. Patterns of orientation and eye preference bias in V2 and V3 showed a substantial degree of spatial correlation with the corresponding patterns in V1, suggesting that response biases in these areas originate in V1. Together, these findings indicate that multivariate classification results need not reflect the underlying columnar organization of neuronal response selectivities in early visual areas.NEW & NOTEWORTHY Large-scale response biases can account for decoding of orientation and eye of origin in human early visual areas V1-V3. For eye of origin this pattern is a nasotemporal bias; for orientation it is a radial bias. Differences in decoding performance across areas and stimulus features are not well predicted by differences in columnar-scale organization of each feature. Large-scale biases in extrastriate areas are spatially correlated with those in V1, suggesting biases originate in primary visual cortex.


Assuntos
Mapeamento Encefálico , Imageamento por Ressonância Magnética , Acoplamento Neurovascular/fisiologia , Orientação/fisiologia , Detecção de Sinal Psicológico/fisiologia , Córtex Visual/diagnóstico por imagem , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Oxigênio/sangue , Reconhecimento Visual de Modelos/fisiologia , Estimulação Luminosa , Máquina de Vetores de Suporte , Córtex Visual/fisiologia , Vias Visuais/diagnóstico por imagem , Vias Visuais/fisiologia , Adulto Jovem
15.
Blood ; 125(12): 1890-900, 2015 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-25645357

RESUMO

Polycomb repressive complex 2 (PRC2) plays a key role in hematopoietic stem and progenitor cell (HSPC) function. Analyses of mouse mutants harboring deletions of core components have implicated PRC2 in fine-tuning multiple pathways that instruct HSPC behavior, yet how PRC2 is targeted to specific genomic loci within HSPCs remains unknown. Here we use short hairpin RNA-mediated knockdown to survey the function of PRC2 accessory factors that were defined in embryonic stem cells (ESCs) by testing the competitive reconstitution capacity of transduced murine HSPCs. We find that, similar to the phenotype observed upon depletion of core subunit Suz12, depleting Jarid2 enhances the competitive transplantation capacity of both fetal and adult mouse HSPCs. Furthermore, we demonstrate that depletion of JARID2 enhances the in vitro expansion and in vivo reconstitution capacity of human HSPCs. Gene expression profiling revealed common Suz12 and Jarid2 target genes that are enriched for the H3K27me3 mark established by PRC2. These data implicate Jarid2 as an important component of PRC2 that has a central role in coordinating HSPC function.


Assuntos
Regulação Neoplásica da Expressão Gênica , Complexo Repressor Polycomb 2/metabolismo , Animais , Antígenos CD34/metabolismo , Linhagem da Célula , Perfilação da Expressão Gênica , Hematopoese , Células-Tronco Hematopoéticas/citologia , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Fígado/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Fenótipo , RNA Interferente Pequeno/metabolismo , Células-Tronco/citologia
17.
J Neurophysiol ; 114(2): 1211-26, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26063774

RESUMO

Adaptation at early stages of sensory processing can be propagated to downstream areas. Such inherited adaptation is a potential confound for functional magnetic resonance imaging (fMRI) techniques that use selectivity of adaptation to infer neuronal selectivity. However, the relative contributions of inherited and intrinsic adaptation at higher cortical stages, and the impact of inherited adaptation on downstream processing, remain unclear. Using fMRI, we investigated how adaptation to visual motion direction and orientation influences visually evoked responses in human V1 and extrastriate visual areas. To dissociate inherited from intrinsic adaptation, we quantified the spatial specificity of adaptation for each visual area as a measure of the receptive field sizes of the area where adaptation originated, predicting that adaptation originating in V1 should be more spatially specific than adaptation intrinsic to extrastriate visual cortex. In most extrastriate visual areas, the spatial specificity of adaptation did not differ from that in V1, suggesting that adaptation originated in V1. Only in one extrastriate area-MT-was the spatial specificity of direction-selective adaptation significantly broader than in V1, consistent with a combination of inherited V1 adaptation and intrinsic MT adaptation. Moreover, inherited adaptation effects could be both facilitatory and suppressive. These results suggest that adaptation at early visual processing stages can have widespread and profound effects on responses in extrastriate visual areas, placing important constraints on the use of fMRI adaptation techniques, while also demonstrating a general experimental strategy for systematically dissociating inherited from intrinsic adaptation by fMRI.


Assuntos
Adaptação Fisiológica/fisiologia , Córtex Visual/fisiologia , Percepção Visual/fisiologia , Adolescente , Adulto , Mapeamento Encefálico , Circulação Cerebrovascular/fisiologia , Humanos , Imageamento por Ressonância Magnética , Modelos Neurológicos , Testes Neuropsicológicos , Oxigênio/sangue , Estimulação Luminosa/métodos , Tempo de Reação , Vias Visuais/fisiologia , Adulto Jovem
18.
Blood ; 121(19): 3838-42, S1-15, 2013 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-23509159

RESUMO

In an attempt to discover novel growth factors for hematopoietic stem and progenitor cells (HSPCs), we have assessed cytokine responses of cord blood (CB)-derived CD34(+) cells in a high-content growth factor screen. We identify the immunoregulatory chemokine (C-C motif) ligand 28 (CCL28) as a novel growth factor that directly stimulates proliferation of primitive hematopoietic cells from different ontogenetic origins. CCL28 enhances the functional progenitor cell content of cultured cells by stimulating cell cycling and induces gene expression changes associated with survival. Importantly, addition of CCL28 to cultures of purified putative hematopoietic stem cells (HSCs) significantly increases the ability of the cells to long-term repopulate immunodeficient mice compared with equivalent input numbers of fresh cells. Together, our findings identify CCL28 as a potent growth-promoting factor with the ability to support the in vitro and in vivo functional properties of cultured human hematopoietic cells.


Assuntos
Proliferação de Células , Quimiocinas CC/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Quimiocinas CC/genética , Quimiocinas CC/metabolismo , Quimiocinas CC/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Recém-Nascido , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Células-Tronco/fisiologia
19.
Foodborne Pathog Dis ; 12(3): 244-52, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25658711

RESUMO

Listeria monocytogenes is a foodborne pathogen responsible for a severe disease known as listeriosis. The European Centre for Disease Prevention and Control (ECDC) coordinates a network of national public health laboratories (NPHLs) in charge of typing clinical strains. In food, it is the European Union Reference Laboratory for L. monocytogenes (EURL Lm), which manages a network of National Reference Laboratories (NRLs). A pulsed-field gel electrophoresis (PFGE) standard operating procedure (EURL SOP) has been used routinely at the EURL Lm since 2007. The EURL Lm has recommended that NRLs use the EURL SOP, whereas the Statens Serum Institut (SSI), under contract for ECDC, requested that NPHLs use Halpins' SOP (HSOP) published in 2010 for the PulseNet USA network. An update of Halpins' SOP (uHSOP) was published in 2013. To facilitate the exchange of profiles among human and food European reference laboratories, it is crucial to ensure that the PFGE profiles obtained with these different SOPs are comparable. The aim here was to compare the EURL SOP with HSOP and uHSOP. The panel comprised 114 well-characterized SSI/EURL strains. All were characterized at the EURL using both the EURL SOP and uHSOP. Seventy of the 114 strains were also characterized at the SSI using HSOP. The EURL SOP and uHSOP produced indistinguishable combined (ApaI/AscI) profiles for the 114 strains tested. The EURL SOP and HSOP produced indistinguishable combined profiles for 69 of the 70 strains tested. One strain displayed for the AscI profile an additional low-intensity band at 184 kbp with HSOP. For this strain, SSI and EUR Lm had already observed the same profile from NPHLs and NRLs. However, this deviation is minor as it accounted for about 1% of all the 114 combined profiles. This study should facilitate the exchange of reproducible PFGE profiles among human and food reference laboratories.


Assuntos
Eletroforese em Gel de Campo Pulsado/normas , Listeria monocytogenes/classificação , Listeria monocytogenes/isolamento & purificação , Listeriose/microbiologia , Técnicas de Tipagem Bacteriana , Europa (Continente) , União Europeia , Microbiologia de Alimentos , Humanos , Listeriose/epidemiologia , Sorotipagem
20.
Curr Opin Hematol ; 21(4): 283-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24811164

RESUMO

PURPOSE OF REVIEW: The molecular principles regulating hematopoietic stem cells (HSCs) remain incompletely defined. In this review, we will discuss how RNA interference (RNAi) screening has emerged as a new and powerful tool to molecularly dissect various functional aspects of both normal and malignant HSCs, and how this may ultimately enable the discovery of novel therapeutic targets for clinical applications. RECENT FINDINGS: Advances in RNAi technology and the creation of short hairpin RNA retroviral and lentiviral vector-libraries have provided tools to perform broad forward genetic screens in primary mammalian hematopoietic cells. Recent studies have identified novel fate determinants in murine HSCs as well as potential targets for ex-vivo expansion of human HSCs. RNAi screens have further unraveled tumor suppressor genes associated with hematopoietic neoplasms as well as candidate therapeutic targets in leukemic cells. SUMMARY: RNAi screening is a feasible tool to discover novel molecules that regulate both normal and malignant HSCs, thus increasing our general understanding of the physiology and pathophysiology of hematopoiesis. Moreover, RNAi technology holds great promise for the discovery of specific targets for therapeutic interventions.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Ensaios de Triagem em Larga Escala , Interferência de RNA , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Animais , Diferenciação Celular/genética , Sobrevivência Celular/genética , Haploinsuficiência/genética , Células-Tronco Hematopoéticas/citologia , Humanos , Leucemia/genética , Leucemia/terapia , Camundongos , Terapia de Alvo Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA