Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-37546948

RESUMO

Most human pancreatic ductal adenocarcinoma (PDAC) are not infiltrated with cytotoxic T cells and are highly resistant to immunotherapy. Over 90% of PDAC have oncogenic KRAS mutations, and phosphoinositide 3-kinases (PI3Ks) are direct effectors of KRAS. Our previous study demonstrated that ablation of Pik3ca in KPC (KrasG12D; Trp53R172H; Pdx1-Cre) pancreatic cancer cells induced host T cells to infiltrate and completely eliminate the tumors in a syngeneic orthotopic implantation mouse model. Now, we show that implantation of Pik3ca-/- KPC (named αKO) cancer cells induces clonal expansion of cytotoxic T cells infiltrating the pancreatic tumors. To identify potential molecules that can regulate the activity of these anti-tumor T cells, we conducted an in vivo genome-wide gene-deletion screen using αKO cells implanted in the mouse pancreas. The result shows that deletion of propionyl-CoA carboxylase subunit B gene (Pccb) in αKO cells (named p-αKO) leads to immune evasion, tumor progression and death of host mice. Surprisingly, p-αKO tumors are still infiltrated with clonally expanded CD8+ T cells but they are inactive against tumor cells. However, blockade of PD-L1/PD1 interaction reactivated these clonally expanded T cells infiltrating p-αKO tumors, leading to slower tumor progression and improve survival of host mice. These results indicate that Pccb can modulate the activity of cytotoxic T cells infiltrating some pancreatic cancers and this understanding may lead to improvement in immunotherapy for this difficult-to-treat cancer.

2.
Invest Ophthalmol Vis Sci ; 64(13): 37, 2023 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-37870847

RESUMO

Purpose: The lens epithelium maintains the overall health of the organ. We used single-cell RNA sequencing (scRNA-seq) technology to assess transcriptional heterogeneity between cells in the postnatal day 2 (P2) epithelium and identify distinct epithelial cell subtypes. Analysis of these data was used to better understand lens growth, differentiation, and homeostasis on P2. Methods: scRNA-seq on P2 mouse lenses was performed using the 10x Genomics Chromium Single Cell 3' Kit (v3.1) and short-read Illumina sequencing. Sequence alignment and preprocessing of data were conducted using 10x Genomics Cell Ranger software. Seurat was employed for preprocessing, quality control, dimensionality reduction, and cell clustering, and Monocle was utilized for trajectory analysis to understand the developmental progression of the lens cells. CellChat and GO analyses were used to explore cell-cell communication networks and signaling interactions. Results: Lens epithelial cells (LECs) were divided into seven subclusters, classified by specific gene markers. The expression of crystallin, cell-cycle, and metabolic genes was not uniform, indicating distinct functional roles of LECs. Trajectory analysis predicted a bifurcation of differentiating and cycling cells from an Igfbp5+ progenitor pool. We also identified heterogeneity in signaling molecules and pathways, suggesting that cycling and progenitor subclusters have prominent roles in coordinating crosstalk. Conclusions: scRNA-seq corroborated many known markers of epithelial differentiation and proliferation while providing further insight into the pathways and genes directing these processes. Interestingly, we demonstrated that the developing epithelium can be divided into distinct subpopulations. These clusters reflect the transcriptionally diverse roles of the epithelium in proliferation, signaling, and maintenance.


Assuntos
Cristalino , Animais , Camundongos , Cristalino/metabolismo , Epitélio , Células Epiteliais/metabolismo , Diferenciação Celular , Análise de Sequência de RNA
3.
FASEB J ; 34(9): 12663-12676, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32761888

RESUMO

Phospholipase Cß1 is activated by Gαq to generate calcium signals in response to hormones and neurotransmitters. Besides carrying out this plasma membrane function, PLCß1 has a cytosolic population that helps to drive the differentiation of PC12 cells by inhibiting a nuclease that promotes RNA-induced silencing (C3PO). Here, we show that down-regulating PLCß1 or reducing its cytosolic population by activating Gαq to localize it to the plasma membrane returns differentiated PC12 and SK-N-SH cells to an undifferentiated state. In this state, PC12 cells have a spherical morphology, resume proliferation, and express the stem cell transcription factors nanog and Oct4. Similar changes are seen when C3PO is down-regulated. This return to a stem-like state is accompanied by shifts in multiple miR populations. Surprisingly, de-differentiation can be induced by extended stimulation of Gαq where cells return to a spherical morphology and levels of specific miRs return to their undifferentiated values. In complementary studies, we followed the real-time hydrolysis of a fluorescent-tagged miR in cells where PLCß1 or C3PO were down-regulated in PC12 cells and find substantial differences in miR processing in the undifferentiated and differentiated states. Taken together, our studies suggest that PLCß1, through its ability to regulate C3PO and endogenous miR populations, mediates the differentiation of two types of cultured neuronal cells.


Assuntos
Desdiferenciação Celular , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Fosfolipase C beta/metabolismo , Complexo de Inativação Induzido por RNA/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , MicroRNAs/metabolismo , Células PC12 , Interferência de RNA , Ratos , Transdução de Sinais
4.
Cell Signal ; 24(5): 1109-14, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22286107

RESUMO

α-Synuclein is a conserved protein that is a key component in neurodegenerative plaques [1,2]. α-Synuclein binds strongly to phospholipase Cß (PLCß) and promotes Ca2+ release in cells. Here, we show that expression of α-synuclein increases the cellular level of PLCß1 in two neuronal cell lines: PC12 and SK-N-S-SH. The increase in PLCß1 is not accompanied by changes in the level of RNA or in ubiquitination. Instead, we find that α-synuclein protects PLCß1 from trypsin digestion and from degradation by the Ca(+2) activated protease calpain. Calpain removes the C-terminal region of the enzyme which mediates activation by Gα(q). We find that in SK-N-SH cells, α-synuclein reduced degradation of PLCß1 by calpain during Ca2+ signaling allowing the enzyme to remain sensitive to Gα(q) activation. Taken together, our studies show that α-synuclein protects the integrity of PLCß1 and its ability to be activated by Gα(q), which may in turn impact Ca2+ signaling.


Assuntos
Fosfolipase C beta/metabolismo , alfa-Sinucleína/fisiologia , Animais , Calcimicina/farmacologia , Calpaína/antagonistas & inibidores , Calpaína/química , Calpaína/metabolismo , Ativadores de Enzimas/farmacologia , Células HEK293 , Humanos , Células PC12 , Fragmentos de Peptídeos/química , Fosfolipase C beta/química , Fosfolipase C beta/genética , Ligação Proteica , Proteólise , Ratos , Transcrição Gênica , Tripsina/química , Ubiquitinação , alfa-Sinucleína/química , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
5.
Circ Arrhythm Electrophysiol ; 4(5): 753-60, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21828312

RESUMO

BACKGROUND: After the recent cloning of light-sensitive ion channels and their expression in mammalian cells, a new field, optogenetics, emerged in neuroscience, allowing for precise perturbations of neural circuits by light. However, functionality of optogenetic tools has not been fully explored outside neuroscience, and a nonviral, nonembryogenesis-based strategy for optogenetics has not been shown before. METHODS AND RESULTS: We demonstrate the utility of optogenetics to cardiac muscle by a tandem cell unit (TCU) strategy, in which nonexcitable cells carry exogenous light-sensitive ion channels, and, when electrically coupled to cardiomyocytes, produce optically excitable heart tissue. A stable channelrhodopsin2 (ChR2)-expressing cell line was developed, characterized, and used as a cell delivery system. The TCU strategy was validated in vitro in cell pairs with adult canine myocytes (for a wide range of coupling strengths) and in cardiac syncytium with neonatal rat cardiomyocytes. For the first time, we combined optical excitation and optical imaging to capture light-triggered muscle contractions and high-resolution propagation maps of light-triggered electric waves, found to be quantitatively indistinguishable from electrically triggered waves. CONCLUSIONS: Our results demonstrate feasibility to control excitation and contraction in cardiac muscle by light, using the TCU approach. Optical pacing in this case uses less energy, offers superior spatiotemporal control and remote access and can serve not only as an elegant tool in arrhythmia research but may form the basis for a new generation of light-driven cardiac pacemakers and muscle actuators. The TCU strategy is extendable to (nonviral) stem cell therapy and is directly relevant to in vivo applications.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Luz , Contração Muscular/fisiologia , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Comunicação Celular/fisiologia , Channelrhodopsins , Técnicas de Cocultura , Cães , Estimulação Elétrica , Estudos de Viabilidade , Células HEK293 , Humanos , Rim/citologia , Rim/metabolismo , Miócitos Cardíacos/citologia , Proteínas do Tecido Nervoso/genética , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Transfecção
6.
FASEB J ; 17(2): 330-2, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12490549

RESUMO

The ether-a-go-go-related genes (erg) are expressed in tissues other than heart and brain, in which human erg (HERG) K+ channels are known to regulate the repolarization of heart action potentials and neuronal spike-frequency accommodation. We provide evidence that erg1 transcripts and ERG proteins are present in rat chromaffin cells in which we could isolate a K+ current that was biophysically and pharmacologically similar to the ERG current. Firing frequency and catecholamine release were analyzed at the single-cell level by means of perforated patch-clamp and carbon fiber electrochemical detection. It was found that the blocking of ERG, KATP, and KCa channels led to hyperexcitability and an increase in catecholamine release. Combined immunocytochemical experiments with antibodies directed against phenylethanolamine N-methyltransferase and ERG channels suggested expression of these channels in epinephrine- but not in norepinephrine-containing cells. It is concluded that, in addition to being crucial in regulating the QT period in the heart, ERG channels play a role in modulating epinephrine, a fundamental neurotransmitter shaping cardiac function. This finding suggests that the sudden death phenotype associated with LQT2 syndrome mutations may be the result of an emotionally triggered increase in epinephrine in a long-QT running heart.


Assuntos
Células Cromafins/efeitos dos fármacos , Epinefrina/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Animais , Benzimidazóis/farmacologia , Cálcio/metabolismo , Catecolaminas/metabolismo , Células Cromafins/metabolismo , Células Cromafins/fisiologia , Relação Dose-Resposta a Droga , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Expressão Gênica , Histamina/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Nicotina/farmacologia , Técnicas de Patch-Clamp , Canais de Potássio/genética , Canais de Potássio/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Ratos , Sulfanilamidas/farmacologia
7.
J Neurosci ; 22(9): 3414-25, 2002 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11978818

RESUMO

Native rat lactotrophs express thyrotrophin-releasing hormone-dependent K+ currents consisting of fast and slow deactivating components that are both sensitive to the class III anti-arrhythmic drugs that block the eag-related gene (ERG) K+ current (I(ERG)). Here we describe in MMQ prolactin-releasing pituitary cells the isolation of the slowly deactivating long-lasting component (I(ERGS)), which, unlike the fast component (I(ERGF)), is insensitive to verapamil 2 microm but sensitive to a novel scorpion toxin (ErgTx-2) that hardly affects I(ERGF). The time constants of I(ERGS) activation, deactivation, and recovery from inactivation are more than one order of magnitude greater than in I(ERGF), and the voltage-dependent inactivation is left-shifted by approximately 25 mV. The very slow MMQ firing frequency (approximately 0.2 Hz) investigated in perforated patch is increased approximately four times by anti-arrhythmic agents, by ErgTx-2, and by the abrupt I(ERGS) deactivation. Prolactin secretion in the presence of anti-arrhythmics is three- to fourfold higher in comparison with controls. We provide evidence from I(ERGS) and I(ERGF) simulations in a firing model cell to indicate that only I(ERGS) has an accommodating role during the experimentally observed very slow firing. Thus, we suggest that I(ERGS) potently modulates both firing and prolactin release in lactotroph cells.


Assuntos
Proteínas de Transporte de Cátions , Proteínas de Membrana Transportadoras , Hipófise/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Potássio/metabolismo , Prolactina/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Antiarrítmicos/farmacologia , Encéfalo/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular , Simulação por Computador , Relação Dose-Resposta a Droga , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Modelos Neurológicos , Técnicas de Patch-Clamp , Hipófise/citologia , Hipófise/efeitos dos fármacos , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/genética , RNA/genética , RNA/metabolismo , Ratos , Venenos de Escorpião/isolamento & purificação , Venenos de Escorpião/farmacologia , Tretinoína/metabolismo , Verapamil/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA