Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 149
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 175(6): 1546-1560.e17, 2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30500537

RESUMO

Mammalian folate metabolism is comprised of cytosolic and mitochondrial pathways with nearly identical core reactions, yet the functional advantages of such an organization are not well understood. Using genome-editing and biochemical approaches, we find that ablating folate metabolism in the mitochondria of mammalian cell lines results in folate degradation in the cytosol. Mechanistically, we show that QDPR, an enzyme in tetrahydrobiopterin metabolism, moonlights to repair oxidative damage to tetrahydrofolate (THF). This repair capacity is overwhelmed when cytosolic THF hyperaccumulates in the absence of mitochondrially produced formate, leading to THF degradation. Unexpectedly, we also find that the classic antifolate methotrexate, by inhibiting its well-known target DHFR, causes even more extensive folate degradation in nearly all tested cancer cell lines. These findings shed light on design features of folate metabolism, provide a biochemical basis for clinically observed folate deficiency in QDPR-deficient patients, and reveal a hitherto unknown and unexplored cellular effect of methotrexate.


Assuntos
Carbono/metabolismo , Citosol/metabolismo , Formiatos/metabolismo , Mitocôndrias/metabolismo , Neoplasias/metabolismo , Tetra-Hidrofolatos/metabolismo , Citosol/patologia , Células HCT116 , Células HeLa , Humanos , Células MCF-7 , Metotrexato/farmacocinética , Metotrexato/farmacologia , Mitocôndrias/patologia , Proteínas Mitocondriais/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Tetra-Hidrofolato Desidrogenase/metabolismo
2.
Nat Rev Mol Cell Biol ; 18(11): 671-684, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28852221

RESUMO

Fatty acids are the most efficient substrates for energy production in vertebrates and are essential components of the lipids that form biological membranes. Synthesis of triacylglycerols from non-esterified free fatty acids (FFAs) combined with triacylglycerol storage represents a highly efficient strategy to stockpile FFAs in cells and prevent FFA-induced lipotoxicity. Although essentially all vertebrate cells have some capacity to store and utilize triacylglycerols, white adipose tissue is by far the largest triacylglycerol depot and is uniquely able to supply FFAs to other tissues. The release of FFAs from triacylglycerols requires their enzymatic hydrolysis by a process called lipolysis. Recent discoveries thoroughly altered and extended our understanding of lipolysis. This Review discusses how cytosolic 'neutral' lipolysis and lipophagy, which utilizes 'acid' lipolysis in lysosomes, degrade cellular triacylglycerols as well as how these pathways communicate, how they affect lipid metabolism and energy homeostasis and how their dysfunction affects the pathogenesis of metabolic diseases. Answers to these questions will likely uncover novel strategies for the treatment of prevalent metabolic diseases.


Assuntos
Tecido Adiposo Branco/metabolismo , Citosol/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Lipólise , Lisossomos/metabolismo , Triglicerídeos/metabolismo , Tecido Adiposo Branco/patologia , Animais , Citosol/patologia , Metabolismo Energético , Humanos , Lisossomos/patologia , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia , Doenças Metabólicas/terapia
3.
Nature ; 579(7799): 433-437, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32132706

RESUMO

Mitochondrial fidelity is tightly linked to overall cellular homeostasis and is compromised in ageing and various pathologies1-3. Mitochondrial malfunction needs to be relayed to the cytosol, where an integrated stress response is triggered by the phosphorylation of eukaryotic translation initiation factor 2α (eIF2α) in mammalian cells4,5. eIF2α phosphorylation is mediated by the four eIF2α kinases GCN2, HRI, PERK and PKR, which are activated by diverse types of cellular stress6. However, the machinery that communicates mitochondrial perturbation to the cytosol to trigger the integrated stress response remains unknown1,2,7. Here we combine genome engineering and haploid genetics to unbiasedly identify genes that affect the induction of C/EBP homologous protein (CHOP), a key factor in the integrated stress response. We show that the mitochondrial protease OMA1 and the poorly characterized protein DELE1, together with HRI, constitute the missing pathway that is triggered by mitochondrial stress. Mechanistically, stress-induced activation of OMA1 causes DELE1 to be cleaved into a short form that accumulates in the cytosol, where it binds to and activates HRI via its C-terminal portion. Obstruction of this pathway can be beneficial or adverse depending on the type of mitochondrial perturbation. In addition to the core pathway components, our comparative genetic screening strategy identifies a suite of additional regulators. Together, these findings could be used to inform future strategies to modulate the cellular response to mitochondrial dysfunction in the context of human disease.


Assuntos
Citosol/metabolismo , Citosol/patologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Mitocondriais/metabolismo , Ativação Enzimática , Fator de Iniciação 2 em Eucariotos/metabolismo , Genoma Humano/genética , Humanos , Metaloendopeptidases/metabolismo , Mitocôndrias/enzimologia , Fosforilação , Ligação Proteica , Estresse Fisiológico , Fator de Transcrição CHOP/metabolismo , eIF-2 Quinase/metabolismo
4.
Circulation ; 141(1): 42-66, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31887080

RESUMO

BACKGROUND: Sporadic aortic aneurysm and dissection (AAD), caused by progressive aortic smooth muscle cell (SMC) loss and extracellular matrix degradation, is a highly lethal condition. Identifying mechanisms that drive aortic degeneration is a crucial step in developing an effective pharmacologic treatment to prevent disease progression. Recent evidence has indicated that cytosolic DNA and abnormal activation of the cytosolic DNA sensing adaptor STING (stimulator of interferon genes) play a critical role in vascular inflammation and destruction. Here, we examined the involvement of this mechanism in aortic degeneration and sporadic AAD formation. METHODS: The presence of cytosolic DNA in aortic cells and activation of the STING pathway were examined in aortic tissues from patients with sporadic ascending thoracic AAD. The role of STING in AAD development was evaluated in Sting-deficient (Stinggt/gt) mice in a sporadic AAD model induced by challenging mice with a combination of a high-fat diet and angiotensin II. We also examined the direct effects of STING on SMC death and macrophage activation in vitro. RESULTS: In human sporadic AAD tissues, we observed the presence of cytosolic DNA in SMCs and macrophages and significant activation of the STING pathway. In the sporadic AAD model, Stinggt/gt mice showed significant reductions in challenge-induced aortic enlargement, dissection, and rupture in both the thoracic and abdominal aortic regions. Single-cell transcriptome analysis revealed that aortic challenge in wild-type mice induced the DNA damage response, the inflammatory response, dedifferentiation and cell death in SMCs, and matrix metalloproteinase expression in macrophages. These changes were attenuated in challenged Stinggt/gt mice. Mechanistically, nuclear and mitochondrial DNA damage in SMCs and the subsequent leak of DNA to the cytosol activated STING signaling, which induced cell death through apoptosis and necroptosis. In addition, DNA from damaged SMCs was engulfed by macrophages in which it activated STING and its target interferon regulatory factor 3, which directly induced matrix metalloproteinase-9 expression. We also found that pharmacologically inhibiting STING activation partially prevented AAD development. CONCLUSIONS: Our findings indicate that the presence of cytosolic DNA and subsequent activation of cytosolic DNA sensing adaptor STING signaling represent a key mechanism in aortic degeneration and that targeting STING may prevent sporadic AAD development.


Assuntos
Dissecção Aórtica/metabolismo , Ruptura Aórtica/metabolismo , Citosol/metabolismo , DNA/metabolismo , Proteínas de Membrana/metabolismo , Transdução de Sinais , Dissecção Aórtica/genética , Dissecção Aórtica/patologia , Animais , Ruptura Aórtica/genética , Ruptura Aórtica/patologia , Citosol/patologia , DNA/genética , Feminino , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout
5.
Cancer Sci ; 112(1): 133-143, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33067904

RESUMO

To select the most efficient chemical to induce apoptosis in leukemia cells, a multidrug screen was applied on bone marrow mononuclear cells from chronic myeloid leukemia (CML) patients. Oprozomib (Cpd 21) was chosen for the subsequent experiments. The isobaric tags for relative and absolute quantitation (iTRAQ) was then performed to identify the responsible pathway relative to apoptosis and the results showed that endoplasmic reticulum (ER) chaperones were upregulated. Apoptosis was attributed to a joint effect of calcium leakage andPERK and IRE1α phosphorylation. The PERK branch was responsible for the first wave of cell death that occurred within 24 hours. The later wave of apoptosis was mediated by IRE1α, which transmit apoptotic signals through the ASK-JNK-BIM axis. Release of Ca2+ from ER into cytosol resulted in activation of calpain, which, in turn, cleaved caspase-12. Our data also explained the selective killing effects of oprozomib on CML cells, which relied on proteasome activity. The present study demonstrated that prolonged inhibition of proteasome to trigger unfolded protein response could be an alternative strategy for treating CML in light of tyrosine kinase inhibitors resistance.


Assuntos
Morte Celular/efeitos dos fármacos , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Oligopeptídeos/farmacologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Apoptose/genética , Cálcio/metabolismo , Morte Celular/genética , Linhagem Celular Tumoral , Citosol/efeitos dos fármacos , Citosol/patologia , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/genética , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/genética , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Resposta a Proteínas não Dobradas/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
6.
J Virol ; 94(11)2020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-32161173

RESUMO

The nonenveloped polyomavirus simian virus 40 (SV40) must penetrate the host endoplasmic reticulum (ER) membrane to enter the cytosol in order to promote infection. How this is accomplished is not entirely clear. Here, we demonstrate that the cytosolic chaperone Ubiquilin4 (Ubqln4) binds directly to the ER membrane J proteins B12 and B14. Strategically localized at the ER-cytosol interface, Ubqln4 captures SV40 emerging from the ER, thereby facilitating escape of the virus from the ER into the cytosol, which leads to infection. Strikingly, Ubqln4 engages the J proteins in a J-domain-independent manner, in contrast to the previously reported Hsc70-Hsp105-SGTA-Bag2 cytosolic complex that also mediates SV40 ER-to-cytosol transport. Our results also reveal that the H domain and STI1 motif (1-2) of Ubqln4 support J protein binding, essential for SV40 infection. Together, these data further clarify the molecular basis by which a nonenveloped virus escapes a host membrane during infectious entry.IMPORTANCE How a nonenveloped virus escapes from a host membrane to promote infection remains enigmatic. In the case of the nonenveloped polyomavirus SV40, penetration of the ER membrane to reach the cytosol is a decisive virus infection step. In this study, we found a new host factor called Ubqln4 that facilitates escape of SV40 from the ER into the cytosol, thereby providing a path for the virus to enter the nucleus to cause infection.


Assuntos
Proteínas de Transporte/metabolismo , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas Nucleares/metabolismo , Infecções por Polyomavirus/metabolismo , Vírus 40 dos Símios/metabolismo , Motivos de Aminoácidos , Transporte Biológico Ativo/genética , Proteínas de Transporte/genética , Linhagem Celular , Citosol/patologia , Citosol/virologia , Retículo Endoplasmático/genética , Retículo Endoplasmático/patologia , Retículo Endoplasmático/virologia , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/genética , Infecções por Polyomavirus/genética , Infecções por Polyomavirus/patologia , Domínios Proteicos , Vírus 40 dos Símios/genética
7.
J Pineal Res ; 70(1): e12677, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32621295

RESUMO

This commentary reviews the concept of the circadian melatonin rhythm playing an essential role in reducing the development of diseases such as solid tumors which adopt cytosolic aerobic glycolysis (Warburg effect) to support their enhanced metabolism. Experimental data show that solid mammary tumors depend on aerobic glycolysis during the day but likely revert to mitochondrial oxidative phosphorylation at night for ATP production. This conversion of diseased cells during the day to a healthier phenotype at night occurs under control of the circulating melatonin rhythm. When the nocturnal melatonin rise is inhibited by light exposure at night, cancer cells function in the diseased state 24/7. The ability of melatonin to switch cancer cells as well as other diseased cells, for example, Alzheimer disease, fibrosis, hyperactivation of macrophages, etc, from aerobic glycolysis to mitochondrial oxidative phosphorylation may be a basic protective mechanism to reduce pathologies.


Assuntos
Ritmo Circadiano , Citosol/metabolismo , Glicólise , Melatonina/metabolismo , Mitocôndrias/metabolismo , Neoplasias/metabolismo , Fosforilação Oxidativa , Animais , Citosol/patologia , Humanos , Mitocôndrias/patologia , Neoplasias/patologia , Fatores de Tempo , Efeito Warburg em Oncologia
8.
Molecules ; 26(12)2021 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-34208178

RESUMO

The heat shock protein (HSP) 70 is considered the main hallmark in preclinical studies to stain the peri-infarct region defined area penumbra in preclinical models of brain ischemia. This protein is also considered as a potential disease modifier, which may improve the outcome of ischemic damage. In fact, the molecule HSP70 acts as a chaperonine being able to impact at several level the homeostasis of neurons. Despite being used routinely to stain area penumbra in light microscopy, the subcellular placement of this protein within area penumbra neurons, to our knowledge, remains undefined. This is key mostly when considering studies aimed at deciphering the functional role of this protein as a determinant of neuronal survival. The general subcellular placement of HSP70 was grossly reported in studies using confocal microscopy, although no direct visualization of this molecule at electron microscopy was carried out. The present study aims to provide a direct evidence of HSP70 within various subcellular compartments. In detail, by using ultrastructural morphometry to quantify HSP70 stoichiometrically detected by immuno-gold within specific organelles we could compare the compartmentalization of the molecule within area penumbra compared with control brain areas. The study indicates that two cell compartments in control conditions own a high density of HSP70, cytosolic vacuoles and mitochondria. In these organelles, HSP70 is present in amount exceeding several-fold the presence in the cytosol. Remarkably, within area penumbra a loss of such a specific polarization is documented. This leads to the depletion of HSP70 from mitochondria and mostly cell vacuoles. Such an effect is expected to lead to significant variations in the ability of HSP70 to exert its physiological roles. The present findings, beyond defining the neuronal compartmentalization of HSP70 within area penumbra may lead to a better comprehension of its beneficial/detrimental role in promoting neuronal survival.


Assuntos
Isquemia Encefálica/metabolismo , Citosol/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Vacúolos/metabolismo , Animais , Isquemia Encefálica/patologia , Morte Celular/fisiologia , Citosol/patologia , Modelos Animais de Doenças , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Microscopia Eletrônica de Varredura , Mitocôndrias/patologia , Neurônios/patologia , Vacúolos/patologia
9.
Neurochem Res ; 45(6): 1354-1364, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31280399

RESUMO

Motor neuron disease (MND) is a progressive neurodegenerative disease with no effective treatment. One of the principal pathological hallmarks is the deposition of TAR DNA binding protein 43 (TDP-43) in cytoplasmic inclusions. TDP-43 aggregation occurs in both familial and sporadic MND; however, the mechanism of endogenous TDP-43 aggregation in disease is incompletely understood. This study focused on the induction of cytoplasmic accumulation of endogenous TDP-43 in the motor neuronal cell line NSC-34. The endoplasmic reticulum (ER) stressor tunicamycin induced casein kinase 1 (CK1)-dependent cytoplasmic accumulation of endogenous TDP-43 in differentiated NSC-34 cells, as seen by immunocytochemistry. Immunoblotting showed that induction of ER stress had no effect on abundance of TDP-43 or phosphorylated TDP-43 in the NP-40/RIPA soluble fraction. However, there were significant increases in abundance of TDP-43 and phosphorylated TDP-43 in the NP-40/RIPA-insoluble, urea-soluble fraction, including high molecular weight species. In all cases, these increases were lowered by CK1 inhibition. Thus ER stress signalling, as induced by tunicamycin, causes CK1-dependent phosphorylation of TDP-43 and its consequent cytosolic accumulation.


Assuntos
Caseína Quinase I/biossíntese , Citosol/metabolismo , Proteínas de Ligação a DNA/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Corpos de Inclusão/metabolismo , Neurônios Motores/metabolismo , Antibacterianos/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Citosol/efeitos dos fármacos , Citosol/patologia , Relação Dose-Resposta a Droga , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Indução Enzimática/fisiologia , Humanos , Corpos de Inclusão/efeitos dos fármacos , Corpos de Inclusão/patologia , Doença dos Neurônios Motores/induzido quimicamente , Doença dos Neurônios Motores/metabolismo , Doença dos Neurônios Motores/patologia , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Tunicamicina/toxicidade
10.
J Biol Chem ; 293(40): 15581-15593, 2018 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-30143534

RESUMO

PolyQ-expanded huntingtin (mHtt) variants form aggregates, termed inclusion bodies (IBs), in individuals with and models of Huntington's disease (HD). The role of IB versus diffusible mHtt in neurotoxicity remains unclear. Using a ponasterone (PA)-inducible cell model of HD, here we evaluated the effects of heat shock on the appearance and functional outcome of Htt103QExon1-EGFP expression. Quantitative image analysis indicated that 80-90% of this mHtt protein initially appears as "diffuse" signals in the cytosol, with IBs forming at high mHtt expression. A 2-h heat shock during the PA induction reduced the diffuse signal, but greatly increased mHtt IB formation in both cytosol and nucleus. Dose- and time-dependent mHtt expression suggested that nucleated polymerization drives IB formation. RNA-mediated knockdown of heat shock protein 70 (HSP70) and heat shock cognate 70 protein (HSC70) provided evidence for their involvement in promoting diffuse mHtt to form IBs. Reporter gene assays assessing the impacts of diffuse versus IB mHtt showed concordance of diffuse mHtt expression with the repression of heat shock factor 1, cAMP-responsive element-binding protein (CREB), and NF-κB activity. CREB repression was reversed by heat shock coinciding with mHtt IB formation. In an embryonic striatal neuron-derived HD model, the chemical chaperone sorbitol similarly promoted the structuring of diffuse mHtt into IBs and supported cell survival under stress. Our results provide evidence that mHtt IB formation is a chaperone-supported cellular coping mechanism that depletes diffusible mHtt conformers, alleviates transcription factor dysfunction, and promotes neuron survival.


Assuntos
Fatores de Transcrição de Choque Térmico/genética , Resposta ao Choque Térmico , Proteína Huntingtina/genética , Doença de Huntington/genética , Corpos de Inclusão/metabolismo , Neurônios/metabolismo , Animais , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Citosol/patologia , Ecdisterona/análogos & derivados , Ecdisterona/farmacologia , Embrião de Mamíferos , Regulação da Expressão Gênica , Proteínas de Choque Térmico HSC70/genética , Proteínas de Choque Térmico HSC70/metabolismo , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Fatores de Transcrição de Choque Térmico/metabolismo , Proteína Huntingtina/metabolismo , Doença de Huntington/induzido quimicamente , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Corpos de Inclusão/química , Corpos de Inclusão/efeitos dos fármacos , Modelos Biológicos , Mutação , NF-kappa B/genética , NF-kappa B/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Células PC12 , Cultura Primária de Células , Ratos , Sorbitol/farmacologia
11.
Anal Chem ; 91(14): 8772-8776, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31184112

RESUMO

A multicellular tumor aggregate, known as a spheroid, is an indispensable tool to study cancer biology. Owing to its three-dimensional organization, a spheroid exhibits an inherent gradient of nutrients, oxygen, and metabolites within itself. The spheroid provides culture conditions that resemble the microenvironment of certain cancer cells and causes these cells to acquire characteristics relevant to tumors in our body. However, site-specific gene expression analysis in an intact spheroid with single-cell resolution has not been explored. Recently, some types of electrochemical syringes were developed to extract cellular materials from living single cells for transcriptomic analysis. Here, we investigated whether an electrochemical syringe could be used to evaluate site-specific gene expression in a spheroid. A small amount of cytosol (roughly 540-1480 fL, less than the volume of a single cell) was successfully collected from the first, second, and third layers of the spheroid using an electrochemical syringe without causing damage to the spheroid architecture. We found that the CCNB1 and CCNA2 expression levels were different between the surface and the average of the entire spheroid, indicating that there are heterogeneous cellular functions across different regions of the spheroid. This method provides opportunities to improve our understanding of spatial gene expression of single cells in a three-dimensional environment.


Assuntos
Citosol/patologia , Neoplasias/patologia , Análise de Célula Única , Manejo de Espécimes , Esferoides Celulares/patologia , Citosol/metabolismo , Técnicas Eletroquímicas/instrumentação , Desenho de Equipamento , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Neoplasias/genética , Análise de Célula Única/instrumentação , Manejo de Espécimes/instrumentação , Esferoides Celulares/metabolismo , Seringas , Microambiente Tumoral
12.
FASEB J ; 32(5): 2891-2898, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29401590

RESUMO

Cells have developed lineage-specific mechanisms to control proliferation and drive morphologic changes upon differentiation. A hallmark of differentiation is the assembly of signaling molecules that transduce extracellular signals, such as the production of the G protein-regulated enzyme phospholipase Cß (PLCß), which generates calcium signals from sensory stimuli. We found that in most cancerous cell lines there is positive correlation between PLCß1 levels and cell proliferation. In cells of neuronal lineage, however, reducing PLCß1 levels increases the rate of proliferation. Using a combination of biochemical and biophysical methods, we find that, in the G1 phase, a cytosolic population of PLCß1 associates with cyclin-dependent kinase 16 (CDK16), a neuron-specific enzyme that is activated by cyclin Y to inactivate the antioncogenic protein p27Kip1. Binding of PLCß1 directly inhibits CDK16 activity and in turn reduces the ability of cells to enter the S phase. Activation of Gαq by carbachol causes movement of PLCß from the cytosol to the plasma membrane, reducing its association with CDK16. Similarly, the overexpression of activated Gαq moves PLCß1 to the membrane, reverses G1 arrest, and promotes proliferation, thereby connecting external stimuli with cell proliferation. Our results present a model in which the transient high expression of PLCß1 that occurs at the onset of differentiation arrests cells in the G1 phase through its association with CDK16 and allows CDK16 to transition to its postmitotic function of neurite outgrowth and trafficking of synaptic vesicles. The novel role of PLCß1 in neuronal cell proliferation offers a unique interaction that can be manipulated to guide cells into a neuronal phenotype or to develop therapies for neuroblastomas.-Garwain, O., Valla, K., Scarlata, S. Phospholipase Cß1 regulates proliferation of neuronal cells.


Assuntos
Fase G1 , Regulação Enzimológica da Expressão Gênica , Neuritos/enzimologia , Fosfolipase C beta/biossíntese , Fase S , Animais , Membrana Celular/enzimologia , Membrana Celular/genética , Membrana Celular/patologia , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Citosol/enzimologia , Citosol/patologia , Neuritos/patologia , Neuroblastoma/enzimologia , Neuroblastoma/genética , Neuroblastoma/patologia , Neuroblastoma/terapia , Células PC12 , Fosfolipase C beta/genética , Ratos
13.
Int J Mol Sci ; 20(3)2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30717258

RESUMO

Primary liver cancer comprises a diverse group of liver tumors. The heterogeneity of these tumors is seen as one of the obstacles to finding an effective therapy. The Hippo pathway, with its downstream transcriptional co-activator Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), has a decisive role in the carcinogenesis of primary liver cancer. Therefore, we examined the expression pattern of YAP and TAZ in 141 patients with hepatocellular carcinoma keratin 19 positive (HCC K19⁺), hepatocellular carcinoma keratin 19 negative (HCC K19-), combined hepatocellular⁻cholangiocarcinoma carcinoma (cHCC-CCA), or cholangiocarcinoma (CCA). All cHCC-CCA and CCA patients showed high expression levels for YAP and TAZ, while only some patients of the HCC group were positive. Notably, we found that a histoscore of both markers is useful in the challenging diagnosis of cHCC-CCA. In addition, positivity for YAP and TAZ was observed in the hepatocellular and cholangiocellular components of cHCC-CCA, which suggests a single cell origin in cHCC-CCA. Within the K19- HCC group, our results demonstrate that the expression of YAP is a statistically significant predictor of poor prognosis when observed in the cytoplasm. Nuclear expression of TAZ is an even more specific and independent predictor of poor disease-free survival and overall survival of K19- HCC patients. Our results thus identify different levels of YAP/TAZ expression in various liver cancers that can be used for diagnostics.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias dos Ductos Biliares/genética , Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/genética , Colangiocarcinoma/genética , Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias Hepáticas/genética , Fosfoproteínas/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Idoso , Neoplasias dos Ductos Biliares/diagnóstico , Neoplasias dos Ductos Biliares/mortalidade , Neoplasias dos Ductos Biliares/patologia , Biomarcadores Tumorais/metabolismo , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/mortalidade , Carcinoma Hepatocelular/patologia , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Colangiocarcinoma/diagnóstico , Colangiocarcinoma/mortalidade , Colangiocarcinoma/patologia , Citosol/metabolismo , Citosol/patologia , Feminino , Heterogeneidade Genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Estimativa de Kaplan-Meier , Queratina-19/deficiência , Queratina-19/genética , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , Fosfoproteínas/metabolismo , Prognóstico , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Transdução de Sinais , Transativadores , Fatores de Transcrição , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Proteínas de Sinalização YAP
14.
Biochim Biophys Acta Mol Basis Dis ; 1863(11): 2964-2972, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28754452

RESUMO

Dilated cardiomyopathy (DCM) is cardiac disease characterized by increased left ventricular chamber volume and decreased systolic function. DCM patient-specific human induced-pluripotent stem cells-derived cardiomyocytes (DCM-hiPSC-CMs) were generated. We found that uniaxial stretch elicited a cytosolic [Ca2+]i rise in hiPSC-CMs. Compared to control-hiPSC-CMs, DCM-hiPSC-CMs displayed a greater magnitude of [Ca2+]i responses to the cell stretch of 10-15% elongation in length. This stretch-induced [Ca2+]i rise was abolished by removal of extracellular Ca2+ and markedly attenuated by TRPV4 inhibitors HC-067047 and RN-1734. Application of nifedipine and tranilast also reduced the [Ca2+]i response but to a lesser degree. Moreover, the augmented [Ca2+]i was decreased by cytochalasin D treatment. Taken together, our study for the first time demonstrated an abnormal TRPV4-related mechanosensitive Ca2+ signaling in DCM-hiPSC-CMs.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Cardiomiopatia Dilatada/metabolismo , Citosol/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Mecanotransdução Celular , Miócitos Cardíacos/metabolismo , Canais de Cátion TRPV/metabolismo , Cardiomiopatia Dilatada/patologia , Citosol/patologia , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Masculino , Miócitos Cardíacos/patologia
15.
J Neurooncol ; 135(3): 443-452, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28849427

RESUMO

A unique feature in several non-CNS-tumors is the overexpression of heat shock protein 70 (Hsp70, HSPA1A) in the cytosol, but also its unusual plasma membrane expression and release. Although in gliomas, cytosolic Hsp70 levels are not associated with histological grading, the role of membrane bound and released Hsp70 is still completely unknown. Membrane bound as well as cytosolic Hsp70 can be detected in viable tumor cells with the monoclonal antibody (mAb) cmHsp70.1. Herein, we analysed membrane bound Hsp70 levels in primary and secondary gliomas of different grades and on isolated glioma subpopulations (endothelial cells, CD133-positive cells, primary cultures) by immunohistochemistry and flow cytometry using cmHsp70.1 mAb. Extracellular Hsp70 was determined by a commercial Hsp70 sandwich ELISA (R&D) in plasma samples of glioblastoma patients and healthy volunteers. We found an overexpression of Hsp70 in primary glioblastomas compared to low-grade, anaplastic, or secondary gliomas as determined by immunohistochemistry. Especially in flow cytometry, a strong plasma membrane Hsp70 expression was only observed in primary but not secondary glioblastomas. Within the heterogeneous tumor mass, CD133-positive tumor-initiating and primary glioblastoma cells showed a high membrane Hsp70 expression density, whereas endothelial cells, isolated from glioblastoma tissues only showed a weak staining pattern. Also in plasma samples, secreted Hsp70 protein was significantly increased in patients harbouring primary glioblastomas compared to those with secondary and low grade glioblastomas. Taken together, we show for the first time that cytosolic, membrane bound and extracellular Hsp70 is uniquely overexpressed in primary glioblastomas.


Assuntos
Neoplasias Encefálicas/metabolismo , Membrana Celular/metabolismo , Citosol/metabolismo , Espaço Extracelular/metabolismo , Glioblastoma/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Antígeno AC133/metabolismo , Biomarcadores/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/cirurgia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Membrana Celular/patologia , Estudos de Coortes , Citosol/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Epilepsia/metabolismo , Epilepsia/patologia , Epilepsia/cirurgia , Feminino , Glioblastoma/genética , Glioblastoma/patologia , Glioblastoma/terapia , Humanos , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Curva ROC , Sarcoma de Células Pequenas
16.
Neurobiol Dis ; 85: 1-10, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26459111

RESUMO

Expression of the frontotemporal dementia-related tau mutation, P301L, at physiological levels in adult mouse brain (KI-P301L mice) results in overt hypophosphorylation of tau and age-dependent alterations in axonal mitochondrial transport in peripheral nerves. To determine the effects of P301L tau expression in the central nervous system, we examined the kinetics of mitochondrial axonal transport and tau phosphorylation in primary cortical neurons from P301L knock-in (KI-P301L) mice. We observed a significant 50% reduction in the number of mitochondria in the axons of cortical neurons cultured from KI-P301L mice compared to wild-type neurons. Expression of murine P301L tau did not change the speed, direction of travel or likelihood of movement of mitochondria. Notably, the angle that defines the orientation of the mitochondria in the axon, and the volume of individual moving mitochondria, were significantly increased in neurons expressing P301L tau. We found that murine tau phosphorylation in KI-P301L mouse neurons was diminished and the ability of P301L tau to bind to microtubules was also reduced compared to tau in wild-type neurons. The P301L mutation did not influence the ability of murine tau to associate with membranes in cortical neurons or in adult mouse brain. We conclude that P301L tau is associated with mitochondrial changes and causes an early reduction in murine tau phosphorylation in neurons coupled with impaired microtubule binding of tau. These results support the association of mutant tau with detrimental effects on mitochondria and will be of significance for the pathogenesis of tauopathies.


Assuntos
Axônios/metabolismo , Mitocôndrias/metabolismo , Proteínas tau/metabolismo , Animais , Axônios/patologia , Membrana Celular/metabolismo , Membrana Celular/patologia , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Citosol/metabolismo , Citosol/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microtúbulos/metabolismo , Mitocôndrias/patologia , Mutação , Fosforilação , Ratos , Proteínas tau/genética
17.
Acta Neuropathol ; 132(1): 77-91, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26839082

RESUMO

Cell-to-cell transmission of α-synuclein (αS) aggregates has been proposed to be responsible for progressive αS pathology in Parkinson disease (PD) and related disorders, including dementia with Lewy bodies. In support of this concept, a growing body of in vitro and in vivo experimental evidence shows that exogenously introduced αS aggregates can spread into surrounding cells and trigger PD-like pathology. It remains to be determined what factor(s) lead to initiation of αS aggregation that is capable of seeding subsequent propagation. In this study we demonstrate that filamentous αS aggregates form in neurons in response to apoptosis induced by staurosporine or other toxins-6-hydroxy-dopamine and 1-methyl-4-phenylpyridinium (MPP+). Interaction between αS and proaggregant nuclear factor(s) is associated with disruption of nuclear envelope integrity. Knocking down a key nuclear envelop constituent protein, lamin B1, enhances αS aggregation. Moreover, in vitro and in vivo experimental models demonstrate that aggregates released upon cell breakdown can be taken up by surrounding cells. Accordingly, we suggest that at least some αS aggregation might be related to neuronal apoptosis or loss of nuclear membrane integrity, exposing cytosolic α-synuclein to proaggregant nuclear factors. These findings provide new clues to the pathogenesis of PD and related disorders that can lead to novel treatments of these disorders. Specifically, finding ways to limit the effects of apoptosis on αS aggregation, deposition, local uptake and subsequent propagation might significantly impact progression of disease.


Assuntos
Apoptose/fisiologia , Lamina Tipo B/metabolismo , Neurônios/metabolismo , Membrana Nuclear/metabolismo , Agregação Patológica de Proteínas/metabolismo , 1-Metil-4-fenilpiridínio/toxicidade , Animais , Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular Tumoral , Citosol/efeitos dos fármacos , Citosol/metabolismo , Citosol/patologia , Modelos Animais de Doenças , Humanos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/patologia , Membrana Nuclear/efeitos dos fármacos , Membrana Nuclear/patologia , Oxidopamina/toxicidade , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Agregação Patológica de Proteínas/patologia , Estaurosporina/toxicidade
18.
Tumour Biol ; 37(2): 2473-80, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26383528

RESUMO

High-grade gliomas are considered the most malignant of brain tumors and have a poor prognosis. In a previous study, we showed that LIM domain only 2 (LMO2) regulates glioma stem cell properties and tumor angiogenesis and gave rise to highly invasive glioma xenografts. Glioma invasion in the surrounding parenchymal tissues is a major hurdle with respect to eliminating glioma by surgery. Invasive glioma cells are considered one of the main culprits for the recurrence of tumors after therapies. In the current study, we focused on determining the molecular mechanism(s) by which LMO2 regulates glioma cell migration and invasion. Forced expression of LMO2 in human U87MG glioma cells led to glioma invasion, as determined by in vivo xenograft assays and enhanced in vitro migration and invasion. LMO2 was associated with increased levels of cytosolic p27(Kip1) protein. LMO2 possibly induced the stabilization and augmented interactions between p27(Kip1) and RhoA. We knocked down the expression of p27(Kip1), which led to a decrease in LMO2-driven glioma cell migration and invasion. Taken together, our findings indicate that LMO2 promotes glioma cell migration and invasion by increasing the levels of cytosolic p27(Kip1).


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Movimento Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Citosol/metabolismo , Glioma/metabolismo , Glioma/patologia , Proteínas com Domínio LIM/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular , Linhagem Celular Tumoral , Citosol/patologia , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Proteína rhoA de Ligação ao GTP/metabolismo
19.
Cerebellum ; 15(4): 509-17, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26374457

RESUMO

Telomerase reverse transcriptase (TERT) is the catalytic subunit of telomerase, an enzyme that elongates telomeres at the ends of chromosomes during DNA replication. Recently, it was shown that TERT has additional roles in cell survival, mitochondrial function, DNA repair, and Wnt signaling, all of which are unrelated to telomeres. Here, we demonstrate that TERT is enriched in Purkinje neurons, but not in the granule cells of the adult mouse cerebellum. TERT immunoreactivity in Purkinje neurons is present in the nucleus, mitochondria, and cytoplasm. Furthermore, TERT co-localizes with mitochondrial markers, and immunoblot analysis of protein extracts from isolated mitochondria and synaptosomes confirmed TERT localization in mitochondria. TERT expression in Purkinje neurons increased significantly in response to two stressors: a sub-lethal dose of X-ray radiation and exposure to a high glutamate concentration. While X-ray radiation increased TERT levels in the nucleus, glutamate exposure elevated TERT levels in mitochondria. Our findings suggest that in mature Purkinje neurons, TERT is present both in the nucleus and in mitochondria, where it may participate in adaptive responses of the neurons to excitotoxic and radiation stress.


Assuntos
Citosol/enzimologia , Ácido Glutâmico/toxicidade , Mitocôndrias/enzimologia , Células de Purkinje/enzimologia , Lesões Experimentais por Radiação/enzimologia , Telomerase/metabolismo , Animais , Núcleo Celular/enzimologia , Núcleo Celular/patologia , Núcleo Celular/efeitos da radiação , Citosol/patologia , Citosol/efeitos da radiação , Dano ao DNA/fisiologia , Dano ao DNA/efeitos da radiação , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Imunofluorescência , Immunoblotting , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/patologia , Mitocôndrias/efeitos da radiação , Células de Purkinje/patologia , Células de Purkinje/efeitos da radiação , Lesões Experimentais por Radiação/patologia , Estresse Fisiológico/fisiologia , Estresse Fisiológico/efeitos da radiação , Telomerase/genética , Técnicas de Cultura de Tecidos , Raios X/efeitos adversos
20.
J Pharmacol Sci ; 132(2): 166-170, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27802908

RESUMO

Dextromethorphan (DM) administered at supra-antitussive doses produce psychotoxic and neurotoxic effects in humans. We administered DM (80 mg/kg) to rats intraperitoneally to determine the ultrastructural change induced by DM, because intraperitoneal route is sensitive for the behavioral responses. Treatment with DM resulted in mitochondrial dysfunction and formation of myelinoid bodies in the hippocampus. MK-801 [(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate] attenuated DM-induced cytosolic oxidative burdens. However, neither MK-801 nor naloxone affected DM-induced mitochondrial dysfunction and formation of myelinoid bodies, indicating that the neurotoxic mechanism needs to be further elucidated. Therefore, the spectrum of toxicological effects associated with DM need to be reassessed.


Assuntos
Antitussígenos/toxicidade , Dextrometorfano/toxicidade , Hipocampo/efeitos dos fármacos , Hipocampo/ultraestrutura , Bainha de Mielina/ultraestrutura , Animais , Antitussígenos/administração & dosagem , Citosol/efeitos dos fármacos , Citosol/patologia , Citosol/ultraestrutura , Dextrometorfano/administração & dosagem , Hipocampo/patologia , Injeções Intraperitoneais , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Mitocôndrias/ultraestrutura , Bainha de Mielina/patologia , Ratos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa