Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.658
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(14)2023 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-37511222

RESUMO

The aim of this review article is to collate recent contributions of proteomic studies to cystic fibrosis transmembrane conductance regulator (CFTR) biology. We summarize advances from these studies and create an accessible resource for future CFTR proteomic efforts. We focus our attention on the CFTR interaction network at the cell surface, thus generating a CFTR 'surfaceome'. We review the main findings about CFTR interactions and highlight several functional categories amongst these that could lead to the discovery of potential biomarkers and drug targets for CF.


Assuntos
Membrana Celular , Regulador de Condutância Transmembrana em Fibrose Cística , Fibrose Cística , Proteômica , Humanos , Membrana Celular/fisiologia , Fibrose Cística/genética , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Transporte de Íons , Mutação , Transdução de Sinais
2.
Cryobiology ; 112: 104552, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37301358

RESUMO

Maintenance of cells within a volume range compatible with their functional integrity is a critical determinant of cell survival after cryopreservation, and quantifying this osmotically induced damage is a part of the rational design of improved cryopreservation protocols. The degree that cells tolerate osmotic stress significantly impacts applicable cryoprotocols, but there has been little research on the time dependence of this osmotic stress. Additionally, the flavonoid silymarin has been shown to be hepatoprotective. Therefore, here we test the hypotheses that osmotic damage is time-dependent and that flavonoid inclusion reduces osmotic damage. In our first experiment, cells were exposed to a series of anisosmotic solutions of graded hypo- and hypertonicity for 10-40 min, resulting in a conclusion that osmotically induced damage is time dependent. In the next experiment, adherent cells preincubated with silymarin at the concentration of 10-4 mol/L and 10-5 mol/L showed a significant increase in cell proliferation and metabolic activity after osmotic stress compared to untreated matched controls. For instance, when adherent cells preincubated with 10-5 mol/L silymarin were tested, resistance to osmotic damage and a significant increase (15%) in membrane integrity was observed in hypo-osmotic media and a 22% increase in hyperosmotic conditions. Similarly, significant protection from osmotic damage was observed in suspended HepG2 cells in the presence of silymarin. Our study concludes that osmotic damage is time dependent, and the addition of silymarin leads to elevated resistance to osmotic stress and a potential increase in the cryosurvival of HepG2 cells.


Assuntos
Silimarina , Espermatozoides , Masculino , Humanos , Espermatozoides/fisiologia , Membrana Celular/fisiologia , Silimarina/farmacologia , Silimarina/metabolismo , Células Hep G2 , Suspensões , Criopreservação/métodos , Pressão Osmótica
3.
Int J Mol Sci ; 23(9)2022 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-35563465

RESUMO

We propose a novel mechanism by which cancer cells can modulate the oxygen concentration within the nucleus, potentially creating low nuclear oxygen conditions without the need of an hypoxic micro-environment and suited for allowing cancer cells to resist chemo- and radio-therapy. The cells ability to alter intra-cellular oxygen conditions depends on the amount of cholesterol present within the cellular membranes, where high levels of cholesterol can yield rigid membranes that slow oxygen diffusion. The proposed mechanism centers on the competition between (1) the diffusion of oxygen within the cell and across cellular membranes that replenishes any consumed oxygen and (2) the consumption of oxygen in the mitochondria, peroxisomes, endoplasmic reticulum (ER), etc. The novelty of our work centers around the assumption that the cholesterol content of a membrane can affect the oxygen diffusion across the membrane, reducing the cell ability to replenish the oxygen consumed within the cell. For these conditions, the effective diffusion rate of oxygen becomes of the same order as the oxygen consumption rate, allowing the cell to reduce the oxygen concentration of the nucleus, with implications to the Warburg Effect. The cellular and nucleus oxygen content is indirectly evaluated experimentally for bladder (T24) cancer cells and during the cell cycle, where the cells are initially synchronized using hydroxeaurea (HU) at the late G1-phase/early S-phase. The analysis of cellular and nucleus oxygen concentration during cell cycle is performed via (i) RT-qPCR gene analysis of hypoxia inducible transcription factors (HIF) and prolyl hydroxylases (PHD) and (ii) radiation clonogenic assay every 2 h, after release from synchronization. The HIF/PHD genes allowed us to correlate cellular oxygen with oxygen concentration in the nucleus that is obtained from the cells radiation response, where the amount DNA damage due to radiation is directly related to the amount of oxygen present in the nucleus. We demonstrate that during the S-phase cells can become hypoxic in the late S-phase/early G2-phase and therefore the radiation resistance increases 2- to 3-fold.


Assuntos
Núcleo Celular , Colesterol , Hipóxia , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral/metabolismo , Linhagem Celular Tumoral/fisiologia , Membrana Celular/metabolismo , Membrana Celular/fisiologia , Núcleo Celular/metabolismo , Colesterol/metabolismo , Humanos , Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Oxigênio/metabolismo , Prolil Hidroxilases/metabolismo , Tolerância a Radiação/fisiologia , Fase S
4.
Cell Rep ; 38(1): 110187, 2022 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-34986345

RESUMO

Candida albicans is both a commensal and an opportunistic fungal pathogen. Invading hyphae of C. albicans secrete candidalysin, a pore-forming peptide toxin. To prevent cell death, epithelial cells must protect themselves from direct damage induced by candidalysin and by the mechanical forces exerted by expanding hyphae. We identify two key Ca2+-dependent repair mechanisms employed by epithelial cells to withstand candidalysin-producing hyphae. Using camelid nanobodies, we demonstrate candidalysin secretion directly into the invasion pockets induced by elongating C. albicans hyphae. The toxin induces oscillatory increases in cytosolic [Ca2+], which cause hydrolysis of PtdIns(4,5)P2 and loss of cortical actin. Epithelial cells dispose of damaged membrane regions containing candidalysin by an Alg-2/Alix/ESCRT-III-dependent blebbing process. At later stages, plasmalemmal tears induced mechanically by invading hyphae are repaired by exocytic insertion of lysosomal membranes. These two repair mechanisms maintain epithelial integrity and prevent mucosal damage during both commensal growth and infection by C. albicans.


Assuntos
Candida albicans/metabolismo , Candidíase/patologia , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Proteínas Fúngicas/metabolismo , Lisossomos/metabolismo , Mucosa/fisiologia , Animais , Cálcio/metabolismo , Linhagem Celular , Membrana Celular/fisiologia , Células Epiteliais/metabolismo , Exocitose/fisiologia , Proteínas Fúngicas/genética , Interações Hospedeiro-Patógeno , Humanos , Hifas/crescimento & desenvolvimento , Camundongos , Mucosa/citologia , Mucosa/microbiologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Células RAW 264.7
5.
Dev Cell ; 57(2): 228-245.e6, 2022 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-35016014

RESUMO

Although overwhelming plasma membrane integrity loss leads to cell lysis and necrosis, cells can tolerate a limited level of plasma membrane damage, undergo ESCRT-III-mediated repair, and survive. Here, we find that cells which undergo limited plasma membrane damage from the pore-forming actions of MLKL, GSDMD, perforin, or detergents experience local activation of PKCs through Ca2+ influx at the damage sites. S660-phosphorylated PKCs subsequently activate the TAK1/IKKs axis and RelA/Cux1 complex to trigger chemokine expressions. We observe that in late-stage cancers, cells with active MLKL show expression of CXCL8. Similar expression induction is also found in ischemia-injured kidneys. Chemokines generated in this manner are also indispensable for recruiting immune cells to the dead and dying cells. This plasma membrane integrity-sensing pathway is similar to the well-established yeast cell wall integrity signaling pathway at molecular level, and this suggests an evolutionary conserved mechanism to respond to the cellular barrier damage.


Assuntos
Membrana Celular/metabolismo , Quimiocinas/fisiologia , Proteína Quinase C/fisiologia , Animais , Apoptose/fisiologia , Membrana Celular/fisiologia , Quimiocinas/genética , Quimiocinas/imunologia , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Necrose/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Fosforilação , Proteína Quinase C/metabolismo , Proteínas Quinases/metabolismo , Proteínas Quinases/fisiologia , Transdução de Sinais
6.
PLoS Comput Biol ; 17(11): e1009520, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34723961

RESUMO

Regulation of cytosolic calcium (Ca2+) dynamics is fundamental to microglial function. Temporal and spatial Ca2+ fluxes are induced from a complicated signal transduction pathway linked to brain ionic homeostasis. In this paper, we develop a novel biophysical model of Ca2+ and sodium (Na+) dynamics in human microglia and evaluate the contribution of purinergic receptors (P2XRs) to both intracellular Ca2+ and Na+ levels in response to agonist/ATP binding. This is the first comprehensive model that integrates P2XRs to predict intricate Ca2+ and Na+ transient responses in microglia. Specifically, a novel compact biophysical model is proposed for the capture of whole-cell patch-clamp currents associated with P2X4 and P2X7 receptors, which is composed of only four state variables. The entire model shows that intricate intracellular ion dynamics arise from the coupled interaction between P2X4 and P2X7 receptors, the Na+/Ca2+ exchanger (NCX), Ca2+ extrusion by the plasma membrane Ca2+ ATPase (PMCA), and Ca2+ and Na+ leak channels. Both P2XRs are modelled as two separate adenosine triphosphate (ATP) gated Ca2+ and Na+ conductance channels, where the stoichiometry is the removal of one Ca2+ for the hydrolysis of one ATP molecule. Two unique sets of model parameters were determined using an evolutionary algorithm to optimise fitting to experimental data for each of the receptors. This allows the proposed model to capture both human P2X7 and P2X4 data (hP2X7 and hP2X4). The model architecture enables a high degree of simplicity, accuracy and predictability of Ca2+ and Na+ dynamics thus providing quantitative insights into different behaviours of intracellular Na+ and Ca2+ which will guide future experimental research. Understanding the interactions between these receptors and other membrane-bound transporters provides a step forward in resolving the qualitative link between purinergic receptors and microglial physiology and their contribution to brain pathology.


Assuntos
Cálcio/metabolismo , Microglia/metabolismo , Modelos Biológicos , Receptores Purinérgicos P2X/fisiologia , Trifosfato de Adenosina/metabolismo , Algoritmos , Membrana Celular/fisiologia , Humanos , Hidrólise , Potenciais da Membrana , Sódio/metabolismo , Trocador de Sódio e Cálcio/metabolismo
7.
Int J Mol Sci ; 22(21)2021 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-34768998

RESUMO

Mechanical cues are crucial for survival, adaptation, and normal homeostasis in virtually every cell type. The transduction of mechanical messages into intracellular biochemical messages is termed mechanotransduction. While significant advances in biochemical signaling have been made in the last few decades, the role of mechanotransduction in physiological and pathological processes has been largely overlooked until recently. In this review, the role of interactions between the cytoskeleton and cell-cell/cell-matrix adhesions in transducing mechanical signals is discussed. In addition, mechanosensors that reside in the cell membrane and the transduction of mechanical signals to the nucleus are discussed. Finally, we describe two examples in which mechanotransduction plays a significant role in normal physiology and disease development. The first example is the role of mechanotransduction in the proliferation and metastasis of cancerous cells. In this system, the role of mechanotransduction in cellular processes, including proliferation, differentiation, and motility, is described. In the second example, the role of mechanotransduction in a mechanically active organ, the gastrointestinal tract, is described. In the gut, mechanotransduction contributes to normal physiology and the development of motility disorders.


Assuntos
Membrana Celular/fisiologia , Citoesqueleto/fisiologia , Mecanotransdução Celular/fisiologia , Animais , Núcleo Celular/fisiologia , Adesões Focais/fisiologia , Humanos
8.
Adv Drug Deliv Rev ; 179: 114006, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34655662

RESUMO

Nanoparticle-based therapeutics have the potential to change the paradigm of how we approach the diagnosis and treatment of human disease. Employing naturally derived cell membranes as a surface coating has created a powerful new approach by which nanoparticles can be functionalized towards a wide range of biomedical applications. By using membranes derived from different cell sources, the resulting nanoparticles inherit properties that can make them well-suited for a variety of tasks. In recent years, stimuli-responsive platforms with the ability to release payloads on demand have received increasing attention due to their improved delivery, reduced side effects, and precision targeting. Nanoformulations have been developed to respond to external stimuli such as magnetic fields, ultrasound, and radiation, as well as local stimuli such as pH gradients, redox potentials, and other chemical conditions. Here, an overview of the novel cell membrane coating platform is provided, followed by a discussion of stimuli-responsive platforms that leverage this technology.


Assuntos
Biomimética/métodos , Membrana Celular/fisiologia , Nanopartículas/química , Células Sanguíneas/fisiologia , Portadores de Fármacos , Humanos , Concentração de Íons de Hidrogênio , Campos Magnéticos , Radioterapia/métodos , Ultrassonografia/métodos
9.
Nat Methods ; 18(10): 1239-1246, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34608318

RESUMO

In spite of their great importance in biology, methods providing access to spontaneous molecular interactions with and on biological membranes have been sparse. The recent advent of mass photometry to quantify mass distributions of unlabeled biomolecules landing on surfaces raised hopes that this approach could be transferred to membranes. Here, by introducing a new interferometric scattering (iSCAT) image processing and analysis strategy adapted to diffusing particles, we enable mass-sensitive particle tracking (MSPT) of single unlabeled biomolecules on a supported lipid bilayer. We applied this approach to the highly nonlinear reaction cycles underlying MinDE protein self-organization. MSPT allowed us to determine the stoichiometry and turnover of individual membrane-bound MinD/MinDE protein complexes and to quantify their size-dependent diffusion. This study demonstrates the potential of MSPT to enhance our quantitative understanding of membrane-associated biological systems.


Assuntos
Adenosina Trifosfatases/metabolismo , Fenômenos Biofísicos , Proteínas de Ciclo Celular/metabolismo , Membrana Celular/fisiologia , Proteínas de Escherichia coli/metabolismo , Bicamadas Lipídicas/metabolismo , Adenosina Trifosfatases/química , Proteínas de Ciclo Celular/química , Membrana Celular/metabolismo , Escherichia coli , Proteínas de Escherichia coli/química , Bicamadas Lipídicas/química
10.
Biochemistry ; 60(37): 2761-2772, 2021 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-34492183

RESUMO

Glutathione peroxidase 4 (GPx4) serves as the only enzyme that protects membranes through the reduction of lipid hydroperoxides, preventing membrane oxidative damage and cell death through ferroptosis. Recently, GPx4 has gained attention as a therapeutic target for cancer through inhibition and as a target for inflammatory diseases through activation. In addition, GPx4 isoforms perform several distinct moonlighting functions including cysteine cross-linking of protamines during sperm cell chromatin remodeling, a function for which molecular and structural details are undefined. Despite the importance in biology, disease, and potential for drug development, little is known about GPx4 functional interactions at high resolution. This study presents the first NMR assignments of GPx4, and the electrostatic interaction of GPx4 with the membrane is characterized. Mutagenesis reveals the cationic patch residues that are key to membrane binding and stabilization. The cationic patch is observed to be important in binding headgroups of highly anionic cardiolipin. A novel lipid binding site is observed adjacent to the catalytic site and may enable protection of lipid-headgroups from oxidative damage. Arachidonic acid is also found to engage with GPx4, while cholesterol did not display any interaction. The cationic patch residues were also found to enable DNA binding, the first observation of this interaction. Electrostatic DNA binding explains a mechanism for the nuclear isoform of GPx4 to target DNA-bound protamines and to potentially reduce oxidatively damaged DNA. Together, these results highlight the importance of electrostatics in the function of GPx4 and illuminate how the multifunctional enzyme is able to fill multiple biological roles.


Assuntos
Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/ultraestrutura , Sítios de Ligação , Domínio Catalítico , Morte Celular , Membrana Celular/metabolismo , Membrana Celular/fisiologia , DNA/metabolismo , DNA/fisiologia , Ferroptose , Glutationa Peroxidase/metabolismo , Humanos , Peróxidos Lipídicos/metabolismo , Imageamento por Ressonância Magnética/métodos , Lipídeos de Membrana/metabolismo , Lipídeos de Membrana/fisiologia , Estresse Oxidativo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/fisiologia , Ligação Proteica , Isoformas de Proteínas/metabolismo , Eletricidade Estática
11.
Adv Sci (Weinh) ; 8(21): e2102519, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34495564

RESUMO

Nanomaterial-induced endothelial leakiness (NanoEL) is an interfacial phenomenon denoting the paracellular transport of nanoparticles that is pertinent to nanotoxicology, nanomedicine and biomedical engineering. While the NanoEL phenomenon is complementary to the enhanced permeability and retention effect in terms of their common applicability to delineating the permeability and behavior of nanoparticles in tumoral environments, these two effects significantly differ in scope, origin, and manifestation. In the current study, the descriptors are fully examined of the NanoEL phenomenon elicited by generic citrate-coated gold nanoparticles (AuNPs) of changing size and concentration, from microscopic gap formation and actin reorganization down to molecular signaling pathways and nanoscale interactions of AuNPs with VE-cadherin and its intra/extracellular cofactors. Employing synergistic in silico methodologies, for the first time the molecular and statistical mechanics of cadherin pair disruption, especially in response to AuNPs of the smallest size and highest concentration are revealed. This study marks a major advancement toward establishing a comprehensive NanoEL framework for complementing the understanding of the transcytotic pathway and for guiding the design and application of future nanomedicines harnessing the myriad functions of the mammalian vasculature.


Assuntos
Ouro/química , Nanopartículas Metálicas/química , Animais , Antígenos CD/química , Antígenos CD/metabolismo , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/metabolismo , Caderinas/química , Caderinas/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Ácido Cítrico/química , Dimerização , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Nanopartículas Metálicas/toxicidade , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Suínos
12.
Toxicol Lett ; 351: 145-154, 2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34509610

RESUMO

Fe3O4 nanoparticles are widely used in the diagnosis and treatment of diseases due to their superparamagnetism, but their toxicity in vivo, which can result in apoptosis or autophagy, cannot be ignored. It has been reported that polydopamine (PDA) modification can reduce the toxicity of Fe3O4 and increase its biocompatibility. However, more research is warranted to further improve the modification method. We therefore developed a new method to coat Fe3O4@PDA nanoparticles with the mesenchymal stem cell membrane (MSCM) and evaluated the toxicity of the modified particles in the lungs of mice. We found that the MSCM modification significantly reduced lung injury induced by Fe3O4 particles in mice. Compared with Fe3O4@PDA nanoparticles, co-modification with MSCM and PDA significantly reduced autophagy and apoptosis in mouse lung tissue, and reduced activation of autophagy mediated by the AMPK-ULK1 pathway axis. Thus, co-modification with MSCM and PDA prevents Fe3O4-induced pulmonary toxicity in mice by inhibiting autophagy, apoptosis, and oxidative stress.


Assuntos
Adenilato Quinase/metabolismo , Membrana Celular/efeitos dos fármacos , Compostos Férricos/toxicidade , Indóis/farmacologia , Pneumopatias/induzido quimicamente , Células-Tronco Mesenquimais/efeitos dos fármacos , Polímeros/farmacologia , Adenilato Quinase/genética , Animais , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Membrana Celular/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos ICR , Estresse Oxidativo/efeitos dos fármacos
13.
Sci Rep ; 11(1): 19141, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34580330

RESUMO

Plasma membrane repair mechanisms are activated within seconds post-injury to promote rapid membrane resealing in eukaryotic cells and prevent cell death. However, less is known about the regeneration phase that follows and how cells respond to injury in the short-term. Here, we provide a genome-wide study into the mRNA expression profile of MCF-7 breast cancer cells exposed to injury by digitonin, a mild non-ionic detergent that permeabilizes the plasma membrane. We focused on the early transcriptional signature and found a time-dependent increase in the number of differentially expressed (> twofold, P < 0.05) genes (34, 114 and 236 genes at 20-, 40- and 60-min post-injury, respectively). Pathway analysis highlighted a robust and gradual three-part transcriptional response: (1) prompt activation of immediate-early response genes, (2) activation of specific MAPK cascades and (3) induction of inflammatory and immune pathways. Therefore, plasma membrane injury triggers a rapid and strong stress and immunogenic response. Our meta-analysis suggests that this is a conserved transcriptome response to plasma membrane injury across different cell and injury types. Taken together, our study shows that injury has profound effects on the transcriptome of wounded cells in the regeneration phase (subsequent to membrane resealing), which is likely to influence cellular status and has been previously overlooked.


Assuntos
Membrana Celular/fisiologia , Regulação da Expressão Gênica , Regeneração/genética , Animais , Biologia Computacional , Humanos , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/imunologia , Células MCF-7 , RNA-Seq , Regeneração/imunologia
14.
Immunol Res ; 69(6): 496-519, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34410575

RESUMO

The SARS-CoV-2 S protein on the membrane of infected cells can promote receptor-dependent syncytia formation, relating to extensive tissue damage and lymphocyte elimination. In this case, it is challenging to obtain neutralizing antibodies and prevent them through antibodies effectively. Considering that, in the current study, structural domain search methods are adopted to analyze the SARS-CoV-2 S protein to find the fusion mechanism. The results show that after the EF-hand domain of S protein bound to calcium ions, S2 protein had CaMKII protein activities. Besides, the CaMKII_AD domain of S2 changed S2 conformation, facilitating the formation of HR1-HR2 six-helix bundles. Apart from that, the Ca2+-ATPase of S2 pumped calcium ions from the virus cytoplasm to help membrane fusion, while motor structures of S drove the CaATP_NAI and CaMKII_AD domains to extend to the outside and combined the viral membrane and the cell membrane, thus forming a calcium bridge. Furthermore, the phospholipid-flipping-ATPase released water, triggering lipid mixing and fusion and generating fusion pores. Then, motor structures promoted fusion pore extension, followed by the cytoplasmic contents of the virus being discharged into the cell cytoplasm. After that, the membrane of the virus slid onto the cell membrane along the flowing membrane on the gap of the three CaATP_NAI. At last, the HR1-HR2 hexamer would fall into the cytoplasm or stay on the cell membrane. Therefore, the CaMKII_like system of S protein facilitated membrane fusion for further inducing syncytial multinucleated giant cells.


Assuntos
COVID-19/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , ATPases Transportadoras de Cálcio/metabolismo , Células Gigantes/metabolismo , Fusão de Membrana/fisiologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Sequência de Aminoácidos , Cálcio/metabolismo , Membrana Celular/fisiologia , Membrana Celular/virologia , Células Gigantes/virologia , Humanos , SARS-CoV-2 , Alinhamento de Sequência , Internalização do Vírus
15.
mBio ; 12(4): e0177021, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34399625

RESUMO

The plasma membrane (PM) must be overcome by viruses during entry and release. Furthermore, the PM represents the cellular communication compartment and the immune system interface. Hence, viruses have evolved sophisticated strategies to remodel the PM, for instance to avoid immune sensing and clearance of infected cells. We performed a comprehensive analysis of cell surface dysregulation by two human-pathogenic viruses, human cytomegalovirus (HCMV) and human immunodeficiency virus type 1 (HIV-1), in primary macrophages, which are classical antigen-presenting cells and orchestrators of the immune system. Scanning ion conductance microscopy revealed a loss of roughness and an overall smooth phenotype of HCMV-infected macrophages, in contrast to HIV-1 infection. This phenotype was also evident on the molecular level. When we screened for cell surface receptors modulated by HCMV, 42 of 332 receptors tested were up- or downregulated, whereas HIV-1 affected only 7 receptors. In particular CD164, CD84, and CD180 were targeted by HCMV. Mechanistically, HCMV induced transcriptional silencing of these receptors in an interferon (IFN)-independent manner, and expression was reduced not only by lab-adapted HCMV but also by clinical HCMV isolates. Altogether, our plasma membrane profiling of human macrophages provides clues to understand how viruses evade the immune system and identified novel cell surface receptors targeted by HCMV. IMPORTANCE The PM is a key component that viruses have to cope with. It is a barrier for infection and egress and is critically involved in antiviral immune signaling. We hence asked the question how two immunomodulatory viruses, HIV-1 and HCMV, dysregulate this compartment in infected macrophages, relevant in vivo targets of both viruses. We employed a contact-free microscopic technique to image the PM of infected cells and performed a phenotypic flow cytometry-based screen to identify receptor modulations on a molecular level. Our results show that HIV-1 and HCMV differentially manipulate the PM of macrophages. While HIV-1-mediated changes are relatively subtle, HCMV induces major alterations of the PM. We identify novel immune receptors manipulated by HCMV and define mechanisms of how HCMV interferes with receptor expression. Altogether, our study reveals differential strategies of how two human-pathogenic viruses manipulate infected cells and identifies potential novel pathways of HCMV immune evasion.


Assuntos
Membrana Celular/fisiologia , Membrana Celular/virologia , Citomegalovirus/imunologia , HIV-1/imunologia , Evasão da Resposta Imune , Macrófagos/imunologia , Macrófagos/virologia , Células Cultivadas , Citomegalovirus/patogenicidade , HIV-1/patogenicidade , Humanos , Transdução de Sinais , Células THP-1
16.
Nat Rev Endocrinol ; 17(9): 560-571, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34194011

RESUMO

The pharmacological treatment of pituitary tumours is based on the use of stable analogues of somatostatin and dopamine. The analogues bind to somatostatin receptor types 2 and 5 (SST2 and SST5) and dopamine receptor type 2 (DRD2), respectively, and generate signal transduction cascades in cancerous pituitary cells that culminate in the inhibition of hormone secretion, cell growth and invasion. Drug resistance occurs in a subset of patients and can involve different steps at different stages, such as following receptor activation by the agonist or during the final biological responses. Although the expression of somatostatin and dopamine receptors in cancer cells is a prerequisite for these drugs to reach a biological effect, their presence does not guarantee the success of the therapy. Successful therapy also requires the proper functioning of the machinery of signal transduction and the finely tuned regulation of receptor desensitization, internalization and intracellular trafficking. The present Review provides an updated overview of the molecular factors underlying the pharmacological resistance of pituitary tumours. The Review discusses the experimental evidence that supports a role for receptors and intracellular proteins in the function of SSTs and DRD2 and their clinical importance.


Assuntos
Adenoma/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/fisiologia , Neoplasias Hipofisárias/tratamento farmacológico , Adenoma/patologia , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neoplasias Hipofisárias/patologia , Transdução de Sinais/efeitos dos fármacos
17.
Elife ; 102021 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-34212861

RESUMO

The Drosophila polarity protein Crumbs is essential for the establishment and growth of the apical domain in epithelial cells. The protein Yurt limits the ability of Crumbs to promote apical membrane growth, thereby defining proper apical/lateral membrane ratio that is crucial for forming and maintaining complex epithelial structures such as tubes or acini. Here, we show that Yurt also increases Myosin-dependent cortical tension downstream of Crumbs. Yurt overexpression thus induces apical constriction in epithelial cells. The kinase aPKC phosphorylates Yurt, thereby dislodging the latter from the apical domain and releasing apical tension. In contrast, the kinase Pak1 promotes Yurt dephosphorylation through activation of the phosphatase PP2A. The Pak1-PP2A module thus opposes aPKC function and supports Yurt-induced apical constriction. Hence, the complex interplay between Yurt, aPKC, Pak1, and PP2A contributes to the functional plasticity of Crumbs. Overall, our data increase our understanding of how proteins sustaining epithelial cell polarization and Myosin-dependent cell contractility interact with one another to control epithelial tissue architecture.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Proteínas de Membrana/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Proteína Quinase C/metabolismo , Quinases Ativadas por p21/metabolismo , Animais , Membrana Celular/fisiologia , Citoesqueleto/fisiologia , Drosophila/embriologia , Drosophila/genética , Proteínas de Drosophila/genética , Células Epiteliais/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas de Membrana/genética , Miosinas/genética , Miosinas/metabolismo , Fosfoproteínas Fosfatases/genética , Proteína Quinase C/genética , Quinases Ativadas por p21/genética
18.
J Mol Cell Biol ; 13(6): 395-408, 2021 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-34143183

RESUMO

Cell polarity is essential for spatially regulating of physiological processes in metazoans by which hormonal stimulation‒secretion coupling is precisely coupled for tissue homeostasis and organ communications. However, the molecular mechanisms underlying epithelial cell polarity establishment remain elusive. Here, we show that septin cytoskeleton interacts with catenin complex to organize a functional domain to separate apical from basal membranes in polarized epithelial cells. Using polarized epithelial cell monolayer as a model system with transepithelial electrical resistance as functional readout, our studies show that septins are essential for epithelial cell polarization. Our proteomic analyses discovered a novel septin‒catenin complex during epithelial cell polarization. The functional relevance of septin‒catenin complex was then examined in three-dimensional (3D) culture in which suppression of septins resulted in deformation of apical lumen in cysts, a hallmark seen in polarity-deficient 3D cultures and animals. Mechanistically, septin cytoskeleton stabilizes the association of adherens catenin complex with actin cytoskeleton, and depletion or disruption of septin cytoskeleton liberates adherens junction and polarity complexes into the cytoplasm. Together, these findings reveal a previously unrecognized role for septin cytoskeleton in the polarization of the apical‒basal axis and lumen formation in polarized epithelial cells.


Assuntos
Citoesqueleto de Actina/metabolismo , Cateninas/metabolismo , Polaridade Celular/fisiologia , Células Epiteliais/metabolismo , Septinas/metabolismo , Citoesqueleto de Actina/fisiologia , Células CACO-2 , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/fisiologia , Células Epiteliais/fisiologia , Humanos , Morfogênese/fisiologia , Proteômica/métodos
19.
Opt Express ; 29(8): 11976-11986, 2021 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-33984967

RESUMO

The fluidity of the cell membrane is closely related to cancer metastasis/invasion. To test the relationship of membrane fluidity and invasiveness, we first demonstrated that transfection of small RNA miR-92b-3p can significantly increase invasiveness of the small cell lung cancer cell line SHP77. Then optical tweezers were used to measure membrane fluidity. This study employed continuous and step-like stretching methods to examine fluidity changes in SHP77 cell membranes before and after miR-92b-3p transfection. A newly developed physical model was used to derive the effective viscosity and static tension of the cell membrane from relaxation curves obtained via step-like stretching. Experiments showed that invasiveness and fluidity increased significantly after miR-92b-3p transfection. This study paved the way toward a better understanding of cancer cell invasion and membrane mechanical characteristics.


Assuntos
Neoplasias Pulmonares/patologia , Fluidez de Membrana/fisiologia , Pinças Ópticas , Carcinoma de Pequenas Células do Pulmão/patologia , Linhagem Celular Tumoral , Membrana Celular/fisiologia , Vetores Genéticos , Humanos , Lentivirus/genética , Neoplasias Pulmonares/genética , MicroRNAs/genética , Invasividade Neoplásica , Reação em Cadeia da Polimerase em Tempo Real , Carcinoma de Pequenas Células do Pulmão/genética , Transfecção
20.
Yakugaku Zasshi ; 141(5): 661-665, 2021.
Artigo em Japonês | MEDLINE | ID: mdl-33952748

RESUMO

Although the concept of a drug delivery system (DDS) is usually applied to conventional drug therapy, it is also important for cell-based therapy. The surface manipulation of living cells represents a powerful tool for controlling cell behaviors in the body, such as enhancement of cell-cell interactions, targeted delivery of cells, and protection from immunological rejection. Functional groups, including amines, thiols, and carbonyls, offer excellent opportunities for chemical modification through the formation of covalent bonds with exogenous molecules. Non-natural reactive groups introduced by metabolic labeling were recently utilized for targeted chemical modification. On the other hand, noncovalent strategies are also available; two major examples are electrostatic interaction with a negative charge on the cell surface and hydrophobic insertion or interaction with the cell membrane. In this study, we analyzed factors affecting cell surface modifications using PEG-lipid and succeeded in enhancing the efficacy of modification by cyclodextrin. Then, mesenchymal stem cells (MSCs), whose therapeutic effect has been demonstrated at the clinical stage and which have been clinically used as a drug, were decorated with PEG-lipid conjugates having a targeted ligand such as peptide or scFv, which are recognized by ICAM1. The peptide or scFv decoration enhanced the cell adhesion of MSCs on cytokine treated-endothelial cells. This technique will prompt the targeted delivery of MSCs to intended therapy sites, and underscores the promise of cell surface engineering as a tool for improving cell-based therapy.


Assuntos
Membrana Celular/fisiologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Sistemas de Liberação de Medicamentos , Adesão Celular , Comunicação Celular , Engenharia Celular , Membrana Celular/metabolismo , Ciclodextrinas/farmacologia , Humanos , Interações Hidrofóbicas e Hidrofílicas , Molécula 1 de Adesão Intercelular/fisiologia , Células-Tronco Mesenquimais , Polietilenoglicóis , Eletricidade Estática
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA