Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30.906
Filtrar
1.
Cell Mol Life Sci ; 81(1): 402, 2024 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-39276234

RESUMO

The excessive inflammation caused by the prolonged activation of Toll-like receptor 4 (TLR4) and its downstream signaling pathways leads to sepsis. CD14-mediated endocytosis of TLR4 is the key step to control the amount of TLR4 on cell membrane and the activity of downstream pathways. The actin cytoskeleton is necessary for receptor-mediated endocytosis, but its role in TLR4 endocytosis remains elusive. Here we show that Tropomodulin 1 (Tmod1), an actin capping protein, inhibited lipopolysaccharide (LPS)-induced TLR4 endocytosis and intracellular trafficking in macrophages. Thus it resulted in increased surface TLR4 and the upregulation of myeloid differentiation factor 88 (MyD88)-dependent pathway and the downregulation of TIR domain-containing adaptor-inducing interferon-ß (TRIF)-dependent pathway, leading to the enhanced secretion of inflammatory cytokines, such as TNF-α and IL-6, and the reduced secretion of cytokines, such as IFN-ß. Macrophages deficient with Tmod1 relieved the inflammatory response in LPS-induced acute lung injury mouse model. Mechanistically, Tmod1 negatively regulated LPS-induced TLR4 endocytosis and inflammatory response through modulating the activity of CD14/Syk/PLCγ2/IP3/Ca2+ signaling pathway, the reorganization of actin cytoskeleton, and the membrane tension. Therefore, Tmod1 is a key regulator of inflammatory response and immune functions in macrophages and may be a potential target for the treatment of excessive inflammation and sepsis.


Assuntos
Endocitose , Inflamação , Lipopolissacarídeos , Macrófagos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Receptor 4 Toll-Like , Tropomodulina , Animais , Receptor 4 Toll-Like/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Macrófagos/metabolismo , Macrófagos/imunologia , Inflamação/metabolismo , Inflamação/patologia , Tropomodulina/metabolismo , Tropomodulina/genética , Citocinas/metabolismo , Células RAW 264.7 , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Receptores de Lipopolissacarídeos/metabolismo , Masculino , Camundongos Knockout , Citoesqueleto de Actina/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/patologia
3.
Parasites Hosts Dis ; 62(3): 281-293, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39218627

RESUMO

We previously reported that leukotriene B4 (LTB4) contained in Trichomonas vaginalis-derived secretory products (TvSP) play an essential role in interleukin-8 (IL-8) production in human mast cell line (HMC-1 cells) via LTB4 receptor (BLT)-mediated Nuclear Factor-kappa B (NF-кB) activation. Dynamin, a GTPase, has been known to be involved in endocytosis of receptors for signaling of production of cytokine or chemokines. In the present study, we investigated the role of dynamin-mediated BLT1 endocytosis in TvSP-induced IL-8 production. When HMC-1 cells were transfected with BLT1 or BLT2 siRNA, TvSP-induced IL-8 production was significantly inhibited compared with that in cells transfected with control siRNA. In addition, pretreatment of HMC-1 cells with a dynamin inhibitor (Dynasore) reduced IL-8 production induced by TvSP or LTB4. TvSP- or LTB4- induced phosphorylation of NF-кB was also attenuated by pretreatment with Dynasore. After exposing HMC-1 cells to TvSP or LTB4, BLT1 was translocated from the intracellular compartments to the plasma membrane within 30 min. At 60 min after stimulation with TvSP or LTB4, BLT1 remigrated from the cell surface to intracellular areas. Pretreatment of HMC-1 cells with dynamin-2 siRNA blocked internalization of BLT1 induced by TvSP or LTB4. Co-immunoprecipitation experiments revealed that dynamin-2 strongly interacted with BLT1 60 min after stimulation with TvSP or LTB4. These results suggest that T. vaginalis-secreted LTB4 induces IL-8 production in HMC-1 cells via dynamin 2-mediated endocytosis of BLT1 and phosphorylation of NF-кB.


Assuntos
Dinamina II , Endocitose , Interleucina-8 , Receptores do Leucotrieno B4 , Trichomonas vaginalis , Humanos , Interleucina-8/metabolismo , Interleucina-8/genética , Receptores do Leucotrieno B4/metabolismo , Receptores do Leucotrieno B4/genética , Endocitose/efeitos dos fármacos , Dinamina II/metabolismo , Dinamina II/genética , Linhagem Celular , Trichomonas vaginalis/metabolismo , Leucotrieno B4/metabolismo , Mastócitos/metabolismo , Mastócitos/imunologia , NF-kappa B/metabolismo , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/genética
4.
Front Immunol ; 15: 1406643, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39263220

RESUMO

Introduction: Immunogenicity, the unwanted immune response triggered by therapeutic antibodies, poses significant challenges in biotherapeutic development. This response can lead to the production of anti-drug antibodies, potentially compromising the efficacy and safety of treatments. The internalization of therapeutic antibodies into dendritic cells (DCs) is a critical factor influencing immunogenicity. Using monoclonal antibodies, with differences in non-specific cellular uptake, as tools to explore the impact on the overall risk of immunogenicity, this study explores how internalization influences peptide presentation and subsequently T cell activation. Materials and methods: To investigate the impact of antibody internalization on immunogenicity, untargeted toolantibodies with engineered positive or negative charge patches were utilized. Immature monocyte-derived DCs (moDCs), known for their physiologically relevant high endocytic activity, were employed for internalization assays, while mature moDCs were used for MHC-II associated peptide proteomics (MAPPs) assays. In addition to the lysosomal accumulation and peptide presentation, subsequent CD4+ T cell activation has been assessed. Consequently, a known CD4+ T cell epitope from ovalbumin was inserted into the tool antibodies to evaluate T cell activation on a single, shared epitope. Results: Antibodies with positive charge patches exhibited higher rates of lysosomal accumulation and epitope presentation compared to those with negative charge patches or neutral surface charge. Furthermore, a direct correlation between internalization rate and presentation on MHC-II molecules could be established. To explore the link between internalization, peptide presentation and CD4+ T cell activation, tool antibodies containing the same OVA epitope were used. Previous observations were not altered by the insertion of the OVA epitope and ultimately, an enhanced CD4+ T cell response correlated with increased internalization in DCs and peptide presentation. Discussion: These findings demonstrate that the biophysical properties of therapeutic antibodies, particularly surface charge, play a crucial role in their internalization into DCs. Antibodies internalized faster and processed by DCs, are also more prone to be presented on their surface leading to a higher risk of triggering an immune response. These insights underscore the importance of considering antibody surface charge and other properties that enhance cellular accumulation during the preclinical development of biotherapeutics to mitigate immunogenicity risks.


Assuntos
Apresentação de Antígeno , Células Dendríticas , Ativação Linfocitária , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Humanos , Apresentação de Antígeno/imunologia , Ativação Linfocitária/imunologia , Anticorpos Monoclonais/imunologia , Linfócitos T CD4-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Fatores de Risco , Endocitose/imunologia , Ovalbumina/imunologia
5.
Traffic ; 25(9): e12951, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39238078

RESUMO

Mitochondria, the dynamic organelles responsible for energy production and cellular metabolism, have the metabolic function of extracting energy from nutrients and synthesizing crucial metabolites. Nevertheless, recent research unveils that intercellular mitochondrial transfer by tunneling nanotubes, tumor microtubes, gap junction intercellular communication, extracellular vesicles, endocytosis and cell fusion may regulate mitochondrial function within recipient cells, potentially contributing to disease treatment, such as nonalcoholic steatohepatitis, glioblastoma, ischemic stroke, bladder cancer and neurodegenerative diseases. This review introduces the principal approaches to intercellular mitochondrial transfer and examines its role in various diseases. Furthermore, we provide a comprehensive overview of the inhibitors and activators of intercellular mitochondrial transfer, offering a unique perspective to illustrate the relationship between intercellular mitochondrial transfer and diseases.


Assuntos
Mitocôndrias , Humanos , Mitocôndrias/metabolismo , Animais , Comunicação Celular , Vesículas Extracelulares/metabolismo , Transporte Biológico , Endocitose/fisiologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/terapia
6.
Int J Nanomedicine ; 19: 9091-9107, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39258003

RESUMO

Purpose: Castration Resistant Prostate Cancer (CRPC) is characterized by poor prognosis and limited therapeutic options. AgNPs functionalized with glucose (G-AgNPs) were observed cytotoxic to CRPC cell lines (PC-3 and Du-145) and not LNCaP. This study aims to evaluate AgNPs and G-AgNPs' uptake mechanisms in these cells and understand their role in the selective effect against CRPC cells. Methods: Uptake of AgNPs and G-AgNPs was assessed through transmission electron microscopy (TEM). A microRNA (miRNAs) analysis approach was used to uncover the main molecular differences responsible for the endocytic mechanisms' regulation. Caveolin (Cav) 1 and 2 mRNA and protein levels were assessed in the three cell lines. Caveolae-dependent endocytosis was inhibited with genistein or siCav1- and siCav2- in PC-3 and Du-145 and resazurin assay was used to evaluate viability after AgNPs and G-AgNPs administration. Caveolae-dependent endocytosis was induced with Cav1+ and Cav2+ plasmids in LNCaP, resazurin assay was used to evaluate viability after AgNPs and G-AgNPs administration and TEM to assess their location. Results: AgNPs and G-AgNPs were not uptaked by LNCaP. miRNA analysis revealed 37 upregulated and 90 downregulated miRNAs. Functional enrichment analysis of miRNAs' targets resulted in enrichment of terms related to endocytosis and caveolae. We observed that Cav1 and Cav2 are not expressed in LNCaP. Inhibiting caveolae-dependent endocytosis in Du-145 and PC-3 led to a significative reduction of cytotoxic capacity of AgNPs and G-AgNPs and induction of caveolae-dependent endocytosis in LNCaP lead to a significative increase as well as their uptake by cells. Conclusion: This study shows the potential of these AgNPs as a new therapeutic approach directed to CRPC patients, uncovers caveolae-dependent endocytosis as the uptake mechanism of these AgNPs and highlights deregulation of Cav1 and Cav2 expression as a key difference in hormone sensitive and resistant PCa cells which may be responsible for drug resistance.


Assuntos
Cavéolas , Caveolina 1 , Endocitose , Nanopartículas Metálicas , MicroRNAs , Neoplasias de Próstata Resistentes à Castração , Prata , Masculino , Humanos , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Neoplasias de Próstata Resistentes à Castração/metabolismo , Cavéolas/metabolismo , Cavéolas/efeitos dos fármacos , Prata/química , Prata/farmacologia , Prata/farmacocinética , Caveolina 1/metabolismo , Caveolina 1/genética , Nanopartículas Metálicas/química , Linhagem Celular Tumoral , MicroRNAs/metabolismo , MicroRNAs/genética , Sobrevivência Celular/efeitos dos fármacos , Caveolina 2/metabolismo , Caveolina 2/genética , Antineoplásicos/farmacologia , Células PC-3
7.
J Neurosci ; 44(38)2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39147590

RESUMO

Ribbon synapses of inner hair cells (IHCs) are uniquely designed for ultrafast and indefatigable neurotransmission of the sound. The molecular machinery ensuring the efficient, compensatory recycling of the synaptic vesicles (SVs), however, remains elusive. This study showed that hair cell knock-out of murine Dmxl2, whose human homolog is responsible for nonsyndromic sensorineural hearing loss DFNA71, resulted in auditory synaptopathy by impairing synaptic endocytosis and recycling. The mutant mice in the C57BL/6J background of either sex had mild hearing loss with severely diminished wave I amplitude of the auditory brainstem response. Membrane capacitance measurements of the IHCs revealed deficiency in sustained synaptic exocytosis and endocytic membrane retrieval. Consistent with the electrophysiological findings, 3D electron microscopy reconstruction showed reduced reserve pool of SVs and endocytic compartments, while the membrane-proximal and ribbon-associated vesicles remain intact. Our results propose an important role of DMXL2 in hair cell endocytosis and recycling of the SVs.


Assuntos
Endocitose , Células Ciliadas Auditivas Internas , Proteínas do Tecido Nervoso , Vesículas Sinápticas , Animais , Feminino , Masculino , Camundongos , Endocitose/fisiologia , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Exocitose/fisiologia , Células Ciliadas Auditivas Internas/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vesículas Sinápticas/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo
8.
Biochem Soc Trans ; 52(4): 1703-1713, 2024 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-39092762

RESUMO

Evading programmed cell death (PCD) is a hallmark of cancer that allows tumor cells to survive and proliferate unchecked. Endocytosis, the process by which cells internalize extracellular materials, has emerged as a key regulator of cell death pathways in cancer. Many tumor types exhibit dysregulated endocytic dynamics that fuel their metabolic demands, promote resistance to cytotoxic therapies, and facilitate immune evasion. This review examines the roles of endocytosis in apoptotic resistance and immune escape mechanisms utilized by cancer cells. We highlight how inhibiting endocytosis can sensitize malignant cells to therapeutic agents and restore susceptibility to PCD. Strategies to modulate endocytosis for enhanced cancer treatment are discussed, including targeting endocytic regulatory proteins, altering membrane biophysical properties, and inhibiting Rho-associated kinases. While promising, challenges remain regarding the specificity and selectivity of endocytosis-targeting agents. Nonetheless, harnessing endocytic pathways represents an attractive approach to overcome apoptotic resistance and could yield more effective therapies by rendering cancer cells vulnerable to PCD. Understanding the interplay between endocytosis and PCD regulation is crucial for developing novel anticancer strategies that selectively induce tumor cell death.


Assuntos
Apoptose , Endocitose , Neoplasias , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Animais
9.
Cells ; 13(15)2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39120284

RESUMO

Mitochondria are crucial for cellular ATP production. They are highly dynamic organelles, whose morphology and function are controlled through mitochondrial fusion and fission. The specific roles of mitochondria in podocytes, the highly specialized cells of the kidney glomerulus, remain less understood. Given the significant structural, functional, and molecular similarities between mammalian podocytes and Drosophila nephrocytes, we employed fly nephrocytes to explore the roles of mitochondria in cellular function. Our study revealed that alterations in the Pink1-Park (mammalian PINK1-PRKN) pathway can disrupt mitochondrial dynamics in Drosophila nephrocytes. This disruption led to either fragmented or enlarged mitochondria, both of which impaired mitochondrial function. The mitochondrial dysfunction subsequently triggered defective intracellular endocytosis, protein aggregation, and cellular damage. These findings underscore the critical roles of mitochondria in nephrocyte functionality.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Endocitose , Mitocôndrias , Dinâmica Mitocondrial , Podócitos , Animais , Podócitos/metabolismo , Podócitos/patologia , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Drosophila melanogaster/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ubiquitina-Proteína Ligases
10.
ACS Nano ; 18(34): 23090-23103, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39143650

RESUMO

Actin- and microtubule (MT)-based transport systems are essential for intracellular transport. During influenza A virus (IAV) infection, MTs provide long tracks for virus trafficking toward the nucleus. However, the role of the actin cytoskeleton in IAV entry and especially the transit process is still ambiguous. Here, by using quantum dot-based single-virus tracking, it was revealed that the actin cytoskeleton was crucial for the virus entry via clathrin-mediated endocytosis (CME). After entry via CME, the virus reached MTs through three different pathways: the virus (1) was driven by myosin VI to move along actin filaments to reach MTs (AF); (2) was propelled by actin tails assembled by an Arp2/3-dependent mechanism to reach MTs (AT); and (3) directly reached MTs without experiencing actin-related movement (NA). Therefore, the NA pathway was the main one and the fastest for the virus to reach MTs. The AT pathway was activated only when plenty of viruses entered the cell. The viruses transported by the AF and AT pathways shared similar moving velocities, durations, and displacements. This study comprehensively visualized the role of the actin cytoskeleton in IAV entry and transport, revealing different pathways for IAV to reach MTs after entry. The results are of great significance for globally understanding IAV infection and the cellular endocytic transport pathway.


Assuntos
Endocitose , Vírus da Influenza A , Microtúbulos , Pontos Quânticos , Pontos Quânticos/química , Microtúbulos/metabolismo , Microtúbulos/virologia , Humanos , Vírus da Influenza A/fisiologia , Internalização do Vírus , Animais , Cães , Células Madin Darby de Rim Canino , Citoesqueleto de Actina/metabolismo
11.
Int J Biol Macromol ; 277(Pt 3): 134517, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39111497

RESUMO

Fucoidan-coated pH sensitive liposomes were designed for targeted delivery of gemcitabine (FU-GEM PSL) to treat pancreatic cancer (PC). FU-GEM PSL had a particle size of 175.3 ± 4.9 nm, zeta potential of -19.0 ± 3.7 mV, encapsulation efficiency (EE) of 74.05 ± 0.17 %, and drug loading (DL) of 21.27 ± 0.05 %. Cell experiments in vitro showed that FU-GEM PSL could increase the release of GEM and drug concentration, and could inhibit tumor cell proliferation by affecting the cell cycle. FU-GEM PSL entered cells through macropinocytosis and caveolin-mediated endocytosis to exert effects. Meanwhile, the expression of P-selectin was detected in human tissues, demonstrating the feasibility of targeting FU. Moreover, combined with animal experiments in vivo, FU-GEM PSL could inhibit the development of PC. Furthermore, anti-tumor experiments in vivo carried on BALB/c mice indicated that FU-GEM PSL had tumor suppression abilities and safety. Therefore, FU-GEM PSL is a promising formulation for PC therapy.


Assuntos
Proliferação de Células , Desoxicitidina , Gencitabina , Lipossomos , Neoplasias Pancreáticas , Polissacarídeos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Desoxicitidina/química , Desoxicitidina/administração & dosagem , Animais , Polissacarídeos/química , Polissacarídeos/farmacologia , Lipossomos/química , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Proliferação de Células/efeitos dos fármacos , Linhagem Celular Tumoral , Camundongos Endogâmicos BALB C , Liberação Controlada de Fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Sistemas de Liberação de Medicamentos , Endocitose/efeitos dos fármacos
12.
Cell Rep ; 43(8): 114649, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39159044

RESUMO

Each cargo in a cell employs a unique set of motor proteins for its transport. To dissect the roles of each type of motor, we developed optogenetic inhibitors of endogenous kinesin-1, -2, -3 and dynein motors and examined their effect on the transport of early endosomes, late endosomes, and lysosomes. While kinesin-1, -3, and dynein transport vesicles at all stages of endocytosis, kinesin-2 primarily drives late endosomes and lysosomes. Transient optogenetic inhibition of kinesin-1 or dynein causes both early and late endosomes to move more processively by relieving competition with opposing motors. Kinesin-2 and -3 support long-range transport, and optogenetic inhibition reduces the distances that their cargoes move. These results suggest that the directionality of transport is controlled through regulating kinesin-1 and dynein activity. On vesicles transported by several kinesin and dynein motors, modulating the activity of a single type of motor on the cargo is sufficient to direct motility.


Assuntos
Dineínas , Cinesinas , Optogenética , Cinesinas/metabolismo , Optogenética/métodos , Dineínas/metabolismo , Humanos , Animais , Endossomos/metabolismo , Lisossomos/metabolismo , Transporte Biológico , Células HeLa , Endocitose
13.
Science ; 385(6711): eado2032, 2024 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-39172837

RESUMO

Clathrin-mediated endocytosis has characteristic features in neuronal dendrites and presynapses, but how membrane proteins are internalized along the axon shaft remains unclear. We focused on clathrin-coated structures and endocytosis along the axon initial segment (AIS) and their relationship to the periodic actin-spectrin scaffold that lines the axonal plasma membrane. A combination of super-resolution microscopy and platinum-replica electron microscopy on cultured neurons revealed that AIS clathrin-coated pits form within "clearings", circular areas devoid of actin-spectrin mesh. Actin-spectrin scaffold disorganization increased clathrin-coated pit formation. Cargo uptake and live-cell imaging showed that AIS clathrin-coated pits are particularly stable. Neuronal plasticity-inducing stimulation triggered internalization of the clathrin-coated pits through polymerization of branched actin around them. Thus, spectrin and actin regulate clathrin-coated pit formation and scission to control endocytosis at the AIS.


Assuntos
Actinas , Axônios , Clatrina , Endocitose , Espectrina , Animais , Humanos , Ratos , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Axônios/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Clatrina/metabolismo , Vesículas Revestidas por Clatrina/metabolismo , Invaginações Revestidas da Membrana Celular/metabolismo , Células HEK293 , Plasticidade Neuronal , Neurônios/metabolismo , Espectrina/metabolismo
14.
Viruses ; 16(8)2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39205315

RESUMO

The efficiency of virus internalization into target cells is a major determinant of infectivity. SARS-CoV-2 internalization occurs via S-protein-mediated cell binding followed either by direct fusion with the plasma membrane or endocytosis and subsequent fusion with the endosomal membrane. Despite the crucial role of virus internalization, the precise kinetics of the processes involved remains elusive. We developed a pipeline, which combines live-cell microscopy and advanced image analysis, for measuring the rates of multiple internalization-associated molecular events of single SARS-CoV-2-virus-like particles (VLPs), including endosome ingression and pH change. Our live-cell imaging experiments demonstrate that only a few minutes after binding to the plasma membrane, VLPs ingress into RAP5-negative endosomes via dynamin-dependent scission. Less than two minutes later, VLP speed increases in parallel with a pH drop below 5, yet these two events are not interrelated. By co-imaging fluorescently labeled nucleocapsid proteins, we show that nucleocapsid release occurs with similar kinetics to VLP acidification. Neither Omicron mutations nor abrogation of the S protein polybasic cleavage site affected the rate of VLP internalization, indicating that they do not confer any significant advantages or disadvantages during this process. Finally, we observe that VLP internalization occurs two to three times faster in VeroE6 than in A549 cells, which may contribute to the greater susceptibility of the former cell line to SARS-CoV-2 infection. Taken together, our precise measurements of the kinetics of VLP internalization-associated processes shed light on their contribution to the effectiveness of SARS-CoV-2 propagation in cells.


Assuntos
COVID-19 , Endossomos , SARS-CoV-2 , Internalização do Vírus , SARS-CoV-2/fisiologia , SARS-CoV-2/metabolismo , Humanos , Cinética , COVID-19/virologia , COVID-19/metabolismo , Endossomos/metabolismo , Endossomos/virologia , Endocitose , Animais , Concentração de Íons de Hidrogênio , Chlorocebus aethiops , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Vero , Membrana Celular/metabolismo , Membrana Celular/virologia , Vírion/metabolismo
15.
J Physiol ; 602(17): 4291-4307, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39106251

RESUMO

ClC-K/barttin channels are involved in the transepithelial transport of chloride in the kidney and inner ear. Their physiological role is crucial in humans because mutations in CLCNKB or BSND, encoding ClC-Kb and barttin, cause Bartter's syndrome types III and IV, respectively. In vitro experiments have shown that an amino acid change in a proline-tyrosine motif in the C-terminus of barttin stimulates ClC-K currents. The molecular mechanism of this enhancement and whether this potentiation has any in vivo relevance remains unknown. We performed electrophysiological and biochemical experiments in Xenopus oocytes and kidney cells co-expressing ClC-K and barttin constructs. We demonstrated that barttin possesses a YxxØ motif and, when mutated, increases ClC-K plasma membrane stability, resulting in larger currents. To address the impact of mutating this motif in kidney physiology, we generated a knock-in mouse. Comparing wild-type (WT) and knock-in mice under a standard diet, we could not observe any difference in ClC-K and barttin protein levels or localization, either in urinary or plasma parameters. However, under a high-sodium low-potassium diet, known to induce hyperplasia of distal convoluted tubules, knock-in mice exhibit reduced hyperplasia compared to WT mice. In summary, our in vitro and in vivo studies demonstrate that the previously identified PY motif is indeed an endocytic YxxØ motif in which mutations cause a gain of function of the channel. KEY POINTS: It is revealed by mutagenesis and functional experiments that a previously identified proline-tyrosine motif regulating ClC-K plasma membrane levels is indeed an endocytic YxxØ motif. Biochemical characterization of mutants in the YxxØ motif in Xenopus oocytes and human embryonic kidney cells indicates that mutants showed increased plasma membrane levels as a result of an increased stability, resulting in higher function of ClC-K channels. Mutation of this motif does not affect barttin protein expression and subcellular localization in vivo. Knock-in mice with a mutation in this motif, under conditions of a high-sodium low-potassium diet, exhibit less hyperplasia in the distal convoluted tubule than wild-type animals, indicating a gain of function of the channel in vivo.


Assuntos
Canais de Cloreto , Endocitose , Xenopus laevis , Animais , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Endocitose/fisiologia , Camundongos , Túbulos Renais Distais/metabolismo , Hiperplasia , Humanos , Feminino , Transportadores de Sulfato/genética , Transportadores de Sulfato/metabolismo , Camundongos Endogâmicos C57BL , Células HEK293 , Oócitos/metabolismo , Proteínas de Transporte de Ânions
16.
J Cell Sci ; 137(16)2024 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-39161994

RESUMO

Clathrin-coated vesicles (CCVs), generated by clathrin-mediated endocytosis (CME), are essential eukaryotic trafficking organelles that transport extracellular and plasma membrane-bound materials into the cell. In this Review, we explore mechanisms of CME in mammals, yeasts and plants, and highlight recent advances in the characterization of endocytosis in plants. Plants separated from mammals and yeast over 1.5 billion years ago, and plant cells have distinct biophysical parameters that can influence CME, such as extreme turgor pressure. Plants can therefore provide a wider perspective on fundamental processes in eukaryotic cells. We compare key mechanisms that drive CCV formation and explore what these mechanisms might reveal about the core principles of endocytosis across the tree of life. Fascinatingly, CME in plants appears to more closely resemble that in mammalian cells than that in yeasts, despite plants being evolutionarily further from mammals than yeast. Endocytic initiation appears to be highly conserved across these three systems, requiring similar protein domains and regulatory processes. Clathrin coat proteins and their honeycomb lattice structures are also highly conserved. However, major differences are found in membrane-bending mechanisms. Unlike in mammals or yeast, plant endocytosis occurs independently of actin, highlighting that mechanistic assumptions about CME across different systems should be made with caution.


Assuntos
Vesículas Revestidas por Clatrina , Endocitose , Mamíferos , Animais , Vesículas Revestidas por Clatrina/metabolismo , Mamíferos/metabolismo , Plantas/metabolismo , Plantas/microbiologia , Humanos , Clatrina/metabolismo , Leveduras/metabolismo
17.
J Agric Food Chem ; 72(36): 19786-19799, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39187786

RESUMO

Bioactive peptides, derived from short protein fragments, are recognized for their neuroprotective properties and potential therapeutic applications in treating central nervous system (CNS) diseases. However, a significant challenge for these peptides is their ability to penetrate the blood-brain barrier (BBB). EVSGPGYSPN (EV-10) peptide, a walnut-derived peptide, has demonstrated promising neuroprotective effects in vivo. This study aimed to investigate the transportability of EV-10 across the BBB, explore its capacity to penetrate this barrier, and elucidate the regulatory mechanisms underlying peptide-induced cellular internalization and transport pathways within the BBB. The results indicated that at a concentration of 100 µM and osmotic time of 4 h, the apparent permeability coefficient of EV-10 was Papp = 8.52166 ± 0.58 × 10-6 cm/s. The penetration efficiency of EV-10 was influenced by time, concentration, and temperature. Utilizing Western blot analysis, immunofluorescence, and flow cytometry, in conjunction with the caveolin (Cav)-specific inhibitor M-ß-CD, we confirmed that EV-10 undergoes transcellular transport through a Cav-dependent endocytosis pathway. Notably, the tight junction proteins ZO-1, occludin, and claudin-5 were not disrupted by EV-10. Throughout its transport, EV-10 was localized within the mitochondria, Golgi apparatus, endoplasmic reticulum, lysosomes, endosomes, and cell membranes. Moreover, Cav-1 overexpression facilitated the release of EV-10 from lysosomes. Evidence of EV-10 accumulation was observed in mouse brains using brain slice scans. This study is the first to demonstrate that Cav-1 can facilitate the targeted delivery of walnut-derived peptide to the brain, laying a foundation for the development of functional foods aimed at CNS disease intervention.


Assuntos
Barreira Hematoencefálica , Juglans , Peptídeos , Juglans/química , Juglans/metabolismo , Barreira Hematoencefálica/metabolismo , Animais , Camundongos , Peptídeos/química , Peptídeos/metabolismo , Peptídeos/farmacologia , Transporte Biológico , Caveolinas/metabolismo , Caveolinas/química , Humanos , Endocitose , Proteínas de Plantas/metabolismo , Proteínas de Plantas/química , Nozes/química , Nozes/metabolismo
18.
EMBO Rep ; 25(9): 4013-4032, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39112792

RESUMO

VE-cadherin is a major component of the cell adhesion machinery which provides integrity and plasticity of the barrier function of endothelial junctions. Here, we analyze whether ubiquitination of VE-cadherin is involved in the regulation of the endothelial barrier in inflammation in vivo. We show that histamine and thrombin stimulate ubiquitination of VE-cadherin in HUVEC, which is completely blocked if the two lysine residues K626 and K633 are replaced by arginine. Similarly, these mutations block histamine-induced endocytosis of VE-cadherin. We describe two knock-in mouse lines with endogenous VE-cadherin being replaced by either a VE-cadherin K626/633R or a VE-cadherin KallR mutant, where all seven lysine residues are mutated. Mutant mice are viable, healthy and fertile with normal expression levels of junctional VE-cadherin. Histamine- or LPS-induced vascular permeability in the skin or lung of both of these mutant mice are clearly and similarly reduced in comparison to WT mice. Additionally, we detect a role of K626/633 for lysosomal targeting. Collectively, our findings identify ubiquitination of VE-cadherin as important for the induction of vascular permeability in the inflamed skin and lung.


Assuntos
Antígenos CD , Caderinas , Permeabilidade Capilar , Inflamação , Ubiquitinação , Animais , Humanos , Camundongos , Antígenos CD/metabolismo , Antígenos CD/genética , Caderinas/metabolismo , Caderinas/genética , Endocitose , Técnicas de Introdução de Genes , Histamina/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Inflamação/metabolismo , Inflamação/genética , Lipopolissacarídeos/farmacologia , Pulmão/metabolismo , Lisossomos/metabolismo , Pele/metabolismo
19.
Int J Biol Macromol ; 278(Pt 1): 134668, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39137851

RESUMO

Immunotoxins (ITs) are recombinant chimeric proteins that combine a protein toxin with a targeting moiety to facilitate the selective delivery of the toxin to cancer cells. Here, we present a novel strategy to enhance the cytosolic access of ITs by promoting their dissociation from target receptors under the reducing conditions of the endocytic pathway. We engineered monobodySS, a human fibronectin type III domain-based monobody with disulfide bond (SS)-containing paratopes, targeting receptors such as EGFR, EpCAM, Her2, and FAP. MonobodySS exhibited SS-dependent target receptor binding with a significant reduction in binding under reducing conditions. We then created monobodySS-based ITs carrying a 25 kDa fragment of Pseudomonas exotoxin A (PE25), termed monobodySS-PE25. These ITs showed dose-dependent cytotoxicity against target receptor-expressing cancer cells and a wider therapeutic window due to higher efficacy at lower doses compared to controls with SS reduction inhibited. ERSS/28-PE25, with a KD of 28 nM for EGFR, demonstrated superior tumor-killing potency compared to ER/21-PE25, which lacks an SS bond, at equivalent and lower doses. In vivo, ERSS/28-PE25 outperformed ER/21-PE25 in suppressing tumor growth in EGFR-overexpressing xenograft mouse models. This study presents a strategy for developing solid tumor-targeting ITs using SS-containing paratopes to enhance cytosolic delivery and antitumor efficacy.


Assuntos
Endocitose , Exotoxinas , Imunotoxinas , Humanos , Imunotoxinas/farmacologia , Imunotoxinas/química , Animais , Endocitose/efeitos dos fármacos , Camundongos , Linhagem Celular Tumoral , Exotoxinas/farmacologia , Exotoxinas/química , Exotoxina A de Pseudomonas aeruginosa , ADP Ribose Transferases/farmacologia , ADP Ribose Transferases/química , Ensaios Antitumorais Modelo de Xenoenxerto , Toxinas Bacterianas/química , Toxinas Bacterianas/farmacologia , Oxirredução/efeitos dos fármacos , Feminino
20.
Development ; 151(20)2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39171380

RESUMO

Delivering molecular tools into oocytes is essential for developmental and reproductive biology. Microinjection, the conventional method, is equipment intensive, often technically challenging and has a low yield, and is impractical in species with delicate oocytes or restricted spawning seasons. To overcome these limitations, we developed VitelloTag, a cost-effective, high-throughput system using vitellogenin-derived fusion proteins to enable efficient cargo delivery via receptor-mediated endocytosis. We demonstrate its utility by delivering Cas9/sgRNA complexes in two distantly related species for gene knockout.


Assuntos
Oócitos , Vitelogeninas , Oócitos/metabolismo , Animais , Vitelogeninas/metabolismo , Vitelogeninas/genética , Feminino , Sistemas CRISPR-Cas/genética , Endocitose , Técnicas de Inativação de Genes , Microinjeções/métodos , Peixe-Zebra/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...