Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.945
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(9): 2175-2193.e21, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38552623

RESUMEN

In addition to long-distance molecular motor-mediated transport, cellular vesicles also need to be moved at short distances with defined directions to meet functional needs in subcellular compartments but with unknown mechanisms. Such short-distance vesicle transport does not involve molecular motors. Here, we demonstrate, using synaptic vesicle (SV) transport as a paradigm, that phase separation of synaptic proteins with vesicles can facilitate regulated, directional vesicle transport between different presynaptic bouton sub-compartments. Specifically, a large coiled-coil scaffold protein Piccolo, in response to Ca2+ and via its C2A domain-mediated Ca2+ sensing, can extract SVs from the synapsin-clustered reserve pool condensate and deposit the extracted SVs onto the surface of the active zone protein condensate. We further show that the Trk-fused gene, TFG, also participates in COPII vesicle trafficking from ER to the ER-Golgi intermediate compartment via phase separation. Thus, phase separation may play a general role in short-distance, directional vesicle transport in cells.


Asunto(s)
Vesículas Cubiertas por Proteínas de Revestimiento , Retículo Endoplásmico , Vesículas Sinápticas , Animales , Vesículas Sinápticas/metabolismo , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Retículo Endoplásmico/metabolismo , Calcio/metabolismo , Aparato de Golgi/metabolismo , Ratas , Transporte Biológico , Terminales Presinápticos/metabolismo , Sinapsinas/metabolismo , Condensados Biomoleculares/metabolismo , Proteínas del Citoesqueleto/metabolismo , Separación de Fases
2.
Cell ; 187(16): 4272-4288.e20, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39013469

RESUMEN

Vesicle trafficking is a fundamental process that allows for the sorting and transport of specific proteins (i.e., "cargoes") to different compartments of eukaryotic cells. Cargo recognition primarily occurs through coats and the associated proteins at the donor membrane. However, it remains unclear whether cargoes can also be selected at other stages of vesicle trafficking to further enhance the fidelity of the process. The WDR11-FAM91A1 complex functions downstream of the clathrin-associated AP-1 complex to facilitate protein transport from endosomes to the TGN. Here, we report the cryo-EM structure of human WDR11-FAM91A1 complex. WDR11 directly and specifically recognizes a subset of acidic clusters, which we term super acidic clusters (SACs). WDR11 complex assembly and its binding to SAC-containing proteins are indispensable for the trafficking of SAC-containing proteins and proper neuronal development in zebrafish. Our studies thus uncover that cargo proteins could be recognized in a sequence-specific manner downstream of a protein coat.


Asunto(s)
Microscopía por Crioelectrón , Transporte de Proteínas , Pez Cebra , Humanos , Animales , Endosomas/metabolismo , Células HEK293 , Células HeLa , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/química , Unión Proteica
3.
Cell ; 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39208797

RESUMEN

In mammalian cells, two phosphatidylserine (PS) synthases drive PS synthesis. Gain-of-function mutations in the Ptdss1 gene lead to heightened PS production, causing Lenz-Majewski syndrome (LMS). Recently, pharmacological inhibition of PSS1 has been shown to suppress tumorigenesis. Here, we report the cryo-EM structures of wild-type human PSS1 (PSS1WT), the LMS-causing Pro269Ser mutant (PSS1P269S), and PSS1WT in complex with its inhibitor DS55980254. PSS1 contains 10 transmembrane helices (TMs), with TMs 4-8 forming a catalytic core in the luminal leaflet. These structures revealed a working mechanism of PSS1 akin to the postulated mechanisms of the membrane-bound O-acyltransferase family. Additionally, we showed that both PS and DS55980254 can allosterically inhibit PSS1 and that inhibition by DS55980254 activates the SREBP pathways, thus enhancing the expression of LDL receptors and increasing cellular LDL uptake. This work uncovers a mechanism of mammalian PS synthesis and suggests that selective PSS1 inhibitors have the potential to lower blood cholesterol levels.

4.
Cell ; 186(15): 3307-3324.e30, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37385249

RESUMEN

The ability to map trafficking for thousands of endogenous proteins at once in living cells would reveal biology currently invisible to both microscopy and mass spectrometry. Here, we report TransitID, a method for unbiased mapping of endogenous proteome trafficking with nanometer spatial resolution in living cells. Two proximity labeling (PL) enzymes, TurboID and APEX, are targeted to source and destination compartments, and PL with each enzyme is performed in tandem via sequential addition of their small-molecule substrates. Mass spectrometry identifies the proteins tagged by both enzymes. Using TransitID, we mapped proteome trafficking between cytosol and mitochondria, cytosol and nucleus, and nucleolus and stress granules (SGs), uncovering a role for SGs in protecting the transcription factor JUN from oxidative stress. TransitID also identifies proteins that signal intercellularly between macrophages and cancer cells. TransitID offers a powerful approach for distinguishing protein populations based on compartment or cell type of origin.


Asunto(s)
Mitocondrias , Proteoma , Proteoma/metabolismo , Mitocondrias/metabolismo , Nucléolo Celular/metabolismo , Espectrometría de Masas/métodos , Regulación de la Expresión Génica
5.
Cell ; 186(17): 3706-3725.e29, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37562402

RESUMEN

The bone marrow in the skull is important for shaping immune responses in the brain and meninges, but its molecular makeup among bones and relevance in human diseases remain unclear. Here, we show that the mouse skull has the most distinct transcriptomic profile compared with other bones in states of health and injury, characterized by a late-stage neutrophil phenotype. In humans, proteome analysis reveals that the skull marrow is the most distinct, with differentially expressed neutrophil-related pathways and a unique synaptic protein signature. 3D imaging demonstrates the structural and cellular details of human skull-meninges connections (SMCs) compared with veins. Last, using translocator protein positron emission tomography (TSPO-PET) imaging, we show that the skull bone marrow reflects inflammatory brain responses with a disease-specific spatial distribution in patients with various neurological disorders. The unique molecular profile and anatomical and functional connections of the skull show its potential as a site for diagnosing, monitoring, and treating brain diseases.


Asunto(s)
Médula Ósea , Enfermedades del Sistema Nervioso , Cráneo , Animales , Humanos , Ratones , Médula Ósea/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Proteínas Portadoras/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología , Tomografía de Emisión de Positrones/métodos , Receptores de GABA/metabolismo , Cráneo/citología , Cráneo/diagnóstico por imagen
6.
Cell ; 184(9): 2412-2429.e16, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33852913

RESUMEN

Cellular versatility depends on accurate trafficking of diverse proteins to their organellar destinations. For the secretory pathway (followed by approximately 30% of all proteins), the physical nature of the vessel conducting the first portage (endoplasmic reticulum [ER] to Golgi apparatus) is unclear. We provide a dynamic 3D view of early secretory compartments in mammalian cells with isotropic resolution and precise protein localization using whole-cell, focused ion beam scanning electron microscopy with cryo-structured illumination microscopy and live-cell synchronized cargo release approaches. Rather than vesicles alone, the ER spawns an elaborate, interwoven tubular network of contiguous lipid bilayers (ER exit site) for protein export. This receptacle is capable of extending microns along microtubules while still connected to the ER by a thin neck. COPII localizes to this neck region and dynamically regulates cargo entry from the ER, while COPI acts more distally, escorting the detached, accelerating tubular entity on its way to joining the Golgi apparatus through microtubule-directed movement.


Asunto(s)
Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Microtúbulos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Transporte Biológico Activo , Células HeLa , Humanos , Transporte de Proteínas
7.
Cell ; 181(3): 637-652.e15, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32272059

RESUMEN

Many cytosolic proteins lacking a signal peptide, called leaderless cargoes, are secreted through unconventional secretion. Vesicle trafficking is a major pathway involved. It is unclear how leaderless cargoes enter into the vesicle. Here, we find a translocation pathway regulating vesicle entry and secretion of leaderless cargoes. We identify TMED10 as a protein channel for the vesicle entry and secretion of many leaderless cargoes. The interaction of TMED10 C-terminal region with a motif in the cargo accounts for the selective release of the cargoes. In an in vitro reconstitution assay, TMED10 directly mediates the membrane translocation of leaderless cargoes into the liposome, which is dependent on protein unfolding and enhanced by HSP90s. In the cell, TMED10 localizes on the endoplasmic reticulum (ER)-Golgi intermediate compartment and directs the entry of cargoes into this compartment. Furthermore, cargo induces the formation of TMED10 homo-oligomers which may act as a protein channel for cargo translocation.


Asunto(s)
Sistemas de Translocación de Proteínas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Transporte Biológico , Línea Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Citosol/metabolismo , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Señales de Clasificación de Proteína , Sistemas de Translocación de Proteínas/fisiología , Transporte de Proteínas/fisiología , Proteínas/metabolismo , Vías Secretoras , Proteínas de Transporte Vesicular/fisiología
8.
Cell ; 183(5): 1325-1339.e21, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33080218

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a recently identified coronavirus that causes the respiratory disease known as coronavirus disease 2019 (COVID-19). Despite the urgent need, we still do not fully understand the molecular basis of SARS-CoV-2 pathogenesis. Here, we comprehensively define the interactions between SARS-CoV-2 proteins and human RNAs. NSP16 binds to the mRNA recognition domains of the U1 and U2 splicing RNAs and acts to suppress global mRNA splicing upon SARS-CoV-2 infection. NSP1 binds to 18S ribosomal RNA in the mRNA entry channel of the ribosome and leads to global inhibition of mRNA translation upon infection. Finally, NSP8 and NSP9 bind to the 7SL RNA in the signal recognition particle and interfere with protein trafficking to the cell membrane upon infection. Disruption of each of these essential cellular functions acts to suppress the interferon response to viral infection. Our results uncover a multipronged strategy utilized by SARS-CoV-2 to antagonize essential cellular processes to suppress host defenses.


Asunto(s)
COVID-19/metabolismo , Interacciones Huésped-Patógeno , Biosíntesis de Proteínas , Empalme del ARN , SARS-CoV-2/metabolismo , Proteínas no Estructurales Virales/metabolismo , Células A549 , Animales , COVID-19/virología , Chlorocebus aethiops , Células HEK293 , Humanos , Interferones/metabolismo , Transporte de Proteínas , ARN Mensajero/metabolismo , ARN Ribosómico 18S/metabolismo , ARN Citoplasmático Pequeño/química , ARN Citoplasmático Pequeño/metabolismo , Partícula de Reconocimiento de Señal/química , Partícula de Reconocimiento de Señal/metabolismo , Células Vero , Proteínas no Estructurales Virales/química
9.
Annu Rev Cell Dev Biol ; 37: 369-389, 2021 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-34196570

RESUMEN

Wnt signaling has multiple functions beyond the transcriptional effects of ß-catenin stabilization. We review recent investigations that uncover new cell physiological effects through the regulation of Wnt receptor endocytosis, Wnt-induced stabilization of proteins (Wnt-STOP), macropinocytosis, increase in lysosomal activity, and metabolic changes. Many of these growth-promoting effects of canonical Wnt occur within minutes and are independent of new protein synthesis. A key element is the sequestration of glycogen synthase kinase 3 (GSK3) inside multivesicular bodies and lysosomes. Twenty percent of human proteins contain consecutive GSK3 phosphorylation motifs, which in the absence of Wnt can form phosphodegrons for polyubiquitination and proteasomal degradation. Wnt signaling by either the pharmacological inhibition of GSK3 or the loss of tumor-suppressor proteins, such as adenomatous polyposis coli (APC) and Axin1, increases lysosomal acidification, anabolic metabolites, and macropinocytosis, which is normally repressed by the GSK3-Axin1-APC destruction complex. The combination of these cell physiological effects drives cell growth.


Asunto(s)
Glucógeno Sintasa Quinasa 3 , Vía de Señalización Wnt , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Lisosomas/metabolismo , Fosforilación , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/fisiología
10.
Annu Rev Biochem ; 88: 487-514, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31220978

RESUMEN

Exosomes are small, single-membrane, secreted organelles of ∼30 to ∼200 nm in diameter that have the same topology as the cell and are enriched in selected proteins, lipids, nucleic acids, and glycoconjugates. Exosomes contain an array of membrane-associated, high-order oligomeric protein complexes, display pronounced molecular heterogeneity, and are created by budding at both plasma and endosome membranes. Exosome biogenesis is a mechanism of protein quality control, and once released, exosomes have activities as diverse as remodeling the extracellular matrix and transmitting signals and molecules to other cells. This pathway of intercellular vesicle traffic plays important roles in many aspects of human health and disease, including development, immunity, tissue homeostasis, cancer, and neurodegenerative diseases. In addition, viruses co-opt exosome biogenesis pathways both for assembling infectious particles and for establishing host permissiveness. On the basis of these and other properties, exosomes are being developed as therapeutic agents in multiple disease models.


Asunto(s)
Exosomas/metabolismo , Animales , Transporte Biológico , Exosomas/inmunología , Exosomas/fisiología , Exosomas/ultraestructura , Matriz Extracelular/metabolismo , Humanos , Neoplasias , Enfermedades Neurodegenerativas , Multimerización de Proteína , Transducción de Señal
11.
Cell ; 177(7): 1682-1699, 2019 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-31199916

RESUMEN

Macroautophagy (herein referred to as autophagy) is an evolutionary ancient mechanism that culminates with the lysosomal degradation of superfluous or potentially dangerous cytosolic entities. Over the past 2 decades, the molecular mechanisms underlying several variants of autophagy have been characterized in detail. Accumulating evidence suggests that most, if not all, components of the molecular machinery for autophagy also mediate autophagy-independent functions. Here, we discuss emerging data on the non-autophagic functions of autophagy-relevant proteins.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia/fisiología , Lisosomas/metabolismo , Animales , Humanos
12.
Cell ; 177(7): 1771-1780.e12, 2019 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-31199917

RESUMEN

Cargo trafficking along microtubules is exploited by eukaryotic viruses, but no such examples have been reported in bacteria. Several large Pseudomonas phages assemble a dynamic, tubulin-based (PhuZ) spindle that centers replicating phage DNA sequestered within a nucleus-like structure. Here, we show that capsids assemble on the membrane and then move rapidly along PhuZ filaments toward the phage nucleus for DNA packaging. The spindle rotates the phage nucleus, distributing capsids around its surface. PhuZ filaments treadmill toward the nucleus at a constant rate similar to the rate of capsid movement and the linear velocity of nucleus rotation. Capsids become trapped along mutant static PhuZ filaments that are defective in GTP hydrolysis. Our results suggest a transport and distribution mechanism in which capsids attached to the sides of filaments are trafficked to the nucleus by PhuZ polymerization at the poles, demonstrating that the phage cytoskeleton evolved cargo-trafficking capabilities in bacteria.


Asunto(s)
Proteínas Bacterianas , Citoesqueleto , ADN Viral , Fagos Pseudomonas , Pseudomonas , Tubulina (Proteína) , Virión , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , ADN Viral/biosíntesis , ADN Viral/genética , Pseudomonas/genética , Pseudomonas/metabolismo , Pseudomonas/virología , Fagos Pseudomonas/genética , Fagos Pseudomonas/metabolismo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Virión/genética , Virión/metabolismo
13.
Cell ; 178(2): 374-384.e15, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31299201

RESUMEN

Multicellular lifestyle requires cell-cell connections. In multicellular cyanobacteria, septal junctions enable molecular exchange between sister cells and are required for cellular differentiation. The structure of septal junctions is poorly understood, and it is unknown whether they are capable of controlling intercellular communication. Here, we resolved the in situ architecture of septal junctions by electron cryotomography of cryo-focused ion beam-milled cyanobacterial filaments. Septal junctions consisted of a tube traversing the septal peptidoglycan. Each tube end comprised a FraD-containing plug, which was covered by a cytoplasmic cap. Fluorescence recovery after photobleaching showed that intercellular communication was blocked upon stress. Gating was accompanied by a reversible conformational change of the septal junction cap. We provide the mechanistic framework for a cell junction that predates eukaryotic gap junctions by a billion years. The conservation of a gated dynamic mechanism across different domains of life emphasizes the importance of controlling molecular exchange in multicellular organisms.


Asunto(s)
Uniones Comunicantes/metabolismo , Anabaena/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Carbonil Cianuro m-Clorofenil Hidrazona/análogos & derivados , Carbonil Cianuro m-Clorofenil Hidrazona/farmacología , Comunicación Celular/efectos de los fármacos , Microscopía por Crioelectrón , Uniones Comunicantes/química , Uniones Comunicantes/ultraestructura , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutagénesis
14.
Cell ; 176(1-2): 56-72.e15, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30612743

RESUMEN

Local translation regulates the axonal proteome, playing an important role in neuronal wiring and axon maintenance. How axonal mRNAs are localized to specific subcellular sites for translation, however, is not understood. Here we report that RNA granules associate with endosomes along the axons of retinal ganglion cells. RNA-bearing Rab7a late endosomes also associate with ribosomes, and real-time translation imaging reveals that they are sites of local protein synthesis. We show that RNA-bearing late endosomes often pause on mitochondria and that mRNAs encoding proteins for mitochondrial function are translated on Rab7a endosomes. Disruption of Rab7a function with Rab7a mutants, including those associated with Charcot-Marie-Tooth type 2B neuropathy, markedly decreases axonal protein synthesis, impairs mitochondrial function, and compromises axonal viability. Our findings thus reveal that late endosomes interact with RNA granules, translation machinery, and mitochondria and suggest that they serve as sites for regulating the supply of nascent pro-survival proteins in axons.


Asunto(s)
Endosomas/fisiología , Biosíntesis de Proteínas/fisiología , Proteínas de Unión al GTP rab/metabolismo , Animales , Axones/metabolismo , Endosomas/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , ARN/metabolismo , ARN Mensajero/metabolismo , ARN Mensajero/fisiología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/fisiología , Ribosomas/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/fisiología , Proteínas de Unión a GTP rab7
15.
Cell ; 179(2): 485-497.e18, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31543266

RESUMEN

Niemann-Pick type C (NPC) proteins are essential for sterol homeostasis, believed to drive sterol integration into the lysosomal membrane before redistribution to other cellular membranes. Here, using a combination of crystallography, cryo-electron microscopy, and biochemical and in vivo studies on the Saccharomyces cerevisiae NPC system (NCR1 and NPC2), we present a framework for sterol membrane integration. Sterols are transferred between hydrophobic pockets of vacuolar NPC2 and membrane-protein NCR1. NCR1 has its N-terminal domain (NTD) positioned to deliver a sterol to a tunnel connecting NTD to the luminal membrane leaflet 50 Å away. A sterol is caught inside this tunnel during transport, and a proton-relay network of charged residues in the transmembrane region is linked to this tunnel supporting a proton-driven transport mechanism. We propose a model for sterol integration that clarifies the role of NPC proteins in this essential eukaryotic pathway and that rationalizes mutations in patients with Niemann-Pick disease type C.


Asunto(s)
Proteínas Portadoras/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Esteroles/metabolismo , Proteínas de Transporte Vesicular/química , Transporte Biológico , Microscopía por Crioelectrón , Cristalografía , Membranas Intracelulares/metabolismo , Lisosomas/metabolismo , Dominios Proteicos , Vacuolas/metabolismo
16.
Annu Rev Cell Dev Biol ; 36: 237-264, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32749865

RESUMEN

Parkinson's disease (PD) is a leading cause of neurodegeneration that is defined by the selective loss of dopaminergic neurons and the accumulation of protein aggregates called Lewy bodies (LBs). The unequivocal identification of Mendelian inherited mutations in 13 genes in PD has provided transforming insights into the pathogenesis of this disease. The mechanistic analysis of several PD genes, including α-synuclein (α-syn), leucine-rich repeat kinase 2 (LRRK2), PTEN-induced kinase 1 (PINK1), and Parkin, has revealed central roles for protein aggregation, mitochondrial damage, and defects in endolysosomal trafficking in PD neurodegeneration. In this review, we outline recent advances in our understanding of these gene pathways with a focus on the emergent role of Rab (Ras analog in brain) GTPases and vesicular trafficking as a common mechanism that underpins how mutations in PD genes lead to neuronal loss. These advances have led to previously distinct genes such as vacuolar protein-sorting-associated protein 35 (VPS35) and LRRK2 being implicated in a common signaling pathway. A greater understanding of these common nodes of vesicular trafficking will be crucial for linking other PD genes and improving patient stratification in clinical trials underway against α-syn and LRRK2 targets.


Asunto(s)
Enfermedad de Parkinson/metabolismo , Animales , Autofagia , Vesículas Citoplasmáticas/metabolismo , Humanos , Mitocondrias/metabolismo , Enfermedad de Parkinson/genética , Agregado de Proteínas , Transporte de Proteínas
17.
Immunity ; 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39151425

RESUMEN

The intestinal microbiota determines immune responses against extraintestinal antigens, including tumor-associated antigens. Indeed, depletion or gross perturbation of the microbiota undermines the efficacy of cancer immunotherapy, thereby compromising the clinical outcome of cancer patients. In this review, we discuss the long-distance effects of the gut microbiota and the mechanisms governing antitumor immunity, such as the translocation of intestinal microbes into tumors, migration of leukocyte populations from the gut to the rest of the body, including tumors, as well as immunomodulatory microbial products and metabolites. The relationship between these pathways is incompletely understood, in particular the significance of the tumor microbiota with respect to the identification of host and/or microbial products that regulate the egress of bacteria and immunocytes toward tumor beds.

18.
Immunity ; 57(3): 541-558.e7, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38442708

RESUMEN

Cancer patients often receive a combination of antibodies targeting programmed death-ligand 1 (PD-L1) and cytotoxic T lymphocyte antigen-4 (CTLA4). We conducted a window-of-opportunity study in head and neck squamous cell carcinoma (HNSCC) to examine the contribution of anti-CTLA4 to anti-PD-L1 therapy. Single-cell profiling of on- versus pre-treatment biopsies identified T cell expansion as an early response marker. In tumors, anti-PD-L1 triggered the expansion of mostly CD8+ T cells, whereas combination therapy expanded both CD4+ and CD8+ T cells. Such CD4+ T cells exhibited an activated T helper 1 (Th1) phenotype. CD4+ and CD8+ T cells co-localized with and were surrounded by dendritic cells expressing T cell homing factors or antibody-producing plasma cells. T cell receptor tracing suggests that anti-CTLA4, but not anti-PD-L1, triggers the trafficking of CD4+ naive/central-memory T cells from tumor-draining lymph nodes (tdLNs), via blood, to the tumor wherein T cells acquire a Th1 phenotype. Thus, CD4+ T cell activation and recruitment from tdLNs are hallmarks of early response to anti-PD-L1 plus anti-CTLA4 in HNSCC.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias de Cabeza y Cuello , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello , Antígeno B7-H1/genética , Antígeno CTLA-4 , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Linfocitos T CD4-Positivos , Microambiente Tumoral
19.
Cell ; 174(3): 659-671.e14, 2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30053425

RESUMEN

The HIV accessory protein Nef counteracts immune defenses by subverting coated vesicle pathways. The 3.7 Å cryo-EM structure of a closed trimer of the clathrin adaptor AP-1, the small GTPase Arf1, HIV-1 Nef, and the cytosolic tail of the restriction factor tetherin suggested a mechanism for inactivating tetherin by Golgi retention. The 4.3 Å structure of a mutant Nef-induced dimer of AP-1 showed how the closed trimer is regulated by the dileucine loop of Nef. HDX-MS and mutational analysis were used to show how cargo dynamics leads to alternative Arf1 trimerization, directing Nef targets to be either retained at the trans-Golgi or sorted to lysosomes. Phosphorylation of the NL4-3 M-Nef was shown to regulate AP-1 trimerization, explaining how O-Nefs lacking this phosphosite counteract tetherin but most M-Nefs do not. These observations show how the higher-order organization of a vesicular coat can be allosterically modulated to direct cargoes to distinct fates.


Asunto(s)
Factor de Transcripción AP-1/ultraestructura , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/ultraestructura , Factor 1 de Ribosilacion-ADP/metabolismo , Factor 1 de Ribosilacion-ADP/ultraestructura , Proteínas Adaptadoras del Transporte Vesicular , Antígeno 2 del Estroma de la Médula Ósea/metabolismo , Antígeno 2 del Estroma de la Médula Ósea/ultraestructura , Clatrina , Aparato de Golgi , Células HEK293 , VIH-1 , Humanos , Transporte de Proteínas/fisiología , Factor de Transcripción AP-1/metabolismo , Factor de Transcripción AP-1/fisiología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/fisiología
20.
Cell ; 172(1-2): 275-288.e18, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29328916

RESUMEN

The neuronal gene Arc is essential for long-lasting information storage in the mammalian brain, mediates various forms of synaptic plasticity, and has been implicated in neurodevelopmental disorders. However, little is known about Arc's molecular function and evolutionary origins. Here, we show that Arc self-assembles into virus-like capsids that encapsulate RNA. Endogenous Arc protein is released from neurons in extracellular vesicles that mediate the transfer of Arc mRNA into new target cells, where it can undergo activity-dependent translation. Purified Arc capsids are endocytosed and are able to transfer Arc mRNA into the cytoplasm of neurons. These results show that Arc exhibits similar molecular properties to retroviral Gag proteins. Evolutionary analysis indicates that Arc is derived from a vertebrate lineage of Ty3/gypsy retrotransposons, which are also ancestors to retroviruses. These findings suggest that Gag retroelements have been repurposed during evolution to mediate intercellular communication in the nervous system.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Exosomas/metabolismo , Productos del Gen gag/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , ARN Mensajero/metabolismo , Animales , Células Cultivadas , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Endocitosis , Femenino , Productos del Gen gag/química , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Neuronas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA