Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Cell ; 182(5): 1170-1185.e9, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32795412

RESUMEN

Loss of the gene (Fmr1) encoding Fragile X mental retardation protein (FMRP) causes increased mRNA translation and aberrant synaptic development. We find neurons of the Fmr1-/y mouse have a mitochondrial inner membrane leak contributing to a "leak metabolism." In human Fragile X syndrome (FXS) fibroblasts and in Fmr1-/y mouse neurons, closure of the ATP synthase leak channel by mild depletion of its c-subunit or pharmacological inhibition normalizes stimulus-induced and constitutive mRNA translation rate, decreases lactate and key glycolytic and tricarboxylic acid (TCA) cycle enzyme levels, and triggers synapse maturation. FMRP regulates leak closure in wild-type (WT), but not FX synapses, by stimulus-dependent ATP synthase ß subunit translation; this increases the ratio of ATP synthase enzyme to its c-subunit, enhancing ATP production efficiency and synaptic growth. In contrast, in FXS, inability to close developmental c-subunit leak prevents stimulus-dependent synaptic maturation. Therefore, ATP synthase c-subunit leak closure encourages development and attenuates autistic behaviors.


Asunto(s)
Adenosina Trifosfato/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Subunidades de Proteína/metabolismo , Animales , Línea Celular , Ciclo del Ácido Cítrico/fisiología , Fibroblastos/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Células HEK293 , Humanos , Ratones , Neuronas/metabolismo , ARN Mensajero , Sinapsis/metabolismo
2.
Cell ; 163(6): 1444-56, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26638073

RESUMEN

The intestinal mucosal barrier controlling the resident microbiome is dependent on a protective mucus layer generated by goblet cells, impairment of which is a hallmark of the inflammatory bowel disease, ulcerative colitis. Here, we show that IL-18 is critical in driving the pathologic breakdown of barrier integrity in a model of colitis. Deletion of Il18 or its receptor Il18r1 in intestinal epithelial cells (Δ/EC) conferred protection from colitis and mucosal damage in mice. In contrast, deletion of the IL-18 negative regulator Il18bp resulted in severe colitis associated with loss of mature goblet cells. Colitis and goblet cell loss were rescued in Il18bp(-/-);Il18r(Δ/EC) mice, demonstrating that colitis severity is controlled at the level of IL-18 signaling in intestinal epithelial cells. IL-18 inhibited goblet cell maturation by regulating the transcriptional program instructing goblet cell development. These results inform on the mechanism of goblet cell dysfunction that underlies the pathology of ulcerative colitis.


Asunto(s)
Colitis Ulcerosa/patología , Colitis Ulcerosa/fisiopatología , Interleucina-18/inmunología , Animales , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/metabolismo , Sulfato de Dextran , Células Endoteliales/metabolismo , Células Epiteliales/citología , Femenino , Células Caliciformes/metabolismo , Células Caliciformes/patología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Subunidad alfa del Receptor de Interleucina-18/genética , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Mucosa Intestinal/fisiopatología , Masculino , Ratones , Transducción de Señal
3.
J Immunol ; 212(7): 1063-1068, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38353614

RESUMEN

Activation of naive CD8-positive T lymphocytes is mediated by dendritic cells that cross-present MHC class I (MHC-I)-associated peptides derived from exogenous Ags. The most accepted mechanism involves the translocation of Ags from phagosomes or endolysosomes into the cytosol, where antigenic peptides generated by cytosolic proteasomes are delivered by the transporter associated with Ag processing (TAP) to the endoplasmic reticulum, or an endocytic Ag-loading compartment, where binding to MHC-I occurs. We have described an alternative pathway where cross-presentation is independent of TAP but remains dependent on proteasomes. We provided evidence that active proteasomes found within the lumen of phagosomes and endolysosomal vesicles locally generate antigenic peptides that can be directly loaded onto trafficking MHC-I molecules. However, the mechanism of active proteasome delivery to the endocytic compartments remained unknown. In this study, we demonstrate that phagosome-associated LC3A/B structures deliver proteasomes into subcellular compartments containing exogenous Ags and that autophagy drives TAP-independent, proteasome-dependent cross-presentation.


Asunto(s)
Reactividad Cruzada , Complejo de la Endopetidasa Proteasomal , Complejo de la Endopetidasa Proteasomal/metabolismo , Presentación de Antígeno , Autofagosomas , Fagosomas/metabolismo , Antígenos de Histocompatibilidad Clase I , Antígenos , Proteínas de Transporte de Membrana/metabolismo , Péptidos/metabolismo
4.
EMBO J ; 38(16): e99266, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31271236

RESUMEN

During MHC-I-restricted antigen processing, peptides generated by cytosolic proteasomes are translocated by the transporter associated with antigen processing (TAP) into the endoplasmic reticulum, where they bind to newly synthesized MHC-I molecules. Dendritic cells and other cell types can also generate MHC-I complexes with peptides derived from internalized proteins, a process called cross-presentation. Here, we show that active proteasomes within cross-presenting cell phagosomes can generate these peptides. Active proteasomes are detectable within endocytic compartments in mouse bone marrow-derived dendritic cells. In TAP-deficient mouse dendritic cells, cross-presentation is enhanced by the introduction of human ß2 -microglobulin, which increases surface expression of MHC-I and suggests a role for recycling MHC-I molecules. In addition, surface MHC-I can be reduced by proteasome inhibition and stabilized by MHC-I-restricted peptides. This is consistent with constitutive proteasome-dependent but TAP-independent peptide loading in the endocytic pathway. Rab-GTPase mutants that restrain phagosome maturation increase proteasome recruitment and enhance TAP-independent cross-presentation. Thus, phagosomal/endosomal binding of peptides locally generated by proteasomes allows cross-presentation to generate MHC-I-peptide complexes identical to those produced by conventional antigen processing.


Asunto(s)
Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase I/química , Complejo de la Endopetidasa Proteasomal/inmunología , Microglobulina beta-2/metabolismo , Animales , Presentación de Antígeno , Células Cultivadas , Reactividad Cruzada , Células Dendríticas/citología , Endocitosis , Humanos , Ratones , Fagosomas/inmunología , Proteolisis , Microglobulina beta-2/genética
5.
PLoS Genet ; 15(11): e1008387, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31738769

RESUMEN

The ubiquitin-proteasome system regulates numerous cellular processes and is central to protein homeostasis. In proliferating yeast and many mammalian cells, proteasomes are highly enriched in the nucleus. In carbon-starved yeast, proteasomes migrate to the cytoplasm and collect in proteasome storage granules (PSGs). PSGs dissolve and proteasomes return to the nucleus within minutes of glucose refeeding. The mechanisms by which cells regulate proteasome homeostasis under these conditions remain largely unknown. Here we show that AMP-activated protein kinase (AMPK) together with endosomal sorting complexes required for transport (ESCRTs) drive a glucose starvation-dependent microautophagy pathway that preferentially sorts aberrant proteasomes into the vacuole, thereby biasing accumulation of functional proteasomes in PSGs. The proteasome core particle (CP) and regulatory particle (RP) are regulated differently. Without AMPK, the insoluble protein deposit (IPOD) serves as an alternative site that specifically sequesters CP aggregates. Our findings reveal a novel AMPK-controlled ESCRT-mediated microautophagy mechanism in the regulation of proteasome trafficking and homeostasis under carbon starvation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Microautofagia/genética , Complejo de la Endopetidasa Proteasomal/genética , Citoplasma/genética , Citoplasma/metabolismo , Glucosa/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Transporte de Proteínas/genética , Saccharomyces cerevisiae/genética , Inanición/genética , Inanición/metabolismo , Ubiquitina/genética , Ubiquitinación/genética , Vacuolas/genética , Vacuolas/metabolismo
6.
J Biol Chem ; 293(52): 19974-19981, 2018 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-30463941

RESUMEN

Human babesiosis is an emerging tick-borne disease caused by apicomplexan parasites of the genus Babesia Clinical cases caused by Babesia duncani have been associated with high parasite burden, severe pathology, and death. In both mice and hamsters, the parasite causes uncontrolled fulminant infections, which ultimately lead to death. Resolving these infections requires knowledge of B. duncani biology, virulence, and susceptibility to anti-infectives, but little is known and further research is hindered by a lack of relevant model systems. Here, we report the first continuous in vitro culture of B. duncani in human red blood cells. We show that during its asexual cycle within human erythrocytes, B. duncani develops and divides to form four daughter parasites with parasitemia doubling every ∼22 h. Using this in vitro culture assay, we found that B. duncani has low susceptibility to the four drugs recommended for treatment of human babesiosis, atovaquone, azithromycin, clindamycin, and quinine, with IC50 values ranging between 500 nm and 20 µm These data suggest that current practices are of limited effect in treating the disease. We anticipate this new disease model will set the stage for a better understanding of the biology of this parasite and will help guide better therapeutic strategies to treat B. duncani-associated babesiosis.


Asunto(s)
Antiparasitarios/farmacología , Babesia/efectos de los fármacos , Babesiosis/tratamiento farmacológico , Babesiosis/parasitología , Eritrocitos/parasitología , Pruebas de Sensibilidad Parasitaria/métodos , Atovacuona/farmacología , Azitromicina/farmacología , Babesia/crecimiento & desarrollo , Técnicas de Cultivo de Célula/métodos , Clindamicina/farmacología , Humanos , Quinina/farmacología
7.
Proc Natl Acad Sci U S A ; 111(5): 1849-54, 2014 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-24449908

RESUMEN

Two classes of proteins that bind to each other and to Golgi membranes have been implicated in the adhesion of Golgi cisternae to each other to form their characteristic stacks: Golgi reassembly and stacking proteins 55 and 65 (GRASP55 and GRASP65) and Golgin of 45 kDa and Golgi matrix protein of 130 kDa. We report here that efficient stacking occurs in the absence of GRASP65/55 when either Golgin is overexpressed, as judged by quantitative electron microscopy. The Golgi stacks in these GRASP-deficient HeLa cells were normal both in morphology and in anterograde cargo transport. This suggests the simple hypothesis that the total amount of adhesive energy gluing cisternae dictates Golgi cisternal stacking, irrespective of which molecules mediate the adhesive process. In support of this hypothesis, we show that adding artificial adhesive energy between cisternae and mitochondria by dimerizing rapamycin-binding domain and FK506-binding protein domains that are attached to cisternal adhesive proteins allows mitochondria to invade the stack and even replace Golgi cisternae within a few hours. These results indicate that although Golgi stacking is a highly complicated process involving a large number of adhesive and regulatory proteins, the overriding principle of a Golgi stack assembly is likely to be quite simple. From this simplified perspective, we propose a model, based on cisternal adhesion and cisternal maturation as the two core principles, illustrating how the most ancient form of Golgi stacking might have occurred using only weak cisternal adhesive processes because of the differential between the rate of influx and outflux of membrane transport through the Golgi.


Asunto(s)
Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Membranas Intracelulares/metabolismo , Adhesividad , Autoantígenos/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas de la Matriz de Golgi , Células HeLa , Humanos , Membranas Intracelulares/ultraestructura , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Modelos Biológicos , Transfección
8.
Proc Natl Acad Sci U S A ; 111(29): 10580-5, 2014 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-24979777

RESUMEN

Mitochondria maintain tight regulation of inner mitochondrial membrane (IMM) permeability to sustain ATP production. Stressful events cause cellular calcium (Ca(2+)) dysregulation followed by rapid loss of IMM potential known as permeability transition (PT), which produces osmotic shifts, metabolic dysfunction, and cell death. The molecular identity of the mitochondrial PT pore (mPTP) was previously unknown. We show that the purified reconstituted c-subunit ring of the FO of the F1FO ATP synthase forms a voltage-sensitive channel, the persistent opening of which leads to rapid and uncontrolled depolarization of the IMM in cells. Prolonged high matrix Ca(2+) enlarges the c-subunit ring and unhooks it from cyclophilin D/cyclosporine A binding sites in the ATP synthase F1, providing a mechanism for mPTP opening. In contrast, recombinant F1 beta-subunit applied exogenously to the purified c-subunit enhances the probability of pore closure. Depletion of the c-subunit attenuates Ca(2+)-induced IMM depolarization and inhibits Ca(2+) and reactive oxygen species-induced cell death whereas increasing the expression or single-channel conductance of the c-subunit sensitizes to death. We conclude that a highly regulated c-subunit leak channel is a candidate for the mPTP. Beyond cell death, these findings also imply that increasing the probability of c-subunit channel closure in a healthy cell will enhance IMM coupling and increase cellular metabolic efficiency.


Asunto(s)
Canales Iónicos/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Subunidades de Proteína/metabolismo , ATPasas de Translocación de Protón/metabolismo , Animales , Calcio/farmacología , Muerte Celular/efectos de los fármacos , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Liposomas/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/química , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Mutación/genética , Conformación Proteica , ATPasas de Translocación de Protón/química , Ratas , Especies Reactivas de Oxígeno/metabolismo
9.
JHEP Rep ; 6(7): 101069, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38966234

RESUMEN

Background & Aims: The lymphatic system plays crucial roles in maintaining fluid balance and immune regulation. Studying the liver lymphatics has been considered challenging, as common lymphatic endothelial cell (LyEC) markers are expressed by other liver cells. Additionally, isolation of sufficient numbers of LyECs has been challenging because of their extremely low abundance (<0.01% of entire liver cell population) in a normal liver. Methods: Potential LyEC markers was identified using our published single-cell RNA sequencing (scRNA-seq) dataset (GSE147581) in mouse livers. Interleukin-7 (IL7) promoter-driven green fluorescent protein knock-in heterozygous mice were used for the validation of IL7 expression in LyECs in the liver, for the development of liver LyEC isolation protocol, and generating liver ischemia/reperfusion (I/R) injury. Scanning electron microscopy was used for the structural analysis of LyECs. Changes in LyEC phenotypes in livers of mice with I/R were determined by RNA-seq analysis. Results: Through scRNA-seq analysis, we have identified IL7 as an exclusive marker for liver LyECs, with no overlap with other liver cell types. Based on IL7 expression in liver LyECs, we have established an LyEC isolation method and observed distinct cell surface structures of LyECs with fenestrae and cellular pores (ranging from 100 to 400 nm in diameter). Furthermore, we identified LyEC genes that undergo alterations during I/R liver injuries. Conclusions: This study not only identified IL7 as an exclusively expressed gene in liver LyECs, but also enhanced our understanding of LyEC structures and demonstrated transcriptomic changes in injured livers. Impact and implications: Understanding the lymphatic system in the liver is challenging because of the absence of specific markers for liver LyEC. This study has identified IL7 as a reliable marker for LyECs, enabling the development of an effective method for their isolation, elucidating their unique cell surface structure, and identifying LyEC genes that undergo changes during liver damage. The development of IL7 antibodies for detecting it in human liver specimens will further advance our understanding of the liver lymphatic system in the future.

10.
Proc Natl Acad Sci U S A ; 107(50): 21511-6, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21115825

RESUMEN

Epsin is a ubiquitin-binding endocytic adaptor, which is highly concentrated at clathrin-coated pits and coordinates acquisition of bilayer curvature with coat recruitment and cargo selection. Epsin is encoded by three distinct genes in mammals. Epsin 1 and 2 have broad tissue distribution with high-level expression in the brain. In contrast, epsin 3 was reported to be expressed primarily in immature keratinocytes. Here, we show that epsin 3 is selectively expressed at high levels in the stomach (including the majority of gastric cancers), where it is concentrated in parietal cells. In these cells, epsin 3 is enriched and colocalized with clathrin around apical canaliculi, the sites that control acidification of the stomach lumen via the exo-endocytosis of vesicles containing the H/K ATPase. Deletion of the epsin 3 gene in mice did not result in obvious pathological phenotypes in either the stomach or other organs, possibly because of overlapping functions of the other two epsins. However, levels of EHD1 and EHD2, two membrane tubulating proteins with a role in endocytic recycling, were elevated in epsin 3 knock-out stomachs, pointing to a functional interplay of epsin 3 with EHD proteins in the endocytic pathway of parietal cells. We suggest that epsin 3 cooperates with other bilayer binding proteins with curvature sensing/generating properties in the specialized traffic and membrane remodeling processes typical of gastric parietal cells.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Polaridad Celular , Endocitosis/fisiología , Mucosa Gástrica/metabolismo , Células Parietales Gástricas/fisiología , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Humanos , Ratones , Ratones Noqueados , Células Parietales Gástricas/ultraestructura , Estómago/anatomía & histología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Distribución Tisular , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
11.
mSphere ; 8(6): e0046023, 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-37847028

RESUMEN

IMPORTANCE: Neurospora is a quintessential tip-growing organism, which is well known for packaging and longitudinal transport of tip-building blocks. Thus far, however, little attention has been paid to the co-essential process of reclamation, that is-taking apart of upstream, older structural elements, otherwise known as "autophagy". We are not yet prepared to set out the chemistry of that elaborate process, but its morphological start alone is worthy of attention. Carbon starvation triggers significant autophagic changes, beginning with prolific vacuolation along the plasma membrane, and eventual filling of 70% (or more) of cytoplasmic volume. Additionally, the Neurospora plasma membrane elaborates a variety of phagophores which themselves often look lytic. These have either dual enclosing membranes, like the familiar autophagosomes, can be doubled and have four wrapping membranes, or can be compounded with multiple membrane layers. These reclamation processes must be accommodated by the mechanism of tip growth.


Asunto(s)
Neurospora crassa , Neurospora crassa/metabolismo , Autofagia , Membrana Celular/metabolismo
12.
Cell Microbiol ; 13(1): 47-61, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20716207

RESUMEN

Anaplasma phagocytophilum causes human granulocytic anaplasmosis, one of the most common tick-borne diseases in North America. This unusual obligate intracellular pathogen selectively persists within polymorphonuclear leucocytes. In this study, using the yeast surrogate model we identified an A. phagocytophilum virulence protein, AptA (A. phagocytophilum toxin A), that activates mammalian Erk1/2 mitogen-activated protein kinase. This activation is important for A. phagocytophilum survival within human neutrophils. AptA interacts with the intermediate filament protein vimentin, which is essential for A. phagocytophilum-induced Erk1/2 activation and infection. A. phagocytophilum infection reorganizes vimentin around the bacterial inclusion, thereby contributing to intracellular survival. These observations reveal a major role for the bacterial protein, AptA, and the host protein, vimentin, in the activation of Erk1/2 during A. phagocytophilum infection.


Asunto(s)
Anaplasma phagocytophilum/patogenicidad , Proteínas Bacterianas/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Transducción de Señal , Factores de Virulencia/metabolismo , Línea Celular , Humanos , Neutrófilos/microbiología , Vimentina/metabolismo
13.
Sci Rep ; 12(1): 14975, 2022 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-36056100

RESUMEN

Retro-2 directly interacts with an ER exit site protein, Sec16A, inhibiting ER exit of a Golgi tSNARE, Syntaxin5, which results in rapid re-distribution of Syntaxin5 to the ER. Recently, it was shown that SARS-CoV-2 infection disrupts the Golgi apparatus within 6-12 h, while its replication was effectively inhibited by Retro-2 in cultured human lung cells. Yet, exactly how Retro-2 may influence ultrastructure of the Golgi apparatus have not been thoroughly investigated. In this study, we characterized the effect of Retro-2 treatment on ultrastructure of the Golgi apparatus using electron microscopy and EM tomography. Our initial results on protein secretion showed that Retro-2 treatment does not significantly influence secretion of either small or large cargos. Ultra-structural study of the Golgi, however, revealed rapid accumulation of COPI-like vesicular profiles in the perinuclear area and a partial disassembly of the Golgi stack under electron microscope within 3-5 h, suggesting altered Golgi organization in these cells. Retro-2 treatment in cells depleted of GRASP65/55, the two well-known Golgi structural proteins, induced complete and rapid disassembly of the Golgi into individual cisterna. Taken together, these results suggest that Retro-2 profoundly alters Golgi structure to a much greater extent than previously anticipated.


Asunto(s)
COVID-19 , Aparato de Golgi , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Humanos , SARS-CoV-2 , Proteínas de Transporte Vesicular/metabolismo
14.
Cell Death Differ ; 29(9): 1874-1887, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35322203

RESUMEN

Mitochondrial ATP synthase is vital not only for cellular energy production but also for energy dissipation and cell death. ATP synthase c-ring was suggested to house the leak channel of mitochondrial permeability transition (mPT), which activates during excitotoxic ischemic insult. In this present study, we purified human c-ring from both eukaryotic and prokaryotic hosts to biophysically characterize its channel activity. We show that purified c-ring forms a large multi-conductance, voltage-gated ion channel that is inhibited by the addition of ATP synthase F1 subcomplex. In contrast, dissociation of F1 from FO occurs during excitotoxic neuronal death suggesting that the F1 constitutes the gate of the channel. mPT is known to dissipate the osmotic gradient across the inner membrane during cell death. We show that ATP synthase c-subunit knock down (KD) prevents the osmotic change in response to high calcium and eliminates large conductance, Ca2+ and CsA sensitive channel activity of mPT. These findings elucidate the gating mechanism of the ATP synthase c-subunit leak channel (ACLC) and suggest how ACLC opening is regulated by cell stress in a CypD-dependent manner.


Asunto(s)
Proteínas de Transporte de Membrana Mitocondrial , ATPasas de Translocación de Protón Mitocondriales , Adenosina Trifosfato/metabolismo , Muerte Celular , Humanos , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , ATPasas de Translocación de Protón Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/metabolismo , ATPasas de Translocación de Protón/metabolismo
15.
Nat Commun ; 13(1): 7637, 2022 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-36496409

RESUMEN

Although mitochondrial activity is critical for angiogenesis, its mechanism is not entirely clear. Here we show that mice with endothelial deficiency of any one of the three nuclear genes encoding for mitochondrial proteins, transcriptional factor (TFAM), respiratory complex IV component (COX10), or redox protein thioredoxin 2 (TRX2), exhibit retarded retinal vessel growth and arteriovenous malformations (AVM). Single-cell RNA-seq analyses indicate that retinal ECs from the three mutant mice have increased TGFß signaling and altered gene expressions associated with vascular maturation and extracellular matrix, correlating with vascular malformation and increased basement membrane thickening in microvesels of mutant retinas. Mechanistic studies suggest that mitochondrial dysfunction from Tfam, Cox10, or Trx2 depletion induces a mitochondrial localization and MAPKs-mediated phosphorylation of SMAD2, leading to enhanced ALK5-SMAD2 signaling. Importantly, pharmacological blockade of ALK5 signaling or genetic deficiency of SMAD2 prevented retinal vessel growth retardation and AVM in all three mutant mice. Our studies uncover a novel mechanism whereby mitochondrial dysfunction via the ALK5-SMAD2 signaling induces retinal vascular malformations, and have therapeutic values for the alleviation of angiogenesis-associated human retinal diseases.


Asunto(s)
Malformaciones Arteriovenosas , Receptor Tipo I de Factor de Crecimiento Transformador beta , Proteína Smad2 , Animales , Ratones , Malformaciones Arteriovenosas/genética , Malformaciones Arteriovenosas/metabolismo , Regulación de la Expresión Génica , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Fosforilación , Transducción de Señal , Proteína Smad2/genética , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo
16.
Microbiome ; 10(1): 173, 2022 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-36253842

RESUMEN

BACKGROUND: Ixodes scapularis is the predominant tick vector of Borrelia burgdorferi, the agent of Lyme disease, in the USA. Molecular interactions between the tick and B. burgdorferi orchestrate the migration of spirochetes from the midgut to the salivary glands-critical steps that precede transmission to the vertebrate host. Over the last decade, research efforts have invoked a potential role for the tick microbiome in modulating tick-pathogen interactions. RESULTS: Using multiple strategies to perturb the microbiome composition of B. burgdorferi-infected nymphal ticks, we observe that changes in the microbiome composition do not significantly influence B. burgdorferi migration from the midgut, invasion of salivary glands, or transmission to the murine host. We also show that within 24 and 48 h of the onset of tick feeding, B. burgdorferi spirochetes are within the peritrophic matrix and epithelial cells of the midgut in preparation for exit from the midgut. CONCLUSIONS: This study highlights two aspects of tick-spirochete interactions: (1) environmental bacteria associated with the tick do not influence spirochete transmission to the mammalian host and (2) the spirochete may utilize an intracellular exit route during migration from the midgut to the salivary glands, a strategy that may allow the spirochete to distance itself from microbiota in the midgut lumen effectively. This may explain in part, the inability of environment-acquired midgut microbiota to significantly influence spirochete transmission. Unraveling a molecular understanding of this exit strategy will be critical to gain new insights into the biology of the spirochete and the tick. Video Abstract.


Asunto(s)
Borrelia burgdorferi , Ixodes , Enfermedad de Lyme , Microbiota , Animales , Borrelia burgdorferi/genética , Ixodes/microbiología , Enfermedad de Lyme/microbiología , Mamíferos , Ratones , Ninfa/microbiología
17.
Sci Rep ; 11(1): 7730, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33833328

RESUMEN

The pigment cell-specific protein PMEL forms a functional amyloid matrix in melanosomes onto which the pigment melanin is deposited. The amyloid core consists of a short proteolytic fragment, which we have termed the core-amyloid fragment (CAF) and perhaps additional parts of the protein, such as the PKD domain. A highly O-glycosylated repeat (RPT) domain also derived from PMEL proteolysis associates with the amyloid and is necessary to establish the sheet-like morphology of the assemblies. Excluded from the aggregate is the regulatory N-terminus, which nevertheless must be linked in cis to the CAF in order to drive amyloid formation. The domain is then likely cleaved away immediately before, during, or immediately after the incorporation of a new CAF subunit into the nascent amyloid. We had previously identified a 21 amino acid long region, which mediates the regulatory activity of the N-terminus towards the CAF. However, many mutations in the respective segment caused misfolding and/or blocked PMEL export from the endoplasmic reticulum, leaving their phenotype hard to interpret. Here, we employ a saturating mutagenesis approach targeting the motif at single amino acid resolution. Our results confirm the critical nature of the PMEL N-terminal region and identify several residues essential for PMEL amyloidogenesis.


Asunto(s)
Aminoácidos/química , Dominios Proteicos , Antígeno gp100 del Melanoma/química , Secuencia de Aminoácidos , Retículo Endoplásmico/metabolismo , Humanos , Melanosomas/metabolismo , Mutación , Pliegue de Proteína , Transporte de Proteínas , Fracciones Subcelulares/metabolismo , Antígeno gp100 del Melanoma/metabolismo
18.
Nat Commun ; 12(1): 504, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33495460

RESUMEN

Cerebral cavernous malformations (CCMs) are vascular abnormalities that primarily occur in adulthood and cause cerebral hemorrhage, stroke, and seizures. CCMs are thought to be initiated by endothelial cell (EC) loss of any one of the three Ccm genes: CCM1 (KRIT1), CCM2 (OSM), or CCM3 (PDCD10). Here we report that mice with a brain EC-specific deletion of Pdcd10 (Pdcd10BECKO) survive up to 6-12 months and develop bona fide CCM lesions in all regions of brain, allowing us to visualize the vascular dynamics of CCM lesions using transcranial two-photon microscopy. This approach reveals that CCMs initiate from protrusion at the level of capillary and post-capillary venules with gradual dissociation of pericytes. Microvascular beds in lesions are hyper-permeable, and these disorganized structures present endomucin-positive ECs and α-smooth muscle actin-positive pericytes. Caveolae in the endothelium of Pdcd10BECKO lesions are drastically increased, enhancing Tie2 signaling in Ccm3-deficient ECs. Moreover, genetic deletion of caveolin-1 or pharmacological blockade of Tie2 signaling effectively normalizes microvascular structure and barrier function with attenuated EC-pericyte disassociation and CCM lesion formation in Pdcd10BECKO mice. Our study establishes a chronic CCM model and uncovers a mechanism by which CCM3 mutation-induced caveolae-Tie2 signaling contributes to CCM pathogenesis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/deficiencia , Encéfalo/metabolismo , Caveolas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Hemangioma Cavernoso del Sistema Nervioso Central/metabolismo , Receptor TIE-2/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Encéfalo/patología , Encéfalo/ultraestructura , Caveolas/ultraestructura , Células Cultivadas , Hemangioma Cavernoso del Sistema Nervioso Central/genética , Humanos , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Pericitos/metabolismo , Receptor TIE-2/genética , Transducción de Señal , Análisis de Supervivencia
19.
Theranostics ; 11(12): 5876-5888, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33897887

RESUMEN

Inflammation plays a major role in the pathogenesis of several vascular pathologies, including abdominal aortic aneurysm (AAA). Evaluating the role of inflammation in AAA pathobiology and potentially outcome in vivo requires non-invasive tools for high-resolution imaging. We investigated the feasibility of X-ray computed tomography (CT) imaging of phagocytic activity using nanoparticle contrast agents to predict AAA outcome. Methods: Uptake of several nanoparticle CT contrast agents was evaluated in a macrophage cell line. The most promising agent, Exitron nano 12000, was further characterized in vitro and used for subsequent in vivo testing. AAA was induced in Apoe-/- mice through angiotensin II (Ang II) infusion for up to 4 weeks. Nanoparticle biodistribution and uptake in AAA were evaluated by CT imaging in Ang II-infused Apoe-/- mice. After imaging, the aortic tissue was harvested and used from morphometry, transmission electron microscopy and gene expression analysis. A group of Ang II-infused Apoe-/- mice underwent nanoparticle-enhanced CT imaging within the first week of Ang II infusion, and their survival and aortic external diameter were evaluated at 4 weeks to address the value of vessel wall CT enhancement in predicting AAA outcome. Results: Exitron nano 12000 showed specific uptake in macrophages in vitro. Nanoparticle accumulation was observed by CT imaging in tissues rich in mononuclear phagocytes. Aortic wall enhancement was detectable on delayed CT images following nanoparticle administration and correlated with vessel wall CD68 expression. Transmission electron microscopy ascertained the presence of nanoparticles in AAA adventitial macrophages. Nanoparticle-induced CT enhancement on images obtained within one week of AAA induction was predictive of AAA outcome at 4 weeks. Conclusions: By establishing the feasibility of CT-based molecular imaging of phagocytic activity in AAA, this study links the inflammatory signal on early time point images to AAA evolution. This readily available technology overcomes an important barrier to cross-sectional, longitudinal and outcome studies, not only in AAA, but also in other cardiovascular pathologies and facilitates the evaluation of modulatory interventions, and ultimately upon clinical translation, patient management.


Asunto(s)
Aneurisma de la Aorta Abdominal/patología , Macrófagos/patología , Fagocitos/patología , Angiotensina II/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Aneurisma de la Aorta Abdominal/metabolismo , Apolipoproteínas E/metabolismo , Modelos Animales de Enfermedad , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fagocitos/metabolismo , Tomografía Computarizada por Rayos X/métodos
20.
FEBS Lett ; 594(19): 3086-3094, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32668013

RESUMEN

The Golgi is surrounded by a ribosome-excluding matrix. Recently, we reported that the cis-Golgi-localized golgin GM130 can phase-separate to form dynamic, liquid-like condensates in vitro and in vivo. Here, we show that the overexpression of each of the remaining cis (golgin160, GMAP210)- and trans (golgin97, golgin245, GCC88, GCC185)-golgins results in novel protein condensates. Focused ion beam scanning electron microscopy (FIB-SEM) images of GM130 condensates reveal a complex internal organization with branching aqueous channels. Pairs of golgins overexpressed in the same cell form distinct juxtaposed condensates. These findings support the hypothesis that, in addition to their established roles as vesicle tethers, phase separation may be a common feature of the golgin family that contributes to Golgi organization.


Asunto(s)
Autoantígenos/metabolismo , Proteínas de la Matriz de Golgi/metabolismo , Autoantígenos/química , Autoantígenos/ultraestructura , Supervivencia Celular , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Proteínas de la Matriz de Golgi/química , Proteínas de la Matriz de Golgi/ultraestructura , Células HeLa , Humanos , Imagen de Lapso de Tiempo , Red trans-Golgi/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA