Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 196
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(17): 4605-4620.e17, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-38959891

RESUMEN

The ability of mitochondria to coordinate stress responses across tissues is critical for health. In C. elegans, neurons experiencing mitochondrial stress elicit an inter-tissue signaling pathway through the release of mitokine signals, such as serotonin or the Wnt ligand EGL-20, which activate the mitochondrial unfolded protein response (UPRMT) in the periphery to promote organismal health and lifespan. We find that germline mitochondria play a surprising role in neuron-to-periphery UPRMT signaling. Specifically, we find that germline mitochondria signal downstream of neuronal mitokines, Wnt and serotonin, and upstream of lipid metabolic pathways in the periphery to regulate UPRMT activation. We also find that the germline tissue itself is essential for UPRMT signaling. We propose that the germline has a central signaling role in coordinating mitochondrial stress responses across tissues, and germline mitochondria play a defining role in this coordination because of their inherent roles in germline integrity and inter-tissue signaling.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Células Germinativas , Mitocondrias , Transducción de Señal , Respuesta de Proteína Desplegada , Animales , Caenorhabditis elegans/metabolismo , Mitocondrias/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Células Germinativas/metabolismo , Neuronas/metabolismo , Serotonina/metabolismo , Proteínas Wnt/metabolismo
2.
Cell ; 187(16): 4289-4304.e26, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-38942015

RESUMEN

Cellular homeostasis is intricately influenced by stimuli from the microenvironment, including signaling molecules, metabolites, and pathogens. Functioning as a signaling hub within the cell, mitochondria integrate information from various intracellular compartments to regulate cellular signaling and metabolism. Multiple studies have shown that mitochondria may respond to various extracellular signaling events. However, it is less clear how changes in the extracellular matrix (ECM) can impact mitochondrial homeostasis to regulate animal physiology. We find that ECM remodeling alters mitochondrial homeostasis in an evolutionarily conserved manner. Mechanistically, ECM remodeling triggers a TGF-ß response to induce mitochondrial fission and the unfolded protein response of the mitochondria (UPRMT). At the organismal level, ECM remodeling promotes defense of animals against pathogens through enhanced mitochondrial stress responses. We postulate that this ECM-mitochondria crosstalk represents an ancient immune pathway, which detects infection- or mechanical-stress-induced ECM damage, thereby initiating adaptive mitochondria-based immune and metabolic responses.


Asunto(s)
Matriz Extracelular , Homeostasis , Mitocondrias , Respuesta de Proteína Desplegada , Matriz Extracelular/metabolismo , Animales , Mitocondrias/metabolismo , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Dinámicas Mitocondriales , Ratones , Transducción de Señal , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/inmunología
3.
Physiol Rev ; 104(2): 533-587, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-37561137

RESUMEN

Lung endothelia in the arteries, capillaries, and veins are heterogeneous in structure and function. Lung capillaries in particular represent a unique vascular niche, with a thin yet highly restrictive alveolar-capillary barrier that optimizes gas exchange. Capillary endothelium surveys the blood while simultaneously interpreting cues initiated within the alveolus and communicated via immediately adjacent type I and type II epithelial cells, fibroblasts, and pericytes. This cell-cell communication is necessary to coordinate the immune response to lower respiratory tract infection. Recent discoveries identify an important role for the microtubule-associated protein tau that is expressed in lung capillary endothelia in the host-pathogen interaction. This endothelial tau stabilizes microtubules necessary for barrier integrity, yet infection drives production of cytotoxic tau variants that are released into the airways and circulation, where they contribute to end-organ dysfunction. Similarly, beta-amyloid is produced during infection. Beta-amyloid has antimicrobial activity, but during infection it can acquire cytotoxic activity that is deleterious to the host. The production and function of these cytotoxic tau and amyloid variants are the subject of this review. Lung-derived cytotoxic tau and amyloid variants are a recently discovered mechanism of end-organ dysfunction, including neurocognitive dysfunction, during and in the aftermath of infection.


Asunto(s)
Pulmón , Insuficiencia Multiorgánica , Humanos , Insuficiencia Multiorgánica/metabolismo , Pulmón/metabolismo , Endotelio Vascular/metabolismo , Amiloide/química , Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo
4.
Cell ; 159(2): 388-401, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25303532

RESUMEN

The maintenance of nuclear compartmentalization by the nuclear envelope and nuclear pore complexes (NPCs) is essential for cell function; loss of compartmentalization is associated with cancers, laminopathies, and aging. We uncovered a pathway that surveils NPC assembly intermediates to promote the formation of functional NPCs. Surveillance is mediated by Heh2, a member of the LEM (Lap2-emerin-MAN1) family of integral inner nuclear membrane proteins, which binds to an early NPC assembly intermediate, but not to mature NPCs. Heh2 recruits the endosomal sorting complex required for transport (ESCRT)-III subunit Snf7 and the AAA-ATPase Vps4 to destabilize and clear defective NPC assembly intermediates. When surveillance or clearance is compromised, malformed NPCs accumulate in a storage of improperly assembled nuclear pore complexes compartment, or SINC. The SINC is retained in old mothers to prevent loss of daughter lifespan, highlighting a continuum of mechanisms to ensure nuclear compartmentalization.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Poro Nuclear/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Transporte Activo de Núcleo Celular , Schizosaccharomyces/citología , Schizosaccharomyces/metabolismo
5.
Development ; 149(4)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35142351

RESUMEN

The zebrafish has become a widely used animal model due, in large part, to its accessibility to and usefulness for high-resolution optical imaging. Although zebrafish research has historically focused mostly on early development, in recent years the fish has increasingly been used to study regeneration, cancer metastasis, behavior and other processes taking place in juvenile and adult animals. However, imaging of live adult zebrafish is extremely challenging, with survival of adult fish limited to a few tens of minutes using standard imaging methods developed for zebrafish embryos and larvae. Here, we describe a new method for imaging intubated adult zebrafish using a specially designed 3D printed chamber for long-term imaging of adult zebrafish on inverted microscope systems. We demonstrate the utility of this new system by nearly day-long observation of neutrophil recruitment to a wound area in living double-transgenic adult casper zebrafish with fluorescently labeled neutrophils and lymphatic vessels, as well as intubating and imaging the same fish repeatedly. We also show that Mexican cavefish can be intubated and imaged in the same way, demonstrating this method can be used for long-term imaging of adult animals from diverse aquatic species.


Asunto(s)
Microscopía Fluorescente/métodos , Pez Cebra/anatomía & histología , Animales , Animales Modificados Genéticamente/inmunología , Animales Modificados Genéticamente/metabolismo , Embrión no Mamífero/anatomía & histología , Embrión no Mamífero/metabolismo , Larva/anatomía & histología , Larva/crecimiento & desarrollo , Larva/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Neutrófilos/citología , Neutrófilos/inmunología , Neutrófilos/patología , Impresión Tridimensional , Imagen de Lapso de Tiempo , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
6.
Development ; 149(5)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35132436

RESUMEN

The pectoral fins of teleost fish are analogous structures to human forelimbs, and the developmental mechanisms directing their initial growth and patterning are conserved between fish and tetrapods. The forelimb vasculature is crucial for limb function, and it appears to play important roles during development by promoting development of other limb structures, but the steps leading to its formation are poorly understood. In this study, we use high-resolution imaging to document the stepwise assembly of the zebrafish pectoral fin vasculature. We show that fin vascular network formation is a stereotyped, choreographed process that begins with the growth of an initial vascular loop around the pectoral fin. This loop connects to the dorsal aorta to initiate pectoral vascular circulation. Pectoral fin vascular development continues with concurrent formation of three elaborate vascular plexuses, one in the distal fin that develops into the fin-ray vasculature and two near the base of the fin in association with the developing fin musculature. Our findings detail a complex, yet highly choreographed, series of steps involved in the development of a complete, functional, organ-specific vascular network.


Asunto(s)
Aletas de Animales/anatomía & histología , Aletas de Animales/crecimiento & desarrollo , Pez Cebra/anatomía & histología , Pez Cebra/crecimiento & desarrollo , Animales
7.
Dev Biol ; 501: 92-103, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37353106

RESUMEN

During embryonic development, primitive and definitive waves of hematopoiesis take place to provide proper blood cells for each developmental stage, with the possible involvement of epigenetic factors. We previously found that lysine-specific demethylase 1 (LSD1/KDM1A) promotes primitive hematopoietic differentiation by shutting down the gene expression program of hemangioblasts in an Etv2/Etsrp-dependent manner. In the present study, we demonstrated that zebrafish LSD1 also plays important roles in definitive hematopoiesis in the development of hematopoietic stem and progenitor cells. A combination of genetic approaches and imaging analyses allowed us to show that LSD1 promotes the egress of hematopoietic stem and progenitor cells into the bloodstream during the endothelial-to-hematopoietic transition. Analysis of compound mutant lines with Etv2/Etsrp mutant zebrafish revealed that, unlike in primitive hematopoiesis, this function of LSD1 was independent of Etv2/Etsrp. The phenotype of LSD1 mutant zebrafish during the endothelial-to-hematopoietic transition was similar to that of previously reported compound knockout mice of Gfi1/Gfi1b, which forms a complex with LSD1 and represses endothelial genes. Moreover, co-knockdown of zebrafish Gfi1/Gfi1b genes inhibited the development of hematopoietic stem and progenitor cells. We therefore hypothesize that the shutdown of the Gfi1/Gfi1b-target genes during the endothelial-to-hematopoietic transition is one of the key evolutionarily conserved functions of LSD1 in definitive hematopoiesis.


Asunto(s)
Células Madre , Pez Cebra , Animales , Ratones , Diferenciación Celular , Hematopoyesis/genética , Histona Demetilasas/genética , Células Madre/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Annu Rev Cell Dev Biol ; 26: 639-65, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19575651

RESUMEN

The vascular and the nervous systems of vertebrates share many features with similar and often overlapping anatomy. The parallels between these two systems extend to the molecular level, where recent work has identified ever-increasing similarities between the molecular mechanisms employed in the specification, differentiation, and patterning of both systems. This review discusses some of the most recent literature on this subject, with particular emphasis on the roles that the Ephrin, Semaphorin, Netrin, and Slit signaling pathways play in vascular development.


Asunto(s)
Vasos Sanguíneos/embriología , Sistema Nervioso/embriología , Animales , Vasos Sanguíneos/metabolismo , Tipificación del Cuerpo , Sistema Cardiovascular/embriología , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Sistema Nervioso/metabolismo , Vertebrados/embriología
9.
J Med Syst ; 48(1): 69, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39042285

RESUMEN

BACKGROUND:  Despite low mortality for elective procedures in the United States and developed countries, some patients have unexpected care escalations (UCE) following post-anesthesia care unit (PACU) discharge. Studies indicate patient risk factors for UCE, but determining which factors are most important is unclear. Machine learning (ML) can predict clinical events. We hypothesized that ML could predict patient UCE after PACU discharge in surgical patients and identify specific risk factors. METHODS: We conducted a single center, retrospective analysis of all patients undergoing non-cardiac surgery (elective and emergent). We collected data from pre-operative visits, intra-operative records, PACU admissions, and the rate of UCE. We trained a ML model with this data and tested the model on an independent data set to determine its efficacy. Finally, we evaluated the individual patient and clinical factors most likely to predict UCE risk. RESULTS: Our study revealed that ML could predict UCE risk which was approximately 5% in both the training and testing groups. We were able to identify patient risk factors such as patient vital signs, emergent procedure, ASA Status, and non-surgical anesthesia time as significant variable. We plotted Shapley values for significant variables for each patient to help determine which of these variables had the greatest effect on UCE risk. Of note, the UCE risk factors identified frequently by ML were in alignment with anesthesiologist clinical practice and the current literature. CONCLUSIONS: We used ML to analyze data from a single-center, retrospective cohort of non-cardiac surgical patients, some of whom had an UCE. ML assigned risk prediction for patients to have UCE and determined perioperative factors associated with increased risk. We advocate to use ML to augment anesthesiologist clinical decision-making, help decide proper disposition from the PACU, and ensure the safest possible care of our patients.


Asunto(s)
Aprendizaje Automático , Humanos , Estudios Retrospectivos , Femenino , Masculino , Persona de Mediana Edad , Factores de Riesgo , Anciano , Periodo Perioperatorio , Adulto , Signos Vitales , Complicaciones Posoperatorias/epidemiología , Medición de Riesgo/métodos , Periodo de Recuperación de la Anestesia
10.
Am J Respir Cell Mol Biol ; 69(3): 340-354, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37201952

RESUMEN

Pulmonary microvascular endothelial cells contribute to the integrity of the lung gas exchange interface, and they are highly glycolytic. Although glucose and fructose represent discrete substrates available for glycolysis, pulmonary microvascular endothelial cells prefer glucose over fructose, and the mechanisms involved in this selection are unknown. 6-Phosphofructo-2-kinase/fructose-2, 6-bisphosphatase 3 (PFKFB3) is an important glycolytic enzyme that drives glycolytic flux against negative feedback and links glycolytic and fructolytic pathways. We hypothesized that PFKFB3 inhibits fructose metabolism in pulmonary microvascular endothelial cells. We found that PFKFB3 knockout cells survive better than wild-type cells in fructose-rich medium under hypoxia. Seahorse assays, lactate and glucose measurements, and stable isotope tracing showed that PFKFB3 inhibits fructose-hexokinase-mediated glycolysis and oxidative phosphorylation. Microarray analysis revealed that fructose upregulates PFKFB3, and PFKFB3 knockout cells increase fructose-specific GLUT5 (glucose transporter 5) expression. Using conditional endothelial-specific PFKFB3 knockout mice, we demonstrated that endothelial PFKFB3 knockout increases lung tissue lactate production after fructose gavage. Last, we showed that pneumonia increases fructose in BAL fluid in mechanically ventilated ICU patients. Thus, PFKFB3 knockout increases GLUT5 expression and the hexokinase-mediated fructose use in pulmonary microvascular endothelial cells that promotes their survival. Our findings indicate that PFKFB3 is a molecular switch that controls glucose versus fructose use in glycolysis and help better understand lung endothelial cell metabolism during respiratory failure.


Asunto(s)
Células Endoteliales , Fructosa , Hexoquinasa , Animales , Ratones , Células Endoteliales/metabolismo , Glucosa/metabolismo , Lactatos , Pulmón/metabolismo , Fructosa/metabolismo
11.
J Biol Chem ; 298(1): 101482, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34896150

RESUMEN

Patients who recover from nosocomial pneumonia oftentimes exhibit long-lasting cognitive impairment comparable with what is observed in Alzheimer's disease patients. We previously hypothesized that the lung endothelium contributes to infection-related neurocognitive dysfunction, because bacteria-exposed endothelial cells release a form(s) of cytotoxic tau that is sufficient to impair long-term potentiation in the hippocampus. However, the full-length lung and endothelial tau isoform(s) have yet to be resolved and it remains unclear whether the infection-induced endothelial cytotoxic tau triggers neuronal tau aggregation. Here, we demonstrate that lung endothelial cells express a big tau isoform and three additional tau isoforms that are similar to neuronal tau, each containing four microtubule-binding repeat domains, and that tau is expressed in lung capillaries in vivo. To test whether infection elicits endothelial tau capable of causing transmissible tau aggregation, the cells were infected with Pseudomonas aeruginosa. The infection-induced tau released from endothelium into the medium-induced neuronal tau aggregation in reporter cells, including reporter cells that express either the four microtubule-binding repeat domains or the full-length tau. Infection-induced release of pathological tau variant(s) from endothelium, and the ability of the endothelial-derived tau to cause neuronal tau aggregation, was abolished in tau knockout cells. After bacterial lung infection, brain homogenates from WT mice, but not from tau knockout mice, initiated tau aggregation. Thus, we conclude that bacterial pneumonia initiates the release of lung endothelial-derived cytotoxic tau, which is capable of propagating a neuronal tauopathy.


Asunto(s)
Enfermedades Pulmonares , Neumonía Bacteriana , Tauopatías , Proteínas tau , Animales , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/microbiología , Disfunción Cognitiva/patología , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Células Endoteliales/patología , Humanos , Pulmón/irrigación sanguínea , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/microbiología , Enfermedades Pulmonares/patología , Ratones , Neumonía Bacteriana/metabolismo , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , Isoformas de Proteínas , Pseudomonas aeruginosa , Tauopatías/genética , Tauopatías/metabolismo , Tauopatías/patología , Proteínas tau/química , Proteínas tau/genética , Proteínas tau/metabolismo
12.
Circ Res ; 128(1): 42-58, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33135960

RESUMEN

RATIONALE: The recent discovery of meningeal lymphatics in mammals is reshaping our understanding of fluid homeostasis and cellular waste management in the brain, but visualization and experimental analysis of these vessels is challenging in mammals. Although the optical clarity and experimental advantages of zebrafish have made this an essential model organism for studying lymphatic development, the existence of meningeal lymphatics has not yet been reported in this species. OBJECTIVE: Examine the intracranial space of larval, juvenile, and adult zebrafish to determine whether and where intracranial lymphatic vessels are present. METHODS AND RESULTS: Using high-resolution optical imaging of the meninges in living animals, we show that zebrafish possess a meningeal lymphatic network comparable to that found in mammals. We confirm that this network is separate from the blood vascular network and that it drains interstitial fluid from the brain. We document the developmental origins and growth of these vessels into a distinct network separated from the external lymphatics. Finally, we show that these vessels contain immune cells and perform live imaging of immune cell trafficking and transmigration in meningeal lymphatics. CONCLUSIONS: This discovery establishes the zebrafish as a important new model for experimental analysis of meningeal lymphatic development and opens up new avenues for probing meningeal lymphatic function in health and disease.


Asunto(s)
Linfangiogénesis , Vasos Linfáticos/fisiología , Meninges/fisiología , Microscopía Confocal , Imagen Óptica , Animales , Animales Modificados Genéticamente , Linfangiogénesis/efectos de los fármacos , Vasos Linfáticos/efectos de los fármacos , Vasos Linfáticos/inmunología , Meninges/inmunología , Infiltración Neutrófila , Neutrófilos/inmunología , Factor C de Crecimiento Endotelial Vascular/farmacología , Pez Cebra/genética
13.
Am J Respir Cell Mol Biol ; 67(3): 275-283, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35348443

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel coronavirus that is currently causing a pandemic and has been termed coronavirus disease (COVID-19). The elderly or those with preexisting conditions like diabetes, hypertension, coronary heart disease, chronic obstructive pulmonary disease, cerebrovascular disease, or kidney dysfunction are more likely to develop severe cases when infected. Patients with COVID-19 admitted to the ICU have higher mortality than non-ICU patients. Critical illness has consistently posed a challenge not only in terms of mortality but also in regard to long-term outcomes of survivors. Patients who survive acute critical illness including, but not limited to, pulmonary and systemic insults associated with acute respiratory distress syndrome, pneumonia, systemic inflammation, and mechanical ventilation, will likely suffer from post-ICU syndrome, a phenomenon of cognitive, psychiatric, and/or physical disability after treatment in the ICU. Post-ICU morbidity and mortality continue to be a cause for concern when considering large-scale studies showing 12-month mortality risks of 11.8-21%. Previous studies have demonstrated that multiple mechanisms, including cytokine release, mitochondrial dysfunction, and even amyloids, may lead to end-organ dysfunction in patients. We hypothesize that COVID-19 infection will lead to post-ICU syndrome via potentially similar mechanisms as other chronic critical illnesses and cause long-term morbidity and mortality in patients. We consider a variety of mechanisms and questions that not only consider the short-term impact of the COVID-19 pandemic but its long-term effects that may not yet be imagined.


Asunto(s)
COVID-19 , Pandemias , Anciano , Cuidados Críticos , Enfermedad Crítica , Humanos , SARS-CoV-2
15.
Angiogenesis ; 25(3): 411-434, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35320450

RESUMEN

The small monomeric GTPase RHOA acts as a master regulator of signal transduction cascades by activating effectors of cellular signaling, including the Rho-associated protein kinases ROCK1/2. Previous in vitro cell culture studies suggest that RHOA can regulate many critical aspects of vascular endothelial cell (EC) biology, including focal adhesion, stress fiber formation, and angiogenesis. However, the specific in vivo roles of RHOA during vascular development and homeostasis are still not well understood. In this study, we examine the in vivo functions of RHOA in regulating vascular development and integrity in zebrafish. We use zebrafish RHOA-ortholog (rhoaa) mutants, transgenic embryos expressing wild type, dominant negative, or constitutively active forms of rhoaa in ECs, pharmacological inhibitors of RHOA and ROCK1/2, and Rock1 and Rock2a/b dgRNP-injected zebrafish embryos to study the in vivo consequences of RHOA gain- and loss-of-function in the vascular endothelium. Our findings document roles for RHOA in vascular integrity, developmental angiogenesis, and vascular morphogenesis in vivo, showing that either too much or too little RHOA activity leads to vascular dysfunction.


Asunto(s)
Pez Cebra , Proteína de Unión al GTP rhoA , Animales , Animales Modificados Genéticamente , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Transducción de Señal , Pez Cebra/genética , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
16.
FASEB J ; 35(10): e21946, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34555226

RESUMEN

Acute respiratory distress syndrome (ARDS) is a life-threatening illness characterized by decreased alveolar-capillary barrier function, pulmonary edema consisting of proteinaceous fluid, and inhibition of net alveolar fluid transport responsible for resolution of pulmonary edema. There is currently no pharmacotherapy that has proven useful to prevent or treat ARDS, and two trials using beta-agonist therapy to treat ARDS demonstrated no effect. Prior studies indicated that IL-8-induced heterologous desensitization of the beta2-adrenergic receptor (ß2 -AR) led to decreased beta-agonist-induced mobilization of cyclic adenosine monophosphate (cAMP). Interestingly, phosphodiesterase (PDE) 4 inhibitors have been used in human airway diseases characterized by low intracellular cAMP levels and increases in specific cAMP hydrolyzing activity. Therefore, we hypothesized that PDE4 would mediate IL-8-induced heterologous internalization of the ß2 -AR and that PDE4 inhibition would restore beta-agonist-induced functions. We determined that CINC-1 (a functional IL-8 analog in rats) induces internalization of ß2 -AR from the cell surface, and arrestin-2, PDE4, and ß2 -AR form a complex during this process. Furthermore, we determined that cAMP associated with the plasma membrane was adversely affected by ß2 -AR heterologous desensitization. Additionally, we determined that rolipram, a PDE4 inhibitor, reversed CINC-1-induced derangements of cAMP and also caused ß2 -AR to successfully recycle back to the cell surface. Finally, we demonstrated that rolipram could reverse CINC-1-mediated inhibition of beta-agonist-induced alveolar fluid clearance in a murine model of trauma-shock. These results indicate that PDE4 plays a role in CINC-1-induced heterologous internalization of the ß2 -AR; PDE4 inhibition reverses these effects and may be a useful adjunct in particular ARDS patients.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Interleucina-8/inmunología , Receptores Adrenérgicos beta 2/metabolismo , Animales , Líquido del Lavado Bronquioalveolar , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Quimiocina CXCL1/metabolismo , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/farmacología , Regulación hacia Abajo/efectos de los fármacos , Masculino , Ratones , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de Fosfodiesterasa 4/farmacología , beta-Arrestina 1/metabolismo
17.
FASEB J ; 35(9): e21807, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34384141

RESUMEN

Pneumonia causes short- and long-term cognitive dysfunction in a high proportion of patients, although the mechanism(s) responsible for this effect are unknown. Here, we tested the hypothesis that pneumonia-elicited cytotoxic amyloid and tau variants: (1) are present in the circulation during infection; (2) lead to impairment of long-term potentiation; and, (3) inhibit long-term potentiation dependent upon tau. Cytotoxic amyloid and tau species were recovered from the blood and the hippocampus following pneumonia, and they were present in the extracorporeal membrane oxygenation oxygenators of patients with pneumonia, especially in those who died. Introduction of immunopurified blood-borne amyloid and tau into either the airways or the blood of uninfected animals acutely and chronically impaired hippocampal information processing. In contrast, the infection did not impair long-term potentiation in tau knockout mice and the amyloid- and tau-dependent disruption in hippocampal signaling was less severe in tau knockout mice. Moreover, the infection did not elicit cytotoxic amyloid and tau variants in tau knockout mice. Therefore, pneumonia initiates a tauopathy that contributes to cognitive dysfunction.


Asunto(s)
Neumonía/complicaciones , Tauopatías/etiología , Adulto , Anciano , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Amiloide/metabolismo , Animales , Disfunción Cognitiva/etiología , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Femenino , Hipocampo/metabolismo , Humanos , Potenciación a Largo Plazo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Neumonía/metabolismo , Ratas , Tauopatías/metabolismo , Adulto Joven , Proteínas tau/metabolismo
18.
Am J Respir Cell Mol Biol ; 65(6): 630-645, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34251286

RESUMEN

Low tidal volume ventilation protects the lung in mechanically ventilated patients. The impact of the accompanying permissive hypoxemia and hypercapnia on endothelial cell recovery from injury is poorly understood. CA (carbonic anhydrase) IX is expressed in pulmonary microvascular endothelial cells (PMVECs), where it contributes to CO2 and pH homeostasis, bioenergetics, and angiogenesis. We hypothesized that CA IX is important for PMVEC survival and that CA IX expression and release from PMVECs are increased during infection. Although the plasma concentration of CA IX was unchanged in human and rat pneumonia, there was a trend toward increasing CA IX in the bronchoalveolar fluid of mechanically ventilated critically ill patients with pneumonia and a significant increase in CA IX in the lung tissue lysates of pneumonia rats. To investigate the functional implications of the lung CA IX increase, we generated PMVEC cell lines harboring domain-specific CA IX mutations. By using these cells, we found that infection promotes intracellular (IC) expression, release, and MMP (metalloproteinase)-mediated extracellular cleavage of CA IX in PMVECs. IC domain deletion uniquely impaired CA IX membrane localization. Loss of the CA IX IC domain promoted cell death after infection, suggesting that the IC domain has an important role in PMVEC survival. We also found that hypoxia improves survival, whereas hypercapnia reverses the protective effect of hypoxia, during infection. Thus, we report 1) that CA IX increases in the lungs of pneumonia rats and 2) that the CA IX IC domain and hypoxia promote PMVEC survival during infection.


Asunto(s)
Anhidrasa Carbónica IX/metabolismo , Células Endoteliales/enzimología , Pulmón/enzimología , Neumonía Bacteriana/enzimología , Infecciones por Pseudomonas/enzimología , Pseudomonas aeruginosa/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Hipoxia de la Célula , Humanos , Masculino , Ratas , Ratas Endogámicas F344
19.
Angiogenesis ; 24(2): 251-269, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33449300

RESUMEN

Endothelial cells display an extraordinary plasticity both during development and throughout adult life. During early development, endothelial cells assume arterial, venous, or lymphatic identity, while selected endothelial cells undergo additional fate changes to become hematopoietic progenitor, cardiac valve, and other cell types. Adult endothelial cells are some of the longest-lived cells in the body and their participation as stable components of the vascular wall is critical for the proper function of both the circulatory and lymphatic systems, yet these cells also display a remarkable capacity to undergo changes in their differentiated identity during injury, disease, and even normal physiological changes in the vasculature. Here, we discuss how endothelial cells become specified during development as arterial, venous, or lymphatic endothelial cells or convert into hematopoietic stem and progenitor cells or cardiac valve cells. We compare findings from in vitro and in vivo studies with a focus on the zebrafish as a valuable model for exploring the signaling pathways and environmental cues that drive these transitions. We also discuss how endothelial plasticity can aid in revascularization and repair of tissue after damage- but may have detrimental consequences under disease conditions. By better understanding endothelial plasticity and the mechanisms underlying endothelial fate transitions, we can begin to explore new therapeutic avenues.


Asunto(s)
Diferenciación Celular , Células Endoteliales/metabolismo , Neovascularización Fisiológica , Heridas y Lesiones/metabolismo , Pez Cebra/metabolismo , Animales , Arterias/metabolismo , Células Madre Hematopoyéticas , Humanos , Vasos Linfáticos/metabolismo , Venas/metabolismo , Heridas y Lesiones/terapia
20.
FASEB J ; 34(2): 3305-3317, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31916311

RESUMEN

Pulmonary edema associated with increased vascular permeability is a severe complication of Pseudomonas (P.) aeruginosa-induced acute lung injury. The mechanisms underlying P aeruginosa-induced vascular permeability are not well understood. In the present study, we investigated the role of neuronal Wiskott Aldrich syndrome protein (N-WASP) in modulating P aeruginosa-induced vascular permeability. Using lung microvascular endothelial and alveolar epithelial cells, we demonstrated that N-WASP downregulation attenuated P aeruginosa-induced actin stress fiber formation and prevented paracellular permeability. P aeruginosa-induced dissociation between VE-cadherin and ß-catenin, but increased association between N-WASP and VE-cadherin, suggesting a role for N-WASP in promoting P aeruginosa-induced adherens junction rupture. P aeruginosa increased N-WASP-Y256 phosphorylation, which required the activation of Rho GTPase and focal adhesion kinase. Increased N-WASP-Y256 phosphorylation promotes N-WASP and integrin αVß6 association as well as TGF-ß-mediated permeability across alveolar epithelial cells. Inhibition of N-WASP-Y256 phosphorylation by N-WASP-Y256F overexpression blocked N-WASP effects in P aeruginosa-induced actin stress fiber formation and increased paracellular permeability. In vivo, N-WASP knockdown attenuated the development of pulmonary edema and improved survival in a mouse model of P aeruginosa pneumonia. Together, our data demonstrate that N-WASP plays an essential role in P aeruginosa-induced vascular permeability and pulmonary edema through the modulation of actin cytoskeleton dynamics.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Permeabilidad Capilar , Pulmón/metabolismo , Neumonía/metabolismo , Infecciones por Pseudomonas/metabolismo , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Uniones Adherentes/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Cadherinas/metabolismo , Células Cultivadas , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Integrinas/metabolismo , Pulmón/microbiología , Ratones , Pseudomonas aeruginosa/patogenicidad , Ratas , Factor de Crecimiento Transformador beta/metabolismo , Proteína del Síndrome de Wiskott-Aldrich/genética , beta Catenina/metabolismo , Proteínas de Unión al GTP rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA