Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Infect Dis ; 229(3): 876-887, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-37671668

RESUMEN

Mycobacterium tuberculosis (Mtb)-specific γ9δ2 T cells secrete granzyme A (GzmA) protective against intracellular Mtb growth. However, GzmA-enzymatic activity is unnecessary for pathogen inhibition, and the mechanisms of GzmA-mediated protection remain unknown. We show that GzmA homodimerization is essential for opsonization of mycobacteria, altered uptake into human monocytes, and subsequent pathogen clearance within the phagolysosome. Although monomeric and homodimeric GzmA bind mycobacteria, only homodimers also bind cluster of differentiation 14 (CD14) and Toll-like receptor 4 (TLR4). Without access to surface-expressed CD14 and TLR4, GzmA fails to inhibit intracellular Mtb. Upregulation of Rab11FIP1 was associated with inhibitory activity. Furthermore, GzmA colocalized with and was regulated by protein disulfide isomerase AI (PDIA1), which cleaves GzmA homodimers into monomers and prevents Mtb inhibitory activity. These studies identify a previously unrecognized role for homodimeric GzmA structure in opsonization, phagocytosis, and elimination of Mtb in human monocytes, and they highlight PDIA1 as a potential host-directed therapy for prevention and treatment of tuberculosis, a major human disease.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Humanos , Granzimas/metabolismo , Monocitos/metabolismo , Receptor Toll-Like 4/metabolismo , Tuberculosis/microbiología
2.
Peptides ; 150: 170718, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34954230

RESUMEN

Connecting peptide, or C-peptide, is a part of the insulin prohormone and is essential for the proper folding and processing of the mature insulin peptide. C-peptide is released from the same beta cell secretory granules as insulin in equimolar amounts. However, due to their relative stabilities in plasma, the two peptides are detected in the circulation at ratios of approximately 4:1 to 6:1 (C-peptide to insulin), depending on metabolic state. C-peptide binds specifically to human cell membranes and induces intracellular signaling cascades, likely through an interaction with the G protein coupled receptor, GPR146. C-peptide has been shown to exert protective effects against the vascular, renal, and ocular complications of diabetes. The effects of C-peptide appear to be dependent upon the presence of insulin and the absolute, extracellular concentration of glucose. In this study, we employed HEK293 cells to further examine the interactive effects of C-peptide, insulin, and glucose on cell signaling. We observed that C-peptide's cellular effects are dampened significantly when cells are exposed to physiologically relevant concentrations of both insulin and C-peptide. Likewise, the actions of C-peptide on cFos and GPR146 mRNA expressions were affected by changes in extracellular glucose concentration. In particular, C-peptide induced significant elevations in cFos expression in the setting of high (25 mmol) extracellular glucose concentration. These data indicate that future experimentation on the actions of C-peptide should control for the presence or absence of insulin and the concentration of glucose. Furthermore, these findings should be considered prior to the development of C-peptide-based therapeutics for the treatment of diabetes-associated complications.


Asunto(s)
Complicaciones de la Diabetes , Insulina , Glucemia , Péptido C , Glucosa/farmacología , Células HEK293 , Humanos , Insulina/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
3.
Cell Mol Neurobiol ; 42(8): 2909-2918, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34773542

RESUMEN

Emerging evidence implicates the sphingosine-1-phosphate receptor subtype 1 (S1PR1) in the development of neuropathic pain. Continued investigation of the signaling pathways downstream of S1PR1 are needed to support development of S1PR1 antagonists. In rodents, intrathecal (i.th.) injection of SEW2871, a selective S1PR1 agonist, activates the nod-like receptor family, pyrin domain containing 3 inflammasome, increases interleukin-1ß (IL-1ß) and causes behavioral hypersensitivity. I.th. injection of a IL-1ß receptor antagonist blocks SEW2871-induced hypersensitivity, suggesting that IL-1ß contributes to S1PR1's actions. Interestingly, previous studies have suggested that IL-1ß increases the expression/activity of adenosine kinase (ADK), a key regulator of adenosine signaling at its receptors (ARs). Increased ADK expression reduces adenosine signaling whereas inhibiting ADK restores the action of adenosine. Here, we show that SEW287-induced behavioral hypersensitivity is associated with increased expression of ADK in astrocytes of the dorsal horn of the spinal cord. Moreover, the ADK inhibitor, ABT702, blocks SEW2871-induced hypersensitivity. These findings link ADK activation to S1PR1. If SEW2871-induced pain is mediated by IL-1ß, which in turn activates ADK and leads to mechano-allodynia, then blocking ADK should attenuate IL-1ß effects. In support of this idea, recombinant rat (rrIL-1ß)-induced allodynia was blocked by at least 90% with ABT702, functionally linking ADK to IL-1ß. Moreover, the selective A3AR antagonist, MRS1523, prevents the ability of ABT702 to block SEW2871 and IL-1ß-induced allodynia, implicating A3AR signaling in the beneficial effects exerted by ABT702. Our findings provide novel mechanistic insight into how S1PR1 signaling in the spinal cord produces hypersensitivity through IL1-ß and ADK activation.


Asunto(s)
Adenosina Quinasa , Inflamasomas , Adenosina , Animales , Hiperalgesia/metabolismo , Interleucina-1beta/metabolismo , Proteínas NLR , Oxadiazoles , Ratas , Ratas Sprague-Dawley , Receptores de Esfingosina-1-Fosfato , Asta Dorsal de la Médula Espinal/metabolismo , Tiofenos
4.
Front Immunol ; 12: 701227, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34489949

RESUMEN

Neutrophils are the most abundant white blood cells recruited to the sites of infection and inflammation. During neutrophil activation, myeloperoxidase (MPO) is released and converts hydrogen peroxide to hypochlorous acid (HOCl). HOCl reacts with plasmalogen phospholipids to liberate 2-chlorofatty aldehyde (2-ClFALD), which is metabolized to 2-chlorofatty acid (2-ClFA). 2-ClFA and 2-ClFALD are linked with inflammatory diseases and induce endothelial dysfunction, neutrophil extracellular trap formation (NETosis) and neutrophil chemotaxis. Here we examine the neutrophil-derived chlorolipid production in the presence of pathogenic E. coli strain CFT073 and non-pathogenic E. coli strain JM109. Neutrophils cocultured with CFT073 E. coli strain and JM109 E. coli strain resulted in 2-ClFALD production. 2-ClFA was elevated only in CFT073 coculture. NETosis is more prevalent in CFT073 cocultures with neutrophils compared to JM109 cocultures. 2-ClFA and 2-ClFALD were both shown to have significant bactericidal activity, which is more severe in JM109 E. coli. 2-ClFALD metabolic capacity was 1000-fold greater in neutrophils compared to either strain of E. coli. MPO inhibition reduced chlorolipid production as well as bacterial killing capacity. These findings indicate the chlorolipid profile is different in response to these two different strains of E. coli bacteria.


Asunto(s)
Escherichia coli/inmunología , Ácidos Grasos/metabolismo , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Peroxidasa/inmunología , Células Cultivadas , Trampas Extracelulares/inmunología , Humanos , Neutrófilos/enzimología
5.
Mo Med ; 118(4): 352-357, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34373671

RESUMEN

G protein-coupled receptors (GPCRs) transmit the signals of a variety of hormones and neurotransmitters and are targets of more than 30% of all FDA-approved drugs. We developed an approach for identifying the endogenous ligands for a family of orphan GPCRs that enables the development of novel therapeutics for the potential treatment of a wide variety of disorders including pain, diabetes, appetitive behaviors, infertility and obesity. With this approach, we have deorphanized five previously orphaned GPCRs.


Asunto(s)
Obesidad , Humanos , Ligandos
6.
Physiol Behav ; 235: 113380, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33705816

RESUMEN

The existence of the peptide encoded by the cocaine- and amphetamine-regulated transcript (Cartpt) has been recognized since 1981, but it was not until 1995, that the gene encoding CART peptide (CART) was identified. With the availability of the predicted protein sequence of CART investigators were able to identify sites of peptide localization, which then led to numerous approaches attempting to clarify CART's multiple pharmacologic effects and even provide evidence of potential physiologic relevance. Although not without controversy, a picture emerged of the importance of CART in ingestive behaviors, reward behaviors and even pain sensation. Despite the wealth of data hinting at the significance of CART, in the absence of an identified receptor, the full potential for this peptide or its analogs to be developed into therapeutic agents remained unrealized. There was evidence favoring the action of CART via a G protein-coupled receptor (GPCR), but despite multiple attempts the identity of that receptor eluded investigators until recently. Now with the identification of the previously orphaned GPCR, GPR160, as a receptor for CART, focus on this pluripotent neuropeptide will in all likelihood experience a renaissance and the potential for the development of pharmcotherapies targeting GPR160 seems within reach.


Asunto(s)
Cocaína , Neuropéptidos , Conducta Alimentaria , Proteínas del Tejido Nervioso/genética , Recompensa
7.
J Clin Invest ; 131(7)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33621215

RESUMEN

The A3 adenosine receptor (A3AR) has emerged as a therapeutic target with A3AR agonists to tackle the global challenge of neuropathic pain, and investigation into its mode of action is essential for ongoing clinical development. Immune cell A3ARs, and their activation during pathology, modulate cytokine release. Thus, the use of immune cells as a cellular substrate for the pharmacological action of A3AR agonists is enticing, but unknown. The present study discovered that Rag-KO mice lacking T and B cells, as compared with WT mice, are insensitive to the anti-allodynic effects of A3AR agonists. Similar findings were observed in interleukin-10 and interleukin-10 receptor knockout mice. Adoptive transfer of CD4+ T cells from WT mice infiltrated the dorsal root ganglion (DRG) and restored A3AR agonist-mediated anti-allodynia in Rag-KO mice. CD4+ T cells from Adora3-KO or Il10-KO mice did not. Transfer of CD4+ T cells from WT mice, but not Il10-KO mice, into Il10-KO mice or Adora3-KO mice fully reinstated the anti-allodynic effects of A3AR activation. Notably, A3AR agonism reduced DRG neuron excitability when cocultured with CD4+ T cells in an IL-10-dependent manner. A3AR action on CD4+ T cells infiltrated in the DRG decreased phosphorylation of GluN2B-containing N-methyl-D-aspartate receptors at Tyr1472, a modification associated with regulating neuronal hypersensitivity. Our findings establish that activation of A3AR on CD4+ T cells to release IL-10 is required and sufficient evidence for the use of A3AR agonists as therapeutics.


Asunto(s)
Agonistas del Receptor de Adenosina A3/farmacología , Linfocitos T CD4-Positivos/inmunología , Ganglios Espinales/inmunología , Interleucina-10/inmunología , Neuralgia/tratamiento farmacológico , Neuronas/inmunología , Receptor de Adenosina A3/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/patología , Linfocitos T CD4-Positivos/patología , Ganglios Espinales/patología , Interleucina-10/genética , Ratones , Ratones Noqueados , Neuralgia/genética , Neuralgia/inmunología , Neuralgia/patología , Neuronas/patología , Receptor de Adenosina A3/genética
8.
Am J Physiol Regul Integr Comp Physiol ; 320(3): R236-R249, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33206556

RESUMEN

Recent work identified Gpr160 as a candidate receptor for cocaine- and amphetamine-regulated transcript peptide (CARTp) and described its role in pain modulation. The aims of the present study were to determine if Gpr160 is required for the CARTp's ability to reduce food intake and water intake and to initially identify the distribution of Gpr160-like immunoreactivity (Gpr160ir) in the rat brain. A passive immunoneutralization approach targeting Gpr160 was used to block the behavioral effects of a pharmacological dose of CARTp in the fourth cerebroventricle (4V) of rats and to determine the importance of endogenously produced CARTp in the control of ingestive behaviors. Passive immunoneutralization of Gpr160 in the 4V blocked the actions of CARTp to inhibit food intake and water intake. Blockade of Gpr160 in the 4V, independent of pharmacological CART treatment, caused an increase in both overnight food intake and water intake. The decrease in food intake, but not water intake, caused by central injection of CARTp was demonstrated to be interrupted by prior administration of a glucagon-like peptide 1 (GLP-1) receptor antagonist. Gpr160ir was observed in several, distinct sites throughout the rat brain, where CARTp staining has been described. Importantly, Gpr160ir was observed to be present in both neuronal and nonneuronal cell types. These data support the hypothesis that Gpr160 is required for the anorexigenic actions of central CARTp injection and extend these findings to water drinking. Gpr160ir was observed in both neuronal and nonneuronal cell types in regions known to be important in the multiple pharmacological effects of CARTp, identifying those areas as targets for future compromise of function studies.


Asunto(s)
Depresores del Apetito/farmacología , Tronco Encefálico/efectos de los fármacos , Conducta de Ingestión de Líquido/efectos de los fármacos , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Proteínas del Tejido Nervioso/farmacología , Receptores Acoplados a Proteínas G/agonistas , Animales , Tronco Encefálico/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Masculino , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo
9.
J Pharmacol Exp Ther ; 375(2): 367-375, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32913007

RESUMEN

Neuropathic pain is a debilitating public health concern for which novel non-narcotic therapeutic targets are desperately needed. Using unbiased transcriptomic screening of the dorsal horn spinal cord after nerve injury we have identified that Gpr183 (Epstein-Barr virus-induced gene 2) is upregulated after chronic constriction injury (CCI) in rats. GPR183 is a chemotactic receptor known for its role in the maturation of B cells, and the endogenous ligand is the oxysterol 7α,25-dihydroxycholesterol (7α,25-OHC). The role of GPR183 in the central nervous system is not well characterized, and its role in pain is unknown. The profile of commercially available probes for GPR183 limits their use as pharmacological tools to dissect the roles of this receptor in pathophysiological settings. Using in silico modeling, we have screened a library of 5 million compounds to identify several novel small-molecule antagonists of GPR183 with nanomolar potency. These compounds are able to antagonize 7α,25-OHC-induced calcium mobilization in vitro with IC50 values below 50 nM. In vivo intrathecal injections of these antagonists during peak pain after CCI surgery reversed allodynia in male and female mice. Acute intrathecal injection of the GPR183 ligand 7α,25-OHC in naïve mice induced dose-dependent allodynia. Importantly, this effect was blocked using our novel GPR183 antagonists, suggesting spinal GPR183 activation as pronociceptive. These studies are the first to reveal a role for GPR183 in neuropathic pain and identify this receptor as a potential target for therapeutic intervention. SIGNIFICANCE STATEMENT: We have identified several novel GPR183 antagonists with nanomolar potency. Using these antagonists, we have demonstrated that GPR183 signaling in the spinal cord is pronociceptive. These studies are the first to reveal a role for GPR183 in neuropathic pain and identify it as a potential target for therapeutic intervention.


Asunto(s)
Neuralgia/metabolismo , Oxiesteroles/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Médula Espinal/metabolismo , Animales , Femenino , Células HL-60 , Humanos , Masculino , Ratones , Neuralgia/tratamiento farmacológico , Neuralgia/patología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Transducción de Señal , Médula Espinal/patología
10.
IEEE J Biomed Health Inform ; 24(9): 2523-2534, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32750953

RESUMEN

Telepathology aims to replace the pathology operations performed on-site, but current systems are limited by their prohibitive cost, or by the adopted underlying technologies. In this work, we contribute to overcoming these limitations by bringing the recent advances of edge computing to reduce latency and increase local computation abilities to the pathology ecosystem. In particular, this paper presents LiveMicro, a system whose benefit is twofold: on one hand, it enables edge computing driven digital pathology computations, such as data-driven image processing on a live capture of the microscope. On the other hand, our system allows remote pathologists to diagnosis in collaboration in a single virtual microscope session, facilitating continuous medical education and remote consultation, crucial for under-served and remote hospital or private practice. Our results show the benefits and the principles underpinning our solution, with particular emphasis on how the pathologists interact with our application. Additionally, we developed simple yet effective diagnosis-aided algorithms to demonstrate the practicality of our approach.


Asunto(s)
Consulta Remota , Telepatología , Ecosistema , Humanos , Procesamiento de Imagen Asistido por Computador , Microscopía
11.
J Clin Invest ; 130(5): 2587-2592, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31999650

RESUMEN

Treating neuropathic pain is challenging and novel non-opioid-based medicines are needed. Using unbiased receptomics, transcriptomic analyses, immunofluorescence, and in situ hybridization, we found that the expression of the orphan GPCR Gpr160 and GPR160 increased in the rodent dorsal horn of the spinal cord following traumatic nerve injury. Genetic and immunopharmacological approaches demonstrated that GPR160 inhibition in the spinal cord prevented and reversed neuropathic pain in male and female rodents without altering normal pain response. GPR160 inhibition in the spinal cord attenuated sensory processing in the thalamus, a key relay in the sensory discriminative pathways of pain. We also identified cocaine- and amphetamine-regulated transcript peptide (CARTp) as a GPR160 ligand. Inhibiting endogenous CARTp signaling in spinal cord attenuated neuropathic pain, whereas exogenous intrathecal CARTp evoked painful hypersensitivity through GPR160-dependent ERK and cAMP response element-binding protein (CREB). Our findings de-orphanize GPR160, identify it as a determinant of neuropathic pain and potential therapeutic target, and provide insights into its signaling pathways. CARTp is involved in many diseases including depression and reward and addiction; de-orphanization of GPR160 is a major step forward understanding the role of CARTp signaling in health and disease.


Asunto(s)
Neuralgia/etiología , Neuralgia/fisiopatología , Receptores Acoplados a Proteínas G/fisiología , Animales , Línea Celular , Femenino , Humanos , Ligandos , Masculino , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuralgia/genética , Células PC12 , ARN Interferente Pequeño/genética , Ratas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Nervio Ciático/lesiones , Nervio Ciático/fisiopatología , Transducción de Señal , Médula Espinal/metabolismo , Regulación hacia Arriba
12.
Proc Natl Acad Sci U S A ; 116(21): 10557-10562, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31068460

RESUMEN

Neuropathic pain afflicts millions of individuals and represents a major health problem for which there is limited effective and safe therapy. Emerging literature links altered sphingolipid metabolism to nociceptive processing. However, the neuropharmacology of sphingolipid signaling in the central nervous system in the context of chronic pain remains largely unexplored and controversial. We now provide evidence that sphingosine-1-phosphate (S1P) generated in the dorsal horn of the spinal cord in response to nerve injury drives neuropathic pain by selectively activating the S1P receptor subtype 1 (S1PR1) in astrocytes. Accordingly, genetic and pharmacological inhibition of S1PR1 with multiple antagonists in distinct chemical classes, but not agonists, attenuated and even reversed neuropathic pain in rodents of both sexes and in two models of traumatic nerve injury. These S1PR1 antagonists retained their ability to inhibit neuropathic pain during sustained drug administration, and their effects were independent of endogenous opioid circuits. Moreover, mice with astrocyte-specific knockout of S1pr1 did not develop neuropathic pain following nerve injury, thereby identifying astrocytes as the primary cellular substrate of S1PR1 activity. On a molecular level, the beneficial reductions in neuropathic pain resulting from S1PR1 inhibition were driven by interleukin 10 (IL-10), a potent neuroprotective and anti-inflammatory cytokine. Collectively, our results provide fundamental neurobiological insights that identify the cellular and molecular mechanisms engaged by the S1PR1 axis in neuropathic pain and establish S1PR1 as a target for therapeutic intervention with S1PR1 antagonists as a class of nonnarcotic analgesics.


Asunto(s)
Astrocitos/metabolismo , Neuralgia/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Sulfonas/uso terapéutico , Triazoles/uso terapéutico , Animales , Evaluación Preclínica de Medicamentos , Femenino , Interleucina-10/metabolismo , Masculino , Ratones , Neuralgia/tratamiento farmacológico , Neuralgia/etiología , Ratas Sprague-Dawley , Receptores de Esfingosina-1-Fosfato/antagonistas & inhibidores , Sulfonas/farmacología , Triazoles/farmacología
13.
Bioconjug Chem ; 30(1): 34-46, 2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30562006

RESUMEN

Macroporous cell-laden hydrogels have recently gained recognition for a wide range of biomedical and bioengineering applications. There are various approaches to create porosity in hydrogels, including lyophilization or foam formation. However, many do not allow a precise control over pore size or are not compatible with in situ cell encapsulation. Here, we developed novel templated macroporous hydrogels by encapsulating uniform degradable hydrogel microspheres produced via microfluidics into a hydrogel slab. The microspheres degraded completely leaving macropores behind. Microsphere degradation was dependent on the incubation medium, microsphere size, microsphere confinement in the hydrogel as well as cell encapsulation. Uniquely, the degradable microspheres were biocompatible and when laden with cells, the cells were deposited in the macropores upon microsphere degradation and formed multicellular aggregates. The hydrogel-encapsulated cell aggregates were used in a small drug screen to demonstrate the relevance of cell-matrix interactions for multicellular spheroid drug responsiveness. Hydrogel-grown spheroid cultures are increasingly important in applications such as in vitro tumor, hepatocellular, and neurosphere cultures and drug screening; hence, the templated cell aggregate-laden hydrogels described here would find utility in various applications.


Asunto(s)
Hidrogeles/química , Polietilenglicoles/química , Esferoides Celulares/química , Línea Celular Tumoral , Supervivencia Celular , Medios de Cultivo , Humanos , Microfluídica , Microesferas , Porosidad
14.
Brain Pathol ; 28(6): 806-821, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30062819

RESUMEN

BACKGROUND: Mutations in the three-prime repair exonuclease 1 (TREX1) gene have been associated with neurological diseases, including Retinal Vasculopathy with Cerebral Leukoencephalopathy (RVCL). However, the endogenous expression of TREX1 in human brain has not been studied. METHODS: We produced a rabbit polyclonal antibody (pAb) to TREX1 to characterize TREX1 by Western blotting (WB) of cell lysates from normal controls and subjects carrying an RVCL frame-shift mutation. Dual staining was performed to determine cell types expressing TREX1 in human brain tissue. TREX1 distribution in human brain was further evaluated by immunohistochemical analyses of formalin-fixed, paraffin-embedded samples from normal controls and patients with RVCL and ischemic stroke. RESULTS: After validating the specificity of our anti-TREX1 rabbit pAb, WB analysis was utilized to detect the endogenous wild-type and frame-shift mutant of TREX1 in cell lysates. Dual staining in human brain tissues from patients with RVCL and normal controls localized TREX1 to a subset of microglia and macrophages. Quantification of immunohistochemical staining of the cerebral cortex revealed that TREX1+ microglia were primarily in the gray matter of normal controls (22.7 ± 5.1% and 5.5 ± 1.9% of Iba1+ microglia in gray and white matter, respectively) and commonly in association with the microvasculature. In contrast, in subjects with RVCL, the TREX1+ microglia were predominantly located in the white matter of normal appearing cerebral cortex (11.8 ± 3.1% and 38.9 ± 5.8% of Iba1+ microglia in gray and white matter, respectively). The number of TREX1+ microglia was increased in ischemic brain lesions in central nervous system of RVCL and stroke patients. CONCLUSIONS: TREX1 is expressed by a subset of microglia in normal human brain, often in close proximity to the microvasculature, and increases in the setting of ischemic lesions. These findings suggest a role for TREX1+ microglia in vessel homeostasis and response to ischemic injury.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Encéfalo/metabolismo , Exodesoxirribonucleasas/metabolismo , Exodesoxirribonucleasas/fisiología , Microglía/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiología , Anciano , Animales , Anticuerpos/metabolismo , Encéfalo/patología , Exodesoxirribonucleasas/genética , Femenino , Mutación del Sistema de Lectura , Células HEK293 , Células HeLa , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/patología , Homeostasis/fisiología , Humanos , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Fosfoproteínas/genética , Conejos/inmunología , Enfermedades de la Retina/genética , Enfermedades de la Retina/patología , Enfermedades Vasculares/genética , Enfermedades Vasculares/patología
15.
Peptides ; 106: 45-48, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29933026

RESUMEN

Using a series of classical protein purification techniques, coupled with more modern molecular approaches, a family of neuropeptides, the Phoenixins, was identified to be produced in brain and heart, and to bind selectively in pituitary gland, ovary and brain. These same binding sites were revealed, using a novel receptor identification strategy, to express the orphan G protein-coupled receptor, GPR173, the expression of which was required for the actions of phoenixin both in vivo and in vitro. In fact, studies using small interfering RNA molecules to compromise GPR173 expression revealed the physiologic relevance of the initially reported pharmacologic actions of the peptides. Those include not only the reproductive actions of the peptides in brain and pituitary gland, but also a CNS site of action in the maintenance of fluid and electrolyte homeostasis. Additional pharmacologic actions of the phoenixins have been described and the race is on to establish the physiologic relevance of those actions as well as the therapeutic potential of phoenixin analogs.


Asunto(s)
Hormonas Hipotalámicas/fisiología , Neuropéptidos/fisiología , Receptores de Péptidos/metabolismo , Animales , Encéfalo/fisiología , Modelos Animales de Enfermedad , Femenino , Corazón/fisiología , Humanos , Ovario/fisiología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/genética
17.
J Lipid Res ; 59(8): 1424-1432, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29739865

RESUMEN

Neutrophils form neutrophil extracellular traps (NETs), which have been implicated in microcirculatory plugging. NET formation (NETosis) involves the fusion of granule and nuclear contents, which are then released in the extracellular space. Myeloperoxidase (MPO) plays a major role in NETosis leading to the dissociation of DNA from histones. During neutrophil activation, MPO is released and activated to convert hydrogen peroxide and chloride to hypochlorous acid (HOCl). HOCl targets plasmalogens leading to the production of the chlorinated lipids, 2-chlorofatty aldehyde and 2-chlorofatty acid (2-ClFA). Here, we tested the hypothesis that 2-ClFAs are important lipid mediators of NETosis. Human neutrophils treated with physiological levels of 2-ClFAs formed NETs, characterized by MPO association with DNA and neutrophil elastase (NE) redistribution to the perinuclear area. 2-ClFA-induced NETs reduced Escerichia coli colony forming units. 2-ClFA-induced NETosis is calcium- and protein arginine deiminase 4-dependent. Interestingly, unlike PMA, 2-ClFA initiates the NETosis process without neutrophil activation and degranulation. Furthermore, 2-ClFA elicits NETosis in bone-marrow derived neutrophils from MPO-deficient mice. Taken together, these findings suggest 2-ClFA as an MPO product that triggers the NETosis pathway following neutrophil activation.


Asunto(s)
Trampas Extracelulares/efectos de los fármacos , Trampas Extracelulares/metabolismo , Ácidos Grasos/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Neutrófilos/citología , Calcio/metabolismo , ADN/metabolismo , Ácidos Grasos/química , Voluntarios Sanos , Humanos , Neutrófilos/efectos de los fármacos , Arginina Deiminasa Proteína-Tipo 4 , Desiminasas de la Arginina Proteica/metabolismo
18.
Am J Physiol Regul Integr Comp Physiol ; 314(4): R623-R628, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29364701

RESUMEN

The newly described hypothalamic peptide, phoenixin, is produced in the hypothalamus and adenohypophysis, where it acts to control reproductive hormone secretion. Both phoenixin and its receptor GPR173 are expressed in the hypothalamic supraoptic (SON) and paraventricular (PVN) nuclei, suggesting additional, nonreproductive effects of the peptide to control vasopressin (AVP) or oxytocin (OT) secretion. Hypothalamo-neurohypophysial explants released AVP but not OT in response to phoenixin. Intracerebroventricular administration of phoenixin into conscious, unrestrained male and female rats significantly increased circulating AVP, but not OT, levels in plasma, and it increased immediate early gene expression in the supraoptic nuclei of male rats. Bath application of phoenixin in hypothalamic slice preparations resulted in depolarization of PVN neurons, indicating a direct, neural action of phoenixin in the hypothalamus. Our results suggest that the newly described, hypothalamic peptide phoenixin, in addition to its effects on hypothalamic and pituitary mechanisms controlling reproduction, may contribute to the physiological mechanisms regulating fluid and electrolyte homeostasis.


Asunto(s)
Arginina Vasopresina/metabolismo , Hormonas Hipotalámicas/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Hormonas Peptídicas/fisiología , Animales , Arginina Vasopresina/sangre , Femenino , Regulación de la Expresión Génica , Genes fos , Hormonas Hipotalámicas/administración & dosificación , Hormonas Hipotalámicas/fisiología , Sistema Hipotálamo-Hipofisario/metabolismo , Técnicas In Vitro , Inyecciones Intraventriculares , Masculino , Potenciales de la Membrana , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas Sprague-Dawley , Vías Secretoras/efectos de los fármacos , Núcleo Supraóptico/efectos de los fármacos , Núcleo Supraóptico/metabolismo
19.
Development ; 144(17): 3080-3094, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28760814

RESUMEN

The formation of the proper number of nephrons requires a tightly regulated balance between renal progenitor cell self-renewal and differentiation. The molecular pathways that regulate the transition from renal progenitor to renal vesicle are not well understood. Here, we show that Sall1interacts with the nucleosome remodeling and deacetylase complex (NuRD) to inhibit premature differentiation of nephron progenitor cells. Disruption of Sall1-NuRD in vivo in knock-in mice (ΔSRM) resulted in accelerated differentiation of nephron progenitors and bilateral renal hypoplasia. Transcriptional profiling of mutant kidneys revealed a striking pattern in which genes of the glomerular and proximal tubule lineages were either unchanged or upregulated, and those in the loop of Henle and distal tubule lineages were downregulated. These global changes in gene expression were accompanied by a significant decrease in THP-, NKCC2- and AQP1-positive loop of Henle nephron segments in mutant ΔSRM kidneys. These findings highlight an important function of Sall1-NuRD interaction in the regulation of Six2-positive multipotent renal progenitor cells and formation of the loop of Henle.


Asunto(s)
Asa de la Nefrona/embriología , Asa de la Nefrona/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Células Madre Multipotentes/citología , Organogénesis , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Biomarcadores/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Proliferación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Ontología de Genes , Homocigoto , Túbulos Renales/metabolismo , Asa de la Nefrona/anomalías , Ratones , Células Madre Multipotentes/metabolismo , Mutación/genética , Organogénesis/genética , Unión Proteica/genética , Factores de Transcripción/química , Uréter/embriología , Uréter/metabolismo
20.
Biofabrication ; 9(2): 025019, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28516893

RESUMEN

Electrohydrodynamic spraying (EHS) has recently gained popularity for microencapsulation of cells for applications in cell delivery and tissue engineering. Some of the polymers compatible with EHS are alginate, chitosan, and other similar natural polymers, which are subject to ionotropic or physical gelation. It is desirable to further extend the use of the EHS technique beyond such polymers for wider biofabrication applications. Here, building upon our previous work of making PEG microspheres via EHS, we utilized the principles of EHS to fabricate cell-laden polyethylene glycol (PEG) hydrogel microspheres. The gelation of PEG hydrogel microspheres was achieved by forming covalent crosslinks between multiarm PEG acrylate and dithiol crosslinkers via Michael-type addition. We conducted a detailed investigation of the critical parameters of EHS, such as the applied voltage, inner needle diameter (i.d. needle), and flow rate, to obtain PEG microspheres with high cell viability and tightly-controlled diameters in the range of 70-300 µm. The polydispersity of cell-laden PEG hydrogel microspheres as measured by % coefficient of variation was between 6% and 23% for all conditions tested. We established that our method was compatible with different cell types and that all tested cell types could be encapsulated at high densities of 106-109 and ≥90% encapsulation efficiency. We observed cell aggregation within the hydrogel microspheres at applied voltage >5 kV. Since PEG is a synthetic polymer devoid of cell attachment sites, we could overcome this limitation by tethering Arg-Gly-Asp-Ser (RGDS) peptide to the PEG hydrogel microspheres; upon RGDS tethering, we observed uniform cell dispersion. The microencapsulated cells could be cultured in the PEG hydrogel microspheres of different sizes for up to one week without significant loss in cell viability. In conclusion, the EHS technique developed here could be used to generate cell-laden PEG hydrogel microspheres of controlled sizes for potential applications in cell delivery and organoid cultures.


Asunto(s)
Células Inmovilizadas/citología , Electroquímica/métodos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Microesferas , Polietilenglicoles/química , Animales , Recuento de Células , Línea Celular , Supervivencia Celular , Humanos , Microscopía Electrónica de Rastreo , Tamaño de la Partícula , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA