Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Small ; 20(4): e2306270, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37702136

RESUMEN

Persistent and uncontrolled inflammation is the root cause of various debilitating diseases. Given that interleukin-1 receptor-associated kinase 4 (IRAK4) is a critical modulator of inflammation, inhibition of its activity with selective drug molecules (IRAK4 inhibitors) represents a promising therapeutic strategy for inflammatory disorders. To exploit the full potential of this treatment approach, drug carriers for efficient delivery of IRAK4 inhibitors to inflamed tissues are essential. Herein, the first nanoparticle-based platform for the targeted systemic delivery of a clinically tested IRAK4 inhibitor, PF-06650833, with limited aqueous solubility (57 µg mL-1 ) is presented. The developed nanocarriers increase the intrinsic aqueous dispersibility of this IRAK4 inhibitor by 40 times. A targeting peptide on the surface of nanocarriers significantly enhances their accumulation after intravenous injection in inflamed tissues of mice with induced paw edema and ulcerative colitis when compared to non-targeted counterparts. The delivered IRAK4 inhibitor markedly abates inflammation and dramatically suppresses paw edema, mitigates colitis symptoms, and reduces proinflammatory cytokine levels in the affected tissues. Importantly, repeated injections of IRAK4 inhibitor-loaded nanocarriers have no acute toxic effect on major organs of mice. Therefore, the developed nanocarriers have the potential to significantly improve the therapeutic efficacy of IRAK4 inhibitors for different inflammatory diseases.


Asunto(s)
Colitis , Quinasas Asociadas a Receptores de Interleucina-1 , Ratones , Animales , Quinasas Asociadas a Receptores de Interleucina-1/química , Citocinas , Inflamación/tratamiento farmacológico , Edema
2.
Small ; 18(44): e2204436, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36098251

RESUMEN

This study presents the first messenger RNA (mRNA) therapy for metastatic ovarian cancer and cachexia-induced muscle wasting based on lipid nanoparticles that deliver follistatin (FST) mRNA predominantly to cancer clusters following intraperitoneal administration. The secreted FST protein, endogenously synthesized from delivered mRNA, efficiently reduces elevated activin A levels associated with aggressive ovarian cancer and associated cachexia. By altering the cancer cell phenotype, mRNA treatment prevents malignant ascites, delays cancer progression, induces the formation of solid tumors, and preserves muscle mass in cancer-bearing mice by inhibiting negative regulators of muscle mass. Finally, mRNA therapy provides synergistic effects in combination with cisplatin, increasing the survival of mice and counteracting muscle atrophy induced by chemotherapy and cancer-associated cachexia. The treated mice develop few nonadherent tumors that are easily resected from the peritoneum. Clinically, this nanomedicine-based mRNA therapy can facilitate complete cytoreduction, target resistance, improve resilience during aggressive chemotherapy, and improve survival in advanced ovarian cancer.


Asunto(s)
Nanopartículas , Neoplasias Ováricas , Humanos , Femenino , Caquexia/tratamiento farmacológico , Caquexia/metabolismo , Folistatina/metabolismo , Folistatina/farmacología , Folistatina/uso terapéutico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Neoplasias Ováricas/complicaciones , Neoplasias Ováricas/terapia , Músculo Esquelético/metabolismo
3.
Brain Behav Immun ; 97: 102-118, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34245812

RESUMEN

Lipocalin 2 (LCN2) is a pleiotropic molecule that is induced in the central nervous system (CNS) in several acute and chronic pathologies. The acute induction of LCN2 evolved as a beneficial process, aimed at combating bacterial infection through the sequestration of iron from pathogens, while the role of LCN2 during chronic, non-infectious disease remains unclear, and recent studies suggest that LCN2 is neurotoxic. However, whether LCN2 is sufficient to induce behavioral and cognitive alterations remains unclear. In this paper, we sought to address the role of cerebral LCN2 on cognition in both acute and chronic settings. We demonstrate that LCN2 is robustly induced in the CNS during both acute and chronic inflammatory conditions, including LPS-based sepsis and cancer cachexia. In vivo, LPS challenge results in a global induction of LCN2 in the central nervous system, while cancer cachexia results in a distribution specific to the vasculature. Similar to these in vivo observations, in vitro modeling demonstrated that both glia and cerebral endothelium produce and secrete LCN2 when challenged with LPS, while only cerebral endothelium secrete LCN2 when challenged with cancer-conditioned medium. Chronic, but not short-term, cerebral LCN2 exposure resulted in reduced hippocampal neuron staining intensity, an increase in newborn neurons, microglial activation, and increased CNS immune cell infiltration, while gene set analyses suggested these effects were mediated through melanocortin-4 receptor independent mechanisms. RNA sequencing analyses of primary hippocampal neurons revealed a distinct transcriptome associated with prolonged LCN2 exposure, and ontology analysis was suggestive of altered neurite growth and abnormal spatial learning. Indeed, LCN2-treated hippocampal neurons display blunted neurite processes, and mice exposed to prolonged cerebral LCN2 levels experienced a reduction in spatial reference memory as indicated by Y-maze assessment. These findings implicate LCN2 as a pathologic mediator of cognitive decline in the setting of chronic disease.


Asunto(s)
Disfunción Cognitiva , Neuronas , Animales , Hipocampo/metabolismo , Lipocalina 2 , Ratones , Neuroglía/metabolismo , Neuronas/metabolismo
4.
Glia ; 68(7): 1479-1494, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32039522

RESUMEN

Microglia in the mediobasal hypothalamus (MBH) respond to inflammatory stimuli and metabolic perturbations to mediate body composition. This concept is well studied in the context of high fat diet induced obesity (HFDO), yet has not been investigated in the context of cachexia, a devastating metabolic syndrome characterized by anorexia, fatigue, and muscle catabolism. We show that microglia accumulate specifically in the MBH early in pancreatic ductal adenocarcinoma (PDAC)-associated cachexia and assume an activated morphology. Furthermore, we observe astrogliosis in the MBH and hippocampus concurrent with cachexia initiation. We next show that circulating immune cells resembling macrophages infiltrate the MBH. PDAC-derived factors induced microglia to express a transcriptional profile in vitro that was distinct from that induced by lipopolysaccharide (LPS). Microglia depletion through CSF1-R antagonism resulted in accelerated cachexia onset and increased anorexia, fatigue, and muscle catabolism during PDAC. This corresponded with increased hypothalamic-pituitary-adrenal (HPA) axis activation. CSF1-R antagonism had little effect on inflammatory response in the circulation, liver, or tumor. These findings demonstrate that microglia are protective against PDAC cachexia and provide mechanistic insight into this function.


Asunto(s)
Caquexia/metabolismo , Hipotálamo/metabolismo , Microglía/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Caquexia/inmunología , Metabolismo Energético/fisiología , Gliosis/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Ratones , Obesidad/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas
5.
Brain Behav Immun ; 82: 338-353, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31499172

RESUMEN

Toll-like receptors 7 and 8 (TLR7 and TLR8) are endosomal pattern recognition receptors that detect a variety of single-stranded RNA species. While TLR7/8 agonists have robust therapeutic potential, clinical utility of these agents is limited by sickness responses associated with treatment induction. To understand the kinetics and mechanism of these responses, we characterized the acute and chronic effects of TLR7 stimulation. Single-cell RNA-sequencing studies, RNAscope, and radiolabeled in situ hybridization demonstrate that central nervous system gene expression of TLR7 is exclusive to microglia. In vitro studies demonstrate that microglia are highly sensitive to TLR7 stimulation, and respond in a dose-dependent manner to the imidazoquinoline R848. In vivo, both intraperitoneal (IP) and intracerebroventricular (ICV) R848 induce acute sickness responses including hypophagia, weight loss, and decreased voluntary locomotor activity, associated with increased CNS pro-inflammatory gene expression and changes to glial morphology. However, chronic daily IP R848 resulted in rapid tachyphylaxis of behavioral and molecular manifestations of illness. In microglial in vitro assays, pro-inflammatory transcriptional responses rapidly diminished in the context of repeated R848. In addition to TLR7 desensitization, we found that microglia become partially refractory to lipopolysaccharide (LPS) following R848 pretreatment, associated with induction of negative regulators A20 and Irak3. Similarly, mice pre-treated with R848 demonstrate reduced sickness responses, hypothalamic inflammation, and hepatic inflammation in response to LPS. These data combined demonstrate that TLR7 stimulation induces acute behavioral and molecular evidence of sickness responses. Following prolonged dosing, R848 induces a refractory state to both TLR7 and TLR4 activation, consistent with induced immune tolerance.


Asunto(s)
Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/inmunología , Microglía/inmunología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/inmunología , Animales , Conducta Animal , Células Cultivadas , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/inmunología , Citocinas/inmunología , Femenino , Imidazoles/farmacología , Tolerancia Inmunológica/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Lipopolisacáridos/farmacología , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Taquifilaxis/inmunología , Receptor Toll-Like 7/genética , Receptor Toll-Like 8/agonistas , Receptor Toll-Like 8/genética , Receptor Toll-Like 8/inmunología
6.
Brain Behav Immun ; 73: 364-374, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29852290

RESUMEN

Hypothalamic inflammation is a key component of acute sickness behavior and cachexia, yet mechanisms of inflammatory signaling in the central nervous system remain unclear. Previous work from our lab and others showed that while MyD88 is an important inflammatory signaling pathway for sickness behavior, MyD88 knockout (MyD88KO) mice still experience sickness behavior after inflammatory stimuli challenge. We found that after systemic lipopolysaccharide (LPS) challenge, MyD88KO mice showed elevated expression of several cytokine and chemokine genes in the hypothalamus. We therefore assessed the role of an additional inflammatory signaling pathway, TRIF, in acute inflammation (LPS challenge) and in a chronic inflammatory state (cancer cachexia). TRIFKO mice resisted anorexia and weight loss after peripheral (intraperitoneal, IP) or central (intracerebroventricular, ICV) LPS challenge and in a model of pancreatic cancer cachexia. Compared to WT mice, TRIFKO mice showed attenuated upregulation of Il6, Ccl2, Ccl5, Cxcl1, Cxcl2, and Cxcl10 in the hypothalamus after IP LPS treatment, as well as attenuated microglial activation and neutrophil infiltration into the brain after ICV LPS treatment. Lastly, we found that TRIF was required for Ccl2 upregulation in the hypothalamus and induction of the catabolic genes, Mafbx, Murf1, and Foxo1 in gastrocnemius during pancreatic cancer. In summary, our results show that TRIF is an important inflammatory signaling mediator of sickness behavior and cachexia and presents a novel therapeutic target for these conditions.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/fisiología , Caquexia/fisiopatología , Conducta de Enfermedad/efectos de los fármacos , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Encéfalo/metabolismo , Citocinas/metabolismo , Femenino , Hipotálamo/metabolismo , Conducta de Enfermedad/fisiología , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Neoplasias/metabolismo , Transducción de Señal/efectos de los fármacos
7.
J Neuroinflammation ; 14(1): 133, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28668091

RESUMEN

BACKGROUND: During acute infections and chronic illnesses, the pro-inflammatory cytokine interleukin-1ß (IL-1ß) acts within the brain to elicit metabolic derangements and sickness behaviors. It is unknown which cells in the brain are the proximal targets for IL-1ß with respect to the generation of these illness responses. We performed a series of in vitro experiments to (1) investigate which brain cell populations exhibit inflammatory responses to IL-1ß and (2) examine the interactions between different IL-1ß-responsive cell types in various co-culture combinations. METHODS: We treated primary cultures of murine brain microvessel endothelial cells (BMEC), astrocytes, and microglia with PBS or IL-1ß, and then performed qPCR to measure inflammatory gene expression or immunocytochemistry to evaluate nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation. To evaluate whether astrocytes and/or BMEC propagate inflammatory signals to microglia, we exposed microglia to astrocyte-conditioned media and co-cultured endothelial cells and glia in transwells. Treatment groups were compared by Student's t tests or by ANOVA followed by Bonferroni-corrected t tests. RESULTS: IL-1ß increased inflammatory gene expression and NF-κB activation in primary murine-mixed glia, enriched astrocyte, and BMEC cultures. Although IL-1ß elicited minimal changes in inflammatory gene expression and did not induce the nuclear translocation of NF-κB in isolated microglia, these cells were more robustly activated by IL-1ß when co-cultured with astrocytes and/or BMEC. We observed a polarized endothelial response to IL-1ß, because the application of IL-1ß to the abluminal endothelial surface produced a more complex microglial inflammatory response than that which occurred following luminal IL-1ß exposure. CONCLUSIONS: Inflammatory signals are detected, amplified, and propagated through the CNS via a sequential and reverberating signaling cascade involving communication between brain endothelial cells and glia. We propose that the brain's innate immune response differs depending upon which side of the blood-brain barrier the inflammatory stimulus arises, thus allowing the brain to respond differently to central vs. peripheral inflammatory insults.


Asunto(s)
Encéfalo/metabolismo , Células Endoteliales/metabolismo , Interleucina-1beta/farmacología , Neuroglía/metabolismo , Transducción de Señal/fisiología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/efectos de los fármacos , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microvasos/efectos de los fármacos , Microvasos/metabolismo , Neuroglía/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
8.
FASEB J ; 27(9): 3572-82, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23733748

RESUMEN

Cachexia is a wasting condition defined by skeletal muscle atrophy in the setting of systemic inflammation. To explore the site at which inflammatory mediators act to produce atrophy in vivo, we utilized mice with a conditional deletion of the inflammatory adaptor protein myeloid differentiation factor 88 (MyD88). Although whole-body MyD88-knockout (wbMyD88KO) mice resist skeletal muscle atrophy in response to LPS, muscle-specific deletion of MyD88 is not protective. Furthermore, selective reexpression of MyD88 in the muscle of wbMyD88KO mice via electroporation fails to restore atrophy gene induction by LPS. To evaluate the role of glucocorticoids as the inflammation-induced mediator of atrophy in vivo, we generated mice with targeted deletion of the glucocorticoid receptor in muscle (mGRKO mice). Muscle-specific deletion of the glucocorticoid receptor affords a 71% protection against LPS-induced atrophy compared to control animals. Furthermore, mGRKO mice exhibit 77% less skeletal muscle atrophy than control animals in response to tumor growth. These data demonstrate that glucocorticoids are a major determinant of inflammation-induced atrophy in vivo and play a critical role in the pathogenesis of endotoxemic and cancer cachexia.


Asunto(s)
Caquexia/etiología , Caquexia/metabolismo , Carcinoma Pulmonar de Lewis/fisiopatología , Glucocorticoides/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Animales , Western Blotting , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Inmunohistoquímica , Hibridación in Situ , Mediadores de Inflamación/farmacología , Lipopolisacáridos/farmacología , Ratones , Ratones Noqueados , Músculo Esquelético/efectos de los fármacos , Atrofia Muscular/inducido químicamente , Atrofia Muscular/genética , Factor 88 de Diferenciación Mieloide/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
9.
ACS Nano ; 18(36): 24842-24859, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39186628

RESUMEN

mRNA therapeutics encapsulated in lipid nanoparticles (LNPs) offer promising avenues for treating various diseases. While mRNA vaccines anticipate immunogenicity, the associated reactogenicity of mRNA-loaded LNPs poses significant challenges, especially in protein replacement therapies requiring multiple administrations, leading to adverse effects and suboptimal therapeutic outcomes. Historically, research has primarily focused on the reactogenicity of mRNA cargo, leaving the role of LNPs understudied in this context. Adjuvanticity and pro-inflammatory characteristics of LNPs, originating at least in part from ionizable lipids, may induce inflammation, activate toll-like receptors (TLRs), and impact mRNA translation. Knowledge gaps remain in understanding LNP-induced TLR activation and its impact on induction of animal sickness behavior. We hypothesized that ionizable lipids in LNPs, structurally resembling lipid A from lipopolysaccharide, could activate TLR4 signaling via MyD88 and TRIF adaptors, thereby propagating LNP-associated reactogenicity. Our comprehensive investigation utilizing gene ablation studies and pharmacological receptor manipulation proves that TLR4 activation by LNPs triggers distinct physiologically meaningful responses in mice. We show that TLR4 and MyD88 are essential for reactogenic signal initiation, pro-inflammatory gene expression, and physiological outcomes like food intake and body weight─robust metrics of sickness behavior in mice. The application of the TLR4 inhibitor TAK-242 effectively reduces the reactogenicity associated with LNPs by mitigating TLR4-driven inflammatory responses. Our findings elucidate the critical role of the TLR4-MyD88 axis in LNP-induced reactogenicity, providing a mechanistic framework for developing safer mRNA therapeutics and offering a strategy to mitigate adverse effects through targeted inhibition of this pathway.


Asunto(s)
Conducta de Enfermedad , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide , Nanopartículas , Receptor Toll-Like 4 , Animales , Nanopartículas/química , Ratones , Receptor Toll-Like 4/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Conducta de Enfermedad/efectos de los fármacos , Lípidos/química , Transducción de Señal/efectos de los fármacos , Masculino , Liposomas
10.
J Neurosci ; 31(31): 11376-86, 2011 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-21813697

RESUMEN

In response to illness, animals subvert normal homeostasis and divert their energy utilization to fight infection. An important and unexplored feature of this response is the suppression of physical activity and foraging behavior in the setting of negative energy balance. Inflammatory signaling in the hypothalamus mediates the febrile and anorectic responses to disease, but the mechanism by which locomotor activity (LMA) is suppressed has not been described. Lateral hypothalamic orexin (Ox) neurons link energy status with LMA, and deficiencies in Ox signaling lead to hypoactivity and hypophagia. In the present work, we examine the effect of endotoxin-induced inflammation on Ox neuron biology and LMA in rats. Our results demonstrate a vital role for diminished Ox signaling in mediating inflammation-induced lethargy. This work defines a specific population of inflammation-sensitive, arousal-associated Ox neurons and identifies a proximal neural target for inflammatory signaling to Ox neurons, while eliminating several others.


Asunto(s)
Inflamación/complicaciones , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Letargia/tratamiento farmacológico , Letargia/etiología , Neuronas/metabolismo , Neuropéptidos/metabolismo , Análisis de Varianza , Animales , Adaptación a la Oscuridad/efectos de los fármacos , Modelos Animales de Enfermedad , Vías de Administración de Medicamentos , Ensayo de Inmunoadsorción Enzimática/métodos , Privación de Alimentos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Inflamación/inducido químicamente , Inyecciones Intraventriculares/métodos , Interleucina-1beta/farmacología , Interleucina-6/sangre , Péptidos y Proteínas de Señalización Intracelular/farmacología , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/fisiología , Letargia/patología , Masculino , Hormonas Estimuladoras de los Melanocitos/farmacología , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Inhibidor NF-kappaB alfa , Trasplante de Neoplasias/métodos , Neuronas/efectos de los fármacos , Neuropéptidos/farmacología , Neurotensina/genética , Orexinas , Fotoperiodo , Polisacáridos/efectos adversos , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Receptores de Corticotropina/antagonistas & inhibidores , Receptores de Interleucina-11/genética , Receptores de Interleucina-11/metabolismo , Receptores OSM-LIF/genética , Receptores OSM-LIF/metabolismo
11.
J Biol Chem ; 286(47): 40771-81, 2011 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-21984834

RESUMEN

The melanocortin-3 receptor (MC3R) gene is pleiotropic, influencing body composition, natriuresis, immune function, and entrainment of circadian rhythms to nutrient intake. MC3Rs are expressed in hypothalamic and limbic regions of the brain and in peripheral tissues. To investigate the roles of central MC3Rs, we inserted a "lox-stop-lox" (LoxTB) 5' of the translation initiation codon of the mouse Mc3r gene and reactivated transcription using neuron-specific Cre transgenic mice. As predicted based on earlier observations of Mc3r knock-out mice, Mc3r(TB/TB) mice displayed reduced lean mass, increased fat mass, and accelerated diet-induced obesity. Surprisingly, rescuing Mc3r expression in the nervous system using the Nestin-Cre transgene only partially rescued obesity in chow-fed conditions and had no impact on the accelerated diet-induced obesity phenotype. The ventromedial hypothalamus (VMH), a critical node in the neural networks regulating feeding-related behaviors and metabolic homeostasis, exhibits dense Mc3r expression relative to other brain regions. To target VMH MC3R expression, we used the steroidogenic factor-1 Cre transgenic mouse. Although restoring VMH MC3R signaling also had a modest impact on obesity, marked improvements in metabolic homeostasis were observed. VMH MC3R signaling was not sufficient to rescue the lean mass phenotype or the regulation of behaviors anticipating food anticipation. These results suggest that actions of MC3Rs impacting on energy homeostasis involve both central and peripheral sites of action. The impact of central MC3Rs on behavior and metabolism involves divergent pathways; VMH MC3R signaling improves metabolic homeostasis but does not significantly impact on the expression of behaviors anticipating nutrient availability.


Asunto(s)
Membrana Celular/metabolismo , Metabolismo Energético/genética , Homeostasis/genética , Receptor de Melanocortina Tipo 3/genética , Receptor de Melanocortina Tipo 3/metabolismo , Alelos , Animales , Encéfalo/citología , Encéfalo/metabolismo , Línea Celular , Codón/genética , Femenino , Técnicas de Inactivación de Genes , Sitios Genéticos/genética , Genotipo , Masculino , Metaboloma/genética , Ratones , Ratones Transgénicos , Obesidad/genética , Fenotipo , Receptor de Melanocortina Tipo 3/deficiencia
12.
Am J Physiol Endocrinol Metab ; 303(12): E1446-58, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23047987

RESUMEN

Animals exhibit a rapid and sustained anorexia when fed a diet that is deficient in a single indispensable amino acid (IAA). The chemosensor for IAA deficiency resides within the anterior piriform cortex (APC). Although the cellular and molecular mechanisms by which the APC detects IAA deficiency are well established, the efferent neural pathways that reduce feeding in response to an IAA-deficient diet remain to be fully characterized. In the present work, we investigated whether 1) central melanocortin signaling is involved in IAA deficiency-induced anorexia (IAADA) and 2) IAADA engages other key appetite-regulating neuronal populations in the hypothalamus. Rats and mice that consumed a valine-deficient diet (VDD) for 2-3 wk exhibited marked reductions in food intake, body weight, fat and lean body mass, body temperature, and white adipose tissue leptin gene expression, as well as a paradoxical increase in brown adipose tissue uncoupling protein-1 mRNA. Animals consuming the VDD had altered hypothalamic gene expression, typical of starvation. Pharmacological and genetic blockade of central melanocortin signaling failed to increase long-term food intake in this model. Chronic IAA deficiency was associated with a marked upregulation of corticotropin-releasing hormone expression in the lateral hypothalamus, particularly in the parasubthalamic nucleus, an area heavily innervated by efferent projections from the APC. Our observations indicate that the hypothalamic melanocortin system plays a minor role in acute, but not chronic, IAADA and suggest that the restraint on feeding is analogous to that observed after chronic dehydration.


Asunto(s)
Anorexia/etiología , Anorexia/metabolismo , Hipotálamo/metabolismo , Vías Nerviosas/metabolismo , Neuronas/metabolismo , Transducción de Señal , Valina/deficiencia , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Anorexia/patología , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Femenino , Regulación de la Expresión Génica , Hipotálamo/patología , Canales Iónicos/genética , Canales Iónicos/metabolismo , Leptina/genética , Leptina/metabolismo , Masculino , Melanocortinas/metabolismo , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Vías Nerviosas/patología , Neuronas/patología , Especificidad de Órganos , Ratas , Ratas Sprague-Dawley , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Proteína Desacopladora 1 , Valina/metabolismo
13.
Mol Metab ; 58: 101441, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35031523

RESUMEN

OBJECTIVE: Cancer cachexia is a devastating chronic condition characterized by involuntary weight loss, muscle wasting, abnormal fat metabolism, anorexia, and fatigue. However, the molecular mechanisms underlying this syndrome remain poorly understood. In particular, the hypothalamus may play a central role in cachexia, given that it has direct access to peripheral signals because of its anatomical location and attenuated blood-brain barrier. Furthermore, this region has a critical role in regulating appetite and metabolism. METHODS: To provide a detailed analysis of the hypothalamic response to cachexia, we performed single-cell RNA-seq combined with RNA-seq of the medial basal hypothalamus (MBH) in a mouse model for pancreatic cancer. RESULTS: We found many cell type-specific changes, such as inflamed endothelial cells, stressed oligodendrocyes and both inflammatory and moderating microglia. Lcn2, a newly discovered hunger suppressing hormone, was the highest induced gene. Interestingly, cerebral treatment with LCN2 not only induced many of the observed molecular changes in cachexia but also affected gene expression in food-intake decreasing POMC neurons. In addition, we found that many of the cachexia-induced molecular changes found in the hypothalamus mimic those at the primary tumor site. CONCLUSION: Our data reveal that multiple cell types in the MBH are affected by tumor-derived factors or host factors that are induced by tumor growth, leading to a marked change in the microenvironment of neurons critical for behavioral, metabolic, and neuroendocrine outputs dysregulated during cachexia. The mechanistic insights provided in this study explain many of the clinical features of cachexia and will be useful for future therapeutic development.


Asunto(s)
Caquexia , Neoplasias Pancreáticas , Animales , Caquexia/metabolismo , Células Endoteliales/metabolismo , Redes Reguladoras de Genes , Hipotálamo/metabolismo , Ratones , Neoplasias Pancreáticas/complicaciones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Análisis de Secuencia de ARN , Microambiente Tumoral , Neoplasias Pancreáticas
14.
J Cachexia Sarcopenia Muscle ; 12(5): 1312-1332, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34231343

RESUMEN

BACKGROUND: Cancer cachexia is a metabolic disorder characterized by the progressive loss of fat and lean mass that results in significant wasting, ultimately leading to reduced quality of life and increased mortality. Effective therapies for cachexia are lacking, potentially owing to the mismatch in clinically relevant models of cachexia. Specifically, cachexia observed in a clinical setting is commonly associated with advanced or late-stage cancers that are metastatic, yet pre-clinical metastatic models of cachexia are limited. Furthermore, the prevalence of cachexia in head and neck cancer patients is high, yet few pre-clinical models of head and neck cancer cachexia exist. In addition to these shortcomings, cachexia is also heterogeneous among any given cancer, whereas patients with similar disease burden may experience significantly different degrees of cachexia symptoms. In order to address these issues, we characterize a metastatic model of human papilloma virus (HPV) positive head and neck squamous cell carcinoma that recapitulates the cardinal clinical and molecular features of cancer cachexia. METHODS: Male and female C57BL/6 mice were implanted subcutaneously with oropharyngeal squamous cell carcinoma cells stably transformed with HPV16 E6 and E7 together with hRas and luciferase (mEERL) that metastasizes to the lungs (MLM). We then robustly characterize the physiologic, behavioural, and molecular signatures during tumour development in two MLM subclones. RESULTS: Mice injected with MLM tumour cells rapidly developed primary tumours and eventual metastatic lesions to the lungs. MLM3, but not MLM5, engrafted mice progressively lost fat and lean mass during tumour development despite the absence of anorexia (P < 0.05). Behaviourally, MLM3-implanted mice displayed decreased locomotor behaviours and impaired nest building (P < 0.05). Muscle catabolism programmes associated with cachexia, including E3 ubiquitin ligase and autophagy up-regulation, along with progressive adipose wasting and accompanying browning gene signatures, were observed. Tumour progression also corresponded with hypothalamic and peripheral organ inflammation, as well as an elevation in neutrophil-to-lymphocyte ratio (P < 0.05). Finally, we characterize the fat and lean mass sparing effects of voluntary wheel running on MLM3 cachexia (P < 0.05). CONCLUSIONS: This syngeneic MLM3 allograft model of metastatic cancer cachexia is reliable, consistent, and readily recapitulates key clinical and molecular features and heterogeneity of cancer cachexia. Because few metastatic models of cachexia exist-even though cachexia often accompanies metastatic progression-we believe this model more accurately captures cancer cachexia observed in a clinical setting and thus is well suited for future mechanistic studies and pre-clinical therapy development for this crippling metabolic disorder.


Asunto(s)
Caquexia , Neoplasias de Cabeza y Cuello , Animales , Caquexia/etiología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora , Calidad de Vida
15.
Nat Commun ; 12(1): 2057, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33824339

RESUMEN

Lipocalin 2 (LCN2) was recently identified as an endogenous ligand of the type 4 melanocortin receptor (MC4R), a critical regulator of appetite. However, it remains unknown if this molecule influences appetite during cancer cachexia, a devastating clinical entity characterized by decreased nutrition and progressive wasting. We demonstrate that LCN2 is robustly upregulated in murine models of pancreatic cancer, its expression is associated with reduced food consumption, and Lcn2 deletion is protective from cachexia-anorexia. Consistent with LCN2's proposed MC4R-dependent role in cancer-induced anorexia, pharmacologic MC4R antagonism mitigates cachexia-anorexia, while restoration of Lcn2 expression in the bone marrow is sufficient in restoring the anorexia feature of cachexia. Finally, we observe that LCN2 levels correlate with fat and lean mass wasting and is associated with increased mortality in patients with pancreatic cancer. Taken together, these findings implicate LCN2 as a pathologic mediator of appetite suppression during pancreatic cancer cachexia.


Asunto(s)
Apetito , Caquexia/complicaciones , Lipocalina 2/metabolismo , Neoplasias Pancreáticas/complicaciones , Adulto , Anciano , Anciano de 80 o más Años , Animales , Anorexia/sangre , Anorexia/complicaciones , Barrera Hematoencefálica/patología , Médula Ósea/patología , Caquexia/sangre , Línea Celular Tumoral , Modelos Animales de Enfermedad , Conducta Alimentaria , Femenino , Eliminación de Gen , Humanos , Lipocalina 2/sangre , Masculino , Ratones Noqueados , Persona de Mediana Edad , Modelos Biológicos , Músculos/patología , Neutrófilos/patología , Tamaño de los Órganos , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/genética , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/metabolismo , Regulación hacia Arriba
16.
Elife ; 92020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32391790

RESUMEN

Weight loss and anorexia are common symptoms in cancer patients that occur prior to initiation of cancer therapy. Inflammation in the brain is a driver of these symptoms, yet cellular sources of neuroinflammation during malignancy are unknown. In a mouse model of pancreatic ductal adenocarcinoma (PDAC), we observed early and robust myeloid cell infiltration into the brain. Infiltrating immune cells were predominately neutrophils, which accumulated at a unique central nervous system entry portal called the velum interpositum, where they expressed CCR2. Pharmacologic CCR2 blockade and genetic deletion of Ccr2 both resulted in significantly decreased brain-infiltrating myeloid cells as well as attenuated cachexia during PDAC. Lastly, intracerebroventricular blockade of the purinergic receptor P2RX7 during PDAC abolished immune cell recruitment to the brain and attenuated anorexia. Our data demonstrate a novel function for the CCR2/CCL2 axis in recruiting neutrophils to the brain, which drives anorexia and muscle catabolism.


Weight loss, decreased appetite and fatigue are symptoms of a wasting disorder known as cachexia, which is common in several serious diseases such as AIDS, chronic lung disease and heart failure. Up to 80 percent of people with advanced cancer also develop cachexia, and there are no effective treatments. It is not known how cachexia develops, but symptoms like appetite loss and fatigue are controlled by the brain. One theory is that the brain may be responding to a malfunctioning immune response that causes inflammation. While the brain was thought to be protected from this, new research has shown that it is possible for cells from the immune system to reach the brain in some conditions. To find out if this also happens in cancer, Burfeind et al. studied mice that had been implanted with pancreatic cancer cells and were showing signs of cachexia. Samples from the mice's brains showed that immune cells known as neutrophils were present and active. A protein known as CCR2 was found in higher levels in the brains of these mice. This protein is involved in the movement of neutrophil cells through the body. To see what effect this protein had, Burfeind et al. gave the mice a drug that blocks CCR2. This prevented the neutrophils from entering the brain and reduced the symptoms of cachexia in the mice. To further confirm the role of CCR2, the mice were genetically modified so that they could not produce the protein. This reduced the number of neutrophils seen in the brain but not in the rest of the body. This suggests that a drug targeting CCR2 could help to reduce the symptoms of cachexia, without disrupting the normal immune response away from the brain. This approach would still need to be tested in clinical trials before it is possible to know how effective it might be in humans.


Asunto(s)
Encéfalo/fisiopatología , Caquexia/etiología , Carcinoma Ductal Pancreático/patología , Células Mieloides/metabolismo , Neoplasias Pancreáticas/patología , Animales , Anorexia/etiología , Carcinoma Ductal Pancreático/complicaciones , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Femenino , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/inmunología , Infiltración Neutrófila , Neutrófilos/metabolismo , Neoplasias Pancreáticas/complicaciones , Receptores CCR2/genética , Receptores CCR2/metabolismo , Pérdida de Peso
17.
Peptides ; 30(2): 210-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19028534

RESUMEN

Inflammation-associated cachexia is associated with multiple chronic diseases and involves activation of appetite regulating centers in the arcuate nucleus of the hypothalamus (ARH). The nucleus of the solitary tract (NTS) in the brainstem has also been implicated as an important nucleus involved in appetite regulation. We set out to determine whether the NTS may be involved in inflammation-associated anorexia by injecting IL-1 beta into the 4th ventricle and assessing food intake and NTS neuronal activation. Injection of IL-1 beta produced a decrease in food intake at 3 and 12h after injection which was ameliorated at the 12h time point by a sub-threshold dose of agouti-related peptide (AgRP). Investigation into neuron types in the NTS revealed that IL-1 beta injection was associated with an increase in c-Fos activity in NTS neurons expressing tyrosine hydroxylase (TH). Additionally, injection of IL-1 beta into the 4th ventricle did not produce c-Fos activation of neurons expressing pro-opiomelanocortin (POMC) in the ARH, cells known to be involved in producing anorexia in response to systemic inflammation. Double-label in situ hybridization revealed that TH neurons did not express IL-1 receptor I (IL1-RI) transcript, demonstrating that c-Fos activation of TH neurons in this setting was not via direct stimulation of IL-1 beta on TH neurons themselves. We conclude that IL-1 beta injection into the 4th ventricle produces anorexia and is accompanied by an increase in activation in TH neurons in the NTS. This provides evidence that the brainstem may be an important mediator of anorexia in the setting of inflammation.


Asunto(s)
Proteína Relacionada con Agouti/farmacología , Anorexia/inducido químicamente , Interleucina-1beta/administración & dosificación , Neuronas/enzimología , Núcleo Solitario/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Animales , Anorexia/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Tronco Encefálico/metabolismo , Hipotálamo/metabolismo , Inflamación/metabolismo , Inyecciones Intraventriculares , Interleucina-1beta/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Ratas , Ratas Sprague-Dawley , Núcleo Solitario/citología
19.
Nat Commun ; 10(1): 4682, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31615993

RESUMEN

A priority in cancer research is to innovate therapies that are not only effective against tumor progression but also address comorbidities such as cachexia that limit quality and quantity of life. We demonstrate that TLR7/8 agonist R848 induces anti-tumor responses and attenuates cachexia in murine models of pancreatic ductal adenocarcinoma (PDAC). In vivo, tumors from two of three cell lines were R848-sensitive, resulting in smaller tumor mass, increased immune complexity, increased CD8+ T-cell infiltration and activity, and decreased Treg frequency. R848-treated mice demonstrated improvements in behavioral and molecular cachexia manifestations, resulting in a near-doubling of survival duration. Knockout mouse studies revealed that stromal, not neoplastic, TLR7 is requisite for R848-mediated responses. In patient samples, we found Tlr7 is ubiquitously expressed in stroma across all stages of pancreatic neoplasia, but epithelial Tlr7 expression is relatively uncommon. These studies indicate immune-enhancing approaches including R848 may be useful in PDAC and cancer-associated cachexia.


Asunto(s)
Caquexia , Carcinoma Ductal Pancreático/metabolismo , Imidazoles/farmacología , Neoplasias Intraductales Pancreáticas/metabolismo , Neoplasias Pancreáticas/metabolismo , Microambiente Tumoral/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Ingestión de Alimentos/efectos de los fármacos , Expresión Génica , Humanos , Locomoción/efectos de los fármacos , Ratones , Ratones Noqueados , Neoplasias Intraductales Pancreáticas/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Análisis de Secuencia de ARN , Tasa de Supervivencia , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/agonistas , Carga Tumoral , Microambiente Tumoral/inmunología
20.
Endocrinology ; 149(10): 4837-45, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18583425

RESUMEN

Agouti-related protein (AgRP) is an orexigenic neuropeptide produced by neurons in the hypothalamic arcuate nucleus (ARC) that is a key component of central neural circuits that control food intake and energy expenditure. Disorders in energy homeostasis, characterized by hypophagia and increased metabolic rate, frequently develop in animals with either acute or chronic diseases. Recently, studies have demonstrated that proopiomelanocortin-expressing neurons in the ARC are activated by the proinflammatory cytokine IL-1beta. In the current study, we sought to determine whether inflammatory processes regulate the expression of AgRP mRNA and to characterize the response of AgRP neurons to IL-1beta. Here, we show by real-time RT-PCR and in situ hybridization analysis that AgRP mRNA expression in rodents is increased in models of acute and chronic inflammation. AgRP neurons were found to express the type I IL-1 receptor, and the percentage of expression was significantly increased after peripheral administration of lipopolysaccharide. Furthermore, we demonstrate that IL-1beta inhibits the release of AgRP from hypothalamic explants. Collectively, these data indicate that proinflammatory signals decrease the secretion of AgRP while increasing the transcription of the AgRP gene. These observations suggest that AgRP neurons may participate with ARC proopiomelanocortin neurons in mediating the anorexic and metabolic responses to acute and chronic disease processes.


Asunto(s)
Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Núcleo Arqueado del Hipotálamo/fisiología , Inflamación/fisiopatología , Transcripción Genética/inmunología , Animales , Antiinflamatorios no Esteroideos/farmacología , Núcleo Arqueado del Hipotálamo/citología , Trasplante de Tejido Encefálico , Enfermedad Crónica , Modelos Animales de Enfermedad , Inflamación/inducido químicamente , Inflamación/inmunología , Interleucina-1beta/metabolismo , Ketorolaco/farmacología , Fallo Renal Crónico/inmunología , Fallo Renal Crónico/fisiopatología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología , Neoplasias/fisiopatología , Neuronas/fisiología , Prostaglandinas/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , ARN Mensajero/genética , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Receptores de Interleucina-1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA