RESUMEN
The mammalian target of rapamycin (mTOR) pathway plays a key role in determining immune cells function through modulation of their metabolic status. By specific deletion of Rictor in CD11c+ myeloid cells (referred to here as CD11cRicΔ/Δ), we investigated the role of mTOR complex 2 (mTORC2) signaling in dendritic cells (DCs) function in mice. We showed that upon dextran sulfate sodium-induced colitis, the lack of mTORC2 signaling CD11c+ cells diminishes the colitis score and abrogates DC migration to the mesenteric lymph nodes, thereby diminishing the infiltration of T helper 17 cells in the lamina propria and subsequent inflammation. These findings corroborate with the abrogation of cytoskeleton organization and the decreased activation of Rac1 and Cdc42 GTPases observed in CD11c+-mTORC2-deficient cells. Meta-analysis on colonic samples from ulcerative colitis patients revealed increased gene expression of proinflammatory cytokines, which coincided with augmented expression of the mTOR pathway, a positive correlation between the DC marker ITGAX and interleukin-6, the expression of RICTOR, and CDC42. Together, this work proposes that targeting mTORC2 on DCs offers a key to hamper inflammatory responses, and this way, ameliorates the progression and severity of intestinal inflammatory diseases.
Asunto(s)
Movimiento Celular , Colitis , Células Dendríticas , Sulfato de Dextran , Diana Mecanicista del Complejo 2 de la Rapamicina , Células Mieloides , Transducción de Señal , Animales , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Colitis/patología , Colitis/inducido químicamente , Colitis/inmunología , Células Mieloides/metabolismo , Células Mieloides/inmunología , Sulfato de Dextran/toxicidad , Proteína Asociada al mTOR Insensible a la Rapamicina/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina/genética , Antígeno CD11c/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Humanos , Proteína de Unión al GTP rac1/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Ratones Noqueados , Neuropéptidos , Antígenos CD11RESUMEN
Little is known about the effects of high-fat diet (HFD)-induced obesity on resident colonic lamina propria (LP) macrophages (LPMs) function and metabolism. Here, we report that obesity and diabetes resulted in increased macrophage infiltration in the colon. These macrophages exhibited the residency phenotype CX3CR1hiMHCIIhi and were CD4-TIM4-. During HFD, resident colonic LPM exhibited a lipid metabolism gene expression signature that overlapped that used to define lipid-associated macrophages (LAMs). Via single-cell RNA sequencing, we identified a sub-cluster of macrophages, increased in HFD, that were responsible for the LAM signature. Compared to other macrophages in the colon, these cells were characterized by elevated glycolysis, phagocytosis, and efferocytosis signatures. CX3CR1hiMHCIIhi colonic resident LPMs had fewer lipid droplets (LDs) and decreased triacylglycerol (TG) content compared to equivalent cells in lean mice and exhibited increased phagocytic capacity, suggesting that HFD induces adaptive responses in LPMs to limit bacterial translocation.
RESUMEN
Obesity-induced insulin resistance (OIR) has been associated with an increased prevalence of neurodegenerative disorders such as multiple sclerosis. Obesity results in increased blood-brain barrier (BBB) permeability, specifically in the hypothalamic regions associated with the control of caloric intake. In obesity, the chronic state of low-grade inflammation has been implicated in several chronic autoimmune inflammatory disorders. However, the mechanisms that connect the inflammatory profile of obesity with the severity of experimental autoimmune encephalomyelitis (EAE) are poorly defined. In this study, we show that obese mice are more susceptible to EAE, presenting a worse clinical score with more severe pathological changes in the spinal cord when compared with control mice. Analysis of immune infiltrates at the peak of the disease shows that high-fat diet (HFD)- and control (chow)-fed groups do not present any difference in innate or adaptive immune cell compartments, indicating the increased severity occurs prior to disease onset. In the setting of worsening EAE in HFD-fed mice, we observed spinal cord lesions in myelinated regions and (blood brain barrier) BBB disruption. We also found higher levels of pro-inflammatory monocytes, macrophages, and IFN-γ+CD4+ T cells in the HFD-fed group compared to chow-fed animals. Altogether, our results indicate that OIR promotes BBB disruption, allowing the infiltration of monocytes/macrophages and activation of resident microglia, ultimately promoting CNS inflammation and exacerbation of EAE.
Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Ratones , Animales , Esclerosis Múltiple/patología , Barrera Hematoencefálica/patología , Inflamación/patología , Permeabilidad , Obesidad/complicaciones , Ratones Endogámicos C57BLRESUMEN
Invariant natural killer T (iNKT) cells are a distinct population of lymphocytes characterized by their reactivity to glycolipids presented by CD1d. iNKT cells are found throughout the body, and little is known about their tissue-specific metabolic regulation. Here, we show that splenic and hepatic iNKT cells are metabolically comparable and rely on glycolytic metabolism to support their activation. Deletion of the pyruvate kinase M2 (Pkm2) gene in splenic and hepatic iNKT cells impairs their response to specific stimulation and their ability to mitigate acute liver injury. In contrast, adipose tissue (AT) iNKT cells exhibit a distinctive immunometabolic profile, with AMP-activated protein kinase (AMPK) being necessary for their function. AMPK deficiency impairs AT-iNKT physiology, blocking their capacity to maintain AT homeostasis and their ability to regulate AT inflammation during obesity. Our work deepens our understanding on the tissue-specific immunometabolic regulation of iNKT cells, which directly impacts the course of liver injury and obesity-induced inflammation.
Asunto(s)
Proteínas Quinasas Activadas por AMP , Células T Asesinas Naturales , Inflamación , Hígado , Metaboloma , Obesidad , Animales , RatonesRESUMEN
Obesity is a major concern for global health care systems. Systemic low-grade inflammation in obesity is a major risk factor for insulin resistance. Leptin is an adipokine secreted by the adipose tissue that functions by controlling food intake, leading to satiety. Leptin levels are increased in obesity. Here, we show that leptin enhances the effects of LPS in macrophages, intensifying the production of cytokines, glycolytic rates, and morphological and functional changes in the mitochondria through an mTORC2-dependent, mTORC1-independent mechanism. Leptin also boosts the effects of IL-4 in macrophages, leading to increased oxygen consumption, expression of macrophage markers associated with a tissue repair phenotype, and wound healing. In vivo, hyperleptinemia caused by diet-induced obesity increases the inflammatory response by macrophages. Deletion of leptin receptor and subsequently of leptin signaling in myeloid cells (ObR-/-) is sufficient to improve insulin resistance in obese mice and decrease systemic inflammation. Our results indicate that leptin acts as a systemic nutritional checkpoint to regulate macrophage fitness and contributes to obesity-induced inflammation and insulin resistance. Thus, specific interventions aimed at downstream modulators of leptin signaling may represent new therapeutic targets to treat obesity-induced systemic inflammation.
Asunto(s)
Resistencia a la Insulina , Leptina , Tejido Adiposo/metabolismo , Animales , Inflamación/metabolismo , Leptina/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismoRESUMEN
Natural killer T (NKT) cells are an innate-like T cell subset that recognize lipid antigens presented by CD1d-expressing antigen presenting cells (APCs), such as dendritic cells, macrophages, and B cells. They can be subdivided into two different subsets according to the variation in αß TCR chains: type I and type II NKT cells. Type I, also called invariant NKT cells (iNKT), express restricted TCRs with an invariant α-chain (Vα24-Jα18 in humans and Vα14-Jα18 in mice) and limited ß-chains. Here we have established a protocol in which iNKT cells are isolated from a donor wild-type mouse and transferred into iNKT KO (Jα18-/-) mouse. Below we will explore the methods for cell sorting of splenic iNKTs, iNKT cells transfer, and detection of transferred cells into the liver using flow cytometry technique.
Asunto(s)
Células T Asesinas Naturales , Animales , Células Presentadoras de Antígenos , Antígenos CD1d , Citometría de Flujo , Ratones , Receptores de Antígenos de Linfocitos T , Subgrupos de Linfocitos TRESUMEN
The understanding of how different cell types adapt their metabolism in the face of challenges has been attracting the attention of researchers for many years. Recently, immunologists also started to focus on how the metabolism of immune cells can impact the way that immunity drives its responses. The presence of a pathogen or damage in a tissue changes severely the way that the immune cells need to respond. When activated, immune cells usually shift their metabolism from a high energy demanding status using mitochondria respiration to a glycolytic based rapid ATP production. The diminished amount of respiration leads to changes in the mitochondrial membrane potential and, consequently, generation of reactive oxygen species. Here, we show how flow cytometry can be used to track changes in mitochondrial mass, membrane potential and superoxide (ROS) production in live immune cells. â This protocol suggests a quick way of evaluating mitochondrial fitness using flow cytometry. We propose using the probes MitoTraker Green and MitoTracker Red/ MitoSOX at the same time. This way, it is possible to evaluate different parameters of mitochondrial biology in living cells. â Flow cytometry is a highly used tool by immunologists. With the advances of studies focusing on the metabolism of immune cells, a simplified application of flow cytometry for mitochondrial studies and screenings is a helpful clarifying method for immunology.
RESUMEN
Macrophages are essential components of the immune system. Macrophages can be derived from the bone marrow of mice with either recombinant M-CSF or L929 supernatant. Recent literature considers recombinant M-CSF- and L929-derived macrophages as equals, even though L929-derived macrophages are exposed to other substances secreted in the L929 supernatant, and not only M-CSF. Thus, we decided to perform a comparative analysis of both inflammatory and metabolic profiles of macrophages differentiated under the aforementioned conditions, which is relevant for standardization and interpretation of in vitro studies. We observed that, when treated with LPS, L929macs secrete lower levels of proinflammatory cytokines (TNF-α, IL-6, IL12) and present higher glycolysis and oxygen consumption when compared with M-CSFmacs. L929macs also have increased mitochondrial mass, with higher percentage of dysfunctional mitochondria. This sort of information can help direct further studies towards a more specific approach for macrophage generation.
Asunto(s)
Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Metaboloma , Metabolómica , Animales , Biomarcadores , Línea Celular , Citocinas/metabolismo , Metabolismo Energético , Mediadores de Inflamación/metabolismo , Metabolómica/métodos , RatonesRESUMEN
BACKGROUND: The gut microbiota is a key element to support host homeostasis and the development of the immune system. The relationship between the microbiota and immunity is a 2-way road, in which the microbiota contributes to the development/function of immune cells and immunity can affect the composition of microbes. In this context, natural killer T cells (NKT cells) are distinct T lymphocytes that play a role in gut immunity and are influenced by gut microbes. In our work, we investigated the involvement of invariant NKT cells (iNKT) in intestinal homeostasis. RESULTS: We found that iNKT-deficient mice (iNKT-KO) had reduced levels of fecal IgA and an altered composition of the gut microbiota, with increased Bacteroidetes. The absence of iNKT cells also affected TGF-ß1 levels and plasma cells, which were significantly reduced in knockout (KO) mice. In addition, when submitted to dextran sodium sulfate colitis, iNKT-KO mice had worsening of colitis when compared with wild-type (WT) mice. To further address iNKT cell contribution to intestinal homeostasis, we adoptively transferred iNKT cells to KO mice, and they were submitted to colitis. Transfer of iNKT cells improved colitis and restored fecal IgA levels and gut microbiota. CONCLUSIONS: Our results indicate that intestinal NKT cells are important modulators of intestinal homeostasis and that gut microbiota composition may be a potential target in the management of inflammatory bowel diseases.
Asunto(s)
Microbioma Gastrointestinal/inmunología , Homeostasis/inmunología , Inmunoglobulina A/análisis , Intestinos/inmunología , Células T Asesinas Naturales/inmunología , Animales , Colitis/inducido químicamente , Colitis/inmunología , Colitis/microbiología , Sulfato de Dextran , Modelos Animales de Enfermedad , Heces/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones NoqueadosRESUMEN
SUMMARY OBJECTIVE Exploring the use of forecasting models and simulation tools to estimate demand and reduce the waiting time of patients in Emergency Departments (EDs). METHODS The analysis was based on data collected in May 2013 in the ED of Recanto das Emas, Federal District, Brasil, which uses a Manchester Triage System. A total of 100 consecutive patients were included: 70 yellow (70%) and 30 green (30%). Flow patterns, observed waiting time, and inter-arrival times of patients were collected. Process maps, demand, and capacity data were used to build a simulation, which was calibrated against the observed flow times. What-if analysis was conducted to reduce waiting times. RESULTS Green and yellow patient arrival-time patterns were similar, but inter-arrival times were 5 and 38 minutes, respectively. Wait-time was 14 minutes for yellow patients, and 4 hours for green patients. The physician staff comprised four doctors per shift. A simulation predicted that allocating one more doctor per shift would reduce wait-time to 2.5 hours for green patients, with a small impact in yellow patients' wait-time. Maintaining four doctors and allocating one doctor exclusively for green patients would reduce the waiting time to 1.5 hours for green patients and increase it in 15 minutes for yellow patients. The best simulation scenario employed five doctors per shift, with two doctors exclusively for green patients. CONCLUSION Waiting times can be reduced by balancing the allocation of doctors to green and yellow patients and matching the availability of doctors to forecasted demand patterns. Simulations of EDs' can be used to generate and test solutions to decrease overcrowding.
RESUMO OBJETIVO Explorar o uso de modelos de previsão e ferramentas de simulação para estimar a demanda e reduzir o tempo de espera dos pacientes em Departamentos de Emergência (DE). METODOLOGIA A análise foi baseada em dados coletados em maio de 2013, no DE do Recanto das Emas, Distrito Federal, Brasil, que utiliza o Protocolo de Manchester como sistema de triagem. Um total de 100 pacientes consecutivos foram incluídos: 70 amarelos (70%) e 30 verdes (30%). Padrões de fluxo, tempo de espera observado e tempos entre as chegadas dos pacientes foram registrados. Mapas de processo, demanda e dados de capacidade foram utilizados na construção de uma simulação que foi calibrada de acordo com o fluxo observado. Uma análise do tipo "e se..." foi conduzida para reduzir os tempos de espera. RESULTADOS Os padrões de tempo de chegada para pacientes verdes e amarelos foram semelhantes, mas os tempos entre chegadas foram 5 e 38 minutos, respectivamente. O tempo de espera foi de 14 minutos para pacientes amarelos e 4 horas para pacientes verdes. A equipe médica era composta por quatro médicos por turno. Uma simulação previu que a inclusão de mais um médico por turno reduziria o tempo de espera para 2,5 horas para pacientes verdes, com um impacto pequeno no tempo de espera dos pacientes amarelos. A manutenção de quatro médicos e a inclusão de um médico exclusivamente para pacientes verdes reduziria o tempo de espera para 1,5 horas para pacientes verdes e aumentaria em 15 minutos para os pacientes amarelos. O melhor cenário simulado utilizou cinco médicos por plantão, com dois médicos exclusivos para pacientes verdes. CONCLUSÃO Os tempos de espera podem ser reduzidos equilibrando a distribuição de médicos para pacientes verdes e amarelos e relacionando a disponibilidade dos médicos aos padrões de demanda previstos. Simulações de DE podem ser utilizadas para gerar e testar soluções para diminuir a superlotação.
Asunto(s)
Humanos , Simulación por Computador , Aglomeración , Listas de Espera , Servicio de Urgencia en Hospital/estadística & datos numéricos , Modelos Teóricos , Factores de Tiempo , Algoritmos , Brasil , Proyectos Piloto , Reproducibilidad de los Resultados , Predicción , Evaluación en Enfermería/métodosRESUMEN
Over the past years, systemic derived cues that regulate cellular metabolism have been implicated in the regulation of immune responses. Ghrelin is an orexigenic hormone produced by enteroendocrine cells in the gastric mucosa with known immunoregulatory roles. The mechanism behind the function of ghrelin in immune cells, such as macrophages, is still poorly understood. Here, we explored the hypothesis that ghrelin leads to alterations in macrophage metabolism thus modulating macrophage function. We demonstrated that ghrelin exerts an immunomodulatory effect over LPS-activated peritoneal macrophages, as evidenced by inhibition of TNF-α and IL-1ß secretion and increased IL-12 production. Concomitantly, ghrelin increased mitochondrial membrane potential and increased respiratory rate. In agreement, ghrelin prevented LPS-induced ultrastructural damage in the mitochondria. Ghrelin also blunted LPS-induced glycolysis. In LPS-activated macrophages, glucose deprivation did not affect ghrelin-induced IL-12 secretion, whereas the inhibition of pyruvate transport and mitochondria-derived ATP abolished ghrelin-induced IL-12 secretion, indicating a dependence on mitochondrial function. Ghrelin pre-treatment of metabolic activated macrophages inhibited the secretion of TNF-α and enhanced IL-12 levels. Moreover, ghrelin effects on IL-12, and not on TNF-α, are dependent on mitochondria elongation, since ghrelin did not enhance IL-12 secretion in metabolic activated mitofusin-2 deficient macrophages. Thus, ghrelin affects macrophage mitochondrial metabolism and the subsequent macrophage function.
Asunto(s)
Ghrelina/farmacología , Interleucina-12/genética , Interleucina-1beta/genética , Macrófagos Peritoneales/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética , Adenosina Trifosfato/genética , Animales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ghrelina/química , Glucólisis/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/patología , Lipopolisacáridos/toxicidad , Macrófagos Peritoneales/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Óxido Nítrico/genética , Transducción de Señal/genéticaRESUMEN
OBJECTIVE: Exploring the use of forecasting models and simulation tools to estimate demand and reduce the waiting time of patients in Emergency Departments (EDs). METHODS: The analysis was based on data collected in May 2013 in the ED of Recanto das Emas, Federal District, Brasil, which uses a Manchester Triage System. A total of 100 consecutive patients were included: 70 yellow (70%) and 30 green (30%). Flow patterns, observed waiting time, and inter-arrival times of patients were collected. Process maps, demand, and capacity data were used to build a simulation, which was calibrated against the observed flow times. What-if analysis was conducted to reduce waiting times. RESULTS: Green and yellow patient arrival-time patterns were similar, but inter-arrival times were 5 and 38 minutes, respectively. Wait-time was 14 minutes for yellow patients, and 4 hours for green patients. The physician staff comprised four doctors per shift. A simulation predicted that allocating one more doctor per shift would reduce wait-time to 2.5 hours for green patients, with a small impact in yellow patients' wait-time. Maintaining four doctors and allocating one doctor exclusively for green patients would reduce the waiting time to 1.5 hours for green patients and increase it in 15 minutes for yellow patients. The best simulation scenario employed five doctors per shift, with two doctors exclusively for green patients. CONCLUSION: Waiting times can be reduced by balancing the allocation of doctors to green and yellow patients and matching the availability of doctors to forecasted demand patterns. Simulations of EDs' can be used to generate and test solutions to decrease overcrowding.
Asunto(s)
Simulación por Computador , Aglomeración , Servicio de Urgencia en Hospital/estadística & datos numéricos , Modelos Teóricos , Listas de Espera , Algoritmos , Brasil , Predicción , Humanos , Evaluación en Enfermería/métodos , Proyectos Piloto , Reproducibilidad de los Resultados , Factores de TiempoRESUMEN
Inflammatory bowel diseases (IBDs) affect millions of people worldwide and their frequencies in developed countries have increased since the twentieth century. In this context, there is an intensive search for therapies that modulate inflammation and provide tissue regeneration in IBDs. Recently, the immunomodulatory activity of adipose tissue-derived mesenchymal stromal cells (ADMSCs) has been demonstrated to play an important role on several immune cells in different conditions of inflammatory and autoimmune diseases. In this study, we explored the immunomodulatory potential of ADMSC in a classical model of DSS-induced colitis. First, we found that treatment of mice with ADMSC ameliorated the severity of DSS-induced colitis, reducing colitis pathological score and preventing colon shortening. Moreover, a prominent reduction of pro-inflammatory cytokines levels (i.e., IFN-γ, TNF-α, IL-6 and MCP-1) was observed in the colon of animals treated with ADMSC. We also observed a significant reduction in the frequencies of macrophages (F4/80+CD11b+) and dendritic cells (CD11c+CD103+) in the intestinal lamina propria of ADMSC-treated mice. Finally, we detected the up-regulation of immunoregulatory-associated molecules in intestine of mice treated with ADMSCs (i.e., elevated arginase-1 and IL-10). Thus, this present study demonstrated that ADMSC modulates the overall gut inflammation (cell activation and recruitment) in experimental colitis, providing support to the further development of new strategies in the treatment of intestinal diseases.
Asunto(s)
Colitis/metabolismo , Colitis/patología , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Inflamación/patología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Colon/metabolismo , Colon/patología , Citocinas/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/patología , Modelos Animales de Enfermedad , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BLRESUMEN
In the past decade, several reports have appointed the importance of mitochondria in the immune response. Our understanding of mitochondria evolved from a simple supplier of energy into a platform necessary for immunorregulation. Proinflammatory responses are associated with enhanced glycolytic activity and breakdown of the TCA cycle. Mitochondrial reactive species of oxygen (mROS) are key regulators of classically activated macrophages, with substantial impact in the anti-microbicidal activity and pro-inflammatory cytokine secretion of macrophages. The inflammasome activation in macrophages is dependent on mROS production and mitochondrial regulation and mitochondrial dynamics and functionality direct impact inflammatory responses. Alternative activated macrophage metabolism relies on fatty acid oxidation, and the mechanism responsible for this phenotype is not fully elucidated. Thus, cellular metabolism and mitochondria function is a key immunoregulatory feature of macrophage biology. In this review, we will provide insights into recently reported evidences of mitochondria-related metabolic nodes, which are important for macrophage physiology.
Asunto(s)
Inflamación/patología , Macrófagos/inmunología , Mitocondrias/metabolismo , Animales , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamasomas/metabolismo , Inflamación/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Dinámicas Mitocondriales , NADP/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Succinato Deshidrogenasa/metabolismoRESUMEN
Pulmonary fibrosis is a result of an abnormal wound healing in lung tissue triggered by an excessive accumulation of extracellular matrix proteins, loss of tissue elasticity, and debit of ventilatory function. NKT cells are a major source of Th1 and Th2 cytokines and may be crucial in the polarization of M1/M2 macrophages in pulmonary fibrogenesis. Although there appears to be constant scientific progress in that field, pulmonary fibrosis still exhibits no current cure. From these facts, we hypothesized that NKT cells could influence the development of pulmonary fibrosis via modulation of macrophage activation. Wild type (WT) and NKT type I cell-deficient mice (Jα18-/-) were subjected to the protocol of bleomycin-induced pulmonary fibrosis with or without treatment with NKT cell agonists α-galactosylceramide and sulfatide. The participation of different cell populations, collagen deposition, and protein levels of different cytokines involved in inflammation and fibrosis was evaluated. The results indicate a benign role of NKT cells in Jα18-/- mice and in wild-type α-galactosylceramide-sulfatide-treated groups. These animals presented lower levels of collagen deposition, fibrogenic molecules such as TGF-ß and vimentin and improved survival rates. In contrast, WT mice developed a Th2-driven response augmenting IL-4, 5, and 13 protein synthesis and increased collagen deposition. Furthermore, the arginase-1 metabolic pathway was downregulated in wild-type NKT-activated and knockout mice indicating lower activity of M2 macrophages in lung tissue. Hence, our data suggest that NKT cells play a protective role in this experimental model by down modulating the Th2 milieu, inhibiting M2 polarization and finally preventing fibrosis.
Asunto(s)
Bleomicina/farmacología , Macrófagos/fisiología , Células T Asesinas Naturales/fisiología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/fisiopatología , Animales , Colágeno/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Galactosilceramidas/farmacología , Inflamación/metabolismo , Pulmón/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/efectos de los fármacos , Células T Asesinas Naturales/metabolismo , Fenotipo , Fibrosis Pulmonar/metabolismo , Células TH1/metabolismo , Células Th2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Vimentina/metabolismoRESUMEN
Natural killer T (NKT) cells are a subset of lymphocytes that reacts to glycolipids presented by CD1d. Invariant NKT cells (iNKT) correspond to >90% of the total population of NKTs and reacts to α-galactosylceramide (αGalCer). αGalCer promotes a complex mixture of Th1 and Th2 cytokines, as interferon (IFN)-γ and interleukin (IL)-4. NKT cells and IFN-γ are known to participate in some models of renal diseases, but further studies are still necessary to elucidate their mechanisms. The aim of our study was to analyze the participation of iNKT cells in an experimental model of tubule-interstitial nephritis. We used 8-wk-old C57BL/6j, Jα18KO and IFN-γKO mice. They were fed a 0.25% adenine diet for 10 d. Both adenine-fed wild-type (WT) and Jα18KO mice exhibited renal dysfunction, but adenine-fed Jα18KO mice presented higher expression of kidney injury molecule-1 (KIM-1), tumor necrosis factor (TNF)-α and type I collagen. To analyze the role of activated iNKT cells in our model, we administered αGalCer in WT mice during adenine ingestion. After αGalCer injection, we observed a significant reduction in serum creatinine, proinflammatory cytokines and renal fibrosis. However, this improvement in renal function was not observed in IFN-γKO mice after αGalCer treatment and adenine feeding, illustrating that this cytokine plays a role in our model. Our findings may suggest that IFN-γ production is one of the factors contributing to improved renal function after αGalCer administration.
Asunto(s)
Galactosilceramidas/administración & dosificación , Interferón gamma/genética , Nefritis/tratamiento farmacológico , Insuficiencia Renal/tratamiento farmacológico , Adenina/toxicidad , Animales , Antígenos CD1d/biosíntesis , Antígenos CD1d/genética , Colágeno Tipo I/biosíntesis , Receptor Celular 1 del Virus de la Hepatitis A , Humanos , Interleucina-4/biosíntesis , Interleucina-4/genética , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Células T Asesinas Naturales/efectos de los fármacos , Células T Asesinas Naturales/inmunología , Nefritis/inducido químicamente , Nefritis/genética , Nefritis/patología , Insuficiencia Renal/inducido químicamente , Insuficiencia Renal/genética , Insuficiencia Renal/patología , Factor de Necrosis Tumoral alfa/biosíntesisRESUMEN
Obesity is an important worldwide challenge that must be faced in most developed and developing countries because of unhealthy nutritional habits. The consequences of obesity and being overweight are observed in different organs, but the kidney is one of the most affected. Excess adipose tissue causes hemodynamic alterations in the kidney that can result in renal disease. However, obesity is also commonly associated with other comorbidities such as chronic inflammation, hypertension and diabetes. This association of several aggravating factors is still a matter of concern in clinical and basic research because the pathophysiologic mechanisms surrounding chronic kidney disease development in obese patients remain unclear. This review will discuss the consequences of obesity in the context of renal injury.
RESUMEN
Focal and segmental glomerulosclerosis (FSGS) is one of the most important renal diseases related to end-stage renal failure. Bradykinin has been implicated in the pathogenesis of renal inflammation, whereas the role of its receptor 2 (B2RBK; also known as BDKRB2) in FSGS has not been studied. FSGS was induced in wild-type and B2RBK-knockout mice by a single intravenous injection of Adriamycin (ADM). In order to further modulate the kinin receptors, the animals were also treated with the B2RBK antagonist HOE-140 and the B1RBK antagonist DALBK. Here, we show that the blockage of B2RBK with HOE-140 protects mice from the development of FSGS, including podocyte foot process effacement and the re-establishment of slit-diaphragm-related proteins. However, B2RBK-knockout mice were not protected from FSGS. These opposite results were due to B1RBK expression. B1RBK was upregulated after the injection of ADM and this upregulation was exacerbated in B2RBK-knockout animals. Furthermore, treatment with HOE-140 downregulated the B1RBK receptor. The blockage of B1RBK in B2RBK-knockout animals promoted FSGS regression, with a less-inflammatory phenotype. These results indicate a deleterious role of both kinin receptors in an FSGS model and suggest a possible cross-talk between them in the progression of disease.
Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/patología , Receptores de Bradiquinina/fisiología , Animales , Bradiquinina/análogos & derivados , Bradiquinina/farmacología , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Ratones , Ratones Noqueados , Receptores de Bradiquinina/efectos de los fármacos , Receptores de Bradiquinina/genéticaRESUMEN
Platelet-activating factor (PAF) is a lipid mediator with important pro-inflammatory effects, being synthesized by several cell types including kidney cells. Although there is evidence of its involvement in acute renal dysfunction, its role in progressive kidney injury is not completely known. In the present study, we investigated the role of PAF receptor (PAFR) in an experimental model of chronic renal disease. Wild-type (WT) and PAFR knockout (KO) mice underwent unilateral ureter obstruction (UUO), and at kill time, urine and kidney tissue was collected. PAFR KO animals compared with WT mice present: (a) less renal dysfunction, evaluated by urine protein/creatinine ratio; (b) less fibrosis evaluated by collagen deposition, type I collagen, Lysyl Oxidase-1 (LOX-1) and transforming growth factor ß (TGF-ß) gene expression, and higher expression of bone morphogenetic protein 7 (BMP-7) (3.3-fold lower TGF-ß/BMP-7 ratio); (c) downregulation of extracellular matrix (ECM) and adhesion molecule-related machinery genes; and (d) lower levels of pro-inflammatory cytokines. These indicate that PAFR engagement by PAF or PAF-like molecules generated during UUO potentiates renal dysfunction and fibrosis and might promote epithelial-to-mesenchymal transition (EMT). Also, early blockade of PAFR after UUO leads to a protective effect, with less fibrosis deposition. In conclusion, PAFR signaling contributes to a pro-inflammatory environment in the model of obstructive nephropathy, favoring the fibrotic process, which lately will generate renal dysfunction and progressive organ failure.
Asunto(s)
Riñón/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Insuficiencia Renal Crónica/metabolismo , Animales , Azepinas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Riñón/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Nefritis/metabolismo , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Insuficiencia Renal Crónica/patología , Triazoles , Obstrucción UreteralRESUMEN
The aim of this study was to investigate the role of TLR2, TLR4 and MyD88 in sepsis-induced AKI. C57BL/6 TLR2(-/-), TLR4(-/-) and MyD88(-/-) male mice were subjected to sepsis by cecal ligation and puncture (CLP). Twenty four hours later, kidney tissue and blood samples were collected for analysis. The TLR2(-/-), TLR4(-/-) and MyD88(-/-) mice that were subjected to CLP had preserved renal morphology, and fewer areas of hypoxia and apoptosis compared with the wild-type C57BL/6 mice (WT). MyD88(-/-) mice were completely protected compared with the WT mice. We also observed reduced expression of proinflammatory cytokines in the kidneys of the knockout mice compared with those of the WT mice and subsequent inhibition of increased vascular permeability in the kidneys of the knockout mice. The WT mice had increased GR1(+low) cells migration compared with the knockout mice and decreased in GR1(+high) cells migration into the peritoneal cavity. The TLR2(-/-), TLR4(-/-), and MyD88(-/-) mice had lower neutrophil infiltration in the kidneys. Depletion of neutrophils in the WT mice led to protection of renal function and less inflammation in the kidneys of these mice. Innate immunity participates in polymicrobial sepsis-induced AKI, mainly through the MyD88 pathway, by leading to an increased migration of neutrophils to the kidney, increased production of proinflammatory cytokines, vascular permeability, hypoxia and apoptosis of tubular cells.