Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 343
Filtrar
1.
Life Sci ; 344: 122583, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38508232

RESUMEN

AIMS: Formyl peptide receptor 1 (FPR1), from a G-protein coupled receptor family, was previously well-characterized in immune cells. But the function of FPR1 in osteogenesis and fracture healing was rarely reported. This study, using the FPR1 knockout (KO) mouse, is one of the first studies that try to investigate FPR1 function to osteogenic differentiation of bone marrow-derived stem cells (BMSCs) in vitro and bone fracture healing in vivo. MATERIALS AND METHODS: Primary BMSCs were isolated from both FPR1 KO and wild type (WT) mice. Cloned mouse BMSCs (D1 cells) were used to examine role of FoxO1 in FPR1 regulation of osteogenesis. A closed, transverse fracture at the femoral midshaft was created to compare bone healing between KO and WT mice. Biomechanical and structural properties of femur were compared between healthy WT and KO mice. KEY FINDINGS: FPR1 expression increased significantly during osteogenesis of both primary and cloned BMSCs. Compared to BMSCs from FPR1 KO mice, WT BMSCs displayed considerably higher levels of osteogenic markers as well as mineralization. Osteogenesis by D1 cells was inhibited by either an FPR1 antagonist cFLFLF or a specific inhibitor of FoxO1, AS1842856. In addition, the femur from WT mice had better biomechanical properties than FPR1 KO mice. Furthermore, bone healing in WT mice was remarkably improved compared to FPR1 KO mice analyzed by X-ray and micro-CT. SIGNIFICANCE: These findings indicated that FPR1 played a vital role in osteogenic differentiation and regenerative capacity of fractured bone, probably through the activation of FoxO1 related signaling pathways.


Asunto(s)
Osteogénesis , Receptores de Formil Péptido , Ratones , Animales , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Ratones Noqueados , Curación de Fractura , Fémur/metabolismo , Diferenciación Celular , Células de la Médula Ósea
2.
Curr Med Sci ; 44(1): 187-194, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38300426

RESUMEN

OBJECTIVE: Premature rupture of membranes (PROM) is a common pregnancy disorder that is closely associated with structural weakening of fetal membranes. Studies have found that formyl peptide receptor 1 (FPR1) activates inflammatory pathways and amniotic epithelialmesenchymal transition (EMT), stimulates collagen degradation, and leads to membrane weakening and membrane rupture. The purpose of this study was to investigate the anti-inflammatory and EMT inhibitory effects of FPR1 antagonist (BOC-MLF) to provide a basis for clinical prevention of PROM. METHODS: The relationship between PROM, FPR1, and EMT was analyzed in human fetal membrane tissue and plasma samples using Western blotting, PCR, Masson staining, and ELISA assays. Lipopolysaccharide (LPS) was used to establish a fetal membrane inflammation model in pregnant rats, and BOC-MLF was used to treat the LPS rat model. We detected interleukin (IL)-6 in blood from the rat hearts to determine whether the inflammatory model was successful and whether the anti-inflammatory treatment was effective. We used electron microscopy to analyze the structure and collagen expression of rat fetal membrane. RESULTS: Western blotting, PCR and Masson staining indicated that the expression of FPR1 was significantly increased, the expression of collagen was decreased, and EMT appeared in PROM. The rat model indicated that LPS caused the collapse of fetal membrane epithelial cells, increased intercellular gaps, and decreased collagen. BOC-MLF promoted an increase in fetal membrane collagen, inhibited EMT, and reduced the weakening of fetal membranes. CONCLUSION: The expression of FPR1 in the fetal membrane of PROM was significantly increased, and EMT of the amniotic membrane was obvious. BOC-MLF can treat inflammation and inhibit amniotic EMT.


Asunto(s)
Amnios , Lipopolisacáridos , Embarazo , Femenino , Humanos , Animales , Ratas , Amnios/metabolismo , Lipopolisacáridos/farmacología , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Colágeno/metabolismo , Antiinflamatorios , Transición Epitelial-Mesenquimal
3.
Genome Biol Evol ; 15(10)2023 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-37776517

RESUMEN

The detection of invasive pathogens is critical for host immune defense. Cell surface receptors play a key role in the recognition of diverse microbe-associated molecules, triggering leukocyte recruitment, phagocytosis, release of antimicrobial compounds, and cytokine production. The intense evolutionary forces acting on innate immune receptor genes have contributed to their rapid diversification across plants and animals. However, the functional consequences of immune receptor divergence are often unclear. Formyl peptide receptors (FPRs) comprise a family of animal G protein-coupled receptors which are activated in response to a variety of ligands including formylated bacterial peptides, pathogen virulence factors, and host-derived antimicrobial peptides. FPR activation in turn promotes inflammatory signaling and leukocyte migration to sites of infection. Here we investigate patterns of gene loss, diversification, and ligand recognition among FPRs in primates and carnivores. We find that FPR1, which plays a critical role in innate immune defense in humans, has been lost in New World primates. Amino acid variation in FPR1 and FPR2 among primates and carnivores is consistent with a history of repeated positive selection acting on extracellular domains involved in ligand recognition. To assess the consequences of FPR divergence on bacterial ligand interactions, we measured binding between primate FPRs and the FPR agonist Staphylococcus aureus enterotoxin B, as well as S. aureus FLIPr-like, an FPR inhibitor. We found that few rapidly evolving sites in primate FPRs are sufficient to modulate recognition of bacterial proteins, demonstrating how natural selection may serve to tune FPR activation in response to diverse microbial ligands.


Asunto(s)
Receptores de Formil Péptido , Staphylococcus aureus , Humanos , Animales , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Secuencia de Aminoácidos , Ligandos , Staphylococcus aureus/genética , Bacterias/genética , Bacterias/metabolismo , Receptores Inmunológicos , Primates/metabolismo
4.
World J Gastroenterol ; 29(24): 3793-3806, 2023 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-37426322

RESUMEN

BACKGROUND: Formyl peptide receptor 2 (Fpr2) is an important receptor in host resistance to bacterial infections. In previous studies, we found that the liver of Fpr2-/- mice is the most severely damaged target organ in bloodstream infections, although the reason for this is unclear. AIM: To investigate the role of Fpr2 in liver homeostasis and host resistance to bacterial infections. METHODS: Transcriptome sequencing was performed on the livers of Fpr2-/- and wild-type (WT) mice. Differentially expressed genes (DEGs) were identified in the Fpr2-/- and WT mice, and the biological functions of DEGs were analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) en-richment analysis. Quantitative real time-polymerase chain reaction (qRT-PCR) and western blot (WB) analyses were used to further validate the expression levels of differential genes. Cell counting kit-8 assay was employed to investigate cell survival. The cell cycle detection kit was used to measure the distribution of cell cycles. The Luminex assay was used to analyze cytokine levels in the liver. The serum biochemical indices and the number of neutrophils in the liver were measured, and hepatic histopathological analysis was performed. RESULTS: Compared with the WT group, 445 DEGs, including 325 upregulated genes and 120 downregulated genes, were identified in the liver of Fpr2-/- mice. The enrichment analysis using GO and KEGG showed that these DEGs were mainly related to cell cycle. The qRT-PCR analysis confirmed that several key genes (CycA, CycB1, Cdc20, Cdc25c, and Cdk1) involved in the cell cycle had significant changes. The WB analysis confirmed a decrease in the expression of CDK1 protein. WRW4 (an antagonist of Fpr2) could inhibit the proliferation of HepG2 cells in a concentration dependent manner, with an increase in the number of cells in the G0/G1 phase, and a decrease in the number of cells in the S phase. Serum alanine aminotransferase levels increased in Fpr2-/- mice. The Luminex assay measurements showed that interleukin (IL)-10 and chemokine (C-X-C motif) ligand (CXCL)-1 levels were significantly reduced in the liver of Fpr2-/- mice. There was no difference in the number of neutrophils, serum C-reactive protein levels, and liver pathology between WT and Fpr2-/- mice. CONCLUSION: Fpr2 participates in the regulation of cell cycle and cell proliferation, and affects the expression of IL-10 and CXCL-1, thus playing an important protective role in maintaining liver homeostasis.


Asunto(s)
Receptores de Formil Péptido , Transcriptoma , Animales , Ratones , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Hígado/metabolismo , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo
5.
Oncoimmunology ; 12(1): 2237354, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37492227

RESUMEN

Formyl peptide receptor-1 (FPR1) is a pattern recognition receptor that is mostly expressed by myeloid cells. In patients with colorectal cancer (CRC), a loss-of-function polymorphism (rs867228) in the gene coding for FPR1 has been associated with reduced responses to chemotherapy or chemoradiotherapy. Moreover, rs867228 is associated with accelerated esophageal and colorectal carcinogenesis. Here, we show that dendritic cells from Fpr1-/- mice exhibit reduced migration in response to chemotherapy-treated CRC cells. Moreover, Fpr1-/- mice are particularly susceptible to chronic ulcerative colitis and colorectal oncogenesis induced by the mutagen azoxymethane followed by oral dextran sodium sulfate, a detergent that induces colitis. These experiments were performed after initial co-housing of Fpr1-/- mice and wild-type controls, precluding major Fpr1-driven differences in the microbiota. Pharmacological inhibition of Fpr1 by cyclosporin H also tended to increase intestinal oncogenesis in mice bearing the ApcMin mutation, and this effect was reversed by the anti-inflammatory drug sulindac. We conclude that defective FPR1 signaling favors intestinal tumorigenesis through the modulation of the innate inflammatory/immune response.


Asunto(s)
Colitis , Neoplasias Colorrectales , Animales , Ratones , Carcinogénesis/genética , Colitis/inducido químicamente , Colitis/genética , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/genética , Receptores de Formil Péptido/genética , Transducción de Señal
6.
J Innate Immun ; 15(1): 548-561, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37068475

RESUMEN

Using a global formyl peptide receptor (Fpr) 2 knockout mouse colony, we have reported the modulatory properties of this pro-resolving receptor in polymicrobial sepsis. Herein, we have used a humanized FPR2 (hFPR2) mouse colony, bearing an intact or a selective receptor deficiency in myeloid cells to dwell on the cellular mechanisms. hFPR2 mice and myeloid cell-specific hFPR2 KO (KO) mice were subjected to cecal ligation and puncture (CLP)-induced polymicrobial sepsis. Compared with hFPR2 mice, CLP caused exacerbated cardiac dysfunction (assessed by echocardiography), worsened clinical outcome, and impaired bacterial clearance in KO mice. This pathological scenario was paralleled by increased recruitment of pro-inflammatory monocytes and reduced M2-like macrophages within the KO hearts. In peritoneal exudates of KO mice, we quantified increased neutrophil and MHC II+ macrophage numbers but decreased monocyte/macrophage and MHC II- macrophage recruitment. hFPR2 upregulation was absent in myeloid cells, and local production of lipoxin A4 was reduced in septic KO mice. Administration of the FPR2 agonist annexin A1 (AnxA1) improved cardiac function in hFPR2 septic mice but had limited beneficial effects in KO mice, in which the FPR2 ligand failed to polarize macrophages toward an MHC II- phenotype. In conclusion, FPR2 deficiency in myeloid cells exacerbates cardiac dysfunction and worsens clinical outcome in polymicrobial sepsis. The improvement of cardiac function and the host immune response by AnxA1 is more effective in hFPR2-competent septic mice.


Asunto(s)
Cardiopatías , Sepsis , Ratones , Animales , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/agonistas , Leucocitos , Macrófagos , Sepsis/complicaciones , Ratones Noqueados , Ratones Endogámicos C57BL
7.
Biochem Pharmacol ; 212: 115524, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37001680

RESUMEN

Microglial activation-induced neuroinflammation contributes to onset and progression of sporadic and hereditary Parkinson's disease (PD). Activated microglia secrete pro-inflammatory and neurotoxic IL-1ß, IL-6 and TNF-α, which subsequently promote neurodegeneration. Formyl peptide receptor-1 (FPR1) of CNS microglia functions as pattern recognition receptor and is activated by N-formylated peptides, leading to microglial activation, induction of inflammatory responses and resulting neurotoxicity. In this study, it was hypothesized that FPR1 activation of microglia causes loss of dopaminergic neurons by activating inflammasome and upregulating IL-1ß, IL-6 or TNF-α and that FPR1 antagonist HCH6-1 exerts neuroprotective effect on dopaminergic neurons. FPR1 agonist fMLF induced activation of microglia cells by causing activation of NLRP3 inflammasome and upregulation and secretion of IL-1ß, IL-6 or TNF-α. Conditioned medium (CM) of fMLF-treated microglia cells, which contains neurotoxic IL-1ß, IL-6 and TNF-α, caused apoptotic death of differentiated SH-SY5Y dopaminergic neurons by inducing mitochondrial oxidative stress and activating pro-apoptotic signaling. FPR1 antagonist HCH6-1 prevented fMLF-induced activation of inflammasome and upregulation of pro-inflammatory cytokines in microglia cells. HCH6-1 co-treatment reversed CM of fMLF-treated microglia-induced apoptotic death of dopaminergic neurons. FPR1 antagonist HCH6-1 inhibited rotenone-induced upregulation of microglial marker Iba-1 protein level, cell death of dopaminergic neurons and motor impairment in zebrafish. HCH6-1 ameliorated rotenone-induced microglial activation, upregulation of FPR1 mRNA, activation of NLRP3 inflammasome, cell death of SN dopaminergic neurons and PD motor deficit in mice. Our results suggest that FPR1 antagonist HCH6-1 possesses anti-neuroinflammatory and neuroprotective effects on dopaminergic neurons by inhibiting microglial activation and upregulation of inflammasome activity and pro-inflammatory cytokines.


Asunto(s)
Neuroblastoma , Fármacos Neuroprotectores , Enfermedad de Parkinson , Ratones , Humanos , Animales , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Fármacos Neuroprotectores/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedades Neuroinflamatorias , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Interleucina-6/metabolismo , Rotenona/toxicidad , Rotenona/metabolismo , Pez Cebra , Modelos Animales de Enfermedad , Neuroblastoma/metabolismo , Neuronas Dopaminérgicas , Microglía , Citocinas/metabolismo
8.
Oncoimmunology ; 12(1): 2189823, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36970071

RESUMEN

Formyl peptide receptor-1 (FPR1) is a pathogen recognition receptor involved in the detection of bacteria, in the control of inflammation, as well as in cancer immunosurveillance. A single nucleotide polymorphism in FPR1, rs867228, provokes a loss-of-function phenotype. In a bioinformatic study performed on The Cancer Genome Atlas (TCGA), we observed that homo-or heterozygosity for rs867228 in FPR1 (which affects approximately one-third of the population across continents) accelerates age at diagnosis of specific carcinomas including luminal B breast cancer by 4.9 years. To validate this finding, we genotyped 215 patients with metastatic luminal B mammary carcinomas from the SNPs To Risk of Metastasis (SToRM) cohort. The first diagnosis of luminal B breast cancer occurred at an age of 49.2 years for individuals bearing the dysfunctional TT or TG alleles (n = 73) and 55.5 years for patients the functional GG alleles (n = 141), meaning that rs867228 accelerated the age of diagnosis by 6.3 years (p=0.0077, Mann & Whitney). These results confirm our original observation in an independent validation cohort. We speculate that it may be useful to include the detection of rs867228 in breast cancer screening campaigns for selectively increasing the frequency and stringency of examinations starting at a relatively young age.


Asunto(s)
Neoplasias de la Mama , Carcinoma , Receptores de Formil Péptido , Humanos , Genotipo , Fenotipo , Polimorfismo de Nucleótido Simple , Receptores de Formil Péptido/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología
9.
Ophthalmic Res ; 66(1): 681-691, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36805961

RESUMEN

INTRODUCTION: In proliferative diabetic retinopathy (PDR), retinal neovascularization is the essential pathogenic process that is linked to endothelial-to-mesenchymal transition (EndoMT) induced by high glucose (HG). This pathophysiological process may be regulated by a G-protein-coupled chemoattractant receptor FPR2 (mouse Fpr2), involved in inflammatory cell migration and proliferation. In the current study, we investigated the role of Fpr2 in regulating EndoMT and the underlying mechanisms during diabetic retinopathy progression. METHODS: FPR2 agonist or inhibitor was added to human microvascular endothelial cells (HMECs) exposed to normal glucose or HG. Morphologic, phenotypic, and functional changes of HMECs as well as the formation of microvasculature related to EndoMT were assessed. EndoMT biomarkers were detected in the retinal tissues of diabetic mice and fibrovascular epiretinal membranes (FVMs) from patients with PDR. RESULTS: HG upregulated FPR2 in HMECs, which triggered morphological changes, and the cells acquired mesenchymal phenotype, with enhanced cell migration, viability, and angiogenic process shown by tube formation and aortic ring sprouting. Inhibition of FPR2 attenuated HG-induced EndoMT and endothelial cell migration to form vessel-like tube structures. RNA sequence and protein analysis further revealed that inhibition of FPR2 decreased the expression of genes associated with EndoMT. ERK1/2 and P38 signaling pathway was activated in HMECs, promoting neovascularization in HG-induced EndoMT of HMECs. In vivo, increased expression of mesenchymal markers was detected in the retina of diabetic mice and FVMs from patients with PDR. FPR2 deficiency was associated with diminished EndoMT-related phenotypic changes in the retina of diabetic mice. CONCLUSIONS: FPR2 is actively involved in the progression of EndoMT that may contribute to the pathogenesis of PDR. Thus, FPR2 may be a potential therapeutic target for PDR.


Asunto(s)
Diabetes Mellitus Experimental , Retinopatía Diabética , Humanos , Ratones , Animales , Retinopatía Diabética/patología , Células Endoteliales/metabolismo , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Proteínas de Unión al GTP/metabolismo , Glucosa
10.
Exp Neurol ; 359: 114257, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36279933

RESUMEN

Germinal matrix hemorrhage (GMH) is one of the leading causes of morbidity and mortality in preterm infants in the United States, with little progress made in its clinical management. Blood clots disrupting normal cerebrospinal fluid circulation and absorption after germinal matrix hemorrhage are key contributors towards post-hemorrhagic hydrocephalus development. n-formyl peptide receptor 2 (FPR2), a G-protein-coupled receptor, has been associated with the activation of p-ERK1/2, which in turn promotes the transcription of the DUSP1 gene, which may play a role in CD36 signaling. CD36 scavenger, a transmembrane glycoprotein, plays an essential role in microglia phagocytic blood clot clearance after GMH. FPR2's role in blood clot clearance after hemorrhagic stroke is unknown. We hypothesize that FPR2 activation by FPR2 agonist Annexin A1 (AnxA1) will enhance hematoma resolution via the upregulation of the CD36 signaling pathway, thereby improving short- and long-term neurological outcomes. Bacterial collagenase (0.3 U) was infused intraparenchymally into the right hemispheric ganglionic eminence in P7 rat pups to induce GMH. AnxA1 and FPR2 Inhibitor (Boc2) were given at 1-h post-GMH via intranasal administration. FPR2 CRISPR was given 48-h prior to GMH induction. Short-term neurological deficits were assessed using negative geotaxis test. Hematoma volume was assessed using hemoglobin assay. Protein expression was assessed using western blots. Long-term neurocognitive deficits and motor coordination were assessed using Morris water maze, rotarod, and foot fault tests. We have demonstrated that AnxA1 treatment enhances hematoma resolution and improved short and long-term outcomes. Lastly, FPR2 agonist AnxA1 treatment resulted in the upregulation of the FPR2/p-ERK(1/2)/DUSP1/CD36 signaling pathway.


Asunto(s)
Anexina A1 , Receptores de Formil Péptido , Animales , Humanos , Recién Nacido , Ratas , Anexina A1/genética , Anexina A1/metabolismo , Antígenos CD36/genética , Hemorragia Cerebral/complicaciones , Fosfatasa 1 de Especificidad Dual/metabolismo , Hematoma , Recien Nacido Prematuro , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Transducción de Señal , Quinasas MAP Reguladas por Señal Extracelular
11.
Immun Inflamm Dis ; 10(11): e706, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36301030

RESUMEN

BACKGROUND: Focal cortical dysplasia type IIb (FCDIIb) and tuberous sclerosis complex (TSC) show persistent neuroinflammation, which promotes epileptogenesis and epilepsy progression, suggesting that endogenous resolution of inflammation is inadequate to relieve neuronal network hyperexcitability. To explore the potential roles of formyl peptide receptor 2 (FPR2), which is a key regulator of inflammation resolution, in epilepsy caused by FCDIIb and TSC, we examined the expression and cellular distribution of FPR2. METHOD: The expression of FPR2 and nuclear factor-κB (NF-κB) signaling pathway was examined by real-time PCR, western blots, and analyzed via one-way analysis of variance. The distribution of FPR2 was detected using immunostaining. The expression of resolvin D1 (RvD1, the endogenous ligand of FPR2) was observed via enzyme-linked immunosorbent assay. Pearson's correlation test was used to evaluate the correlation between the expression levels of FPR2 and RvD1 and the clinical variants. RESULTS: The expression of FPR2 was significantly lower in FCDIIb (p = .0146) and TSC (p = .0006) cortical lesions than in controls, as was the expression of RvD1 (FCDIIb: p = .00431; TSC: p = .0439). Weak FPR2 immunoreactivity was observed in dysmorphic neurons (DNs), balloon cells (BCs), and giant cells (GCs) in FCDIIb and TSC tissues. Moreover, FPR2 was mainly distributed in dysplastic neurons; it was sparse in microglia and nearly absent in astrocytes. The NF-κB pathway was significantly activated in patients with FCDIIb and TSC, and the protein level of NF-κB was negatively correlated with the protein level of FPR2 (FCDIIb: p = .00395; TSC: p = .0399). In addition, the protein level of FPR2 was negatively correlated with seizure frequency in FCDIIb (p = .0434) and TSC (p = .0351) patients. CONCLUSION: In summary, these results showed that the expression and specific distribution of FPR2 may be involved in epilepsy caused by FCDIIb and TSC, indicating that downregulation of FPR2 mediated the dysfunction of neuroinflammation resolution in FCDIIb and TSC.


Asunto(s)
Epilepsia , Malformaciones del Desarrollo Cortical , Esclerosis Tuberosa , Humanos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Epilepsia/genética , Epilepsia/metabolismo , Inflamación/patología , Malformaciones del Desarrollo Cortical/metabolismo , Malformaciones del Desarrollo Cortical/patología , FN-kappa B/metabolismo , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/complicaciones , Esclerosis Tuberosa/metabolismo
12.
FASEB J ; 36(11): e22579, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36183323

RESUMEN

Abdominal aortic aneurysm (AAA) formation is characterized by inflammation, leukocyte infiltration, and vascular remodeling. Resolvin D1 (RvD1) is derived from ω-3 polyunsaturated fatty acids and is involved in the resolution phase of chronic inflammatory diseases. The aim of this study was to decipher the protective role of RvD1 via formyl peptide receptor 2 (FPR2) receptor signaling in attenuating abdominal aortic aneurysms (AAA). The elastase-treatment model of AAA in C57BL/6 (WT) mice and human AAA tissue was used to confirm our hypotheses. Elastase-treated FPR2-/- mice had a significant increase in aortic diameter, proinflammatory cytokine production, immune cell infiltration (macrophages and neutrophils), elastic fiber disruption, and decrease in smooth muscle cell α-actin expression compared to elastase-treated WT mice. RvD1 treatment attenuated AAA formation, aortic inflammation, and vascular remodeling in WT mice, but not in FPR2-/- mice. Importantly, human AAA tissue demonstrated significantly decreased FPR2 mRNA expression compared to non-aneurysm human aortas. Mechanistically, RvD1/FPR2 signaling mitigated p47phox phosphorylation and prevented hallmarks of ferroptosis, such as lipid peroxidation and Nrf2 translocation, thereby attenuating HMGB1 secretion. Collectively, this study demonstrates RvD1-mediated immunomodulation of FPR2 signaling on macrophages to mitigate ferroptosis and HMGB1 release, leading to resolution of aortic inflammation and remodeling during AAA pathogenesis.


Asunto(s)
Aneurisma de la Aorta Abdominal , Ferroptosis , Proteína HMGB1 , Actinas/metabolismo , Animales , Aneurisma de la Aorta Abdominal/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/metabolismo , Proteína HMGB1/metabolismo , Humanos , Inflamación/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/metabolismo , Elastasa Pancreática/metabolismo , ARN Mensajero/metabolismo , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina , Remodelación Vascular
13.
J Diabetes Res ; 2022: 9881422, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36117520

RESUMEN

Abdominal obesity coupled with polygenic hereditary defects is considered the initial event in the development of metabolic syndrome (MS). The purpose of this study was to analyse the frequency with which polymorphic loci of adiponectin (ADIPOQ) and leptin (LEP) genes occur in patients with MS and the association between the symptoms of MS and these polymorphisms. DNA was isolated from the whole blood of 207 patients with MS and 100 healthy individuals (control group) using the phenol-chloroform method. Gene polymorphisms were determined using real-time polymerase chain reaction (PCR). The most common variant of the ADIPOQ (rs2241766) gene among MS patients was the GT genotype. The A allele of the LEP (rs7799039) gene was found to be the most frequent in MS patients. The highest systolic blood pressure was found in carriers of the GG genotype of the LEP (rs7799039) gene. The carriers of the ADIPOQ (rs2241766) GT genotype were associated with the highest systolic blood pressure and body mass index (BMI); carriers of the ADIPOQ (rs2241766) GG genotype were associated with the highest diastolic blood pressure, hyperglycaemia, and elevated glycated haemoglobin (HbA1c). The results of this study allowed us to establish the unique gene variants associated with the risk of developing MS in the Crimean population.


Asunto(s)
Adiponectina , Síndrome Metabólico , Receptores de Leptina , Adiponectina/genética , Cloroformo , Hemoglobina Glucada/genética , Humanos , Leptina/genética , Síndrome Metabólico/genética , Fenoles , Polimorfismo de Nucleótido Simple , Receptores de Bombesina/genética , Receptores del Factor Estimulante de Colonias/genética , Receptores de Formil Péptido/genética , Receptores de Leptina/genética , Canal Liberador de Calcio Receptor de Rianodina
14.
Int J Mol Sci ; 23(18)2022 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-36142517

RESUMEN

Formyl peptide receptor (FPR) 2 is known to play a critical role in regulating inflammation, including either the pro-inflammatory or pro-resolving effects. However, its role in neonatal hyperoxia-induced lung injury has not been delineated. In this study, we investigate whether mesenchymal stem cells (MSCs) attenuate hyperoxia-induced neonatal lung injury by regulating FPR2 activity. We observed a significant increase in FPR2 levels in alveolar macrophages (RAW264.7 cells) after H2O2-induced stress, which decreased after MSC treatment. In the H2O2-induction model, increased levels of inflammatory cytokines (IL-1α and TNF-α) were significantly reduced in RAW264.7 cells after treatment with WRW4, an inhibitor of FPR2, or MSCs. Viability of lung epithelial cells and endothelial cells was significantly improved when cultured in the conditioned media of RAW264.7 cells treated with WRW4 or MSCs, compared to when cultured in the conditioned media of control RAW265.7 cells exposed to H2O2. For the in vivo study, wild-type and FPR2 knockout (FPR2-/-) C57/BL6 mouse pups were randomly exposed to 80% oxygen or room air from postnatal day (P) 1 to P14. At P5, 2 × 105 MSCs were transplanted intratracheally. MSCs reduced the elevated FPR2 activity at P7 and improved the decreased FPR2 activity as well as the increased immuno-stained FPR2 activity in alveolar macrophages in hyperoxic lungs at P14. Both FPR2-/- and MSCs similarly attenuated impaired alveolarization and angiogenesis, and increased apoptosis and inflammation of hyperoxic lungs without synergistic effects. Our findings suggest that the protective effects of MSCs in hyperoxic lung injury might be related to indirect modulation of FPR2 activity, at least of alveolar macrophages in neonatal mice.


Asunto(s)
Displasia Broncopulmonar , Hiperoxia , Lesión Pulmonar , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Animales , Ratones , Animales Recién Nacidos , Medios de Cultivo Condicionados , Citocinas , Modelos Animales de Enfermedad , Células Endoteliales , Peróxido de Hidrógeno , Hiperoxia/complicaciones , Inflamación , Pulmón , Lesión Pulmonar/etiología , Lesión Pulmonar/terapia , Oxígeno , Receptores de Formil Péptido/genética , Factor de Necrosis Tumoral alfa
15.
Biochim Biophys Acta Mol Cell Res ; 1869(12): 119356, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36087811

RESUMEN

Formyl peptide receptors (FPRs) are expressed in the cells of the innate immune system and provide binding with pathogen and damage-associated molecular patterns with subsequent activation of the phagocytes for defense reactions such as chemotaxis, secretory degranulation and ROS generation. Probably, FPR2 is one of the unique receptors in the organism; it is able to recognize numerous ligands of different chemical structure, and moreover, these ligands can trigger opposite phagocyte responses promoting either pro- or anti-inflammatory reactions. Therefore, FPR2 and its signaling pathways are of intense research interest. We found only slight activation of ERK1/2 in the response to peptide ligand WKYMVM in the accelerating phase of ROS generation and more intense ERK1/2 phosphorylation in the declining phase of it in mouse bone marrow granulocytes. Lipid agonist BML-111 did not induce significant ERK phosphorylation when applied for 10-1800 s. To some extent co-localization of ERK1/2 and NADPH oxidase subunits was observed even in the intact cells and didn't change under FPR2 stimulation by WKYMVM, while direct PKC activation by PMA resulted to more efficient interaction between ERK1/2 and p47phox/p67phox and their translocation to plasma membrane. We have shown that phosphorylation and activation of ERK1/2 in bone marrow granulocytes depended on FPR2-triggered activity of PI3K and PKC, phosphatase DUSP6, and, the most but not the least, on ROS generation. Since blocking of ROS generation led to a slowdown of ERK activation indicating a significant contribution of ROS to the secondary regulation of ERK activity.


Asunto(s)
Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , NADPH Oxidasas , Receptores de Formil Péptido/metabolismo , Animales , Ligandos , Lípidos , Ratones , NADPH Oxidasas/metabolismo , Péptidos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Receptores de Formil Péptido/genética
16.
Mol Oncol ; 16(16): 2959-2980, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35808840

RESUMEN

Formyl peptide receptors (FPR1, FPR2 and FPR3) are innate immune sensors of pathogen and commensal bacteria and have a role in colonic mucosa homeostasis. We identified FPR1 as a tumour suppressor in gastric cancer cells due to its ability to sustain an inflammation resolution response with antiangiogenic potential. Here, we investigate whether FPR1 exerts similar functions in colorectal carcinoma (CRC) cells. Since it has been shown that the commensal bacterium Lactobacillus rhamnosus GG (LGG) can promote intestinal epithelial homeostasis through FPR1, we explored the possibility that it could induce proresolving and antiangiogenic effects in CRC cells. We demonstrated that pharmacologic inhibition or genetic deletion of FPR1 in CRC cells caused a reduction of proresolving mediators and a consequent upregulation of angiogenic factors. The activation of FPR1 mediates opposite effects. Proresolving, antiangiogenic and homeostatic functions were also observed upon treatment of CRC cells with supernatant of LGG culture, but not of other lactic acid or nonprobiotic bacteria (i.e. Bifidobacterium bifidum or Escherichia coli). These activities of LGG are dependent on FPR1 expression and on the subsequent MAPK signalling activation. Thus, the innate immune receptor FPR1 could be a regulator of the balance between microbiota, inflammation and cancer in CRC models.


Asunto(s)
Neoplasias Colorrectales , Lacticaseibacillus rhamnosus , Probióticos , Humanos , Inflamación , Lacticaseibacillus rhamnosus/metabolismo , Probióticos/farmacología , Probióticos/uso terapéutico , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo
17.
Eur J Oral Sci ; 130(4): e12883, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35808844

RESUMEN

Chronic inflammatory responses can inflict permanent damage to host tissues. Specialized pro-resolving mediators downregulate inflammation but also can have other functions. The aim of this study was to examine whether oral epithelial cells express the receptors FPR2/ALX and DRV1/GPR32, which bind RvD1n-3 DPA , a recently described pro-resolving mediator derived from omega-3 docosapentaenoic acid (DPA), and whether RvD1n-3 DPA exposure induced significant responses in these cells. Gingival biopsies were stained using antibodies to FPR2/ALX and DRV1/GPR32. Expression of FPR2/ALX and DRV1/GPR32 was examined in primary oral epithelial cells by qRT-PCR, flow cytometry, and immunofluorescence. The effect of RvD1n-3 DPA on intracellular calcium mobilization and transcription of beta-defensins 1 and 2, and cathelicidin was evaluated by qRT-PCR. FPR2/ALX and DRV1/GPR32 were expressed by gingival keratinocytes in situ. In cultured oral epithelial cells, FPR2/ALX was detected on the cell surface, whereas FPR2/ALX and DRV1/GPR32 were detected intracellularly. Exposure to RvD1n-3 DPA induced intracellular calcium mobilization, FPR2/ALX internalization, DRV1/GPR32 translocation to the nucleus, and significantly increased expression of genes coding for beta-defensin 1, beta-defensin 2, and cathelicidin. This shows that the signal constituted by RvD1n-3 DPA is recognized by oral keratinocytes and that this can strengthen the antimicrobial and regulatory potential of the oral epithelium.


Asunto(s)
Receptores de Formil Péptido , beta-Defensinas , Calcio , Ácidos Docosahexaenoicos/farmacología , Células Epiteliales/metabolismo , Humanos , Inflamación/patología , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo
18.
Dis Markers ; 2022: 1666240, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35692878

RESUMEN

Exploring the role of neuropeptides in the communication between monocyte subtypes facilitates an investigation of the pathogenesis of Kawasaki disease (KD). We investigated the patterns of interaction between neuropeptide-associated ligands and receptors in monocyte subpopulations in KD patients. Single-cell analysis was employed for the identification of cell subpopulations in KD patients, and monocytes were classified into 3 subpopulations: classical monocytes (CMs), intermediate monocytes (IMs), and nonclassical monocytes (NCMs). Cell-cell communication and differential analyses were used to identify ligand-receptor interactions in monocytes. Five neuropeptide-related genes (SORL1, TNF, SORT1, FPR2, and ANXA1) were involved in cell-cell interactions, wherein FPR2, a neuropeptide receptor, was significantly highly expressed in KD. Weighted gene coexpression network analysis revealed a significant correlation between the yellow module and FPR2 (p < 0.001, CC = 0.43). Using the genes in the yellow module, we constructed a PPI network to assess the possible functions of the FPR2-associated gene network. Gene set enrichment analysis showed that increased FPR2 levels may be involved in immune system regulation. FPR2 in CMs mediates the control of inflammation in KD. The findings of this study may provide a novel target for the clinical treatment of KD.


Asunto(s)
Monocitos , Síndrome Mucocutáneo Linfonodular , Biología Computacional , Humanos , Proteínas Relacionadas con Receptor de LDL , Proteínas de Transporte de Membrana , Monocitos/metabolismo , Síndrome Mucocutáneo Linfonodular/genética , Síndrome Mucocutáneo Linfonodular/patología , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/genética , Receptores de Lipoxina/metabolismo , Receptores de Neuropéptido , Análisis de la Célula Individual
19.
Breast Cancer Res ; 24(1): 25, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35382852

RESUMEN

BACKGROUND: Despite advancements in therapies, brain metastasis in patients with triple negative subtype of breast cancer remains a therapeutic challenge. Activated microglia are often observed in close proximity to, or within, malignant tumor masses, suggesting a critical role that microglia play in brain tumor progression. Annexin-A1 (ANXA1), a glucocorticoid-regulated protein with immune-regulatory properties, has been implicated in the growth and metastasis of many cancers. Its role in breast cancer-microglia signaling crosstalk is not known. METHODS: The importance of microglia proliferation and activation in breast cancer to brain metastasis was evaluated in MMTV-Wnt1 spontaneous mammary tumor mice and BALBc mice injected with 4T1 murine breast cancer cells into the carotid artery using flow cytometry. 4T1 induced-proliferation and migration of primary microglia and BV2 microglia cells were evaluated using 2D and coculture transwell assays. The requirement of ANXA1 in these functions was examined using a Crispr/Cas9 deletion mutant of ANXA1 in 4T1 breast cancer cells as well as BV2 microglia. Small molecule inhibition of the ANXA1 receptor FPR1 and FPR2 were also examined. The signaling pathways involved in these interactions were assessed using western blotting. The association between lymph node positive recurrence-free patient survival and distant metastasis-free patient survival and ANXA1 and FPR1 and FPR2 expression was examined using TCGA datasets. RESULTS: Microglia activation is observed prior to brain metastasis in MMTV-Wnt1 mice with primary and secondary metastasis in the periphery. Metastatic 4T1 mammary cancer cells secrete ANXA1 to promote microglial migration, which in turn, enhances tumor cell migration. Silencing of ANXA1 in 4T1 cells by Crispr/Cas9 deletion, or using inhibitors of FPR1 or FPR2 inhibits microglia migration and leads to reduced activation of STAT3. Finally, elevated ANXA1, FPR1 and FPR2 is significantly associated with poor outcome in lymph node positive patients, particularly, for distant metastasis free patient survival. CONCLUSIONS: The present study uncovered a network encompassing autocrine/paracrine ANXA1 signaling between metastatic mammary cancer cells and microglia that drives microglial recruitment and activation. Inhibition of ANXA1 and/or its receptor may be therapeutically rewarding in the treatment of breast cancer and secondary metastasis to the brain.


Asunto(s)
Anexina A1 , Neoplasias de la Mama , Microglía , Receptores de Formil Péptido , Animales , Anexina A1/genética , Encéfalo/patología , Neoplasias de la Mama/patología , Femenino , Humanos , Ratones , Microglía/metabolismo , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina
20.
Bioengineered ; 13(2): 4028-4038, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35129068

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a progressive degenerative disease, of which smoking is the main causer. We carried out this study with the aim of exploring the underlying mechanism of methylprednisolone (MP) treating the COPD. To stimulate COPD in vitro, cigarette smoke extract (CSE)was employed to induce human bronchial epithelial cells BEAS-2B. With the help of MTT and Tunel assays, the viability and apoptosis of BEAS-2B cells after indicated treatment were assessed. The levels of inflammatory response and oxidative stress were determined by the changes of markers basing on their commercial kits. Additionally, annexin A1 (ANXA1) expressions at both protein and mRNA levels were assessed with Western blot and Reverse transcription­quantitative PCR (RT-qPCR). Moreover, the expressions of apoptosis- and formyl peptide receptor 2 (FPR2) receptors and the adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) pathway-related proteins were determined with Western blot., related proteins and proteins. As a result, MP up-regulated the ANXA1 expression in CSE-induced BEAS-2B cells. MP enhanced the viability but suppressed the apoptosis, inflammatory response and oxidative stress of CSE-induced BEAS-2B cells via regulating FPR2/AMPK pathway, while ANXA1 knockdown exhibited oppositive effects on them. In conclusion, MP up-regulated ANXA1 to inhibit the inflammation, apoptosis and oxidative stress of BEAS-2B cells induced by CSE, alleviating COPD through suppressing the FPR2/AMPK pathway.


Asunto(s)
Anexina A1/genética , Metilprednisolona/farmacología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Receptores de Formil Péptido/genética , Receptores de Lipoxina/genética , Humo/efectos adversos , Proteínas Quinasas Activadas por AMP/genética , Apoptosis/efectos de los fármacos , Bronquios/citología , Línea Celular , Células Epiteliales/efectos de los fármacos , Humanos , Inflamación/metabolismo , Modelos Biológicos , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...