fps-zm1 has been researched along with Disease-Models--Animal* in 11 studies
11 other study(ies) available for fps-zm1 and Disease-Models--Animal
Article | Year |
---|---|
Impairing RAGE signaling promotes survival and limits disease pathogenesis following SARS-CoV-2 infection in mice.
Cellular and molecular mechanisms driving morbidity following SARS-CoV-2 infection have not been well defined. The receptor for advanced glycation end products (RAGE) is a central mediator of tissue injury and contributes to SARS-CoV-2 disease pathogenesis. In this study, we temporally delineated key cell and molecular events leading to lung injury in mice following SARS-CoV-2 infection and assessed efficacy of therapeutically targeting RAGE to improve survival. Early following infection, SARS-CoV-2 replicated to high titers within the lungs and evaded triggering inflammation and cell death. However, a significant necrotic cell death event in CD45- populations, corresponding with peak viral loads, was observed on day 2 after infection. Metabolic reprogramming and inflammation were initiated following this cell death event and corresponded with increased lung interstitial pneumonia, perivascular inflammation, and endothelial hyperplasia together with decreased oxygen saturation. Therapeutic treatment with the RAGE antagonist FPS-ZM1 improved survival in infected mice and limited inflammation and associated perivascular pathology. Together, these results provide critical characterization of disease pathogenesis in the mouse model and implicate a role for RAGE signaling as a therapeutic target to improve outcomes following SARS-CoV-2 infection. Topics: Animals; Benzamides; COVID-19; COVID-19 Drug Treatment; Disease Models, Animal; Lung; Mice; Mice, Transgenic; Receptor for Advanced Glycation End Products; SARS-CoV-2; Signal Transduction; Virus Replication | 2022 |
RAGE mediates airway inflammation via the HDAC1 pathway in a toluene diisocyanate-induced murine asthma model.
Exposure to toluene diisocyanate (TDI) is a significant pathogenic factor for asthma. We previously reported that the receptor for advanced glycation end products (RAGE) plays a key role in TDI-induced asthma. Histone deacetylase (HDAC) has been reported to be important in asthmatic pathogenesis. However, its effect on TDI-induced asthma is not known. The aim of this study was to determine the role of RAGE and HDAC in regulating airway inflammation using a TDI-induced murine asthma model.. BALB/c mice were sensitized and challenged with TDI to establish an asthma model. FPS-ZM1 (RAGE inhibitor), JNJ-26482585 and romidepsin (HDAC inhibitors) were administered intraperitoneally before each challenge. In vitro, the human bronchial epithelial cell line 16HBE was stimulated with TDI-human serum albumin (TDI-HSA). RAGE knockdown cells were constructed and evaluated, and MK2006 (AKT inhibitor) was also used in the experiments.. In TDI-induced asthmatic mice, the expression of RAGE, HDAC1, and p-AKT/t-AKT was upregulated, and these expressions were attenuated by FPS-ZM1. Airway reactivity, Th2 cytokine levels in lymph supernatant, IgE, airway inflammation, and goblet cell metaplasia were significantly increased in the TDI-induced asthmatic mice. These increases were suppressed by JNJ-26482585 and romidepsin. In addition, JNJ-26482585 and romidepsin ameliorated the redistribution of E-cadherin and β-catenin in TDI-induced asthma. In TDI-HSA-stimulated 16HBE cells, knockdown of RAGE attenuated the upregulation of HDAC1 and phospho-AKT (p-AKT). Treatment with the AKT inhibitor MK2006 suppressed TDI-induced HDAC1 expression.. These findings indicate that RAGE modulates HDAC1 expression via the PI3K/AKT pathway, and that inhibition of HDAC prevents TDI-induced airway inflammation. Topics: Animals; Asthma; Benzamides; Cell Line; Cytokines; Depsipeptides; Disease Models, Animal; Histone Deacetylase 1; Humans; Inflammation; Male; Mice; Mice, Inbred BALB C; Phosphatidylinositol 3-Kinases; Receptor for Advanced Glycation End Products; Signal Transduction; Toluene 2,4-Diisocyanate | 2022 |
Receptor for Advanced Glycation End Products (RAGE) Mediates Cognitive Impairment Triggered by Pneumococcal Meningitis.
Pneumococcal meningitis is a life-threatening infection of the central nervous system (CNS), and half of the survivors of meningitis suffer from neurological sequelae. We hypothesized that pneumococcal meningitis causes CNS inflammation via the disruption of the blood-brain barrier (BBB) and by increasing the receptor for advanced glycation end product (RAGE) expression in the brain, which causes glial cell activation, leading to cognitive impairment. To test our hypothesis, 60-day-old Wistar rats were subjected to meningitis by receiving an intracisternal injection of Streptococcus pneumoniae or artificial cerebrospinal fluid as a control group and were treated with a RAGE-specific inhibitor (FPS-ZM1) in saline. The rats also received ceftriaxone 100 mg/kg intraperitoneally, bid, and fluid replacements. Experimental pneumococcal meningitis triggered BBB disruption after meningitis induction, and FPS-ZM1 treatment significantly suppressed BBB disruption. Ten days after meningitis induction, surviving animals were free from infection, but they presented increased levels of TNF-α and IL-1β in the prefrontal cortex (PFC); high expression levels of RAGE, amyloid-β (Aβ Topics: Animals; Benzamides; Blood-Brain Barrier; Blotting, Western; Cognitive Dysfunction; Disease Models, Animal; Interleukin-1beta; Male; Meningitis, Pneumococcal; Morris Water Maze Test; Neuroprotective Agents; Open Field Test; Prefrontal Cortex; Rats; Rats, Wistar; Receptor for Advanced Glycation End Products; Tumor Necrosis Factor-alpha | 2021 |
LL-37 and HMGB1 induce alveolar damage and reduce lung tissue regeneration via RAGE.
The receptor for advanced glycation end-products (RAGE) has been implicated in the pathophysiology of chronic obstructive pulmonary disease (COPD). However, it is still unknown whether RAGE directly contributes to alveolar epithelial damage and abnormal repair responses. We hypothesize that RAGE activation not only induces lung tissue damage but also hampers alveolar epithelial repair responses. The effects of the RAGE ligands LL-37 and HMGB1 were examined on airway inflammation and alveolar tissue damage in wild-type and RAGE-deficient mice and on lung damage and repair responses using murine precision cut lung slices (PCLS) and organoids. In addition, their effects were studied on the repair response of human alveolar epithelial A549 cells, using siRNA knockdown of RAGE and treatment with the RAGE inhibitor FPS-ZM1. We observed that intranasal installation of LL-37 and HMGB1 induces RAGE-dependent inflammation and severe alveolar tissue damage in mice within 6 h, with stronger effects in a mouse strain susceptible for emphysema compared with a nonsusceptible strain. In PCLS, RAGE inhibition reduced the recovery from elastase-induced alveolar tissue damage. In organoids, RAGE ligands reduced the organoid-forming efficiency and epithelial differentiation into pneumocyte-organoids. Finally, in A549 cells, we confirmed the role of RAGE in impaired repair responses upon exposure to LL-37. Together, our data indicate that activation of RAGE by its ligands LL-37 and HMGB1 induces acute lung tissue damage and that this impedes alveolar epithelial repair, illustrating the therapeutic potential of RAGE inhibitors for lung tissue repair in emphysema. Topics: A549 Cells; Alveolar Epithelial Cells; Animals; Antimicrobial Cationic Peptides; Benzamides; Cathelicidins; Cell Line, Tumor; Disease Models, Animal; HMGB1 Protein; Humans; Mice; Mice, Inbred C57BL; Mice, Knockout; Organoids; Pancreatic Elastase; Pulmonary Alveoli; Pulmonary Disease, Chronic Obstructive; Receptor for Advanced Glycation End Products; Regeneration | 2021 |
High-mobility group box 1 induces bone destruction associated with advanced oral squamous cancer via RAGE and TLR4.
Bone destruction of maxillary and mandibular bone by invasive oral squamous cell cancer (OSCC) raises various problems in the management of patients, resulting in poor outcomes and survival. However, the mechanism behind bone destruction by OSCC remains unclear. High-mobility group box 1 (HMGB1), a highly conserved ubiquitous nuclear non-histone DNA-binding protein, has been demonstrated to be secreted by aggressive cancers and regulate osteoclastogenesis, a central player during bone destruction. We therefore reasoned that HMGB1 secreted by OSCCs contributes to bone destruction. Our results showed that HMGB1 is produced by human cell lines of OSCC and promotes osteoclastogenesis via up-regulation of the expression of receptor activator of nuclear factor kappa-Β ligand in osteoblasts and osteocytes, and consequently osteoclastic bone destruction in mice. Further, we found that these actions of HMGB1 are mediated via the receptor for advanced glycation end products and toll-like receptors. These findings suggest that HMGB1 of OSCC and its down-stream signal pathways are potential targets for the treatment of bone destruction associated with advanced OSCC. Topics: Animals; Benzamides; Bone and Bones; Bone Resorption; Cell Line, Tumor; Cell Proliferation; Disease Models, Animal; Head and Neck Neoplasms; HMGB1 Protein; Humans; Ki-67 Antigen; Male; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplasm Invasiveness; Osteoblasts; Osteocytes; Osteogenesis; RANK Ligand; RAW 264.7 Cells; Receptor for Advanced Glycation End Products; Squamous Cell Carcinoma of Head and Neck; Sulfonamides; Toll-Like Receptor 4 | 2020 |
The RAGE signaling pathway is involved in intestinal inflammation and represents a promising therapeutic target for Inflammatory Bowel Diseases.
Inflammatory Bowel Diseases (IBD) are chronic inflammatory conditions of the intestinal tract. IBD are believed to result from an inappropriate immune response against the intestinal flora in genetically predisposed patients. The precise etiology of these diseases is not fully understood, therefore treatments rely on the dampening of symptoms, essentially inflammation, rather than on the cure of the disease. Despite the availability of biologics, such as anti-TNF antibodies, some patients remain in therapeutic failure and new treatments are thus needed. The multiligand receptor for advanced glycation end-products (RAGE) is a pattern recognition receptor implicated in inflammatory reactions and immune system activation. Here, we investigated the role of RAGE in intestinal inflammation and its potential as a therapeutic target in IBD. We showed that RAGE was upregulated in inflamed tissues from IBD patients compared to controls. Rage Topics: Animals; Benzamides; Colon; Dextran Sulfate; Disease Models, Animal; Humans; Inflammation; Inflammatory Bowel Diseases; Intestines; Mice; Mice, Inbred C57BL; Mice, Knockout; Molecular Targeted Therapy; Receptor for Advanced Glycation End Products; Signal Transduction | 2019 |
Targeted inhibition of RAGE reduces amyloid-β influx across the blood-brain barrier and improves cognitive deficits in db/db mice.
To investigate restorative effects of the receptor for advanced glycation end products (RAGE)-specific inhibitor FPS-ZM1 on abnormal amyloid β (Aβ) influx across the blood brain-barrier (BBB) and cognitive deficits in db/db mice.. Aβ influx across the BBB was determined by intra-arterial infusion of. Downregulation of abnormal Aβ influx across the BBB by FPS-ZM1 at higher dosage contributes to reduced neuronal apoptosis, improved hippocampal plasticity and cognitive impairment in db/db mice. Topics: Amyloid beta-Peptides; Animals; bcl-2-Associated X Protein; Benzamides; Blood-Brain Barrier; Brain; Caspase 3; Cognition Disorders; Diabetes Mellitus, Type 2; Disease Models, Animal; Exploratory Behavior; Male; Maze Learning; Mice; Microvessels; Peptide Fragments; Proto-Oncogene Proteins c-bcl-2; Receptor for Advanced Glycation End Products; Receptors, Leptin; Synaptic Transmission | 2018 |
HMGB1 promotes neurovascular remodeling via Rage in the late phase of subarachnoid hemorrhage.
High-mobility group box1 (HMGB1) is a nuclear protein widely expressed in the central nervous system. Extracellular HMGB1 serves as a proinflammatory cytokine and contributes to brain injury during the acute stage post-stroke. Recently, increasing evidence has demonstrated beneficial effects of HMGB1 in some types of brain injury, but little is known about its effects during the late phase of subarachnoid hemorrhage (SAH). This study was designed to explore the potential roles and mechanisms of HMGB1 and its receptor, receptor for advanced glycation end-products (Rage), on brain recovery in the late stage of experimental SAH. Two inhibitors of HMGB1, ethyl pyruvate and glycyrrhizin (EP and GA), and Rage antagonist FPS-ZM1 were used to determine whether HMGB1 promotes brain recovery after SAH. The administration of EP, GA, and FPS-ZM1 effectively reduced HMGB1 and Rage expression. Correspondingly, protein levels of beneficial growth factors (NGF, BDNF, and VEGF) and numbers of BrdU and DCX positive neurons in the cortex were also decreased. The biphasic roles of HMGB1 may be based on the different redox modifications of cysteine residues. In this research, rats injected with two different redox status HMGB1 showed different prognosises at 7-14day after SAH. Recombinant HMGB1 can promote cytokine stimulating activity and aggravate brain injury. However, oxidized HMGB1 was unable to stimulate TNF production but can promote brain recovery by promoting neurotrophin expression. In conclusion, our investigation identified that HMGB1 promotes neurovascular recovery via Rage and may act in the oxidized state in the late stage of SAH. Topics: Animals; Benzamides; Brain; Brain Injuries; Cerebral Cortex; Disease Models, Animal; Doublecortin Protein; Glycyrrhizic Acid; HMGB1 Protein; Male; Neurons; Pyruvates; Rats; Rats, Sprague-Dawley; Receptor for Advanced Glycation End Products; Recombinant Proteins; Signal Transduction; Subarachnoid Hemorrhage; Vascular Remodeling | 2017 |
Effect of microRNA-129-5p targeting HMGB1-RAGE signaling pathway on revascularization in a collagenase-induced intracerebral hemorrhage rat model.
This study aimed at exploring the effect of microRNA-129-5p (miR-129-5p) targeting high mobility group box-1 (HMGB1)-receptor for advanced glycation end-products (RAGE) signaling pathway on the revascularization in a collagenase-induced intracerebral hemorrhage (ICH) rat model.. OX26-pGFAP-IL, an immunoliposome expressing miR-129-5p was constructed. The collagenase-induced ICH rat models were successfully established by 96 Sprague Dawley (SD) rats, which were categorized into the sham group, ICH group, miR-129-5p group, negative control (NC) group, ethyl pyruvate (EP, an inhibitor of HMGB1) group and N-benzyl-4-chloro-N-cyclohe-xylbenzamide (FPS-ZM1, a RAGE receptor antagonist) group. The miR-129-5p expression in the brain tissue homogenate was detected using the quantitative real-time polymerase chain reaction (qRT-PCR) and the protein expressions of HMGB1 and RAGE by Western blotting. Immunohistochemistry (IHC) was used for the detection of the vascular endothelial growth factor (VEGF). Microvessel density (MVD) was also detected.. Compared to the sham group, the ICH, NC, EP and FPS-ZM1 groups had a decrease in miR-129-5p expressions, and an increase in the protein expressions of HMGB1, RAGE and VEGF and MVD. In comparison to the ICH, NC, EP and FPS-ZM1 groups the miR-129-5p group had an elevation in the miRNA-129-5p expressions. The miR-129-5p and EP groups had decreased HMGB1 protein expression and the miR-129-5p, EP and FPS-ZM1 groups had a reduced RAGE protein expression as compared to the ICH group. In comparison to the ICH group, the miR-129-5p, EP, FPS-ZM1 groups had a decline in the VEGF protein expression and MVD.. Our study proved that up-regulation of miR-129-5p might suppress the HMGB1-RAGE signaling pathway to restrain the revascularization of rats with ICH. Topics: Animals; Benzamides; Brain; Cerebral Hemorrhage; Collagenases; Disease Models, Animal; Female; HMGB1 Protein; Male; MicroRNAs; Rats; Rats, Sprague-Dawley; Receptor for Advanced Glycation End Products; Signal Transduction; Up-Regulation; Vascular Endothelial Growth Factor A | 2017 |
HMGB1 may act via RAGE to promote angiogenesis in the later phase after intracerebral hemorrhage.
Following intracerebral hemorrhage (ICH), high-mobility group box 1 protein (HMGB1) may promote vascular remodeling. Whether HMGB1 supports angiogenesis after ICH is unclear, as are the receptors and downstream signaling pathway(s) involved. We used the rat model of collagenase-induced ICH to determine whether HMGB1 acts via the receptor for advanced glycation end-products (RAGE) to upregulate vascular endothelial growth factor (VEGF), a potent mitogen of endothelial cells and key regulator of normal and abnormal angiogenesis in the late phase of injury. At 3d after ICH induction, rats were treated with saline, ethyl pyruvate (EP) or N-benzyl-4-chloro-N-cyclohexylbenzamide (FPS-ZM1). ICH induced the movement of HMGB1 from the nucleus into the cytoplasm. Levels of HMGB1 and RAGE in the ipsilateral striatum increased within a few days of induction and continued to rise for 7-14d afterward. By 14d after induction, levels of VEGF and vessel density were higher than in the Sham group. Administering EP 3 days after ICH induction prevented much of the stroke-induced increases in vessel density and in expression of HMGB1, RAGE, and VEGF. Administering FPS-ZM1 after ICH blocked much of the stroke-induced increases in vessel density and VEGF expression. Our results suggest that after ICH, HMGB1 may upregulate VEGF in the ipsilateral striatum predominantly via RAGE. Hence, targeting the HMGB1/RAGE signaling pathway may help reduce inappropriate angiogenesis after ICH. Topics: Animals; Benzamides; Cerebral Hemorrhage; Collagenases; Corpus Striatum; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Administration Schedule; Functional Laterality; Gene Expression Regulation; Glial Fibrillary Acidic Protein; HMGB1 Protein; Male; Neovascularization, Physiologic; Pyruvates; Rats; Rats, Sprague-Dawley; Receptor for Advanced Glycation End Products; Up-Regulation; Vascular Endothelial Growth Factor A | 2015 |
A multimodal RAGE-specific inhibitor reduces amyloid β-mediated brain disorder in a mouse model of Alzheimer disease.
In Alzheimer disease (AD), amyloid β peptide (Aβ) accumulates in plaques in the brain. Receptor for advanced glycation end products (RAGE) mediates Aβ-induced perturbations in cerebral vessels, neurons, and microglia in AD. Here, we identified a high-affinity RAGE-specific inhibitor (FPS-ZM1) that blocked Aβ binding to the V domain of RAGE and inhibited Aβ40- and Aβ42-induced cellular stress in RAGE-expressing cells in vitro and in the mouse brain in vivo. FPS-ZM1 was nontoxic to mice and readily crossed the blood-brain barrier (BBB). In aged APPsw/0 mice overexpressing human Aβ-precursor protein, a transgenic mouse model of AD with established Aβ pathology, FPS-ZM1 inhibited RAGE-mediated influx of circulating Aβ40 and Aβ42 into the brain. In brain, FPS-ZM1 bound exclusively to RAGE, which inhibited β-secretase activity and Aβ production and suppressed microglia activation and the neuroinflammatory response. Blockade of RAGE actions at the BBB and in the brain reduced Aβ40 and Aβ42 levels in brain markedly and normalized cognitive performance and cerebral blood flow responses in aged APPsw/0 mice. Our data suggest that FPS-ZM1 is a potent multimodal RAGE blocker that effectively controls progression of Aβ-mediated brain disorder and that it may have the potential to be a disease-modifying agent for AD. Topics: Aging; Alzheimer Disease; Amyloid beta-Peptides; Amyloid Precursor Protein Secretases; Animals; Benzamides; Blood-Brain Barrier; Brain; Cerebrovascular Circulation; CHO Cells; Cricetinae; Cricetulus; Disease Models, Animal; Drug Evaluation, Preclinical; Humans; Male; Mice; Mice, Transgenic; Neuroprotective Agents; Peptide Fragments; Psychomotor Performance; Receptor for Advanced Glycation End Products; Receptors, Immunologic; Recombinant Fusion Proteins; Small Molecule Libraries | 2012 |