Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 320
Filter
1.
Int J Immunopathol Pharmacol ; 35: 20587384211032098, 2021.
Article in English | MEDLINE | ID: mdl-34275383

ABSTRACT

Hepatic ischemia-reperfusion injury (IRI) is a major unavoidable clinical problem often accompanying various liver surgery and transplantation. d-Pinitol, a cyclic polyol, exhibits hepatoprotective efficacy. The objective of this study is to determine the possible mechanism of action of pinitol against endoplasmic reticulum (ER) stress regulation-mediated hepatic IRI and compare its effects with thymoquinone (TQ) in experimental rats. Male Sprague Dawley rats were pre-treated orally with either vehicle (DMSO) or d-Pinitol (5, 10, and 20 mg/kg) or TQ (30 mg/kg) for 21 days and subjected to 60 min of partial hepatic ischemia followed by 24 h of reperfusion. Pre-treatment with pinitol (10 and 20 mg/kg) effectively (P < 0.05) protected against IRI-induced hepatic damage reflected by attenuation of elevated oxidative stress and pro-inflammatory cytokines. Additionally, western blot and ELISA analyses suggested that pinitol significantly (P < 0.05) down-regulated expression of endoplasmic reticulum stress apoptotic markers, namely glucose-regulated protein (GRP)-78, CCAAT/enhancer-binding protein homologous protein (CHOP), activating transcription factor (AFT)-4 and -6α, X-box binding protein-1, and caspase-3, 9, and 12. Additionally, pinitol pre-treatment effectively (P < 0.05) improved mitochondrial function and phosphorylation of Extracellular signal-regulated kinase (ERK)-1/2 and p38. Pinitol markedly (P < 0.05) protected hepatic apoptosis determined by flow cytometry. Further, pinitol provided effective (P < 0.05) protection against hepatic histological and ultrastructural aberrations induced by IRI. TQ showed more pronounced protective effect against attenuation of IRI-induced hepatic injury as compared to d-Pinitol. Pinitol offered protection against endoplasmic reticulum stress-mediated phosphorylation of ERK1/2 and p38, thereby inhibiting AFT4-CHOP/GRP78 signaling response and caspase-3 induced hepatocellular apoptosis during hepatic ischemia-reperfusion insults. Thus, Pinitol can be considered as a viable option for the management of hepatic IRI.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Inositol/analogs & derivatives , Liver Diseases/drug therapy , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Animals , Caspase 3/drug effects , Heat-Shock Proteins/drug effects , Inositol/therapeutic use , Liver Diseases/pathology , Male , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Transcription Factor CHOP/drug effects
2.
J Cell Physiol ; 236(12): 8110-8121, 2021 12.
Article in English | MEDLINE | ID: mdl-34101831

ABSTRACT

Even though aberrant mechanistic target of rapamycin (mTOR) signaling is known to cause cardiomyopathy, its underlying mechanism remains poorly understood. Because augmentation of αB-crystallin and hspB2 was presented in the cortical tubers and lymphangioleiomyomatosis of tuberous sclerosis complex patients, we deciphered the role of αB-crystallin and its adjacent duplicate gene, hspB2, in hyperactive mTOR-induced cardiomyopathy. Cardiac Tsc1 deletion (T1-hKO) caused mouse mTOR activation and cardiomyopathy. Overexpression of αB-crystallin and hspB2 was presented in the hearts of these mice. Knockout of αB-crystallin/hspB2 reversed deficient Tsc1-mediated fetal gene expression, mTOR activation, mitochondrial damage, cardiomyocyte vacuolar degeneration, cardiomyocyte size, and fibrosis of T1-hKO mice. These cardiac-Tsc1; αB-crystallin; hspB2 triple knockout (tKO) mice had improved cardiac function, smaller heart weight to body weight ratio, and reduced lethality compared with T1-hKO mice. Even though activated mTOR suppressed autophagy in T1-hKO mice, ablation of αB-crystallin and hspB2 failed to restore autophagy in tKO mice. mTOR inhibitors suppressed αB-crystallin expression in T1-hKO mice and rat cardiomyocyte line H9C2. Starvation of H9C2 cells activated autophagy and suppressed αB-crystallin expression. Since inhibition of autophagy restored αB-crystallin expression in starved H9C2 cells, autophagy is a negative regulator of αB-crystallin expression. mTOR thus stimulates αB-crystallin expression through suppression of autophagy. In conclusion, αB-crystallin and hspB2 play a pivotal role in Tsc1 knockout-related cardiomyopathy and are therapeutic targets of hyperactive mTOR-associated cardiomyopathy.


Subject(s)
Cardiomyopathies/metabolism , Crystallins/metabolism , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Myocytes, Cardiac/metabolism , Animals , Cardiomyopathies/drug therapy , Cardiomyopathies/genetics , HSP27 Heat-Shock Proteins/drug effects , HSP27 Heat-Shock Proteins/genetics , Heat-Shock Proteins/drug effects , MTOR Inhibitors/pharmacology , Mice, Knockout , Myocytes, Cardiac/drug effects , Promoter Regions, Genetic/genetics , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism
3.
J Nanobiotechnology ; 19(1): 9, 2021 Jan 06.
Article in English | MEDLINE | ID: mdl-33407570

ABSTRACT

BACKGROUND: Phototherapy is a promising strategy for cancer therapy by reactive oxygen species (ROS) of photodynamic therapy (PDT) and hyperthermia of photothermal therapy (PTT). However, the therapeutic efficacy was restricted by tumor hypoxia and thermal resistance of increased expression of heat shock protein (Hsp). In this study, we developed albumin nanoparticles to combine hypoxia relief and heat shock protein inhibition to overcome these limitations for phototherapy enhancement. RESULTS: Near-infrared photosensitizer (IR780) and gambogic acid (GA, Hsp90 inhibitor) were encapsulated into albumin nanoparticles via hydrophobic interaction, which was further deposited MnO2 on the surface to form IGM nanoparticles. Both in vitro and in vivo studies demonstrated that IGM could catalyze overexpress of hydrogen peroxide to relive hypoxic tumor microenvironment. With near infrared irradiation, the ROS generation was significantly increase for PDT enhancement. In addition, the release of GA was promoted by irradiation to bind with Hsp90, which could reduce cell tolerance to heat for PTT enhancement. As a result, IGM could achieve better antitumor efficacy with enhanced PDT and PTT. CONCLUSION: This study develops a facile approach to co-deliver IR780 and GA with self-assembled albumin nanoparticles, which could relive hypoxia and suppress Hsp for clinical application of cancer phototherapy.


Subject(s)
Heat-Shock Proteins/drug effects , Hypoxia/drug therapy , Nanoparticles/chemistry , Phototherapy/methods , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Survival , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Hydrophobic and Hydrophilic Interactions , Infrared Rays , Male , Manganese Compounds/chemistry , Mice , Mice, Inbred BALB C , Nanoparticles/therapeutic use , Oxides/chemistry , Photosensitizing Agents/pharmacology , Tumor Microenvironment/drug effects , Xanthones/pharmacology
4.
Genome Med ; 12(1): 101, 2020 11 23.
Article in English | MEDLINE | ID: mdl-33225964

ABSTRACT

BACKGROUND: Heat shock proteins (HSPs), a representative family of chaperone genes, play crucial roles in malignant progression and are pursued as attractive anti-cancer therapeutic targets. Despite tremendous efforts to develop anti-cancer drugs based on HSPs, no HSP inhibitors have thus far reached the milestone of FDA approval. There remains an unmet need to further understand the functional roles of HSPs in cancer. METHODS: We constructed the network for HSPs across ~ 10,000 tumor samples from The Cancer Genome Atlas (TCGA) and ~ 10,000 normal samples from Genotype-Tissue Expression (GTEx), and compared the network disruption between tumor and normal samples. We then examined the associations between HSPs and cancer hallmarks and validated these associations from multiple independent high-throughput functional screens, including Project Achilles and DRIVE. Finally, we experimentally characterized the dual function effects of HSPs in tumor proliferation and metastasis. RESULTS: We comprehensively analyzed the HSP expression landscape across multiple human cancers and revealed a global disruption of the co-expression network for HSPs. Through analyzing HSP expression alteration and its association with tumor proliferation and metastasis, we revealed dual functional effects of HSPs, in that they can simultaneously influence proliferation and metastasis in opposite directions. We experimentally characterized the dual function of two genes, DNAJC9 and HSPA14, in lung cancer cells. We further demonstrated the generalization of this dual direction of associations between HSPs and cancer hallmarks, suggesting the necessity to more carefully evaluate HSPs as therapeutic targets and develop highly specific HSP inhibitors for cancer intervention. CONCLUSIONS: Our study furnishes a holistic view of functional associations of HSPs with cancer hallmarks to aid the development of HSP inhibitors as well as other drugs in cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Heat-Shock Proteins/drug effects , Heat-Shock Proteins/genetics , Neoplasms/drug therapy , Neoplasms/genetics , A549 Cells , Cell Proliferation , Gene Knockdown Techniques , HSP40 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Lung Neoplasms/genetics , Neoplasm Metastasis , Survival Analysis , Transcriptome
5.
Commun Biol ; 3(1): 547, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33005001

ABSTRACT

Rising antibiotic resistance urgently begs for novel targets and strategies for antibiotic discovery. Here, we report that over-activation of the periplasmic DegP protease, a member of the highly conserved HtrA family, can be a viable strategy for antibiotic development. We demonstrate that tripodal peptidyl compounds that mimic DegP-activating lipoprotein variants allosterically activate DegP and inhibit the growth of an Escherichia coli strain with a permeable outer membrane in a DegP-dependent fashion. Interestingly, these compounds inhibit bacterial growth at a temperature at which DegP is not essential for cell viability, mainly by over-proteolysis of newly synthesized proteins. Co-crystal structures show that the peptidyl arms of the compounds bind to the substrate-binding sites of DegP. Overall, our results represent an intriguing example of killing bacteria by activating a non-essential enzyme, and thus expand the scope of antibiotic targets beyond the traditional essential proteins or pathways.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Heat-Shock Proteins/metabolism , Periplasmic Proteins/metabolism , Serine Endopeptidases/metabolism , Binding Sites , Enzyme Activation , Enzyme Activators/pharmacology , Escherichia coli/drug effects , Fluorescence Polarization , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/drug effects , Microbial Sensitivity Tests , Peptides/metabolism , Peptides/pharmacology , Periplasmic Proteins/chemistry , Periplasmic Proteins/drug effects , Protein Structure, Tertiary , Serine Endopeptidases/chemistry , Serine Endopeptidases/drug effects
6.
Int. j. morphol ; 38(5): 1455-1462, oct. 2020. tab, graf
Article in English | LILACS | ID: biblio-1134462

ABSTRACT

SUMMARY: This study aimed to investigate the changes in testis tissue of thioacetamide-induced rats and the effect of melatonin on these changes. Thirty-five male Wistar Albino rats were divided into five groups. Group I; Control (n=7), Group II; Melatonin (Mel) (10 mg/kg) a single dose (i.p)(n=7), Group III; Thioacetamide (TAA) (300 mg/kg) (i.p) 2 times with 24 hour intervals (n=7), Group IV; TAA (300 mg/kg) was administered at 24-hour intervals, afterwards of 10 mg/kg single dose of Mel (n=7), Group V; Mel was administered 10 mg/kg a single dose 24 hours before the administration of TAA (n=7). Testis was evaluated histologically, immunohistochemically (Heat Shock Proteins (HSP) 70 and 90), blood serum testosterone, total antioxidant status(TAS) and total oxidant status(TOS) in tissue. The tissue sections of Group III decreased seminiferous tubule diameters, and germinal epithelium spills were observed. HSP70 and HSP90 expressions were increased. There wasn't a statistically significant change in testosterone levels among the groups. While TAS levels decreased in Group III compared to control, TOS levels didn't change. HSP70 and HSP90 decreased in groups with Mel-treated. Mel was found to have both protective and therapeutic effects. According to our results, the therapeutic effect of Mel in thioacetamide-induced acute testicular injury is greater than its protective effect.


RESUMEN: Este estudio tuvo como objetivo investigar los cambios en el tejido testicular de ratas inducidas por tioacetamida y el efecto de la melatonina en estos cambios. Treinta y cinco ratas macho Wistar Albino se dividieron en cinco grupos. Grupo I; Control (n = 7), Grupo II; Melatonina (Mel) (10 mg / kg) una dosis única (i.p) (n = 7), Grupo III; Tioacetamida (TAA) (300 mg / kg) (i.p) 2 veces con intervalos de 24 horas (n = 7), Grupo IV; TAA (300 mg / kg) se administró a intervalos de 24 horas, luego de una dosis única de 10 mg / kg de Mel (n = 7), Grupo V; Mel recibió 10 mg / kg de una dosis única 24 horas antes de la administración de TAA (n = 7). Los testículos se evaluaron histológicamente, inmunohistoquímicamente (proteínas de choque térmico (PCT) 70 y 90), testosterona en suero sanguíneo, estado antioxidante total (EAT) y estado oxidante total (EOT) en el tejido. En secciones de tejido del Grupo III se observó disminución de los diámetros de los túbulos seminíferos y derrames en el epitelio germinal. Se aumentaron las expresiones HSP70 y HSP90. No hubo un cambio estadísticamente significativo en los niveles de testosterona entre los grupos. Mientras que los niveles de EAT disminuyeron en el Grupo III en comparación con el control, los niveles de EOT no cambiaron. HSP70 y HSP90 disminuyeron en los grupos tratados con Mel. Se descubrió que Mel tenía efectos protectores y terapéuticos. Según nuestros resultados, el efecto terapéutico de Mel en la lesión testicular aguda inducida por tioacetamida es mayor que su efecto protector.


Subject(s)
Animals , Male , Rats , Testis/drug effects , Thioacetamide/toxicity , Melatonin/pharmacology , Antioxidants/pharmacology , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Rats, Wistar , Heat-Shock Proteins/drug effects , Heat-Shock Proteins/metabolism , Melatonin/administration & dosage , Antioxidants/administration & dosage
7.
Mol Med Rep ; 22(4): 2887-2895, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32700746

ABSTRACT

Prostaglandin E2 (PGE2) is involved in numerous physiological and pathological processes of the kidney via its four receptors. A previous study has suggested that a defect in the PGE2 receptor 1 (EP1) gene markedly suppressed the transforming growth factor­ß1 (TGF­ß1)­induced mesangial cell (MC) proliferation and extracellular matrix aggregation. Therefore, the present study aimed to adopt a pharmacological method of specifically suppressing or activating the EP1 receptor to further verify and demonstrate these results. The EP1 receptor antagonist SC­19220 and EP1 receptor agonist 17­phenyl­trinor­PGE2 ethyl amide (17­pt­PGE2) were selectively used to treat five­sixths nephrectomy renal fibrosis model mice and TGF­ß1­stimulated MCs. An Alpha screen PGE2 assay kit, flow cytometry, western blotting and immunohistochemical techniques were adopted to perform in vivo and in vitro experiments. The present results suggested that compared with the control group, the selective EP1 receptor antagonist SC­19220 improved renal function, markedly reduced the plasma blood urea nitrogen and creatinine levels (P<0.05) and alleviated glomerulosclerosis (P<0.05). By contrast, the EP1 receptor agonist 17­pt­PGE2 aggravated renal dysfunction and glomerulosclerosis (P<0.05). To verify the renal protection mechanisms mediated by suppression of the EP1 receptor, the expression levels of endoplasmic reticulum stress (ERS)­related proteins, including chaperone glucose­regulated protein 78 (GRP78), transient receptor potential channel 1 (TRPC1) and protein kinase R­like endoplasmic reticulum kinase (PERK), were further evaluated histologically. The expression of GRP78, TRPC1 and PERK in the antagonist treatment group were markedly downregulated (P<0.05), whereas those in the agonist treatment group were upregulated (P<0.05). The present in vitro experiments demonstrated that, compared with the control group, the EP1 receptor antagonist suppressed the expression of GRP78, TRPC1 and PERK (P<0.05), reduced the production of PGE2 (P<0.05) and decreased the MC apoptosis rate (P<0.05), thus alleviating TGF­ß1­stimulated MC injury. Consequently, consistent with previous results, selectively antagonizing the EP1 receptor improved renal function and mitigated glomerulosclerosis, and its potential mechanism might be associated with the suppression of ERS.


Subject(s)
Dinoprostone/metabolism , Glomerulonephritis/drug therapy , Receptors, Prostaglandin E, EP1 Subtype/agonists , Receptors, Prostaglandin E, EP1 Subtype/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cells, Cultured , Dibenz(b,f)(1,4)oxazepine-10(11H)-carboxylic acid, 8-chloro-, 2-acetylhydrazide/pharmacology , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Glomerulonephritis/etiology , Glomerulonephritis/physiopathology , Heat-Shock Proteins/drug effects , Heat-Shock Proteins/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Male , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mice , Mice, Inbred C57BL , Nephrectomy/adverse effects , Prostaglandin Antagonists/pharmacology , TRPC Cation Channels/drug effects , TRPC Cation Channels/metabolism , Transforming Growth Factor beta1/toxicity , eIF-2 Kinase/drug effects , eIF-2 Kinase/metabolism
8.
Mol Med Rep ; 22(2): 697-706, 2020 08.
Article in English | MEDLINE | ID: mdl-32626931

ABSTRACT

An overwhelming endoplasmic reticulum stress (ERS) and the following unfolded protein response (UPR) can induce hepatic inflammation, fibrosis and hepatocellular carcinoma (HCC). Caudatin, one of the species of C­21 steroidal glycosides mainly isolated from the roots of Cynanchum bungei Decne, exhibits potent anticancer activities in vivo. However, the effect of caudatin on HCC remains unclear. In the present study, a diethylnitrosamine (DEN)­induced HCC model was established. Nodules and tumors in rat livers were monitored by T2­/T1­weighted­magnetic resonance imaging (MRI) using a 1.5 T scanner. Caudatin reduced the number and size of nodules and alleviated the inflammatory foci in the liver. In addition, the hepatic pro­inflammatory levels of interleukin (IL) 6, monocyte chemoattractant protein 1 and IL­1ß were decreased in caudatin­treated rats. The DEN­induced surge in malondialdehyde, aspartate aminotransferase, alanine transaminase and TBIL were alleviated following caudatin treatment. The expression of ERS chaperones glucose­regulated protein, 94 kDa, glucose­regulated protein, 78 kDa and protein disulfide­isomerase A4 and the proliferation marker Ki­67 in liver nodules were all downregulated by caudatin as demonstrated by immunohistochemistry, reverse transcription­quantitative PCR and western blot analysis. Caudatin reduced the cytoprotective ERS sensor activating transcription factor 6­mediated signal transduction and inhibited the PKR­like endoplasmic reticulum kinase/eukaryotic initiation factor 2α/activating transcription factor 4 pathway. However, the effect of caudatin on inositol requiring enzyme 1 signaling was negligible. In conclusion, restoration of the dysregulated UPR program was involved in the antitumor efficacy of caudatin without inducing cumulative hepatotoxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Glycosides/pharmacology , Liver Neoplasms, Experimental/drug therapy , Steroids/pharmacology , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 6/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cytokines/metabolism , Diethylnitrosamine/toxicity , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/metabolism , Glycosides/therapeutic use , Heat-Shock Proteins/drug effects , Liver/diagnostic imaging , Liver/drug effects , Liver Neoplasms, Experimental/blood , Liver Neoplasms, Experimental/diagnostic imaging , Liver Neoplasms, Experimental/pathology , Magnetic Resonance Imaging , Rats, Sprague-Dawley , Signal Transduction/drug effects , Steroids/therapeutic use , Unfolded Protein Response/drug effects , eIF-2 Kinase/metabolism
9.
BMC Microbiol ; 20(1): 112, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32398085

ABSTRACT

BACKGROUND: Listeria monocytogenes (L. monocytogenes) is a global opportunistic intracellular pathogen that can cause many infections, including meningitis and abortion in humans and animals; thus, L. monocytogenes poses a great threat to public safety and the development of the aquaculture industry. The isolation rate of Listeria monocytogenes in fishery products has always been high. And the pore-forming toxin listeriolysin O (LLO) is one of the most important virulence factors of L. monocytogenes. LLO can promote cytosolic bacterial proliferation and help the pathogen evade attacks from the host immune system. In addition, L. monocytogenes infection can trigger a series of severe inflammatory reactions. RESULTS: Here, we further confirmed that morin lacking anti-Listeria activity could inhibit LLO oligomerization. We also found that morin can effectively alleviate the inflammation induced by Listeria in vivo and in vitro and exerted an obvious protective effect on infected cells and mice. CONCLUSIONS: Morin does not possess anti-Listeria activity, neither does it interfere with secretion of LLO. However, morin inhibits oligomerisation of LLO and morin does reduce the inflammation caused during Listeria infection.


Subject(s)
Bacterial Toxins/chemistry , Flavonoids/administration & dosage , Heat-Shock Proteins/chemistry , Hemolysin Proteins/chemistry , Listeria monocytogenes/pathogenicity , Listeriosis/drug therapy , Animals , Cell Line , Disease Models, Animal , Flavonoids/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Heat-Shock Proteins/drug effects , Hemolysin Proteins/drug effects , Humans , Listeria monocytogenes/drug effects , Listeria monocytogenes/enzymology , Listeria monocytogenes/growth & development , Mice , Protein Multimerization/drug effects , Virulence/drug effects
10.
Neurotox Res ; 38(2): 447-460, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32410195

ABSTRACT

Several pathophysiological processes involve Hypoxia conditions, where the nervous system is affected as well. We postulate that the GABAergic system is especially sensitive. Furthermore, drugs improving the resistance to hypoxia have been investigated, such as the neurosteroid dehydroepiandrosterone sulfate (DHEAS) which has shown beneficial effects in hypoxic processes in mammals; however, at the cellular level, its exact mechanism of action has yet to be fully elucidated. Here, we used a chemical hypoxia model through sodium sulfite (SS) exposure in Caenorhabditis elegans (C. elegans), a nematode whose response to hypoxia involves pathways and cellular processes conserved in mammals, and that allows study the direct effect of DHEAS without its conversion to sex hormones. This work aimed to determine the effect of DHEAS on damage to the GABAergic system associated with SS exposure in C. elegans. Worms were subjected to nose touch response (Not Assay) and observed in epifluorescence microscopy. DHEAS decreased the shrinkage response of Not Assay and the level of damage in GABAergic neurons on SS-exposed worms. Also, the enhanced nuclear localization of DAF-16 and consequently the overexpression of chaperone HSP-16.2 by hypoxia were significantly reduced in SS + DHEAS exposed worms. As well, DHEAS increased the survival rate of worms exposed to hydrogen peroxide. These results suggest that hypoxia-caused damage over the GABAergic system was prevented at least partially by DHEAS, probably through non-genomic mechanisms that involve its antioxidant properties related to its chemical structure.


Subject(s)
Antioxidants/pharmacology , Caenorhabditis elegans Proteins/drug effects , Dehydroepiandrosterone Sulfate/pharmacology , Forkhead Transcription Factors/drug effects , GABAergic Neurons/drug effects , Heat-Shock Proteins/drug effects , Hypoxia/metabolism , Sulfites/toxicity , Animals , Behavior, Animal/drug effects , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , Forkhead Transcription Factors/metabolism , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Heat-Shock Proteins/metabolism , Hydrogen Peroxide/toxicity , Hypoxia/pathology , Microscopy, Fluorescence , Oxidants/toxicity , Signal Transduction , Survival Rate
11.
Mol Pharmacol ; 97(6): 402-408, 2020 06.
Article in English | MEDLINE | ID: mdl-32276963

ABSTRACT

The 78-kDa glucose-regulated protein (GRP78), an endoplasmic reticulum (ER) chaperone, is a master regulator of the ER stress. A number of studies revealed that high levels of GRP78 protein in cancer cells confer multidrug resistance (MDR) to therapeutic treatment. Therefore, drug candidate that reduces GRP78 may represent a novel approach to eliminate MDR cancer cells. Our earlier studies showed that a set of 4H-chromene derivatives induced selective cytotoxicity in MDR cancer cells. In the present study, we elucidated its selective mechanism in four MDR cancer cell lines with one lead candidate (CXL146). Cytotoxicity results confirmed the selective cytotoxicity of CXL146 toward the MDR cancer cell lines. We noted significant overexpression of GRP78 in all four MDR cell lines compared with the parental cell lines. Unexpectedly, CXL146 treatment rapidly and dose-dependently reduced GRP78 protein in MDR cancer cell lines. Using human leukemia (HL) 60/mitoxantrone (MX) 2 cell line as the model, we demonstrated that CXL146 treatment activated the unfolded protein response (UPR); as evidenced by the activation of inositol-requiring enzyme 1α, protein kinase R-like ER kinase, and activating transcription factor 6. CXL146-induced UPR activation led to a series of downstream events, including extracellular signal-regulated kinase 1/2 and c-Jun N-terminal kinase activation, which contributed to CXL146-induced apoptosis. Targeted reduction in GRP78 resulted in reduced sensitivity of HL60/MX2 toward CXL146. Long-term sublethal CXL146 exposure also led to reduction in GRP78 in HL60/MX2. These data collectively support GRP78 as the target of CXL146 in MDR treatment. Interestingly, HL60/MX2 upon long-term sublethal CXL146 exposure regained sensitivity to mitoxantrone treatment. Therefore, further exploration of CXL146 as a novel therapy in treating MDR cancer cells is warranted. SIGNIFICANCE STATEMENT: Multidrug resistance is one major challenge to cancer treatment. This study provides evidence that cancer cells overexpress 78-kDa glucose-regulated protein (GRP78) as a mechanism to acquire resistance to standard cancer therapies. A chromene-based small molecule, CXL146, selectively eliminates cancer cells with GRP78 overexpression via activating unfolded protein response-mediated apoptosis. Further characterization indicates that CXL146 and standard therapies complementarily target different populations of cancer cells, supporting the potential of CXL146 to overcome multidrug resistance in cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Drug Resistance, Neoplasm/drug effects , Heat-Shock Proteins/metabolism , CRISPR-Cas Systems , Cell Line, Tumor , Drug Resistance, Multiple/drug effects , Drug Screening Assays, Antitumor , Endoplasmic Reticulum Chaperone BiP , HL-60 Cells , Heat-Shock Proteins/drug effects , Humans , Mitoxantrone/pharmacology
12.
Acta Cir Bras ; 35(1): e202000104, 2020.
Article in English | MEDLINE | ID: mdl-32159588

ABSTRACT

PURPOSE: Glutamine, as an essential part of enteral nutrition and parenteral nutrition agent, has been widely recognized to be a kind of important intestinal mucosa protectant in clinical practice and experimental research. However, the mechanisms of its protective effects are still not fully understand. Consequently, this study aimed to explore the potential mechanism of glutamine on ischemia-reperfusion (I/R) injury induced endoplasmic reticulum (ER) stress in intestine. METHODS: An experimental model of intestinal I/R in rats was established by 1 hour occlusion of the superior mesenteric artery followed by 3 hours of reperfusion. Morphologic changes of intestinal mucosa, apoptosis of epithelial cells, and expression of intestinal Grp78, Gadd153, Caspase-12, ATF4, PERK phosphorylation (P-PERK) and elF2αphosphorylation(P-elF2α) were determined. RESULTS: After I/R, the apoptotic index of intestinal mucosa epithelial cells observably increased with notable necrosis of intestinal mucosa, and the expressions of Grp78, Gadd153, Caspase-12, ATF4, P-PERK and P-elF2αall were increased. However, treatment with glutamine could significantly relieve intestinal I/R injury and apoptosis index. Moreover, glutamine could clearly up-regulate the expression of Grp78, restrain P-PERK and P-elF2α, and reduce ATF4, Gadd153 and Caspase-12 expressions. CONCLUSION: Glutamine may be involved in alleviating ER stress induced intestinal mucosa cells apoptosis.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Glutamine/pharmacology , Intestinal Mucosa/drug effects , Protective Agents/pharmacology , Reperfusion Injury/prevention & control , Activating Transcription Factor 4/drug effects , Animals , Caspase 12/drug effects , Heat-Shock Proteins/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/ultrastructure , Male , Mesenteric Artery, Superior/injuries , Models, Animal , RNA, Messenger/drug effects , Rats, Sprague-Dawley , Transcription Factor CHOP/drug effects , eIF-2 Kinase/drug effects
13.
Article in English | MEDLINE | ID: mdl-32094131

ABSTRACT

Antiretroviral therapy (ART) suppresses HIV-1 replication but fails to cure the infection. The presence of an extremely stable viral latent reservoir, primarily in resting memory CD4+ T cells, remains a major obstacle to viral eradication. The "shock and kill" strategy targets these latently infected cells and boosts immune recognition and clearance, and thus, it is a promising approach for an HIV-1 functional cure. Although some latency-reversing agents (LRAs) have been reported, no apparent clinical progress has been made, so it is still vital to seek novel and effective LRAs. Here, we report that thiostrepton (TSR), a proteasome inhibitor, reactivates latent HIV-1 effectively in cellular models and in primary CD4+ T cells from ART-suppressed individuals ex vivo TSR does not induce global T cell activation, severe cytotoxicity, or CD8+ T cell dysfunction, making it a prospective LRA candidate. We also observed a significant synergistic effect of reactivation when TSR was combined with JQ1, prostratin, or bryostatin-1. Interestingly, six TSR analogues also show reactivation abilities that are similar to or more effective than that of TSR. We further verified that TSR upregulated expression of heat shock proteins (HSPs) in CD4+ T cells, which subsequently activated positive transcriptional elongation factor b (p-TEFb) and NF-κB signals, leading to viral reactivation. In summary, we identify TSR as a novel LRA which could have important significance for applications to an HIV-1 functional cure in the future.


Subject(s)
Anti-HIV Agents/pharmacology , Antiviral Agents/pharmacology , HIV Infections/drug therapy , HIV-1/drug effects , Heat-Shock Proteins/drug effects , NF-kappa B/drug effects , Positive Transcriptional Elongation Factor B/drug effects , Signal Transduction/drug effects , Thiostrepton/pharmacology , Virus Activation/drug effects , Virus Latency/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Cell Line , Drug Synergism , HIV Infections/virology , High-Throughput Screening Assays , Humans , Prospective Studies
14.
Cell Stress Chaperones ; 25(1): 173-191, 2020 01.
Article in English | MEDLINE | ID: mdl-31900865

ABSTRACT

Upregulation of heat shock proteins (HSPs) is an approach to treatment of neurodegenerative disorders with impaired proteostasis. Many neurons, including motor neurons affected in amyotrophic lateral sclerosis (ALS), are relatively resistant to stress-induced upregulation of HSPs. This study demonstrated that histone deacetylase (HDAC) inhibitors enable the heat shock response in cultured spinal motor neurons, in a stress-dependent manner, and can improve the efficacy of HSP-inducing drugs in murine spinal cord cultures subjected to thermal or proteotoxic stress. The effect of particular HDAC inhibitors differed with the stress paradigm. The HDAC6 (class IIb) inhibitor, tubastatin A, acted as a co-inducer of Hsp70 (HSPA1A) expression with heat shock, but not with proteotoxic stress induced by expression of mutant SOD1 linked to familial ALS. Certain HDAC class I inhibitors (the pan inhibitor, SAHA, or the HDAC1/3 inhibitor, RGFP109) were HSP co-inducers comparable to the hydroxyamine arimoclomol in response to proteotoxic stress, but not thermal stress. Regardless, stress-induced Hsp70 expression could be enhanced by combining an HDAC inhibitor with either arimoclomol or with an HSP90 inhibitor that constitutively induced HSPs. HDAC inhibition failed to induce Hsp70 in motor neurons expressing ALS-linked mutant FUS, in which the heat shock response was suppressed; yet SAHA, RGFP109, and arimoclomol did reduce loss of nuclear FUS, a disease hallmark, and HDAC inhibition rescued the DNA repair response in iPSC-derived motor neurons carrying the FUSP525Lmutation, pointing to multiple mechanisms of neuroprotection by both HDAC inhibiting drugs and arimoclomol.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Heat-Shock Proteins/drug effects , Hydroxylamines/pharmacology , Motor Neurons/drug effects , Spinal Cord/drug effects , Amyotrophic Lateral Sclerosis/genetics , Animals , Cells, Cultured , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Response/drug effects , Histone Deacetylase Inhibitors/pharmacology , Mice , Motor Neurons/metabolism , Spinal Cord/metabolism , Transcriptional Activation/drug effects , Up-Regulation/drug effects
15.
Rheumatology (Oxford) ; 59(4): 742-753, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31377809

ABSTRACT

OBJECTIVES: Xerostomia in SS patients has been associated with low quality and quantity of salivary mucins, which are fundamental for the hydration and protection of the oral mucosa. The aim of this study was to evaluate if cytokines induce aberrant mucin expression and whether tauroursodeoxycholic acid (TUDCA) is able to counteract such an anomaly. METHODS: Labial salivary glands from 16 SS patients and 15 control subjects, as well as 3D acini or human submandibular gland cells stimulated with TNF-α or IFN-γ and co-incubated with TUDCA, were analysed. mRNA and protein levels of Mucin 1 (MUC1) and MUC7 were determined by RT-qPCR and western blot, respectively. Co-immunoprecipitation and immunofluorescence assays for mucins and GRP78 [an endoplasmic reticulum (ER)-resident protein] were also performed. mRNA levels of RelA/p65 (nuclear factor-κB subunit), TNF-α, IL-1ß, IL-6, SEL1L and EDEM1 were determined by RT-qPCR, and RelA/p65 localization was evaluated by immunofluorescence. RESULTS: MUC1 is overexpressed and accumulated in the ER of labial salivary gland from SS patients, while MUC7 accumulates throughout the cytoplasm of acinar cells; however, MUC1, but not MUC7, co-precipitated with GRP78. TUDCA diminished the overexpression and aberrant accumulation of MUC1 induced by TNF-α and IFN-γ, as well as the nuclear translocation of RelA/p65, together with the expression of inflammatory and ER stress markers in 3D acini. CONCLUSION: Chronic inflammation alters the secretory process of MUC1, inducing ER stress and affecting the quality of saliva in SS patients. TUDCA showed anti-inflammatory properties decreasing aberrant MUC1 accumulation. Further studies are necessary to evaluate the potential therapeutic effect of TUDCA in restoring glandular homeostasis in SS patients.


Subject(s)
Acinar Cells/drug effects , Endoplasmic Reticulum Stress/drug effects , Mucin-1/drug effects , Salivary Glands, Minor/drug effects , Sjogren's Syndrome/metabolism , Submandibular Gland/drug effects , Taurochenodeoxycholic Acid/pharmacology , Xerostomia/metabolism , Acinar Cells/metabolism , Adult , Aged , Case-Control Studies , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/genetics , Female , Heat-Shock Proteins/drug effects , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Immunoprecipitation , In Vitro Techniques , Interferon-gamma/pharmacology , Interleukin-1beta/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Membrane Proteins/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Middle Aged , Mucin-1/genetics , Mucin-1/metabolism , Mucins/drug effects , Mucins/genetics , Mucins/metabolism , Proteins/drug effects , Proteins/genetics , Proteins/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Salivary Glands, Minor/metabolism , Salivary Proteins and Peptides/drug effects , Salivary Proteins and Peptides/genetics , Salivary Proteins and Peptides/metabolism , Sjogren's Syndrome/genetics , Submandibular Gland/cytology , Submandibular Gland/metabolism , Transcription Factor RelA/drug effects , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Xerostomia/genetics
16.
J Chemother ; 32(1): 7-14, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31631801

ABSTRACT

Escherichia coli is a common pathogen of bacterial biofilm infections. Sub-minimum inhibitory concentration ceftazidime (sub-MIC CAZ) could inhibit the biofilm formation of E. coli. Deletion of the ibpAB genes could increase the extracellular indole concentration of E. coli and then inhibit biofilm formation. Therefore, we speculated that sub-MIC CAZ might inhibit biofilm formation via ibpAB. In this study, the results showed that sub-MIC CAZ could significantly inhibit biofilm formation, swimming motility and the expression of the ibpA gene, while it could increase the expression of tnaA gene and extracellular indole concentration. Knockout of the ibpA gene resulted in a decrease in biofilm formation and swimming motility and an increase in the indole concentration. When treated with sub-MIC CAZ, the tnaA gene expression, indole concentration, biofilm formation and swimming motility of MG1655 ΔibpA were similar to those of the control group. The results indicated that sub-MIC CAZ might inhibit the biofilm formation of E. coli by increasing the extracellular indole concentration and downregulating the ibpA gene.


Subject(s)
Anti-Bacterial Agents/pharmacology , Ceftazidime/pharmacology , Escherichia coli Proteins/drug effects , Escherichia coli/physiology , Gene Expression Regulation, Bacterial/drug effects , Heat-Shock Proteins/drug effects , Biofilms , Indoles/metabolism , Microbial Sensitivity Tests
17.
Cell Stress Chaperones ; 25(1): 47-56, 2020 01.
Article in English | MEDLINE | ID: mdl-31745845

ABSTRACT

Our previous study had shown that chronic corticosterone (CORT) exposure causes excessive fat deposition in chicken liver, yet it remains unknown whether it is associated with inflammation and fibrosis. In general, heat shock proteins (HSPs) are activated in response to acute stress to play a cytoprotective role, and this activation is associated with m6A-mediated post-transcriptional regulation. However, changes of HSPs and the m6A methylation on their mRNAs in response to chronic CORT treatment in chicken liver have not been reported. In this study, chronic CORT exposure induced inflammation and fibrosis in chicken liver, associated with significantly modulated expression of HSPs that was significantly upregulated at mRNA level yet downregulated at protein level. Concurrently, m6A methyltransferases METTL3 content was upregulated together with the level of m6A methylation on HSPs transcripts. The m6A-seq analysis revealed 2-6 significantly (P < 0.05) hypermethylated m6A peaks in the mRNA of 4 different species of HSPs in CORT-treated chicken liver. HSP90B1 transcript had 6 differentially methylated m6A peaks among which peaks on exon 16 and exon 17 showed 3.14- and 4.72-fold of increase, respectively. Mutation of the 8 predicted m6A sites on exon 16 and exon 17 resulted in a significant (P < 0.05) increase in eGFP-fused content of HSP90B1 exon 16 and exon 17 fragment in 293 T cells, indicating a possible role of m6A in post-transcriptional regulation of HSPs. In conclusion, chronic CORT exposure induces inflammation and fibrosis in chicken liver along with an increase in the levels and m6A methylation of several HSPs mRNAs; HSPs levels were however reduced under the indicated conditions. Results presented suggest that the reduction in HSPs levels may be associated with m6A methylation in CORT-exposed chickens.


Subject(s)
Corticosterone/pharmacology , Fibrosis/chemically induced , Gene Expression Regulation/drug effects , Heat-Shock Proteins/drug effects , Inflammation/chemically induced , Animals , Chickens , DNA Methylation/drug effects , Fibrosis/drug therapy , Heat-Shock Proteins/metabolism , Inflammation/drug therapy , Inflammation/genetics , Liver Cirrhosis/drug therapy , Methyltransferases/genetics
18.
Acta cir. bras ; 35(1): e202000104, 2020. graf
Article in English | LILACS | ID: biblio-1088525

ABSTRACT

Abstract Purpose Glutamine, as an essential part of enteral nutrition and parenteral nutrition agent, has been widely recognized to be a kind of important intestinal mucosa protectant in clinical practice and experimental research. However, the mechanisms of its protective effects are still not fully understand. Consequently, this study aimed to explore the potential mechanism of glutamine on ischemia-reperfusion (I/R) injury induced endoplasmic reticulum (ER) stress in intestine. Methods An experimental model of intestinal I/R in rats was established by 1 hour occlusion of the superior mesenteric artery followed by 3 hours of reperfusion. Morphologic changes of intestinal mucosa, apoptosis of epithelial cells, and expression of intestinal Grp78, Gadd153, Caspase-12, ATF4, PERK phosphorylation (P-PERK) and elF2αphosphorylation(P-elF2α) were determined. Results After I/R, the apoptotic index of intestinal mucosa epithelial cells observably increased with notable necrosis of intestinal mucosa, and the expressions of Grp78, Gadd153, Caspase-12, ATF4, P-PERK and P-elF2αall were increased. However, treatment with glutamine could significantly relieve intestinal I/R injury and apoptosis index. Moreover, glutamine could clearly up-regulate the expression of Grp78, restrain P-PERK and P-elF2α, and reduce ATF4, Gadd153 and Caspase-12 expressions. Conclusion Glutamine may be involved in alleviating ER stress induced intestinal mucosa cells apoptosis.


Subject(s)
Animals , Male , Reperfusion Injury/prevention & control , Apoptosis/drug effects , Protective Agents/pharmacology , Endoplasmic Reticulum Stress/drug effects , Glutamine/pharmacology , Intestinal Mucosa/drug effects , RNA, Messenger/drug effects , Rats, Sprague-Dawley , Mesenteric Artery, Superior/injuries , eIF-2 Kinase/drug effects , Models, Animal , Activating Transcription Factor 4/drug effects , Transcription Factor CHOP/drug effects , Caspase 12/drug effects , Heat-Shock Proteins/drug effects , Intestinal Mucosa , Intestinal Mucosa/ultrastructure
19.
Int J Mol Sci ; 20(22)2019 Nov 18.
Article in English | MEDLINE | ID: mdl-31752169

ABSTRACT

Heat shock proteins (HSPs) are evolutionary conserved proteins that work as molecular chaperones and perform broad and crucial roles in proteostasis, an important process to preserve the integrity of proteins in different cell types, in health and disease. Their function in cancer is an important aspect to be considered for a better understanding of disease development and progression. Glioblastoma (GBM) is the most frequent and lethal brain cancer, with no effective therapies. In recent years, HSPs have been considered as possible targets for GBM therapy due their importance in different mechanisms that govern GBM malignance. In this review, we address current evidence on the role of several HSPs in the biology of GBMs, and how these molecules have been considered in different treatments in the context of this disease, including their activities in glioblastoma stem-like cells (GSCs), a small subpopulation able to drive GBM growth. Additionally, we highlight recent works that approach other classes of chaperones, such as histone and mitochondrial chaperones, as important molecules for GBM aggressiveness. Herein, we provide new insights into how HSPs and their partners play pivotal roles in GBM biology and may open new therapeutic avenues for GBM based on proteostasis machinery.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Heat-Shock Proteins/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/drug therapy , Heat-Shock Proteins/drug effects , Humans , Molecular Targeted Therapy
20.
Toxicol Ind Health ; 35(7): 457-465, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31364504

ABSTRACT

Solar ultraviolet (UV) radiation is the main factor of photocarcinogenesis, photoaging, and photosensitivity; thus protection from biological damaging UV radiation is a concern. Sunscreens containing UV filters are the most preferred means of photoprotection but the safety and efficacy of UV filters are in question. Benzophenone (BP) and its derivatives, namely, benzophenone 1 (BP1), is commonly used in sunscreens as a UV blocker. The aim of this study was to assess the effects of BP and BP1 on the differential expression of proteins in human keratinocytes (HaCaT cells) under exposure to ultraviolet A radiation. Photosensitive proteins were screened from HaCaT cells by two-dimensional (2-D) gel electrophoresis, and identification of these differentially expressed proteins was performed by matrix-assisted laser desorption/ionization-time-of-flight (MALDI-TOF)/TOF mass spectrometry. Protein identification was performed using the search program MASCOT and a database made of SUMO and GhJMJ12 amino acid sequences. Our results showed that the proteins involved directly or indirectly in apoptosis are 70 kDa heat shock protein, long-chain specific acyl-CoA dehydrogenase, serine/threonine-protein kinase, and FAM78A protein, which were upregulated in comparison to control HaCaT cells. The expressions of binding immunoglobulin protein, podocalyxin-like protein, actin, cytoplasmic, and calreticulin precursors were downregulated. The altered protein expression indicated that cell growth arrest and apoptosis were potential mechanisms of cytotoxicity and genotoxicity of BPs. The results of 2-D gel electrophoresis followed by mass spectrometry showed expression of novel proteins involved in promoting or initiating apoptotic pathways. Hence, we conclude that BPs should be avoided as a UV blocker from sunscreens because of its potential to promote apoptotic proteins in human skin keratinocytes.


Subject(s)
Benzophenones/pharmacology , Keratinocytes/drug effects , Sunscreening Agents/pharmacology , Ultraviolet Rays , Apoptosis/drug effects , Biomarkers , Electrophoresis, Gel, Two-Dimensional , Heat-Shock Proteins/drug effects , Humans , Keratinocytes/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...