Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Photochem Photobiol Sci ; 23(9): 1771-1782, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39249169

ABSTRACT

Potent antioxidants, like 3-hydroxy flavones, attracted considerable attention due to their excited state intramolecular proton transfer (ESIPT)-based fluorescence behaviour. This article is an interesting demonstration of a series of synthetic 3-hydroxy flavone analogues having high antioxidant activity as molecular rotor-like viscosity probes. Among these flavone analogues, 4'-N,N-dimethylamino-3-hydroxy flavone (3) is the most potent one, showing the twisted intramolecular charge transfer (TICT)-dependent fluoroprobing activity toward the blood viscosity changes associated with diabetes and free fatty acids (FFA)-induced nuclear viscosity changes of MIN6 cells. The TICT dynamics of (3), which instigates its viscosity probing activity, was comprehended with the help of DFT-based computational studies. Abnormal cellular viscosity changes are the pathological traits for various diseases, and non-toxic flavone-based viscosity probes can be useful for diagnosing such pathological conditions.


Subject(s)
Antioxidants , Density Functional Theory , Flavones , Flavones/chemistry , Flavones/pharmacology , Viscosity , Antioxidants/chemistry , Antioxidants/pharmacology , Diabetes Mellitus/drug therapy , Animals , Protons , Mice , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Fluorescent Dyes/chemical synthesis , Fatty Acids, Nonesterified/chemistry , Fatty Acids, Nonesterified/metabolism , Humans
2.
J Cell Sci ; 134(21)2021 11 01.
Article in English | MEDLINE | ID: mdl-34643217

ABSTRACT

Elevated fetuin-A levels, chemokines and islet-resident macrophages are crucial factors associated with obesity-mediated type 2 diabetes (T2D). Here, the aim of the study was to investigate the effect of MIN6 (a mouse insulinoma cell line)-derived fetuin-A (also known as AHSG) in macrophage polarization and decipher the effect of M1 type pro-inflammatory macrophages in commanding over insulin secretion. MIN6 and islet-derived fetuin-A induced expression of the M1 type macrophage markers Emr1 (also known as Adgre1), Cd68 and CD11c (Itgax) (∼1.8 fold) along with increased cytokine secretion. Interestingly, suppression of fetuin-A in MIN6 successfully reduced M1 markers by ∼1.5 fold. MIN6-derived fetuin-A also induced chemotaxis of macrophages in a Boyden chamber chemotaxis assay. Furthermore, high-fat feeding in mice showed elevated cytokine and fetuin-A content in serum and islets, and also migration and polarization of macrophages to the islets, while ß-cells failed to meet the increased insulin demand. Moreover, in MIN6 culture, M1 macrophages sharply decreased insulin secretion by ∼2.8 fold. Altogether our results support an association of fetuin-A with islet inflammation and ß-cell dysfunction, owing to its role as a key chemoattractant and macrophage polarizing factor.


Subject(s)
Diabetes Mellitus, Type 2 , Islets of Langerhans , alpha-2-HS-Glycoprotein , Animals , Inflammation/metabolism , Insulin/metabolism , Insulin Secretion , Macrophages/metabolism , Mice , alpha-2-HS-Glycoprotein/metabolism
3.
Biochem Biophys Res Commun ; 647: 55-61, 2023 03 05.
Article in English | MEDLINE | ID: mdl-36716646

ABSTRACT

Dipeptidyl peptidase 4 (DPP-IV) is a ubiquitous proteolytic enzyme that cleaves incretin hormones, such as glucagon-like peptide 1 (GLP1) and gastric inhibitory protein (GIP), leading to reduced glucose stimulated insulin secretion from the pancreatic beta cells. The functionally active enzyme is present in a membrane bound form in several cell types as well as in a soluble form in the circulation. The present report deals with DPP-IV expression and its regulation in the pancreatic beta cells in presence of free fatty acids (FFAs) and Fetuin-A, a circulatory glycoprotein associated with insulin resistance in humans and animals. FFA and Fetuin-A individually or in combination trigger DPP-IV expression in MIN6 cells. Islets isolated from high fat diet fed (HFD) mice (16 weeks) showed higher levels of DPP-IV expression than standard diet (SD) fed mice. Fetuin-A increased DPP-IV expression in HFD mice (4 weeks). Inhibition of TLR4 or NFkB prevented palmitate-Fetuin-A mediated DPP-IV expression in MIN6. It has been seen that Fetuin-A alone also could trigger DPP-IV expression in MIN6 cells via NFkB. Additionally, palmitate treatment exhibited reduced level of soluble DPP-IV in the media of MIN6 culture, which corroborated with the expression pattern of its protease, KLK5 that cleaves and releases the membrane bound DPP-IV into the secretion. Our results demonstrate that FFA-Fetuin-A upregulates DPP-IV expression in the pancreatic beta cells through the TLR4-NFkB pathway.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors , Insulin-Secreting Cells , Humans , Animals , Mice , Insulin-Secreting Cells/metabolism , alpha-2-HS-Glycoprotein/metabolism , Toll-Like Receptor 4/metabolism , Dipeptidyl Peptidase 4/metabolism , Fatty Acids, Nonesterified/metabolism , Palmitates/metabolism , Insulin/metabolism
4.
J Cell Physiol ; 237(1): 532-550, 2022 01.
Article in English | MEDLINE | ID: mdl-34224584

ABSTRACT

Fetuin-A, a hepato-adipokine, is associated with lipid-mediated islet inflammation and inflicts ß-cell death but the underlying mechanisms are still unclear. In an earlier report, it was shown that fetuin-A promotes lipid-induced insulin resistance by acting as an endogenous ligand of toll like receptor 4. Recently, we have also reported that ß-cells secrete fetuin-A on stimulation by palmitate causing ß-cell dysfunction. The aim of this study was twofold: (a) screening the role of fetuin-A in survival of murine ß-cells, and (b) to validate the effect of fetuin-A release and lipid induced apoptosis in mouse insulinoma cell line MIN6. Excess of lipid and fetuin-A in circulation induced significant deterioration of islet histoarchitecture and impeded insulin secretion by 2.7 ± 0.5-folds in 20 weeks high fat diet mice. Administration of fetuin-A (0.7 mg/g) along with 4 weeks of HFD produced similar results as 20 weeks of high fat feeding. Treating high doses of palmitate alone (0.50 mM) as well as in combination with fetuin-A (100 µg/ml) for 24 h inflicted apoptosis in MIN6 through the mitochondrial pathway. Knockdown of fetuin-A gene partially inhibited palmitate inflicted apoptosis in MIN6 by 1.83 ± 0.25 times, however, fetuin-A when added in the medium caused re-emergence of apoptosis. Notably, apoptosis induced by palmitate conditioned media from MIN6, 3T3L1, and HepG2, was partially inhibited in fetuin-A KD MIN6. These results confirmed the critical role of circulatory fetuin-A and ß-cell secreted fetuin-A in ß-cell dysfunction and apoptosis under hyperlipidemic conditions.


Subject(s)
Insulin-Secreting Cells , alpha-2-HS-Glycoprotein , Animals , Apoptosis , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice , Palmitates/pharmacology , alpha-2-HS-Glycoprotein/genetics , alpha-2-HS-Glycoprotein/metabolism
5.
Arch Biochem Biophys ; 697: 108700, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33271149

ABSTRACT

Chemoprevention failure is considered to be the most emerging problem that makes non-small cell lung cancer (NSCLC) as one of the deadliest malignancies in the world. In NSCLC cells, Nuclear factor erythroid 2-related factor 2 (Nrf2), a redox sensitive transcription factor, promotes cancer cell survival and fosters mechanism for drug resistance. Here we report identification of Kaempferol, a dietary flavonoid, as a potent Nrf2 inhibitor using Nrf2 reporter assay in NSCLC cells (A549 and NCIH460). Kaempferol selectively reduces Nrf2 mRNA and protein levels and lower level of nuclear Nrf2 downregulates transcription of Nrf2 target genes (NQO1, HO1, AKR1C1 and GST). Kaempferol (25 µM) mediated downregulation of GST, NQO1 and HO1 expression is also observed even after stimulation of Nrf2 by tert-butylhydroquinone (tBHQ). Again, Kaempferol incubation does not change the levels of NFκBp65 and phospho NFκBp65, suggesting it hampers Nrf2 signalling pathway in these cells. Nrf2 inhibition by Kaempferol induces ROS accumulation after 48 h of treatment and makes NSCLC cells sensitive to apoptosis at physiological concentration. Taken together, our study demonstrates that Kaempferol is a potent inhibitor of Nrf2 and can be used as a natural sensitizer and anti-cancer agent for lung cancer therapeutics.


Subject(s)
Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/pathology , Down-Regulation/drug effects , Kaempferols/pharmacology , Lung Neoplasms/pathology , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , A549 Cells , Humans , NF-E2-Related Factor 2/genetics , RNA, Messenger/genetics
6.
Chem Res Toxicol ; 33(2): 651-656, 2020 02 17.
Article in English | MEDLINE | ID: mdl-31944672

ABSTRACT

A nuclear-localized fluorescent light-up probe, NucFP-NO2, was designed and synthesized that can detect CO selectively in an aqueous buffer (pH 7.4, 37 °C) through the CO-mediated transformation of the nitro group into an amino-functionalized moiety. This probe triggered a more than 55-fold "turn-on" fluorescence response to CO without using any metal ions, e.g., Pd, Rh, Fe, etc. The enhanced response is highly selective over a variety of relevant reactive oxygen, nitrogen, and sulfur species and also various biologically important cationic, anionic, and neutral species. The detection limit of this probe for CO is as low as 0.18 µM with a linear range of 0-70 µM. Also, this fluorogenic probe is an efficient candidate for monitoring intracellular CO in living cells (RAW 264.7, A549 cells), and the fluorescence signals predominantly localize in the nuclear region.


Subject(s)
Carbon Monoxide/analysis , Cell Nucleus/chemistry , Fluorescent Dyes/analysis , Fluorescent Dyes/chemistry , Naphthalimides/analysis , Naphthalimides/chemistry , A549 Cells , Animals , Cell Survival , Fluorescent Dyes/chemical synthesis , Humans , Mice , Molecular Structure , Naphthalimides/chemical synthesis , Optical Imaging , RAW 264.7 Cells , Spectrometry, Fluorescence
7.
Bioconjug Chem ; 30(5): 1575-1583, 2019 05 15.
Article in English | MEDLINE | ID: mdl-31009567

ABSTRACT

The innocent silicon quantum dots (SQDs) having dual emissive property (blue in VIS and red in NIR), high photostability, and freedom from auto fluorescence are designed and synthesized for the first time using ethylene glycol. A new attempt has been made for direct labeling of Alpha 2-HS-Glycoprotein (Fetuin A) through functionalization of the synthesized dots by EDC coupling. The SQDs were characterized by FTIR, TEM, AFM, XRD, EDX, DLS, and TGA. The chemistry involved in the synthesis and functionalization of dots is elucidated in detail. The synthesized SQDs are suitable for live cell imaging as well as direct labeling of the Fetuin A in the NIR region. The direct labeling technique developed for Fetuin A imaging is robust, more specific, and simple, and reduces the number of incubation and washing steps and produces better quality data compared to the conventional method using Rhodamine B.


Subject(s)
Quantum Dots/chemistry , Silicon/chemistry , alpha-2-HS-Glycoprotein/chemistry , Ethylene Glycol/chemistry , Humans , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
8.
J Fish Biol ; 95(1): 324-334, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30238997

ABSTRACT

The chemosensory nature of the tissue from the dorsal surface of the head (also termed sensory pad; SP) of the amphihaline diadromous fish hilsa Tenualosa ilisha was investigated for odorant receptor (OR), olfactory marker protein (OMP) and G-protein subunits (Gαs-olf, Gαq, Gαo, Gαi3) through immunolocalization and immunoblotting techniques. The immunolocalization of OR, OMP and G-protein subunits showed clear expression of these proteins in the tissues of the SP. Robust expressions of these proteins in the SP were detected with immunoblot analysis. The strong expression of these proteins in the SP indicates that the tissues from this area in riverine T. ilisha may play significant role in chemosensing and signalling through ectopic expression of olfactory receptor proteins which are otherwise reported in olfactory organs in vertebrates. Being migratory in nature, ectopic expression of these receptors in T. ilisha probably helps them to prevent damage to epidermal tissues of the SP, or they may also utilize them as a chemo and mechanosensory tool to optimize chemo-communications during migration.


Subject(s)
Fishes/metabolism , GTP-Binding Proteins/metabolism , Receptors, Odorant/metabolism , Animals , Ectopic Gene Expression , Epidermis/metabolism , Epidermis/ultrastructure , Female , Fishes/genetics , GTP-Binding Proteins/genetics , Head/anatomy & histology , Immunoblotting , Immunohistochemistry , Male , Olfactory Receptor Neurons , Protein Subunits/metabolism , Signal Transduction
9.
Biochem Biophys Res Commun ; 491(4): 1118-1124, 2017 09 30.
Article in English | MEDLINE | ID: mdl-28797566

ABSTRACT

Islets of type 2 diabetes patients display inflammation, elevated levels of cytokines and macrophages. The master regulator of inflammation in the islets is free fatty acids (FFA). It has already been reported that FFA and TLR4 stimulation induces pro-inflammatory factors in the islets. In this report we demonstrate that excess lipid triggers Fetuin-A (FetA) secretion from the pancreatic ß-cells. Palmitate treatment to MIN6 cells showed significantly elevated FetA levels in respect to their controls. Fatty acid induces the FetA gene and protein expression in the pancreatic ß-cells via TLR4 and over-expression of NF-κB. In the NF-κB knocked down MIN6 cells palmitate could not trigger FetA release into the incubation medium. These results suggest that NF-κB mediates palmitate stimulated FetA secretion from the pancreatic ß-cells. Blocking the activity of TLR4 by CLI-095 incubation or TLR4 siRNA restored insulin secretion which confirmed the role of TLR4 in FFA-FetA mediated pancreatic ß-cell dysfunction. Palmitate mediated expression of NF-κB enahnced inflammatory response through expression of cytokines such as IL-1ß and IL-6. These results suggest that FFA mediated FetA secretion from pancreatic ß-cells lead to their dysfunction via FFA-TLR4 pathway. FetA thus creates an inflammatory environment in the pancreatic islets that can become a possible cause behind pancreatic ß-cell dysfunction in chronic hyperlipidemic condition.


Subject(s)
Inflammation/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Palmitates/pharmacology , alpha-2-HS-Glycoprotein/metabolism , Animals , Dose-Response Relationship, Drug , Mice , Structure-Activity Relationship , Tumor Cells, Cultured , alpha-2-HS-Glycoprotein/genetics
10.
Mol Cell Biochem ; 405(1-2): 149-58, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25893736

ABSTRACT

Non-small cell lung cancer (NSCLC) is known to be a difficult cancer to treat because of its poor prognosis, limited option for surgery, and resistance to chemo or radiotherapy. In this study, we have demonstrated that suppression of rictor expression in A549 and H1299 NSCLC cells by mahanine, a carbazole alkaloid, disrupted constitutive activation of mTOR and Akt. Mahanine suppression of rictor gene expression and consequent attenuation of its protein expression affected the inhibition of mTOR (Ser-2481) and Akt (Ser-473) phosphorylation. Since mahanine treatment revealed this new insight of rictor-mTOR relationship, we examined an association between mTOR activation with rictor expression. Interestingly, in rictor knockdown (KD) NSCLC cells, mTOR activation was significantly impaired. Transfection of rictor over-expression vector into the NSCLC cells reversed this situation. In fact, both rictor KD and mahanine treated cells showed considerably depleted phospho-mTOR level. These results indicate that rictor is required to maintain constitutive activation of mTOR in lung cancer cells. When mTOR kinase activity in rictor KD cells was examined with Akt as substrate, a significant reduction of Akt phosphorylation indicated impairment of mTOR kinase potentiality. Disruption of mTOR and Akt activation caused drastic mortality of NSCLC cancer cells through apoptosis. Hence, our study reveals a new dimension in mTOR-rictor relationship, where rictor stands to be a suitable therapeutic target for lung cancer.


Subject(s)
Alkaloids/pharmacology , Apoptosis/drug effects , Carbazoles/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carrier Proteins/metabolism , Lung Neoplasms/drug therapy , TOR Serine-Threonine Kinases/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Humans , Lung Neoplasms/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Signal Transduction/drug effects
11.
J Biol Chem ; 288(39): 28324-30, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-23943623

ABSTRACT

Macrophage infiltration into adipose tissue during obesity and their phenotypic conversion from anti-inflammatory M2 to proinflammatory M1 subtype significantly contributes to develop a link between inflammation and insulin resistance; signaling molecule(s) for these events, however, remains poorly understood. We demonstrate here that excess lipid in the adipose tissue environment may trigger one such signal. Adipose tissue from obese diabetic db/db mice, high fat diet-fed mice, and obese diabetic patients showed significantly elevated fetuin-A (FetA) levels in respect to their controls; partially hepatectomized high fat diet mice did not show noticeable alteration, indicating adipose tissue to be the source of this alteration. In adipocytes, fatty acid induces FetA gene and protein expressions, resulting in its copious release. We found that FetA could act as a chemoattractant for macrophages. To simulate lipid-induced inflammatory conditions when proinflammatory adipose tissue and macrophages create a niche of an altered microenvironment, we set up a transculture system of macrophages and adipocytes; the addition of fatty acid to adipocytes released FetA into the medium, which polarized M2 macrophages to M1. This was further confirmed by direct FetA addition to macrophages. Taken together, lipid-induced FetA from adipocytes is an efficient chemokine for macrophage migration and polarization. These findings open a new dimension for understanding obesity-induced inflammation.


Subject(s)
Adipocytes/cytology , Adipose Tissue/metabolism , Macrophages/cytology , alpha-2-HS-Glycoprotein/metabolism , Aged , Animals , Cell Movement , Endotoxins/metabolism , Female , Humans , Inflammation , Lipids/chemistry , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Obesity/metabolism , Signal Transduction
12.
Toxicol Ind Health ; 30(7): 611-20, 2014 Aug.
Article in English | MEDLINE | ID: mdl-23033210

ABSTRACT

In the present study, we attempted to elucidate the induction of autophagy in rat hepatocytes by a low concentration of mercury. Hepatocytes treated with different doses of mercuric chloride (HgCl2; 1, 2.5, 5 and 10 µM) and at different time intervals (0 min, 30 min, 1 h, 2 h and 4 h) show autophagic cell death only at 5 µM HgCl2 within 30 min of incubation. At 1 and 2.5 µM HgCl2, no cell death is recorded, while apoptosis is found at 10 µM HgCl2, as evidenced by the activation of caspase 3. Autophagic cell death is confirmed by the presence of monodansylcadaverine (MDC) positive hepatocytes which is found to be highest at 1 h. Atg5-Atg12 covalent-conjugation triggers the autophagic pathway within 30 min of 5 µM HgCl2 treatment and continues till 4 h of incubation. In addition, damage-regulated autophagy modulator (DRAM) expression gradually increases from 30 min to 4 h of treatment with mercury and a corresponding linear decrease in p53 has been observed. It is concluded that a low concentration (5 µM HgCl2) of mercury induces autophagy or nonapoptotic programmed cell death following an Atg5-Atg12 covalent-conjugation pathway, which is modulated by DRAM in a p53-dependent manner.


Subject(s)
Autophagy/drug effects , Hepatocytes/drug effects , Mercuric Chloride/toxicity , Animals , Benzimidazoles , Cells, Cultured , Coloring Agents , Dose-Response Relationship, Drug , Ethidium , Fluoresceins , Male , Mercuric Chloride/administration & dosage , Rats , Rats, Sprague-Dawley
13.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167047, 2024 03.
Article in English | MEDLINE | ID: mdl-38296116

ABSTRACT

Dipeptidyl peptidase-4 (DPP-4), a ubiquitous proteolytic enzyme, inhibits insulin secretion from pancreatic beta cells by inactivating circulating incretin hormones GLP-1 and GIP. High circulating levels of DPP-4 is presumed to compromise insulin secretion in people with type 2 diabetes (T2D). Our group recently reported lipid induced DPP-4 expression in pancreatic beta cells, mediated by the TLR4-NFkB pathway. In the present study, we looked at the role of Vildagliptin on pancreatic DPP-4 inhibition, preservation of islet mass and restoration of insulin secretion. MIN6 mouse insulinoma cells incubated with palmitate and fetuin-A, a proinflammatory organokine associated with insulin resistance, showed activation of TLR4-NFkB pathway, which was rescued on Vildagliptin treatment. In addition, Vildagliptin, by suppressing palmitate-fetuin-A mediated DPP-4 expression in MIN6, prevented the secretion of IL-1beta and fetuin-A in the culture media. DPP-4 siRNA abrogated TLR4-NFkB pathway mediated islet cell inflammation. Vildagliptin also reduced palmitate-fetuin-A mediated intracellular lipid accumulation in MIN6 and isolated islets from high fat fed (HFD) mice as observed by Oil O Red staining with downregulation of CD36 and PPARgamma. Vildagliptin also preserved islet mass and rescued insulin secretory defect in HFD mice. Our results suggest that inhibition of DPP-4 by Vildagliptin protects pancreatic beta cells from the deleterious effects of lipid and fetuin-A, preserves insulin secretory functions and improves hyperglycemia.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Humans , Mice , Animals , Vildagliptin/pharmacology , Vildagliptin/metabolism , Insulin-Secreting Cells/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , alpha-2-HS-Glycoprotein/metabolism , Toll-Like Receptor 4/metabolism , Insulin/metabolism , Palmitates/pharmacology
14.
Mol Cell Endocrinol ; 581: 112112, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38000461

ABSTRACT

High amount of fat in the pancreas is linked to poor functioning of ß-cells and raises the risk of type 2 diabetes. Here we report the putative role of a circulatory glycoprotein Fetuin-A, a known obesity marker, in promoting lipid accumulation in ß-cells and its association with Fatty acid translocase/CD36 for lipid storage culminate in ß-cell dysfunction. Additionally, this work reveals regulation of CD36 via Nrf2, a key regulator of oxidative stress, and reduction of lipid accumulation by suppression of Nrf2 that restores ß-cell function. Palmitate (0.50 mM) and Fetuin-A (100 µg/mL) exposure showed high levels of intracellular lipid in MIN6 (mouse insulinoma cells) with a concomitant decrease in insulin secretion. This also increased the expression of important lipogenic factors, like CD36, PGC1α, PPARγ, and SREBP1. Flow cytometry analysis of CD36 membrane localization has been corroborated with an increased accumulation of lipids as indicated by Oil-Red-O staining. Immunoblotting and immunofluorescence of Nrf2 indicated its high expression in palmitate-fetuin-A incubation and translocation in the nucleus. Suppression of Nrf2 by siRNA showed a reduced expression of lipogenic genes, ablation of lipid droplets, decrease in the number of apoptotic cells, and restoration of insulin secretion with a corresponding increase of Pdx1, BETA2, and Ins1 gene expression. Our study thus suggested an important aspect of lipid accumulation in the pancreatic ß-cells contributing to ß-cell dysfunction and demonstrated the role of Fetuin-A in CD36 expression, with a possible way of restoring ß-cell function by targeting Nrf2.


Subject(s)
Diabetes Mellitus, Type 2 , Insulinoma , Pancreatic Neoplasms , Animals , Mice , alpha-2-HS-Glycoprotein/metabolism , CD36 Antigens/metabolism , Insulin/metabolism , NF-E2-Related Factor 2/metabolism , Palmitates/pharmacology
15.
Heliyon ; 10(16): e36154, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39247273

ABSTRACT

Multi-epitope vaccines strategically tackle rapidly mutating viruses by targeting diverse epitopes from different proteins, providing a comprehensive and adaptable immune protection approach for enhanced coverage against various viral variants. This research employs a comprehensive approach that includes the mapping of immune cells activating epitopes derived from the six structural glycoproteins (A29L, A30L, A35R, L1R, M1R, and E8L) of Monkeypox virus (Mpox). A total of 7 T-cells-specific epitopes, 13 B-cells-specific epitopes, and 5 IFN-γ activating epitopes were forecasted within these glycoproteins. The selection process focused on epitopes indicating high immunogenicity and favorable binding affinity with multiple MHC alleles. Following this, a vaccine has been formulated by incorporating the chosen epitopes, alongside adjuvants (PADRE peptide) and various linkers (EAAAK, GPGPG, and AAY). The physicochemical properties and 3D structure of the multi-epitope hybrid vaccine were analysed for characterization. MD simulations were employed to predict the binding stability between the vaccine and various pathogen recognition receptors such as TLRs (TLR1, TLR2, TLR4, and TLR6), as well as both class I and II MHC, achieved through hydrogen bonding and hydrophobic interactions. Through in silico cloning and immune simulation, it was observed that the multi-epitopes vaccine induced a robust memory immune response upon booster doses, forecasting protective immunity upon viral challenge. This protective immunity was characterized by the production of IgM + IgG antibodies, along with release of inflammatory cytokines like IFN-γ, and IL12, and the activation of various immune cells. This study offers valuable insights into the potential of a multi-epitope vaccine targeting the Mpox virus.

16.
Toxicol In Vitro ; 101: 105921, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39179137

ABSTRACT

Recently, non-small cell lung cancer (NSCLC) has been the prime concern of cancer clinicians due to its high mortality rate worldwide. Cisplatin, a platinum derivative, has been used as a therapeutic option for treating metastatic NSCLC for several years. However, acquired, or intrinsic drug resistance to Cisplatin is the major obstacle to the successful treatment outcome of patients. Dysregulation of Nrf2 (nuclear factor erythroid 2-related factor 2) and EGFR (epidermal growth factor receptor) signaling have been associated with cellular proliferation, cancer initiation, progression and confer drug resistance to several therapeutic agents including Cisplatin in various cancers. To dissect the molecular mechanism of EGFR activation in resistant cells, we developed Cisplatin-resistant (CisR) human NSCLC cell lines (A549 and NCIH460) with increasing doses of Cisplatin treatment over a 3-month period. CisR cells demonstrated increased proliferative capacity, clonogenic survivability and drug efflux activity compared to the untreated parental (PT) cells. These resistant cells also showed higher levels of Nrf2 and EGFR expression. Here, we found that Nrf2 upregulates both basal and inducible expression of EGFR in these CisR cells at the transcriptional level. Moreover, genetic inhibition of Nrf2 with siRNA in CisR cells showed increased sensitivity towards the EGFR tyrosine kinase inhibitor (TKIs), AG1478. Our study, therefore suggests the use of Nrf2 inhibitors in combinatorial therapy with EGFR TKIs for the treatment of resistant NSCLC.

17.
Indian J Exp Biol ; 51(8): 623-34, 2013 Aug.
Article in English | MEDLINE | ID: mdl-24228386

ABSTRACT

Achatina fulica C-reactive protein (ACRP) reversed the toxic effects of lead nitrate both in vivo in mice and in vitro in rat hepatocytes restoring the basal level of cell viability, lipid peroxidation, reduced glutathione and superoxides. Cytotoxicity was also significantly ameliorated in rat hepatocytes by in vitro pre-treatments with individual subunits (60, 62, 90 and 110 kDa) of ACRP. Annexin V-Cy3/CFDA dual staining showed significant reduction in the number of apoptotic hepatocytes pre-treated with ACRP. ACRP induced restoration of mitochondrial membrane potential was remarkable. ACRP pre-treatment prevented Pb-induced apoptosis mediated by caspase activation. The antagonistic effect of ACRP may be due to scavenging of reactive oxygen species which maintained the homeostasis of cellular redox potential as well as reduced glutathione status. The results suggest that ACRP crosses the species barrier and it may be utilized as a viable exogenous agent of cytoprotection against heavy metal related toxicity.


Subject(s)
Apoptosis/drug effects , C-Reactive Protein/pharmacology , Cytoprotection/drug effects , Hepatocytes/drug effects , Lead/toxicity , Mitochondria, Liver/drug effects , Mollusca , Nitrates/toxicity , Animals , Blotting, Western , Cell Survival , Glutathione/metabolism , Hazardous Substances/toxicity , Hepatocytes/pathology , Lipid Peroxidation/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria, Liver/pathology , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
18.
Future Virol ; 2023 Mar.
Article in English | MEDLINE | ID: mdl-37064327

ABSTRACT

Diabetic patients are at particular risk of severe COVID-19. Human dipeptidyl peptidase-4 (DPP-4) is a membrane-bound aminopeptidase that regulates insulin release by inactivating incretin. DPP-4 inhibitors (DPP-4is) are therefore used as oral anti-diabetic drugs to restore normal insulin levels. These molecules also have anti-inflammatory and anti-hypertension effects. Recent studies on the interactions of SARS-CoV-2 spike glycoprotein and DPP-4 predict a possible entry route for SARS-CoV-2. Therefore, DPP-4is could be effective at reducing the virus-induced 'cytokine storm', thereby ceasing inflammatory injury to vital organs. Moreover, DPP-4is may interfere with viral entry into host cells. Herein, we have reviewed the efficacy of DPP-4is as potential repurposed drugs to reduce the severity of SARS-CoV-2 infection in patients with diabetes.

19.
Int J Biol Macromol ; 245: 125444, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37385308

ABSTRACT

Highly mutated SARS-CoV-2 is known aetiological factor for COVID-19. Here, we have demonstrated that the receptor binding domain (RBD) of the spike protein can interact with human dipeptidyl peptidase 4 (DPP4) to facilitate virus entry, in addition to the usual route of ACE2-RBD binding. Significant number of residues of RBD makes hydrogen bonds and hydrophobic interactions with α/ß-hydrolase domain of DPP4. With this observation, we created a strategy to combat COVID-19 by circumventing the catalytic activity of DPP4 using its inhibitors. Sitagliptin, linagliptin or in combination disavowed RBD to establish a heterodimer complex with both DPP4 and ACE2 which is requisite strategy for virus entry into the cells. Both gliptins not only impede DPP4 activity, but also prevent ACE2-RBD interaction, crucial for virus growth. Sitagliptin, and linagliptin alone or in combination have avidity to impede the growth of pan-SARS-CoV-2 variants including original SARS-CoV-2, alpha, beta, delta, and kappa in a dose dependent manner. However, these drugs were unable to alter enzymatic activity of PLpro and Mpro. We conclude that viruses hijack DPP4 for cell invasion via RBD binding. Impeding RBD interaction with both DPP4 and ACE2 selectively by sitagliptin and linagliptin is an potential strategy for efficiently preventing viral replication.


Subject(s)
COVID-19 , Humans , Linagliptin/pharmacology , SARS-CoV-2/metabolism , Sitagliptin Phosphate/pharmacology , Dipeptidyl Peptidase 4/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Protein Binding
20.
Mol Cell Endocrinol ; 532: 111314, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33989718

ABSTRACT

Lipid mediated pancreatic ß-cell dysfunction during Type 2 diabetes is known to be regulated by activation of TLR4 (Toll Like Receptor 4) and NF-κB (Nuclear factor kappa B). Recently we have reported that MIN6 cells (mouse insulinoma cells) secrete fetuin-A on stimulation by palmitate that aggravates ß-cell dysfunction, but the mechanism involved in-vivo has not been demonstrated and thus remained unclear. Here we attempted to dissect the role of palmitate and fetuin-A on insulin secretion using high fat diet (HFD) fed mice model. HFD islets showed curtailed insulin secretion after 20 weeks of treatment with activated TLR4-NF-κB pathway. Further treatment of islets with palmitate raised fetuin-A expression by ~2.8 folds and cut down insulin secretion by ~1.4 folds. However, blocking the activity of TLR4, fetuin-A and NF-κB using specific inhibitors or siRNAs not only restored insulin secretion by ~2 folds in standard diet fed mice islets and MIN6 cells but also evoke insulin secretory ability by ~2.3 folds in HFD islets. Altogether this study demonstrated that blocking TLR4, fetuin-A and NF-κB protect pancreatic ß-cells from the negative effects of free fatty acid and fetuin-A and restore insulin secretion.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Diet, High-Fat/adverse effects , Hypoglycemic Agents/pharmacology , Insulin Secretion/drug effects , Insulin-Secreting Cells , NF-kappa B/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , alpha-2-HS-Glycoprotein/antagonists & inhibitors , Animals , Cell Line , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Mice , NF-kappa B/metabolism , Palmitates/adverse effects , Palmitates/pharmacology , Toll-Like Receptor 4/metabolism , alpha-2-HS-Glycoprotein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL