Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Biochem Pharmacol ; 208: 115406, 2023 02.
Article in English | MEDLINE | ID: mdl-36596415

ABSTRACT

SAP97 is a member of the MAGUK family of proteins, but unlike other MAGUK proteins that are selectively expressed in the CNS, SAP97 is also expressed in peripheral organs, like the heart and kidneys. SAP97 has several protein binding cassettes, and this review will describe their involvement in creating SAP97-anchored multiprotein networks. SAP97-anchored networks localized at the inner leaflet of the cell membrane play a major role in trafficking and targeting of membrane G protein-coupled receptors (GPCR), channels, and structural proteins. SAP97 plays a major role in compartmentalizing voltage gated sodium and potassium channels to specific cellular compartments of heart cells. SAP97 undergoes extensive alternative splicing. These splice variants give rise to different SAP97 isoforms that alter its cellular localization, networking, signaling and trafficking effects. Regarding GPCR, SAP97 binds to the ß1-adrenergic receptor and recruits AKAP5/PKA and PDE4D8 to create a multiprotein complex that regulates trafficking and signaling of cardiac ß1-AR. In the kidneys, SAP97 anchored networks played a role in trafficking of aquaporin-2 water channels. Cardiac specific ablation of SAP97 (SAP97-cKO) resulted in cardiac hypertrophy and failure in aging mice. Similarly, instituting transverse aortic constriction (TAC) in young SAP97 c-KO mice exacerbated TAC-induced cardiac remodeling and dysfunction. These findings highlight a critical role for SAP97 in the pathophysiology of a number of cardiac and renal diseases, suggesting that SAP97 is a relevant target for drug discovery.


Subject(s)
Adaptor Proteins, Signal Transducing , Membrane Proteins , Mice , Animals , Adaptor Proteins, Signal Transducing/metabolism , Membrane Proteins/metabolism , Protein Transport/physiology , Signal Transduction/physiology , Multiprotein Complexes/metabolism , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism
2.
Andrologia ; 53(11): e14233, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34535909

ABSTRACT

Chemotherapeutic agents can impair gonadal function triggering infertility. Here, we probed the properties of carnosine as an antioxidant in reproductive disorders caused by the combination of cyclophosphamide, hydroxydaunomycin (doxorubicin), oncovin (vincristine) and prednisone (CHOP); this combination is mostly used in treating non-Hodgkin lymphoma. Animals were distributed into four groups: Group I was the control. Group II received carnosine (250mg kg day-1 , i.p.); Group III received CHOP: cyclophosphamide (27 mg/kg/cycle), doxorubicin (1.8 mg/kg/cycle) and vincristine (0.05 mg/kg /cycle) by i.p. plus oral prednisone (1.47 mg kg-1  day-1 /cycle) for five days. Group IV received carnosine plus CHOP. The study involved 4 cycles each of 3 weeks. Also, we explored the effect of combining carnosine with CHOP on the development of solid Ehrlich carcinoma in mice. CHOP lowered genitals weight, sperm count and motility, testicular function marker enzymes, serum testosterone level and gene expression of 3ß-hydroxysteroid dehydrogenase, 17ß-hydroxysteroid dehydrogenase and steroidogenic acute regulatory protein. Furthermore, CHOP elevated testicular oxidative stress, serum follicle-stimulating hormone, luteinising hormone and triggered DNA damage. Morphometric and histopathological examinations of testicular tissues buttressed the biochemical results. Importantly, administration of carnosine ameliorated CHOP-induced alterations without diminishing CHOP's antineoplastic action. These results indicated that carnosine may ameliorate reproductive disorders induced by CHOP.


Subject(s)
Carnosine , Fertility , Animals , Antineoplastic Combined Chemotherapy Protocols , Carnosine/pharmacology , Cyclophosphamide , Doxorubicin , Fertility/drug effects , Male , Mice , Prednisone , Rats , Vincristine
3.
Exp Gerontol ; 137: 110982, 2020 08.
Article in English | MEDLINE | ID: mdl-32450269

ABSTRACT

Ageing is an unavoidable, universal, biological phenomenon affecting all organisms, which involves variable declines of individuals motor and memory capabilities. This study aimed to investigate the potential ameliorating effects of curcumin C3 complex, Astragalus membranaceus and blueberry on certain age-related biochemical alterations in rat liver. Four groups of rats, aged 12 months-old, were used. The first group; aged control group in which rats were left without any treatment until the age of 17 months. The other three groups received daily by oral gavage for 5 months the following supplements; curcumin C3 complex (110 mg/kg), Astragalus membranaceus (100 mg/kg) and blueberry (100 mg/kg) respectively. Additionally, a fifth group of rats, aged 5 months-old, was used as an adult control group. Our supplements alleviated ageing-induced redox state imbalance and inflammation as evidenced by reduction of hepatic thiobarbituric acid reactive substances and 8-hydroxydeoxyguanosine levels, restoration of total antioxidant capacity and nitric oxide contents, and lessening of lipofuscin deposition. All supplements decreased hepatic interlukin-6 gene expression and serum levels. Notably, Astragalus membranaceus and blueberry upregulated hepatic telomerase reverse transcriptase gene expression and increased telomere length. Our findings recommend the use of these natural hepatoprotective supplements for the elderly to promote healthy ageing and minimize the risk of age-related liver diseases.


Subject(s)
Blueberry Plants , Curcumin , Liver Diseases , Animals , Astragalus propinquus , Curcumin/pharmacology , Rats
4.
J Adv Res ; 21: 141-150, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32071782

ABSTRACT

Lately, long noncoding (lnc) RNAs are increasingly appreciated for their involvement in multiple sclerosis (MS). In inflammation and autoimmunity, a role of apoprotein A1 (ApoA1), mediated by sphingosine 1-phosphate receptors (S1PRs), was reported. However, the epigenetic mechanisms regulating these biomolecules and their role in MS remains elusive. This case control study investigated the role of ApoA1, sphingosine kinase 1 and 2 (SPHK1 & 2), S1PR1 & 5, interferon-γ (IFN-γ) and interleukin 17 (IL17) in MS, beside three lncRNA: APOA1-AS, IFNG-AS1, and RMRP. Expression of SPHKs, S1PRs, and lncRNAs were measured in 72 relapsing-remitting MS patients (37 during relapse and 35 in remission) and 28 controls. Plasma levels of ApoA1, IFN-γ and IL17 were determined. The impact of these parameters on MS activity, relapse rate and patient disability was assessed. APOA1-AS, IFNG-AS1, SPHK1 & 2, and S1PR5 were upregulated in RRMS patients. Differences in ApoA1, SPHK2, and IL17 were observed between relapse and remission. Importantly, ApoA1, SPHK2, and IL17 were related to activity, while S1PR1 and IFN-γ were linked to disability, though, only IFN-γ was associated with relapse rate. Finally, an excellent diagnostic power of IFN-γ, IL17, SPHK1 and APOA1-AS was demonstrated, whereas SPHK2 showed promising prognostic power in predicting relapses.

5.
Am J Physiol Renal Physiol ; 317(2): F375-F387, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31141395

ABSTRACT

Arginine-vasopressin (AVP)-mediated translocation of aquaporin-2 (AQP2) protein-forming water channels from storage vesicles to the membrane of renal collecting ducts is critical for the renal conservation of water. The type-1 PDZ-binding motif (PBM) in AQP2, "GTKA," is a critical barcode for its translocation, but its precise role and that of its interacting protein partners in this process remain obscure. We determined that synapse-associated protein-97 (SAP97), a membrane-associated guanylate kinase protein involved in establishing epithelial cell polarity, was an avid binding partner to the PBM of AQP2. The role of PBM and SAP97 on AQP2 redistribution in response to AVP was assessed in LLC-PK1 renal collecting cells by confocal microscopy and cell surface biotinylation techniques. These experiments indicated that distribution of AQP2 and SAP97 overlapped in the kidneys and LLC-PK1 cells and that knockdown of SAP97 inhibited the translocation of AQP2 in response to AVP. Binding between AQP2 and SAP97 was mediated by specific interactions between the second PDZ of SAP97 and PBM of AQP2. Mechanistically, inactivation of the PBM of AQP2, global delocalization of PKA, or knockdown of SAP97 inhibited AQP2 translocation as well as AVP- and forskolin-mediated phosphorylation of Ser256 in AQP2, which serves as the major translocation barcode of AQP2. These results suggest that the targeting of PKA to the microdomain of AQP2 via SAP97-AQP2 interactions in association with cross-talk between two barcodes in AQP2, namely, the PBM and phospho-Ser256, plays an important role in the translocation of AQP2 in the kidney.


Subject(s)
Aquaporin 2/metabolism , Arginine Vasopressin/pharmacology , Discs Large Homolog 1 Protein/metabolism , Epithelial Cells/drug effects , Kidney Tubules, Proximal/drug effects , PDZ Domains , Animals , Aquaporin 2/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Discs Large Homolog 1 Protein/genetics , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , LLC-PK1 Cells , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Protein Transport , Serine , Swine
6.
Mol Pharmacol ; 95(5): 563-572, 2019 05.
Article in English | MEDLINE | ID: mdl-30894404

ABSTRACT

G protein-coupled receptor family C group 6 member A (GPRC6A) is activated by testosterone and modulates prostate cancer progression. Most humans have a GPRC6A variant that contains a recently evolved KGKY insertion/deletion in the third intracellular loop (ICL3) (designated as GPRC6AICL3_KGKY) that replaces the ancestral KGRKLP sequence (GPRC6AICL3_RKLP) present in all other species. In vitro assays purport that human GPRC6AICL3_KGKY is retained intracellularly and lacks function. These findings contrast with ligand-dependent activation and coupling to mammalian target of rapamycin complex 1 (mTORC1) signaling of endogenous human GPRC6AICL3_KGKY in PC-3 cells. To understand these discrepant results, we expressed mouse (mGPRC6AICL3_KGRKLP), human (hGPRC6AICL3_KGKY), and humanized mouse (mGPRC6AICL3_KGKY) GPRC6A into human embryonic kidney 293 cells. Our results demonstrate that mGPRC6AICL3_KGRKLP acts as a classic G protein-coupled receptor, which is expressed at the cell membrane and internalizes in response to ligand activation by testosterone. In contrast, hGPRC6AICL3_KGKY and humanized mouse mGPRC6AICL3_KGKY are retained intracellularly in ligand naive cells, yet exhibit ß-arrestin-dependent signaling responses, mitogen-activated protein kinase [i.e., extracellular signal-regulated kinase (ERK)], and p70S6 kinase phosphorylation in response to testosterone, indicating that hGPRC6AICL3_KGKY is functional. Indeed, testosterone stimulates time- and dose-dependent activation of ERK, protein kinase B, and mTORC1 signaling in wild-type PC-3 cells that express endogenous GPRC6AICL3_KGKY In addition, testosterone stimulates GPRC6A-dependent cell proliferation in wild-type PC-3 cells and inhibits autophagy by activating mTORC1 effectors eukaryotic translation initiation factor 4E binding protein 1 and Unc-51 like autophagy activating kinase 1. Testosterone activation of GPRC6A has the obligate requirement for calcium in the incubation media. In contrast, in GPRC6A-deficient cells, the effect of testosterone to activate downstream signaling is abolished, indicating that human GPRC6A is required for mediating the effects of testosterone on cell proliferation and autophagy.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Prostatic Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Testosterone/pharmacology , Animals , Autophagy/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Ligands , Male , Mice , PC-3 Cells , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
7.
Article in English | MEDLINE | ID: mdl-30008305

ABSTRACT

Untreated invasive aspergillosis results in high mortality rate in pediatric cancer patients. Voriconazole (VORI), the first line of treatment, requires strict dose monitoring because of its narrow therapeutic index and individual variation in plasma concentration levels. Commonly co-administered drugs; either Esomeprazole (ESO) or Ondansetron (OND) have reported drug-drug interaction with VORI that should adversely alter therapeutic outcomes of the latter. Although VORI, ESO and OND are co-administered to pediatric cancer patients, the combined effect of ESO and OND on the plasma concentration levels of VORI has not been fully explored. In this study, an accurate, reliable and sensitive liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay was developed and validated for simultaneous determination of VORI, ESO, and OND in ultra-low sample volumes (25 µL) of plasma of pediatric cancer patients. Based on the physicochemical properties of the studied drugs and internal standard, liquid-liquid extraction was successfully adopted with methyl t-butyl ether. Consistent and reproducible recovery of the three drugs and the internal standard were calculated using plasma and matrix matched samples (RE% > 72.97%, RSD < 8.29%). Chromatographic separation was carried out using UPLC with C18 column and a mobile phase of acetonitrile:water:methanol (70:25:5 V/V/V) at 0.3 mL/min. Mass spectrometric determination at positive electrospray ionization in the MRM mode was employed. The analysis was achieved within 4 min over a linear concentration range of 1.00-200.00 ng/mL for the three drugs. The assay validity was assessed as per the Food and Drug Administration guidelines for bioanalytical method validation, and satisfactory results were obtained. The accuracy and precision were within the acceptable limits for the three drugs in both quality control and incurred plasma samples. Matrix effect and process efficiency were investigated in neat solvent, post-extraction matrix, and plasma. Correlation of the plasma concentration levels of the three drugs revealed differences from the reported drug-drug interactions. This confirmed the need for simultaneous determination of VORI and co-administered drugs in order to achieve optimal therapeutic outcomes. To achieve this, analysis results of this study, genetic polymorphisms in CYP2C19 and clinical data will be used to establish one model incorporating all possible factors that might lead to variation in therapeutic outcomes.


Subject(s)
Antineoplastic Agents/blood , Chromatography, High Pressure Liquid/methods , Tandem Mass Spectrometry/methods , Voriconazole/blood , Adolescent , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Child , Drug Monitoring , Humans , Linear Models , Neoplasms/drug therapy , Precision Medicine , Reproducibility of Results , Sensitivity and Specificity , Voriconazole/pharmacokinetics , Voriconazole/therapeutic use
8.
Mol Pharmacol ; 94(2): 862-875, 2018 08.
Article in English | MEDLINE | ID: mdl-29848777

ABSTRACT

The ß1-adrenergic receptor (ß1-AR) is a major cardiac G protein-coupled receptor, which mediates cardiac actions of catecholamines and is involved in genesis and treatment of numerous cardiovascular disorders. In mammalian cells, catecholamines induce the internalization of the ß1-AR into endosomes and their removal promotes the recycling of the endosomal ß1-AR back to the plasma membrane; however, whether these redistributive processes occur in terminally differentiated cells is unknown. Compartmentalization of the ß1-AR in response to ß-agonists and antagonists was determined by confocal microscopy in primary adult rat ventricular myocytes (ARVMs), which are terminally differentiated myocytes with unique structures such as transverse tubules (T-tubules) and contractile sarcomeres. In unstimulated ARVMs, the fluorescently labeled ß1-AR was expressed on the external membrane (the sarcolemma) of cardiomyocytes. Exposing ARVMs to isoproterenol redistributed surface ß1-ARs into small (∼225-250 nm) regularly spaced internal punctate structures that overlapped with puncta stained by Di-8 ANEPPS, a membrane-impermeant T-tubule-specific dye. Replacing the ß-agonist with the ß-blocker alprenolol, induced the translocation of the wild-type ß1-AR from these punctate structures back to the plasma membrane. This step was dependent on two barcodes, namely, the type-1 PDZ binding motif and serine at position 312 of the ß1-AR, which is phosphorylated by a pool of cAMP-dependent protein kinases anchored at the type-1 PDZ of the ß1-AR. These data show that redistribution of the ß1-AR in ARVMs from internal structures back to the plasma membrane was mediated by a novel sorting mechanism, which might explain unique aspects of cardiac ß1-AR signaling under normal or pathologic conditions.


Subject(s)
Adrenergic beta-1 Receptor Agonists/pharmacology , Adrenergic beta-1 Receptor Antagonists/pharmacology , Myocytes, Cardiac/cytology , Receptors, Adrenergic, beta-1/metabolism , Alprenolol/pharmacology , Animals , Cell Membrane/metabolism , Cells, Cultured , Humans , Isoproterenol/pharmacology , Myocytes, Cardiac/metabolism , Protein Transport/drug effects , Rats , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/genetics
9.
Oxid Med Cell Longev ; 2017: 9396425, 2017.
Article in English | MEDLINE | ID: mdl-29201275

ABSTRACT

Paraquat (PQ) is one of the most used herbicide worldwide. Its cytotoxicity is attributed to reactive radical generation. Resveratrol (Res) and montelukast (MK) have anti-inflammatory and antioxidant properties. The protective effects of Res, MK, or their combination against PQ-induced acute liver injury have not been investigated before. Therefore, we explored the protective potential of Res and/or MK against PQ hepatic toxicity in a mouse model. Mice were randomly assigned to five groups: group I served as the normal control and group II received a single dose of PQ (50 mg/kg, i.p.). Groups III, IV, and V received PQ plus oral Res (5 mg/kg/day), MK (10 mg/kg/day), and Res/MK combination, respectively. Res and/or MK reduced PQ-induced liver injury, evidenced by normalization of serum total protein, ALT, and AST. Res and/or MK significantly reversed PQ-induced oxidative stress markers glutathione and malondialdehyde. Res and/or MK significantly reduced PQ-induced inflammation reflected in TNF-α levels. Furthermore, Res and/or MK reversed PQ-induced apoptosis assessed by differential expression of p53, Bax, and Bcl-2. Histopathologic examination supported the biochemical findings. Although Res and MK displayed antioxidative, anti-inflammatory, and antiapoptotic activities, their combination was not always synergistic.


Subject(s)
Acetates/pharmacology , Acute Lung Injury/chemically induced , Apoptosis/drug effects , Oxidative Stress/drug effects , Paraquat/toxicity , Quinolines/pharmacology , Stilbenes/pharmacology , Acetates/therapeutic use , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Cyclopropanes , Glutathione/metabolism , Inflammation/prevention & control , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Malondialdehyde/metabolism , Mice , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Quinolines/therapeutic use , Reactive Oxygen Species/metabolism , Resveratrol , Stilbenes/therapeutic use , Sulfides , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
10.
Pharmacogn Mag ; 13(Suppl 3): S607-S612, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29142421

ABSTRACT

BACKGROUND: Several studies have affirmed the effectiveness of some Bauhinia plants as antihyperglycemic agents. OBJECTIVE: We investigated the possible effect of Bauhinia vahlii leaves extract in reducing hyperglycemia and reversing signs of organ damage associated with diabetes in streptozotocin (STZ) rat model. MATERIALS AND METHODS: Both polar fraction of the B. vahlii leaves (defatted ethanolic extract [DEE]) and nonpolar fraction (n-hexane extract) were evaluated in vitro for α-glucosidase inhibition and 2,2-diphenyl-1-picrylhydrazyl radical scavenging potential. DEE was selected for further in vivo studies and was administered at two doses, i.e., 150 or 300 mg/kg to STZ-diabetic rats for 4 weeks. RESULTS: Only DEE exhibited in vitro antioxidant and antihyperglycemic activities and its oral administration at both dose levels resulted in significant reduction in fasting blood glucose and glycated hemoglobin. Furthermore, signs of oxidative stress as indicated by hepatic reduced glutathione, nitric oxide, and malondialdehyde levels were completely reversed. In addition, histopathological examination and measurement of serum aspartate transaminase and alanine transaminase levels showed that DEE protected the liver from signs of liver pathogenesis when compared to diabetic untreated animals and those treated with metformin. Phytochemical analysis of DEE showed high flavonoids content with quercitrin as the major constituent along with other quercetin glycosides. CONCLUSION: This study strongly highlights the possible beneficial effect of B. vahlii leaves extract in relieving hyperglycemia and liver damage in STZ-diabetic rats and recommends further investigation of the value of quercetin derivatives in controlling diabetes and ameliorating liver damage associated with it. SUMMARY: The polar fraction of the Bauhinia vahlii leaves (defatted ethanolic extract [DEE]) exhibited both in vitro antioxidant activity in 2,2-diphenyl-1-picrylhydrazyl scavenging assay and strong α-glucosidase inhibition while the nonpolar fraction (n-hexane extract) failed to show any activity in both assays. DEE was further investigated in streptozotocin-induced diabetic rat model where oral administration of DEE at 2 doses (150 and 300 mg/kg) for 4 weeks resulted in significant reduction in fasting blood glucose and glycated hemoglobin and reversal of oxidative stress signs as indicated by measurement of hepatic reduced glutathione, nitric oxide, and malondialdehyde levels. In addition, histopathological examination and measurement of serum aspartate transaminase and alanine transaminase levels showed that DEE protected the liver from signs of pathogenesis observed in diabetic untreated rats. Phytochemical analysis of DEE showed high flavonoid content with quercitrin as the major constituent (62.9 ± 0.18 mg/mg). Abbreviations used: ALT: Alanine transaminase, AST: Aspartate transaminase, DEE: Defatted ethanol extract, DPPH: 2,2-diphenyl-1-picrylhydrazyl, FBG: Fasting blood glucose, GAE: Gallic acid equivalent, GSH: Reduced glutathione, Hb1Ac: Glycated hemoglobin, HE: Hexane extract MDA: Malondialdehyde, QE: Quercetin equivalent, STZ: Streptozotocin, TAC: Total antioxidant capacity.

11.
Methods Cell Biol ; 142: 67-78, 2017.
Article in English | MEDLINE | ID: mdl-28964341

ABSTRACT

G protein-coupled receptors (GPCRs) are recognized as one of the most fruitful group of therapeutic targets, accounting for more than 40% of all approved pharmaceuticals on the market. Therefore, the search for selective agents that affect GPCR function is of major interest to the pharmaceutical industry. This chapter describes methods for measuring agonist-promoted GPCR trafficking, which involves the internalization of the GPCR and its subsequent recycling back to the plasma membrane or retention and eventual degradation. These pathways will be analyzed by confocal cellular imaging, using the ß1-adrenergic receptor (ß1-AR) as a primary model. A major problem encountered in studying GPCR trafficking is the unavailability of antibodies that would recognize the native receptor in cells or tissues. Therefore, wild-type, point mutants, and ß1-AR chimeras are generated as epitope-tagged proteins, which are stably- or transiently expressed in mammalian cells. GPCR are labeled with a fluorophore-conjugated antibody directed against the N-terminal epitope tag. The trafficking of the fluorophore-tagged GPCR between divergent trafficking pathways that result in retention and eventual degradation or recycling and reinsertion into the plasma membrane can be followed by confocal immunofluorescence microscopy techniques outlined in this review.


Subject(s)
Adrenergic beta-1 Receptor Agonists/pharmacology , Cell Membrane/metabolism , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Protein Transport/drug effects , Receptors, Adrenergic, beta-1/metabolism , Animals , Endosomes/metabolism , Fluorescent Dyes/chemistry , HEK293 Cells , Humans , Mice , MicroRNAs/analysis , MicroRNAs/chemistry , Mutagenesis , Myocytes, Cardiac , RNA Interference , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-1/genetics , Staining and Labeling/methods
12.
Cell Signal ; 36: 42-55, 2017 08.
Article in English | MEDLINE | ID: mdl-28449947

ABSTRACT

Proper signaling by G protein coupled receptors (GPCR) is dependent on the specific repertoire of transducing, enzymatic and regulatory kinases and phosphatases that shape its signaling output. Activation and signaling of the GPCR through its cognate G protein is impacted by G protein-coupled receptor kinase (GRK)-imprinted "barcodes" that recruit ß-arrestins to regulate subsequent desensitization, biased signaling and endocytosis of the GPCR. The outcome of agonist-internalized GPCR in endosomes is also regulated by sequence motifs or "barcodes" within the GPCR that mediate its recycling to the plasma membrane or retention and eventual degradation as well as its subsequent signaling in endosomes. Given the vast number of diverse sequences in GPCR, several trafficking mechanisms for endosomal GPCR have been described. The majority of recycling GPCR, are sorted out of endosomes in a "sequence-dependent pathway" anchored around a type-1 PDZ-binding module found in their C-tails. For a subset of these GPCR, a second "barcode" imprinted onto specific GPCR serine/threonine residues by compartmentalized kinase networks was required for their efficient recycling through the "sequence-dependent pathway". Mutating the serine/threonine residues involved, produced dramatic effects on GPCR trafficking, indicating that they played a major role in setting the trafficking itinerary of these GPCR. While endosomal SNX27, retromer/WASH complexes and actin were required for efficient sorting and budding of all these GPCR, additional proteins were required for GPCR sorting via the second "barcode". Here we will review recent developments in GPCR trafficking in general and the human ß1-adrenergic receptor in particular across the various trafficking roadmaps. In addition, we will discuss the role of GPCR trafficking in regulating endosomal GPCR signaling, which promote biochemical and physiological effects that are distinct from those generated by the GPCR signal transduction pathway in membranes.


Subject(s)
Protein Transport , Receptors, G-Protein-Coupled/metabolism , Animals , Endosomes/metabolism , Feedback, Physiological , Humans , Protein Processing, Post-Translational , Signal Transduction
13.
Sci Rep ; 7: 46468, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28425447

ABSTRACT

Recombinant human myelin basic protein (rhMBP) was previously produced in the milk of transgenic cows. Differences in molecular recognition of either hMBP or rhMBP by surface-immobilized anti-hMBP antibodies were demonstrated. This indicated differences in immunological response between rhMBP and hMBP. Here, the activity of free and controlled release rhMBP poly(ε-caprolactone) nanoparticles (NPs), as a therapeutic vaccine against multiple sclerosis (MS) was demonstrated in experimental autoimmune encephalomyelitis (EAE) animal model. Following optimization of nanoformulation, discrete spherical, rough-surfaced rhMBP NPs with high entrapment efficiency and controlled release pattern were obtained. Results indicated that rhMBP was loaded into and electrostatically adsorbed onto the surface of NPs. Subcutaneous administration of free or rhMBP NPs before EAE-induction reduced the average behavioral score in EAE mice and showed only mild histological alterations and preservation of myelin sheath, with rhMBP NPs showing increased protection. Moreover, analysis of inflammatory cytokines (IFN-γ and IL-10) in mice brains revealed that pretreatment with free or rhMBP NPs significantly protected against induced inflammation. IN CONCLUSION: i) rhMBP ameliorated EAE symptoms in EAE animal model, ii) nanoformulation significantly enhanced efficacy of rhMBP as a therapeutic vaccine and iii) clinical investigations are required to demonstrate the activity of rhMBP NPs as a therapeutic vaccine for MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Myelin Basic Protein/therapeutic use , Nanoparticles/therapeutic use , Recombinant Proteins/therapeutic use , Vaccines/therapeutic use , Animals , Mice , Treatment Outcome
14.
Cell Signal ; 29: 192-208, 2017 01.
Article in English | MEDLINE | ID: mdl-27816670

ABSTRACT

Recycling of the majority of agonist-internalized GPCR is dependent on a type I-PDZ "barcode" in their C-tail. The recycling of wild-type (WT) ß1-AR is also dependent on its default "type-1 PDZ barcode", but trafficking of the ß1-AR is inhibited when PKA or its substrate serine at position 312 (Ser312) are inactivated. We tested the hypothesis that phospho-Ser312 provided a second barcode for ß1-AR sorting from endosomes to the plasma membrane by determining the role of retromer/WASH complexes in ß1-AR trafficking. Recycling of WT ß1-AR or WT ß2-AR was dependent on targeting the retromer to endosomal membranes via SNX3 and rab7a, and on complexing the retromer to the WASH pentamer via the C-tail of FAM21 (FAM21C). These maneuvers however, did not inhibit the recycling of a phospho-Ser312 ß1-AR mimic ((S312D) ß1-AR). Knockdown of the trans-acting PDZ protein sorting nexin27 (SNX27) inhibited the recycling of WT ß1-AR and WT ß2-AR, but had no effect on (S312D) ß1-AR∆PDZ or on phosphorylation of WT ß1-AR by PKA at Ser312. However, depletion of FKBP15, a FAM21C-binding endosomal protein, selectively inhibited WT ß1-AR but not ß2-AR recycling, suggesting divergence might exist in GPCR trafficking roadmaps. These results indicate that two barcodes are involved in sorting WT ß1-AR out of early endosomes. The first and antecedent "barcode" was the "type-1 PDZ", followed by a second reversible "phospho-Ser312" verification "barcode". This organization allows tight regulation of ß1-AR density to signaling intensity in conditions associated with aberrant ß1-AR signaling such as in hypertension and heart failure.


Subject(s)
Cell Membrane/metabolism , Endosomes/metabolism , Multiprotein Complexes/metabolism , PDZ Domains , Phosphoserine/metabolism , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/metabolism , Cell Membrane/drug effects , Down-Regulation/drug effects , Endocytosis/drug effects , Endosomes/drug effects , HEK293 Cells , Humans , Isoproterenol/pharmacology , Membrane Proteins/metabolism , Models, Biological , Multiprotein Complexes/chemistry , Phosphate-Binding Proteins , Phosphatidylinositol Phosphates/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Structure, Secondary , Protein Transport/drug effects , Proteins/metabolism , RNA, Small Interfering/metabolism , Sorting Nexins/metabolism , Structure-Activity Relationship , Tacrolimus Binding Proteins , Wiskott-Aldrich Syndrome Protein Family , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
15.
Mol Neurobiol ; 54(5): 3219-3229, 2017 07.
Article in English | MEDLINE | ID: mdl-27067589

ABSTRACT

Multiple sclerosis (MS) is a demyelinating neurodegenerative disease, representing a major cause of neurological disability in young adults. Resveratrol is a stilbenoid polyphenol, known to pass blood brain barrier and exhibit antioxidant, anti-inflammatory, and neuroprotective effects in several brain injuries. Cuprizone model of MS is particularly beneficial in studying demyelination/remyelination. Our study examined the potential neuroprotective and pro-remyelination effects of resveratrol in cuprizone-intoxicated C57Bl/6 mice. Mice were fed with chow containing 0.7 % cuprizone for 7 days, followed by 3 weeks on 0.2 % cuprizone diet. Resveratrol (250 mg/kg/day, p.o.) was given for 3 weeks starting from the second week. At the end of the experiment, animals were tested on rotarod to evaluate changes in balance and motor coordination. Mice were then sacrificed to measure the brain content of glutathione, lipid peroxidation products, adenosine triphosphate, and phospho-inhibitory subunit of nuclear factor κB-α. The activities of cytochrome oxidase and superoxide dismutase were also assessed. The gene expression of myelin basic protein, 2',3'-cyclic nucleotide 3' phosphodiesterase, oligodendrocyte transcription factor-1 (Olig1), NF-κB p65 subunit, and tumor necrosis factor-α was also estimated. Luxol fast blue/periodic acid-Schiff stained brain sections were blindly scored to assess the myelin status. Resveratrol effectively enhanced motor coordination and balance, reversed cuprizone-induced demyelination, improved mitochondrial function, alleviated oxidative stress, and inhibited NF-κB signaling. Interestingly, resveratrol increased Olig1 expression that is positively correlated to active remyelination. The present study may be the first to indicate a pro-remyelinative effect for resveratrol which might represent a potential additive benefit in treating MS.


Subject(s)
Multiple Sclerosis/chemically induced , Multiple Sclerosis/drug therapy , Remyelination/drug effects , Stilbenes/therapeutic use , Animals , Biomarkers/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Cuprizone , Disease Models, Animal , Inflammation/pathology , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Motor Activity/drug effects , Multiple Sclerosis/pathology , Myelin Sheath/metabolism , Oxidative Stress/drug effects , Resveratrol , Stilbenes/pharmacology
16.
Biochem Pharmacol ; 120: 22-32, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27645110

ABSTRACT

ß1-Adrenergic receptor (ß1-AR) agonists and antagonists are widely used in the treatment of major cardiovascular diseases such as heart failure and hypertension. The ß1-AR like other G protein-coupled receptors (GPCRs) are endocytosed in response to intense agonist activation. Recycling of the agonist-internalized ß1-AR is dependent on its carboxy-terminal type-1 PSD-95/DLG/ZO1 (PDZ) and on phospho-serine312 in the third intracellular loop of the ß1-AR. Progressive elongation of the ß1-AR at its C-tail inactivated the PDZ-biding domain and inhibited the recycling of the ß1-AR. However, fusing a twenty amino acid peptide derived from the multiple cloning region of the mammalian expression vector pCDNA3 to the C-tail of the ß1-AR (ß1-AR[+20]) produced a chimeric ß1-AR that recycled rapidly and efficiently. The ß1-AR[+20] recycled in a type-1 PDZ and phospho-Ser312-independent manner, indicating that this peptide provided a general GPCR recycling signal. Fusing the enhanced yellow fluorescent protein (EYFP) down-stream of ß1-AR[+20] generated a ß1-AR-EYFP chimera that was expressed on the membrane and recycled efficiently after agonist-induced internalization. This construct trafficked in a PDZ-SNX27/retromer-independent manner. We also fused EYFP to the N-terminus of the ß1-AR to created EYFP-WT ß1-AR. This construct recycled in PDZ and SNX27/retromer dependent manner. These ß1-AR-EYFP constructs would be useful for high throughput screening (HTS) programs to identify new entities that would interfere with the recycling of agonist internalized GPCR that traffic in PDZ-dependent vs. PDZ-independent roadmaps.


Subject(s)
Adrenergic beta-1 Receptor Agonists/pharmacology , Drug Discovery/methods , Endocytosis/drug effects , Models, Molecular , Receptors, Adrenergic, beta-1/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Endosomes/drug effects , Endosomes/metabolism , HEK293 Cells , High-Throughput Screening Assays , Humans , Immunoprecipitation , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Oligopeptides/genetics , Oligopeptides/metabolism , PDZ Domains , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphoserine/metabolism , Plasmids/chemistry , Plasmids/genetics , Plasmids/metabolism , RNA Interference , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/genetics , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sorting Nexins/antagonists & inhibitors , Sorting Nexins/chemistry , Sorting Nexins/genetics , Sorting Nexins/metabolism
17.
PLoS One ; 11(6): e0158014, 2016.
Article in English | MEDLINE | ID: mdl-27336155

ABSTRACT

BACKGROUND: Vitamin D has been considered a key player in various malignancies including cutaneous cancers. To date, mycosis fungoides (MF) has been the least studied in relation to vitamin D. Furthermore, the vitamin D receptor (VDR) single nucleotide polymorphisms (SNPs) have not been tackled before in the context of MF, despite their incrimination in numerous diseases. AIM OF STUDY: To assess the role of vitamin D in MF by measuring its serum level, and studying VDR SNPs (TaqI, BsmI, FokI) in different stages of MF. PATIENTS AND METHODS: 48 patients with various stages of MF, and 45 healthy controls were included. Complete history, full clinical examination and a five mm punch skin biopsy were performed to all recruited patients. Venous blood samples were withdrawn from both patients and controls to determine the serum vitamin D level and VDR gene polymorphisms. RESULTS: Serum vitamin D level was significantly lower in patients (5.3-33.7 nmol/L)] compared to controls (8.3-90.1 nmol/L)] (P<0.001). A significant difference was observed between patients and controls regarding the FokI polymorphism only, being higher in patients (P = 0.039). Also Vitamin D serum levels differed significantly in patients with FokI genotypes (P = 0.014). No significant correlations were detected between any of the studied parameters and the demographic and clinical data of the included subjects. CONCLUSION: Depressed vitamin D and FokI polymorphism are potentially involved in the context of MF. VDR gene polymorphisms warrant further larger scale investigations to detect the exact genes involved in the pathogenesis of such an enigmatic disease.


Subject(s)
Mycosis Fungoides/blood , Mycosis Fungoides/genetics , Polymorphism, Single Nucleotide , Receptors, Calcitriol/genetics , Vitamin D/blood , Adolescent , Adult , Aged , Alleles , Case-Control Studies , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Phenotype , Young Adult
18.
PLoS One ; 10(10): e0139493, 2015.
Article in English | MEDLINE | ID: mdl-26425844

ABSTRACT

Hyper- or hypothyroidism can impair testicular function leading to infertility. The present study was designed to examine the protective effect of date palm pollen (DPP) extract on thyroid disorder-induced testicular dysfunction. Rats were divided into six groups. Group I was normal control. Group II received oral DPP extract (150 mg kg(-1)), group III (hyperthyroid group) received intraperitoneal injection of L-thyroxine (L-T4, 300 µg kg(-1); i.p.), group IV received L-T4 plus DPP extract, group V (hypothyroid group) received propylthiouracil (PTU, 10 mg kg(-1); i.p.) and group VI received PTU plus DPP extract. All treatments were given every day for 56 days. L-T4 or PTU lowered genital sex organs weight, sperm count and motility, serum levels of luteinizing hormone (LH), follicle stimulating hormone (FSH) and testosterone (T), testicular function markers and activities of testicular 3ß-hydroxysteroid dehydrogenase (3ß-HSD) and 17ß-hydroxysteroid dehydrogenase (17ß-HSD). Moreover, L-T4 or PTU increased estradiol (E2) serum level, testicular oxidative stress, DNA damage and apoptotic markers. Morphometric and histopathologic studies backed these observations. Treatment with DPP extract prevented LT4- or PTU induced changes. In addition, supplementation of DPP extract to normal rats augmented sperm count and motility, serum levels of LH, T and E2 paralleled with increased activities of 3ß-HSD and 17ß-HSD as well as testicular antioxidant status. These results provide evidence that DPP extract may have potential protective effects on testicular dysfunction induced by altered thyroid hormones.


Subject(s)
Phoeniceae/chemistry , Plant Extracts/pharmacology , Pollen/chemistry , Testicular Diseases/drug therapy , Thyroid Diseases/complications , Animals , Biomarkers/analysis , Male , Organ Size/drug effects , Oxidative Stress/drug effects , Rats , Rats, Wistar , Spermatogenesis/drug effects , Testicular Diseases/etiology , Thyroid Diseases/physiopathology
19.
PLoS One ; 9(6): e99286, 2014.
Article in English | MEDLINE | ID: mdl-24915010

ABSTRACT

Vitiligo is the most common depigmentation disorder of the skin. Oxidative stress is implicated as one of the probable events involved in vitiligo pathogenesis possibly contributing to melanocyte destruction. Evidence indicates that certain genes including those involved in oxidative stress and melanin synthesis are crucial for development of vitiligo. This study evaluates the oxidative stress status, the role of catalase (CAT) and catechol-O-Methyltransferase (COMT) gene polymorphisms in the etiology of generalized vitiligo in Egyptians. Total antioxidant capacity (TAC) and malondialdehyde (MDA) levels as well as CAT exon 9 T/C and COMT 158 G/A polymorphisms were determined in 89 patients and 90 age and sex-matched controls. Our results showed significantly lower TAC along with higher MDA levels in vitiligo patients compared with controls. Meanwhile, genotype and allele distributions of CAT and COMT polymorphisms in cases were not significantly different from those of controls. Moreover, we found no association between both polymorphisms and vitiligo susceptibility. In conclusion, the enhanced oxidative stress with the lack of association between CAT and COMT polymorphisms and susceptibility to vitiligo in our patients suggest that mutations in other genes related to the oxidative pathway might contribute to the etiology of generalized vitiligo in Egyptian population.


Subject(s)
Catalase/genetics , Catechol O-Methyltransferase/genetics , Genetic Predisposition to Disease , Oxidative Stress/genetics , Polymorphism, Single Nucleotide/genetics , Vitiligo/enzymology , Vitiligo/genetics , Adult , Antioxidants/metabolism , Biomarkers/metabolism , Case-Control Studies , Demography , Egypt , Exons/genetics , Female , Gene Frequency/genetics , Humans , Male , Malondialdehyde/metabolism , Risk Factors
20.
J Biol Chem ; 289(4): 2277-94, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24324269

ABSTRACT

The ß1-adrenergic receptor (ß1-AR) is a target for treatment of major cardiovascular diseases, such as heart failure and hypertension. Recycling of agonist-internalized ß1-AR is dependent on type I PSD-95/DLG/ZO1 (PDZ) in the C-tail of the ß1-AR and on protein kinase A (PKA) activity (Gardner, L. A., Naren, A. P., and Bahouth, S. W. (2007) J. Biol. Chem. 282, 5085-5099). We explored the effects of point mutations in the PDZ and in the activity of PKA on recycling of the ß1-AR and its binding to the PDZ-binding protein SAP97. These studies indicated that ß1-AR recycling was inhibited by PKA inhibitors and by mutations in the PDZ that interfered with SAP97 binding. The trafficking effects of short sequences differing in PDZ and SAP97 binding were examined using chimeric mutant ß1-AR. ß1-AR chimera containing the type I PDZ of the ß2-adrenergic receptor that does not bind to SAP97 failed to recycle except when serine 312 was mutated to aspartic acid. ß1-AR chimera with type I PDZ sequences from the C-tails of aquaporin-2 or GluR1 recycled in a SAP97- and PKA-dependent manner. Non-PDZ ß1-AR chimera derived from µ-opioid, dopamine 1, or GluR2 receptors promoted rapid recycling of chimeric ß1-AR in a SAP97- and PKA-independent manner. Moreover, the nature of the residue at position -3 in the PDZ regulated whether the ß1-AR was internalized alone or in complex with SAP97. These results indicate that divergent pathways were involved in trafficking the ß1-AR and provide a roadmap for its trafficking via type I PDZs versus non-PDZs.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Membrane Proteins/metabolism , Receptors, Adrenergic, beta-1/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Discs Large Homolog 1 Protein , HEK293 Cells , Humans , Membrane Proteins/genetics , Protein Structure, Tertiary , Protein Transport/physiology , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...