Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cureus ; 15(10): e46391, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37927736

ABSTRACT

INTRODUCTION: Tofacitinib has emerged as a therapeutic option for axial spondyloarthritis (axSpA) following successful clinical trials. The evidence on the efficacy of tofacitinib generics in the management of axSpA is limited. In India, the usage of conventional synthetic disease-modifying anti-rheumatic drugs (csDMARDs) is commonplace in the management of axSpA. Our primary aim was to identify the csDMARD and non-steroidal anti-inflammatory drug (NSAID)-sparing role of tofacitinib generics in an axSpA population. METHODS: This was a retrospective study in a real-world clinical setting. Data from nine rheumatology centers across India were analyzed for 168 patients with active axSpA who were initiated on generic tofacitinib 5 mg twice daily in conjunction with csDMARDs and NSAIDs, over a duration of six months. Our primary outcome was to evaluate the csDMARD and NSAID-sparing effect of tofacitinib generics, while the secondary outcome assessed safety profiles and efficacy at six months. RESULTS: The median Ankylosing Spondylitis Disease Activity Score (ASDAS) erythrocyte sedimentation rate (ESR) score of the study population was 3.91 (3.26, 4.56). Alongside tofacitinib generics, 121 (72%) patients were co-administered csDMARDs (methotrexate/sulfasalazine/both), and 90 (53.6%) patients were co-administered NSAIDs. The csDMARD, NSAID, and combined csDMARD + NSAID-sparing effects of tofacitinib generics were seen in 85 (50.6%), 156 (92.9%), and 81 (48.2%) patients, respectively. Adverse events were mild and well-tolerated. At six months, 124 (57.9%) patients had attained clinically important improvement in ASDAS ESR score, and the median decrease in ASDAS ESR score was 2.02 (1.18, 2.96). CONCLUSION: This real-world study provides evidence supporting the csDMARD and NSAID-sparing ability of tofacitinib generics in the treatment of axSpA. Tofacitinib generics displayed a good safety profile and showed signals of efficacy as well.

2.
J Organomet Chem ; 9362021 Mar 15.
Article in English | MEDLINE | ID: mdl-33953436

ABSTRACT

Tricarbonylrhenium(I)(α-diimine) complexes are of importance because of their strong cytotoxic and fluorescence properties. Syntheses of such complexes were achieved through a two-step process. First, the pentylcarbonato complexes, fac-(CO)3(α-diimine)ReOC(O)OC5H11 were synthesized through a microwave-assisted reaction of Re2(CO)10, α-diimine, 1-pentanol and CO2 in a few hours. Second, the pentylcarbonato complexes are treated with carboxylic, sulfonic and halo acids to obtain the corresponding carboxylato, sulfonato and halido complexes. This is the first example of conversion of Re2(CO)10 into a rhenium carbonyl complex through microwave-assisted reaction.

3.
Acta Neurobiol Exp (Wars) ; 80(1): 57-65, 2020.
Article in English | MEDLINE | ID: mdl-32214275

ABSTRACT

The catecholamine norepinephrine (NE) links hindbrain metabolic­sensory neurons with downstream gluco­regulatory loci, including the ventromedial hypothalamic nucleus (VMN). Exogenous NE up­regulates VMN expression of glutamate decarboxylase (GAD), biomarker for the gluco­inhibitory transmitter γ­aminobutryic acid (GABA). Brain glycogen phosphorylase (GP)­muscle (GPmm) and ­brain (GPbb) variants are stimulated in vitro by NE or energy deficiency, respectively. Current research investigated whether lactoprivic­driven VMN NE signaling regulates GABA and if VMN GPmm and GPbb profiles react differently to that deficit cue. Male rats were pretreated by caudal fourth ventricle delivery of the selective catecholamine neurotoxin 6­hydroxydopamine (6OHDA) ahead of the monocarboxylate transporter inhibitor alpha­cyano­4­hydroxycinnamic acid (4CIN). Micropunch­dissected VMN tissue was analyzed by Western blot and ELISA to assess NE­dependent 4CIN regulation of GAD and GP variant protein expression and NE activity. 4CIN caused 6OHDA­reversible augmentation of VMN NE content and plasma glucose and counter­regulatory hormone levels. 6OHDA stimulated basal VMN GAD expression, but prevented 4CIN stimulation of this profile. Neurotoxin inhibited or increased baseline VMN GPmm and GPbb levels, respectively, in non­4CIN­injected rats. 6OHDA deterred 4CIN inhibition of GPmm, but did not prevent drug stimulation of GPbb. Results affirm hindbrain lactoprivic regulation of glucostasis. Hindbrain NE exerts opposite effects on VMN GABA transmission during hindbrain lactostasis vs. ­privation. VMN norepinephrine­ vs. energy­sensitive GP variants are subject to dissimilar NE regulation during energy homeostasis, and respond differently to hindbrain lactoprivation.


Subject(s)
Glucose/metabolism , Glycogen/metabolism , Rhombencephalon/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Coumaric Acids/pharmacology , Dopamine beta-Hydroxylase/biosynthesis , Dopamine beta-Hydroxylase/genetics , Enzyme Induction/drug effects , Glutamate Decarboxylase/biosynthesis , Glutamate Decarboxylase/genetics , Glycogen Phosphorylase/metabolism , Isoenzymes/biosynthesis , Isoenzymes/genetics , Male , Norepinephrine/pharmacology , Norepinephrine/physiology , Oxidopamine/toxicity , Rats , Rats, Sprague-Dawley , Rhombencephalon/drug effects
4.
Brain Res Bull ; 157: 41-50, 2020 04.
Article in English | MEDLINE | ID: mdl-31981674

ABSTRACT

Mechanisms that underlie metabolic sensor acclimation to recurring insulin-induced hypoglycemia (RIIH) are unclear. Norepinephrine (NE) regulates ventromedial hypothalamic nucleus (VMN) gluco-stimulatory nitric oxide (NO) and gluco-inhibitory γ-aminobutryic acid (GABA) neuron signaling. Current research addressed the hypothesis that during RIIH, NE suppresses 5'-AMP-activated protein kinase (AMPK) reactivity in both populations and impedes counter-regulation. The brain is postulated to utilize non-glucose substrates, e.g. amino acids glutamine (Gln), glutamate (Glu), and aspartate (Asp), to produce energy during hypoglycemia. A correlated aim investigated whether NE controls pyruvate recycling pathway marker protein (glutaminase, GLT; malic enzyme, ME-1) expression in either metabolic-sensory cell population. Male rats were injected subcutaneously with vehicle or insulin on days 1-3, then pretreated on day 4 by intracerebroventricular delivery of the alpha1-adrenergic receptor (α1-AR) reverse-agonist prazocin (PRZ) or vehicle before final insulin therapy. PRZ prevented acute hypoglycemic augmentation of AMPK activation in each cell group. Antecedent hypoglycemic repression of sensor activity was reversed by PRZ in GABA neurons. During RIIH, nitrergic neurons exhibited α1-AR - dependent up-regulated GLT and α2-AR profiles, while GABA cells showed down-regulated α1-AR. LC-ESI-MS analysis documented a decline in VMN Glu, Gln, and Asp concentrations during acute hypoglycemia, and habituation of the former two profiles to RIIH. PRZ attenuated glucagon and corticosterone secretion during acute hypoglycemia, but reversed decrements in output of both hormones during RIIH. Results implicate adjustments in impact of α1-AR signaling in repressed VMN metabolic-sensory AMPK activation and counter-regulatory dysfunction during RIIH. Antecedent hypoglycemia may up-regulate NO neuron energy yield via α1-AR - mediated up-regulated pyruvate recycling.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Hypoglycemia/metabolism , Insulin/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Hypoglycemia/physiopathology , Hypoglycemic Agents/pharmacology , Male , Norepinephrine/metabolism , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism , Rhombencephalon/metabolism , Ventromedial Hypothalamic Nucleus/cytology
5.
Neuropeptides ; 77: 101962, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31488323

ABSTRACT

Hindbrain energy state shapes hypothalamic control of glucostasis. Dorsal vagal complex (DVC) L-lactate deficiency is a potent glucose-stimulatory signal that triggers neuronal transcriptional activation in key hypothalamic metabolic loci. The energy gauge AMPK is activated in DVC metabolic-sensory A2 noradrenergic neurons by hypoglycemia-associated lactoprivation, but sensor reactivity is diminished by antecedent hypoglycemia (AH). Current research addressed the premise that AH alters hindbrain lactoprivic regulation of hypothalamic metabolic transmitter function. AH did not modify reductions in A2 dopamine-beta-hydroxylase and monocarboxylate-2 (MCT2) protein expression elicited by caudal fourth ventricular delivery of the MCT inhibitor alpha-cyano-4-hydroxycinnamic acid (4CIN), but attenuated 4CIN activation of A2 AMPK. 4CIN constraint of hypothalamic norepinephrine (NE) activity was averted by AH in a site-specific manner. 4CIN induction of Fos immunolabeling in hypothalamic arcuate (ARH), ventromedial (VMN), dorsomedial (DMN) and paraventricular (PVN) nuclei and lateral hypothalamic area (LHA) was avoided by AH. AH affected reactivity of select hypothalamic metabolic neurotransmitter/enzyme marker proteins, e.g. ARH neuropeptide Y, VMN glutamate decarboxylase, DMN RFamide-related peptide-1 and -3, and LHA orexin-A profiles to 4CIN, but did not alleviate drug inhibition of ARH proopiomelanocortin. AH prevented 4CIN augmentation of circulating glucagon, but did not alter hyperglycemic or hypocorticosteronemic responses to that treatment. Results identify hindbrain lactate deficiency as a stimulus for glucagon secretion, and imply that habituation of this critical counter-regulatory hormone to recurring hypoglycemia may involve one or more hypothalamic neurotransmitters characterized here by acclimation to this critical sensory stimulus.


Subject(s)
Hypoglycemia/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Rhombencephalon/metabolism , Animals , Blood Glucose/metabolism , Hypoglycemia/chemically induced , Insulin , Male , Neuropeptide Y/metabolism , Norepinephrine/metabolism , Pro-Opiomelanocortin/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Transcriptional Activation
6.
Neuroscience ; 411: 211-221, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31085279

ABSTRACT

Neural substrates for estrogen regulation of glucose homeostasis remain unclear. Female rat dorsal vagal complex (DVC) A2 noradrenergic neurons are estrogen- and metabolic-sensitive. The ventromedial hypothalamic nucleus (VMN) is a key component of the brain network that governs counter-regulatory responses to insulin-induced hypoglycemia (IIH). Here, the selective estrogen receptor-alpha (ERα) or -beta (ERß) antagonists MPP and PHTPP were administered separately to the caudal fourth ventricle to address the premise that these hindbrain ER variants exert distinctive control of VMN reactivity to IIH in the female sex. Data show that ERα governs hypoglycemic patterns of VMN astrocyte glycogen metabolic enzyme, e.g. glycogen synthase and phosphorylase protein expression, whereas ERß mediates local glycogen breakdown. DVC ERs also regulate VMN neurotransmitter signaling of energy sufficiency [γ-aminobutyric acid] or deficiency [nitric oxide, steroidogenic factor-1] during IIH. Neither hindbrain ER mediates IIH-associated diminution of VMN norepinephrine (NE) content. Both ERs oppose hypoglycemic hyperglucagonemia, while ERß contributes to reduced corticosterone output. Outcomes reveal that input from the female hindbrain to the VMN is critical for energy reserve mobilization, metabolic transmitter signaling, and counter-regulatory hormone secretion during hypoglycemia, and that ERs control those cues. Evidence that VMN NE content is not controlled by hindbrain ERα or -ß implies that these receptors may regulate VMN function via NE-independent mechanisms, or alternatively, that other neurotransmitter signals to the VMN may control local substrate receptivity to NE.


Subject(s)
Glycogen/metabolism , Hypoglycemia/metabolism , Receptors, Estrogen/metabolism , Rhombencephalon/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Estrogen Receptor Antagonists/pharmacology , Female , Nitric Oxide Synthase Type I/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Rats , Rhombencephalon/drug effects , Steroidogenic Factor 1/metabolism , Ventromedial Hypothalamic Nucleus/drug effects
7.
Neuroscience ; 409: 253-260, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30954669

ABSTRACT

Estrogen receptor-alpha (ERα) and -beta (ERß) occur in key elements of the brain gluco-homeostatic network in both sexes, including the hindbrain dorsal vagal complex (DVC), but the influence of distinct receptor populations on this critical function is unclear. The ventromedial hypothalamic nucleus (VMN) maintains glucose balance by integrating nutrient, endocrine, and neurochemical cues, including metabolic sensory information supplied by DVC A2 noradrenergic neurons. Current research utilized the selective ERα and ERß antagonists MPP and PHTPP to characterize effects of DVC ERs on VMN norepinephrine (NE) activity and metabolic neurotransmitter signaling in insulin-induced hypoglycemic (IIH) male rats. Data show that ERß inhibits VMN glycogen synthase and stimulates phosphorylase protein expression, while attenuating hypoglycemic augmentation of glycogen content. Furthermore, both ERs attenuate VMN glucose concentrations during IIH. Hypoglycemic up-regulation of nitric oxide (NO) and brain-derived neurotrophic factor (BDNF) signaling was correspondingly driven by ERα or -ß, whereas GABA and steroidogenic factor-1 were respectively suppressed independently of ER input or by ERß. IIH intensified VMN NE accumulation by ERß-dependent mechanisms, but did not alter NE levels in other gluco-regulatory loci. ERß amplified the magnitude of insulin-induced decline in blood glucose. Both ERs regulate corticosterone, but not glucagon secretion during IIH and oppose hypoglycemic diminution of circulating free fatty acids. These findings identify distinguishing versus common VMN functions targeted by DVC ERα and -ß. Sex differences in hypoglycemic VMN NE accumulation, glycogen metabolism, and transmitter signaling may involve, in part, discrepant regulatory involvement or differential magnitude of impact of these hindbrain ERs.


Subject(s)
Glucose/metabolism , Glycogen/metabolism , Hypoglycemia/metabolism , Receptors, Estrogen/metabolism , Rhombencephalon/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Corticosterone/metabolism , Male , Nitric Oxide/metabolism , Norepinephrine/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/antagonists & inhibitors , Rhombencephalon/drug effects , Ventromedial Hypothalamic Nucleus/drug effects
8.
Mol Cell Neurosci ; 95: 51-58, 2019 03.
Article in English | MEDLINE | ID: mdl-30660767

ABSTRACT

The ventromedial hypothalamic nucleus (VMN) is a critical component of the neural circuitry that regulates glucostasis. Astrocyte glycogen is a vital reserve of glucose and its oxidizable metabolite L-lactate. In hypoglycemic female rats, estradiol-dependent augmentation of VMN glycogen phosphorylase (GP) protein requires hindbrain catecholamine input. Research here investigated the premise that norepinephrine (NE) regulation of VMN astrocyte metabolism shapes local glucoregulatory neurotransmitter signaling in this sex. Estradiol-implanted ovariectomized rats were pretreated by intra-VMN administration of the monocarboxylate transporter inhibitor alpha-cyano-4-hydroxy-cinnamic acid (4CIN) or vehicle before NE delivery to that site. NE caused 4CIN-reversible reduction or augmentation of VMN glycogen synthase and phosphorylase expression. 4CIN prevented NE stimulation of gluco-inhibitory (glutamate decarboxylase65/67) and suppression of gluco-stimulatory (neuronal nitric oxide synthase) neuron marker proteins. These outcomes imply that effects of noradrenergic stimulation of VMN astrocyte glycogen depletion on glucoregulatory transmitter signaling may be mediated, in part, by glycogen-derived substrate fuel provision. NE control of astrocyte glycogen metabolism may involve down-regulated adrenoreceptor (AR), e.g. alpha1 and alpha2, alongside amplified beta1 AR and estrogen receptor-beta signaling. Noradrenergic hypoglycemia was refractory to 4CIN, implying that additional NE-sensitive VMN glucoregulatory neurochemicals may be insensitive to monocarboxylate uptake. Augmentation of circulating free fatty acids by combinatory NE and 4CIN, but not NE alone implies that acute hypoglycemia induced here is an insufficient stimulus for mobilization of these fuels, but is adequate when paired with diminished brain monocarboxylate fuel availability.


Subject(s)
Glucose/metabolism , Glycogen/metabolism , Monocarboxylic Acid Transporters/antagonists & inhibitors , Norepinephrine/pharmacology , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Astrocytes/metabolism , Coumaric Acids/pharmacology , Enzyme Inhibitors/pharmacology , Estrogen Receptor beta/metabolism , Estrogens/deficiency , Fatty Acids/metabolism , Female , Glycogen Synthase/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic/metabolism , Ventromedial Hypothalamic Nucleus/cytology
9.
Brain Res Bull ; 144: 171-179, 2019 01.
Article in English | MEDLINE | ID: mdl-30481553

ABSTRACT

Pharmacologic activation of the hindbrain dorsal vagal complex energy sensor 5'-adenosine monophosphate-activated protein kinase (AMPK) causes site-specific adjustments in hypothalamic AMPK activity. DVC A2 noradrenergic neurons are a likely source of metabolo-sensory cues to downstream network components as they express substrate fuel-sensitive AMPK. This study investigated the hypothesis that DVC AMPK controls hypothalamic sensor, metabolic effector transmitter, and counter-regulatory hormone responses to insulin-induced hypoglycemia. Male rats were injected into the caudal fourth ventricle with the AMPK inhibitor compound C (Ccor vehicle before hypoglycemia. Arcuate (ARH), ventromedial (VMN), and dorsomedial (DMN) nuclei and lateral hypothalamic area (LHA) were micropunch-dissected for norepinephrine ELISA and Western blot analyses. Hypoglycemic stimulation of norepinephrine activity in each site was impeded by compound C. Hypoglycemia caused drug-revocable (ARH) or -refractory (VMN, DMN) reductions in AMPK, alongside hindbrain AMPK-dependent augmentation of phospho-AMPK expression in each location. Compound C prevented hypoglycemic augmentation of gluco-stimulatory ARH neuropeptide Y, VMN neuronal nitric oxide synthase, and LHA orexin-A expression, while hypoglycemic suppression of the catabolic neuron protein markers ARH pro-opiomelanocortin and VMN glutamate decarboxylase65/67 was respectively averted or unaffected by drug treatment. DMN RFamide-related peptide-1 and -3 profiles were correspondingly amplified or suppressed hindbrain AMPK-reliant mechanisms during hypoglycemia. Results show that DVC AMPK is required for hypoglycemic intensification of norepinephrine activity in characterized hypothalamic gluco-regulatory structures, and that this sensor regulates AMPK activation and metabolic effector transmission in those sites.


Subject(s)
AMP-Activated Protein Kinases/physiology , Hypoglycemia/metabolism , AMP-Activated Protein Kinases/metabolism , Adrenergic Neurons , Animals , Hypoglycemia/physiopathology , Hypothalamic Area, Lateral/metabolism , Hypothalamus/metabolism , Male , Neuropeptide Y/metabolism , Neuropeptides/metabolism , Nitric Oxide Synthase Type I/metabolism , Norepinephrine/metabolism , Orexins/metabolism , Rats , Rats, Sprague-Dawley , Rhombencephalon/metabolism , Solitary Nucleus/metabolism , Vagus Nerve/metabolism
10.
Neuropeptides ; 70: 37-46, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29779845

ABSTRACT

BACKGROUND: Caudal dorsomedial hindbrain detection of hypoglycemia-associated lactoprivation regulates glucose counter-regulation in male rats. In females, estradiol (E) determines hypothalamic neuroanatomical and molecular foci of hindbrain energy sensor activation. This study investigated the hypothesis that E signal strength governs metabolic neuropeptide and counter-regulatory hormone responses to hindbrain lactoprivic stimuli in hypoglycemic female rats. METHODS: Ovariectomized animals were implanted with E-filled silastic capsules [30 (E-30) or 300 µg (E-300)/mL] to replicate plasma concentrations at estrous cycle nadir versus peak levels. E-30 and E-300 rats were injected with insulin or vehicle following initiation of continuous caudal fourth ventricular L-lactate infusion. RESULTS: Hypoglycemic hypercorticosteronemia was greater in E-30 versus E-300 animals. Glucagon and corticosterone outflow was correspondingly fully or partially reversed by hindbrain lactate infusion. Insulin-injected rats exhibited lactate-reversible augmentation of norepinephrine (NE) accumulation in all preoptic/hypothalamic structures examined, excluding the dorsomedial hypothalamic nucleus (DMH) where hindbrain lactate infusion either suppressed (E-30) or enhanced (E-300) NE content. Expression profiles of hypoglycemia-reactive metabolic neuropeptides were normalized (with greater efficacy in E-300 animals) by lactate infusion. DMH RFamide-related peptide-1 and -3, arcuate neuropeptide Y and kisspeptin, and ventromedial nucleus nitric oxide synthase protein responses to hypoglycemia were E dosage-dependent. CONCLUSIONS: Distinct physiological patterns of E secretion characteristic of the female rat estrous cycle elicit differential corticosterone outflow during hypoglycemia, and establish both common and different hypothalamic metabolic neurotransmitter targets of hindbrain lactate deficit signaling. Outcomes emphasize a need for insight on systems-level organization, interaction, and involvement of E signal strength-sensitive neuropeptides in counter-regulatory functions.


Subject(s)
Estradiol/pharmacology , Hypoglycemic Agents/pharmacology , Neuropeptides/metabolism , Rhombencephalon/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Estradiol/metabolism , Female , Hypoglycemia/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Insulin/pharmacology , Norepinephrine/metabolism , Rats, Sprague-Dawley , Rhombencephalon/metabolism
11.
J Cancer Sci Ther ; 10(2)2018.
Article in English | MEDLINE | ID: mdl-29707104

ABSTRACT

PURPOSE: Because of the scarcity of suitable brain cancer drugs, researchers are frantically trying to discover novel and highly potent drugs free of side effects and drug-resistance. Rhenium compounds are known to be nontoxic and exhibit no drug resistance. For that reason, we have developed a series of novel rhenium acetylsalicylato (RAC or ASP) complexes to test their cytotoxicity on brain cancer cells. Also we have attempted to explore the DNAbinding properties of these compounds because many drugs either directly or indirectly bind to DNA. METHODS: We have treated the RAC series compounds on human astrocytoma brain cancer cell lines and rat normal brain astrocyte cells and determined the efficacy of these complexes through in vitro cytotoxicity assay. We carried out the DNA-binding study through UV titrations of a RAC compound with DNA. Also we attempted to determine the planarity of the polypyridyl ligands of the RAC series compounds using DFT calculations. RESULTS: RAC6 is more potent than any other RAC series compounds on HTB-12 human astrocytoma cancer cells as well as on Glioblastoma Multiforme D54 cell lines. In fact, The IC-50 value of RAC6 on HTB-12 cancer cells is approximately 2 µM. As expected, the RAC series compounds were not active on normal cells. The DFT calculations on the RAC series compounds were done and suggest that the polypyridyl ligands in the complexes are planar. The UV-titrations of RAC9 with DNA were carried out. It suggests that RAC9 and possibly all RAC series compounds bind to minor grooves of the DNA. CONCLUSION: Because of the very low activity of RAC6 on normal cells and low lC50 value of on astrocytoma (HTB-12) cell lines, it is possible that RAC6 and its derivatives may potentially find application in the treatment of brain cancers. The DFT calculations and UV titrations suggest that RAC series compounds either bind to DNA intercalatively or minor grooves of the DNA or both. However, it is highly premature to make any definite statement in the absence of other techniques.

12.
Mol Cell Biochem ; 441(1-2): 151-163, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28913709

ABSTRACT

Cisplatin and other metal-based drugs often display side effects and tumor resistance after prolonged use. Because rhenium-based anticancer complexes are often less toxic, a novel series of organorhenium complexes were synthesized of the types: XRe(CO)3Z (X = α-diimines and Z = p-toluenesulfonate, 1-naphthalenesulfonate, 2-naphthalenesulfonate, picolinate, nicotinate, aspirinate, naproxenate, flufenamate, ibuprofenate, mefenamate, tolfenamate, N-acetyl-tryptophanate), and their biological properties were examined. Specifically, in hormone-dependent MCF-7 and hormone-independent triple-negative MDA-MB-231 breast cancer cells, the p-toluenesulfonato, 1-naphthalenesulfonato, 2-naphthalenesulfonato, picolinato, nicotinato, acetylsalicylato, flufenamato, ibuprofenato, mefenamato, and N-acetyl-tryptophanato complexes were found to be far more potent than conventional drug cisplatin. DNA-binding studies were performed in each case via UV-Vis titrations, cyclic voltammetry, gel electrophoresis, and viscosity, which suggest DNA partial intercalation interaction, and the structure-activity relationship studies suggest that the anticancer activities increase with the increasing lipophilicities of the compounds, roughly consistent with their DNA-binding activities.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Organometallic Compounds , Rhenium , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Female , Humans , MCF-7 Cells , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Rhenium/chemistry , Rhenium/pharmacology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
13.
Adv Exp Med Biol ; 1043: 359-383, 2017.
Article in English | MEDLINE | ID: mdl-29224103

ABSTRACT

Vital nerve cell functions, including maintenance of transmembrane voltage and information transfer, occur at high energy expense. Inadequate provision of the obligate metabolic fuel glucose exposes neurons to risk of dysfunction or injury. Clinical hypoglycemia rarely occurs in nondiabetic individuals but is an unfortunate regular occurrence in patients with type 1 or advanced insulin-treated type 2 diabetes mellitus. Requisite strict glycemic control, involving treatment with insulin, sulfonylureas, or glinides, can cause frequent episodes of iatrogenic hypoglycemia due to defective counter-regulation, including reduced glycemic thresholds and diminished magnitude of motor responses. Multiple components of the body's far-reaching energy balance regulatory network, including the hindbrain dorsal vagal complex, provide dynamic readout of cellular energetic disequilibrium, signals that are utilized by the hypothalamus to shape counterregulatory autonomic, neuroendocrine, and behavioral outflow toward restoration of glucostasis. The ovarian steroid hormone 17ß-estradiol acts on central substrates to preserve nerve cell energy stability brain-wide, thereby providing neuroprotection against bio-energetic insults such as neurodegenerative diseases and acute brain ischemia. The current review highlights recent evidence implicating estrogen in gluco-regulation in females by control of hindbrain metabolic sensor screening and signaling of hypoglycemia-associated neuro-energetic instability. It is anticipated that new understanding of the mechanistic basis of how estradiol influences metabolic sensory input from this critical brain locus to discrete downstream regulatory network substrates will likely reveal viable new molecular targets for therapeutic simulation of hormone actions that promote positive neuronal metabolic state during acute and recurring hypoglycemia.


Subject(s)
Blood Glucose/metabolism , Brain/metabolism , Energy Metabolism , Estradiol/metabolism , Hypoglycemia/blood , Hypoglycemic Agents/adverse effects , Insulin/adverse effects , Animals , Biomarkers/blood , Brain/drug effects , Brain/pathology , Brain/physiopathology , Energy Metabolism/drug effects , Female , Health Status Disparities , Humans , Hypoglycemia/chemically induced , Hypoglycemia/pathology , Hypoglycemia/physiopathology , Male , Sex Characteristics , Sex Factors , Signal Transduction
14.
Neuropeptides ; 66: 25-35, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28823463

ABSTRACT

Glucose counter-regulatory dysfunction correlates with impaired activation of the hypothalamic metabolic sensor adenosine 5'-monophosphate-activated protein kinase (AMPK). Hypothalamic AMPK is controlled by hindbrain energy status; we examined here whether hindbrain AMPK regulates hypothalamic AMPK and metabolic neurotransmitter maladaptation to recurring insulin-induced hypoglycemia (RIIH). Brain tissue was harvested after single versus serial insulin (I) dosing for Western blot analysis of AMPK, phospho-AMPK (pAMPK), and relevant biosynthetic enzyme/neuropeptide expression in micro-punch dissected arcuate (ARH), ventromedial (VMH), dorsomedial (DMH) nuclei and lateral hypothalamic area (LHA) tissue. The AMPK inhibitor compound c (Cc) or vehicle was administered to the caudal fourth ventricle ahead of antecedent I injections. RIIH caused site-specific elevation (ARH, VMH, LHA) or reduction (DMH) of total AMPK protein versus acute hypoglycemia; Cc respectively exacerbated or attenuated this response in the ARH and VMH. Hindbrain AMPK correspondingly inhibited or stimulated LHA and DMH pAMPK expression during RIIH. RIIH elicited Cc-reversible augmentation of VMH glutamate decarboxylase profiles, but stimulated (ARH pro-opiomelanocortin; LHA orexin-A) or decreased (VMH nitric oxide synthase) other metabolic neurotransmitters without hindbrain sensor involvement. Results demonstrate acclimated up-regulation of total AMPK protein expression in multiple hypothalamic loci during RIIH, and document hindbrain sensor contribution to amplification of this protein profile in the VMH. Concurrent lack of net change in ARH and VMH tissue pAMPK implies adaptive reductions in local sensor activity, which may/may not reflect positive gain in energy state. It remains unclear if 'glucose-excited' VMH GABAergic and/or ARH pro-opiomelanocortin neurons exhibit AMPK habituation to RIIH, and whether diminished sensor activation in these and other mediobasal hypothalamic neurotransmitter populations may contribute to HAAF.


Subject(s)
Adenylate Kinase/metabolism , Hypoglycemia/metabolism , Hypothalamus/metabolism , Rhombencephalon/metabolism , Animals , Blood Glucose/metabolism , Hypoglycemia/chemically induced , Insulin , Male , Neurons/metabolism , Nitric Oxide Synthase Type I/metabolism , Norepinephrine/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley
15.
Int J Rheum Dis ; 20(6): 760-766, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28036132

ABSTRACT

OBJECTIVE: To estimate levels of salivary and serum free light chains (FLCs) and explore its utility as a biomarker in primary Sjögren's syndrome (pSS). METHODS: Patients with pSS classified by American European Consensus group 2002 or American College of Rheumatology 2012 criteria between January 2015 and August 2015 were included. Healthy staff and non-first degree relatives of patients constituted controls. Serum and salivary FLCs were measured by immunoturbidometry using FREELITE™ Human Kappa(κ) and Lambda(λ) Free Kit (Binding site, Birmingham, UK), on a Roche Modular P800. FLCs were compared between cases and controls using the Mann-Whitney U-test. The receiver operator characteristic curve was constructed to analyze the discriminating ability of salivary and serum kappa and lambda FLCs. RESULTS: Salivary and serum FLCs were assayed in 15 patients and 13 patients, respectively, and in 15 controls. Median age of cases and controls was 34 years. Salivary kappa and lambda FLCs were higher in pSS as compared to controls (P < 0.05 and P < 0.001, respectively). Serum kappa and lambda FLCs were also higher in pSS (both P < 0.05). Salivary lambda levels were higher in pSS with ocular signs; serum kappa and lambda levels were higher in those with ocular symptoms. A cut off of ≥ 1.1 mg/L for salivary lambda FLC had a sensitivity and specificity of 73.3% and 93.3%, respectively, for the diagnosis of pSS. Serum kappa FLC ≥ 30 mg/L had a sensitivity and specificity of 92.3% and 73.3%, respectively. CONCLUSION: Serum and salivary FLCs and in particular the latter, are potential biomarkers in pSS. Larger studies are required for validating the findings.


Subject(s)
Immunoglobulin kappa-Chains/analysis , Immunoglobulin lambda-Chains/analysis , Saliva/immunology , Sjogren's Syndrome/diagnosis , Adult , Area Under Curve , Biomarkers/analysis , Biomarkers/blood , Case-Control Studies , Female , Humans , Immunoglobulin kappa-Chains/blood , Immunoglobulin lambda-Chains/blood , Male , Pilot Projects , Predictive Value of Tests , ROC Curve , Reproducibility of Results , Sjogren's Syndrome/blood , Sjogren's Syndrome/immunology
17.
Br J Pharm Res ; 4(3): 362-367, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25419517

ABSTRACT

AIM: To study the efficacy of novel rhenium compounds to treat triple node negative breast cancer. PLACE AND DURATION: Six (6) novel rhenium pentycarbanato compounds (PC1-6) were synthesized and triple node negative breast cancer cell lines HTB-132 and Balb/c mouse kidney cell lines were treated with each of them for 48 hours. The results were analyzed by a common trypan blue cell death assay system and statistically analyzed. PLACE AND DURATION: The compounds were synthesized, analyzed and evaluated at the Department of Chemistryof Morgan State University, Baltimore, Maryland and the Pharmaceutical Sciences Department of Elizabeth City State University campus of the University of North Carolina system. METHODOLOGY: The novel rhenium compounds were synthesized from one-pot reactions of Re2(CO)10 with the corresponding α-diimine ligands in 1-pentanol.The compounds were characterized spectroscopically. The cell lines were cultured by standard cell culture procedure and treated with each of the six compounds in DMSO for 48 hours with a negative control and a DMSO vehicular control along with a cisplatin positive control.The cytotoxicity was evaluated by standard trypan blue assay and the results were statistically analyzed. RESULTS: The trypan blueassay reveals that these compounds have significant cytotoxicity against MDA-MB-468 (HTB-132) triple node negative breast cancer cell lines and are less nephrotoxic than cisplatin. CONCLUSION: The novel rhenium compounds PC 1-6 can potentially find applications in the treatment of highly malignant triple node negative breast cancer.

18.
Mol Cell Biochem ; 398(1-2): 21-30, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25262122

ABSTRACT

Seven organorhenium pentylcarbonate compounds (PC1-PC7) have been synthesized. DNA-binding studies of the PC-series compounds using electronic spectroscopy and gel electrophoresis suggest that the compounds presumably bind to DNA in an intercalative mode. The intrinsic binding constants for PC4, PC6, and PC7 were found to be 1.6 × 10(4), 3.9 × 10(4), and 4.2 × 10(4) M(-1), respectively. The X-ray structure determinations and density functional theory calculations indicate that the polypyridyl ligands in the compounds are nearly planar facilitating DNA binding through an intercalation mechanism. Cytotoxicity studies of 10 µM pentylcarbonate compounds against HTB-12 human astrocytoma brain cancer cells were studied for 48 h. It was observed that each of the pentylcarbonate compounds is active against the cancer cells. However, under analogous conditions, CRL-2005 rat astrocyte normal brain cells are not affected significantly.


Subject(s)
DNA/metabolism , Organometallic Compounds/chemistry , Organometallic Compounds/metabolism , Ruthenium/chemistry , Animals , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , DNA/genetics , Electrophoresis, Agar Gel , Humans , Intercalating Agents/chemistry , Intercalating Agents/metabolism , Intercalating Agents/pharmacology , Molecular Structure , Organometallic Compounds/pharmacology , Rats , Spectrophotometry
19.
J Bioprocess Biotech ; 4(1): 141, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-25221731

ABSTRACT

Despite the tremendous success of cisplatin and other platinum-based anticancer drugs, severe toxicity and resistance to tumors limit their applications. It is believed that the coordination (formation of covalent bond) of the metal (platinum) to the nitrogen bases of DNA cause the ruptures of the cancer as well as normal cells. A search for anticancer drugs with different modes of action resulted in the synthesis of variety of novel compounds. Many of them are in clinical trials now. Recently we synthesized a series of novel rhenium pentylcarbonato compounds (PC1-PC6). The rhenium atom in each compound is coordinated (bonded) to a planar polypyridyl aromatic ligand, thereby forcing each compound to intercalate between the DNA bases. We have investigated the DNA binding properties of one of the PC-series of compounds (PC6) using electronic spectroscopy. The UV absorption titration of PC6 with DNA shows hypochromic effect with concomitant bathochromic shift of the charge transfer band at 290 nm. These results suggest that the compound PC6 binds to DNA through intercalation. It is therefore likely that the other PC-series of compounds will behave in a similar manner. Thus it is expected that these compounds will exhibit negligible or no side effect. We have observed that the PC-series of compounds are strong cytotoxic agents against lymphosarcoma (average GI50 ≈ 2±2.6 µM), PC-3 prostate (average GI50 ≈ 3±2.8 µM) and myeloid leukemia (average GI50 ≈ 3±2.8 µM) cancer cell lines. The average GI50 values of the PC-series of compounds are 2-3 less than the corresponding GI50 values of cisplatin. Also each of the PC-series of compounds exhibits less toxicity than cisplatin in the glomerular mesangial cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...