Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 360(2): 346-355, 2017 02.
Article in English | MEDLINE | ID: mdl-27965369

ABSTRACT

Although the physiologic role of muscarinic receptors in bladder function and the therapeutic efficacy of muscarinic antagonists for the treatment of overactive bladder are well established, the role of ß3-adrenergic receptors (ß3ARs) and their potential as therapeutics is just emerging. In this manuscript, we characterized the pharmacology of a novel ß3AR agonist vibegron (MK-4618, KRP-114V) and explored mechanistic interactions of ß3AR agonism and muscarinic antagonism in urinary bladder function. Vibegron is a potent, selective full ß3AR agonist across species, and it dose dependently increased bladder capacity, decreased micturition pressure, and increased bladder compliance in rhesus monkeys. The relaxation effect of vibegron was enhanced when combined with muscarinic antagonists, but differentially influenced by muscarinic receptor subtype selectivity. The effect was greater when vibegron was co-administered with tolterodine, a nonselective antagonist, compared with coadministration with darifenacin, a selective M3 antagonist. Furthermore, a synergistic effect for bladder strip relaxation was observed with the combination of a ß3AR agonist and tolterodine in contrast to simple additivity with darifenacin. To determine expression in rhesus bladder, we employed a novel ß3AR agonist probe, [3H]MRL-037, that selectively labels ß3 receptors in both urothelium and detrusor smooth muscle. Vibegron administration caused a dose-dependent increase in circulating glycerol and fatty acid levels in rhesus and rat in vivo, suggesting these circulating lipids can be surrogate biomarkers. The translation of our observation to the clinic has yet to be determined, but the combination of ß3AR agonists with M2/M3 antimuscarinics has the potential to redefine the standard of care for the pharmacological treatment of overactive bladder.


Subject(s)
Adrenergic beta-3 Receptor Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Pyrimidinones/pharmacology , Pyrrolidines/pharmacology , Receptors, Adrenergic, beta-3/metabolism , Urinary Bladder, Overactive/drug therapy , Adrenergic beta-3 Receptor Agonists/therapeutic use , Animals , Drug Interactions , Female , Humans , Macaca mulatta , Male , Muscarinic Antagonists/therapeutic use , Muscle, Smooth/drug effects , Muscle, Smooth/physiopathology , Protein Transport/drug effects , Pyrimidinones/therapeutic use , Pyrrolidines/therapeutic use , Rats , Species Specificity , Urinary Bladder/drug effects , Urinary Bladder/physiopathology , Urinary Bladder, Overactive/metabolism , Urinary Bladder, Overactive/physiopathology , Urodynamics/drug effects
2.
J Theor Biol ; 389: 146-58, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26549470

ABSTRACT

We present a theoretical agent-based model of cell evolution under the action of cytotoxic treatments, such as radiotherapy or chemotherapy. The major features of cell cycle and proliferation, cell damage and repair, and chemical diffusion are included. Cell evolution is based on a discrete Markov chain, with cells stepping along a sequence of discrete internal states from 'normal' to 'inactive'. Probabilistic laws are introduced for each type of event a cell can undergo during its life: duplication, arrest, senescence, damage, reparation, or death. We adjust the model parameters on a series of cell irradiation experiments, carried out in a clinical LINAC, in which the damage and repair kinetics of single- and double-strand breaks are followed. Two showcase applications of the model are then presented. In the first one, we reconstruct the cell survival curves from a number of published low- and high-dose irradiation experiments. We reobtain a very good description of the data without assuming the well-known linear-quadratic model, but instead including a variable DSB repair probability. The repair capability of the model spontaneously saturates to an exponential decay at increasingly high doses. As a second test, we attempt to simulate the two extreme possibilities of the so-called 'bystander' effect in radiotherapy: the 'local' effect versus a 'global' effect, respectively activated by the short-range or long-range diffusion of some factor, presumably secreted by the irradiated cells. Even with an oversimplified simulation, we could demonstrate a sizeable difference in the proliferation rate of non-irradiated cells, the proliferation acceleration being much larger for the global than the local effect, for relatively small fractions of irradiated cells in the colony.


Subject(s)
DNA Breaks, Double-Stranded , Drug Therapy/methods , Neoplasms/drug therapy , Neoplasms/radiotherapy , Radiotherapy/methods , Algorithms , Bystander Effect , Calibration , Cell Cycle , Cell Movement , Cell Proliferation , Cell Survival , Computer Simulation , DNA Repair , Diffusion , Dose-Response Relationship, Radiation , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , Infant , Male , Models, Biological , Monte Carlo Method , Poisson Distribution , Probability , Skin/metabolism
3.
Cell Death Dis ; 5: e1577, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25522271

ABSTRACT

Senescence is a non-proliferative state reached by normal cells in response to various stresses, including telomere uncapping, oxidative stress or oncogene activation. In previous reports, we have highlighted that senescent human epidermal keratinocytes have two opposite outcomes: either they die by autophagic programmed cell death or they evade in the form of neoplastic postsenescence emergent (PSNE) cells. Herein, we show that partially reducing macroautophagy in senescent keratinocytes using 3-methyl adenine or anti-Atg5 siRNAs increases the PSNE frequency, suggesting that senescent keratinocytes have to escape autophagic cell death to generate PSNE cells. However, totally inhibiting macroautophagy impairs PSNE and leads to a huge accumulation of oxidative damages, indicating that senescent keratinocytes need to achieve quality-control macroautophagy for PSNE to occur. In accordance, we demonstrate that the progenitors of PSNE cells display a level of macroautophagy slightly lower than that of the average senescent population, which is directly dictated by their level of reactive oxygen species, their level of upregulation of MnSOD, their level of activation of NF-κB transcription factors and their level of dysfunctional mitochondria. Macroautophagy thus has antagonistic roles during senescence, inducing cell death or promoting neoplastic transformation, depending on its level of activation. Taken together, these data suggest that levels of oxidative damages and ensuing macroautophagic activity could be two main determinants of the very initial phases of neoplastic transformation by senescence evasion.


Subject(s)
Autophagy , Cell Transformation, Neoplastic/metabolism , Keratinocytes/cytology , Autophagy-Related Protein 5 , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cellular Senescence , Female , Humans , Keratinocytes/metabolism , Male , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
4.
Horm Metab Res ; 40(6): 386-90, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18401834

ABSTRACT

Numerous data show that malnutrition during early life programs chronic diseases in adulthood. Many of these disorders may result from alterations in the development of neuroendocrine systems, such as the hypothalamo-pituitary-adrenal axis and the sympathoadrenal system. We have previously reported that maternal 50% food restriction during late pregnancy and lactation reduces adrenal weight and impairs chromaffin cell differentiation in male rats at weaning. In addition, maternal undernutrition modifies the expression of several genes involved in proliferation and apoptosis. This study therefore investigated the impact of maternal food restriction on adrenal cell growth in the late postnatal rat. Histological analysis showed that the number of proliferating chromaffin cells assessed by nuclear labelling with BrdU was reduced by 45%, whereas the level of apoptosis visualised by caspase-3 immunoreactivity was increased by 340% in adrenal medulla of offspring from undernourished mothers. In contrast, maternal food restriction did not affect proliferation and apoptosis in cortical cells of rats. These developmental changes were associated with overexpression of TGFbeta2. These data show that perinatal undernutrition impairs the balance between chromaffin cell proliferation and apoptosis. These modifications may lead to "malprogramming" of adrenal medulla development, which could contribute to the pathogenesis of chronic diseases in adulthood.


Subject(s)
Adrenal Medulla/cytology , Apoptosis/physiology , Chromaffin Cells/cytology , Malnutrition/physiopathology , Prenatal Exposure Delayed Effects , Adrenal Medulla/physiology , Animals , Animals, Newborn , Cell Proliferation , Chromaffin Cells/physiology , Female , Hypothalamo-Hypophyseal System/cytology , Hypothalamo-Hypophyseal System/growth & development , Male , Malnutrition/pathology , Pituitary-Adrenal System/cytology , Pituitary-Adrenal System/growth & development , Pregnancy , Rats , Rats, Wistar
5.
Neuroscience ; 149(3): 706-14, 2007 Nov 09.
Article in English | MEDLINE | ID: mdl-17870246

ABSTRACT

Recent findings demonstrate that chemokines, and more specifically CC chemokine ligand 2 (CCL2 or monocyte chemoattractant protein-1), play a major role in pain processing. In the present study, we assess nociceptive responses of mice that overexpressed CCL2 under control of glial fibrillary acidic protein promoter (CCL2 tg). In models of acute nociception CCL2 tg mice demonstrated significantly enhanced nociceptive behavior relative to wild-type controls in responses to both thermal (hot plate) and chemical (formalin test) stimulus modalities. There were no differences in mechanical allodynia in the partial sciatic nerve ligation model, in terms of either magnitude or duration of the allodynic response; however, both groups responded to the maximal extent measurable. In a model of inflammatory pain, elicited by intraplantar administration of complete Freund's adjuvant (CFA), CCL2 tg mice displayed both greater edema and thermal hyperalgesia compared with control mice. In control mice, edema and hyperalgesia returned to baseline values 5-7 days post CFA. However, in CCL2 tg mice, thermal hyperalgesia was significantly different from baseline up to 3 weeks post CFA. Parallel to these enhanced behavioral responses CCL2 serum levels were significantly greater in CCL2 overexpressing mice and remained elevated 7 days post CFA. Consequently, proinflammatory cytokine mRNA expression (IL-1beta, IL-6, and TNFalpha) levels were greater in skin, dorsal root ganglia (DRG), and spinal cord, whereas the anti-inflammatory cytokine (IL-10) level was lower in skin and DRG in CCL2 overexpressing mice than in control mice. Taken together with data from CCR2-deficient mice, these present data confirm a key role of CCL2/CCR2 axis in pain pathways and suggest that inhibiting this axis may result in novel pain therapies.


Subject(s)
Astrocytes/physiology , Chemokine CCL2/biosynthesis , Chemokine CCL2/physiology , Pain/physiopathology , Animals , Astrocytes/metabolism , Chemokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Formaldehyde , Freund's Adjuvant , Ganglia, Spinal/metabolism , Hot Temperature , Inflammation/chemically induced , Inflammation/complications , Inflammation/physiopathology , Male , Mice , Pain Measurement , Peripheral Nerve Injuries , Peripheral Nervous System Diseases/complications , Peripheral Nervous System Diseases/physiopathology , Phenotype , Physical Stimulation , Reaction Time/physiology , Reverse Transcriptase Polymerase Chain Reaction , Schwann Cells/physiology , Spinal Cord/physiology
6.
Neuroscience ; 127(2): 419-30, 2004.
Article in English | MEDLINE | ID: mdl-15262332

ABSTRACT

The cloned mu opioid receptor MOR-1 undergoes alternative splicing. Extensive 3'-splicing downstream from exon 3 leads to a number of C-terminal splice variants that are differentially expressed within the CNS. Recently, 5'-splicing has been observed with eight additional variants containing exon 11, a new exon located approximately 10 kb upstream from exon 1 that is under the control of a different promoter located even further upstream. Three of these variants generate the same protein as MOR-1 itself, but under the control of the new exon 11 promoter. Three variants in which exon 11 is translated have been identified within the brain, including MOR-1G, MOR-1M and MOR-1N. The present paper defines immunohistochemically the distribution of these variants using an exon 11-specific antiserum. The expression of exon 11-like immunoreactivity (-LI) was seen primarily in the olfactory tubercle, caudate-putamen, globus pallidus and substantia nigra. We did not observe exon 11-LI in a number of regions expressing MOR-1. Within the caudate-putamen, the general pattern of labeling was diffuse, in contrast to the pattern seen with an exon 4-generated antiserum that labels MOR-1 itself. However, we did observe in the caudate-putamen co-expression of exon 4- and exon 11-LI in cells that were apposed to dopaminergic terminals. These results provide new insights regarding the potential physiological significance of these exon 11-containing variants.


Subject(s)
Alternative Splicing/genetics , Brain/metabolism , Exons/genetics , Promoter Regions, Genetic/genetics , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Animals , Antibody Specificity , Brain/cytology , Cell Line , Corpus Striatum/cytology , Corpus Striatum/metabolism , Dopamine/metabolism , Gene Expression Regulation/genetics , Humans , Immunohistochemistry , Male , Mice , Olfactory Pathways/cytology , Olfactory Pathways/metabolism , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Protein Isoforms/genetics , Protein Isoforms/metabolism , Substantia Nigra/cytology , Substantia Nigra/metabolism
7.
Eur J Neurosci ; 16(6): 1075-82, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12383236

ABSTRACT

The present study characterizes the relationship between the endogenous mu opioid peptides endomorphin-1 (EM-1) and endomorphin-2 (EM-2) and several splice variants of the cloned mu opioid receptor (MOR-1) encoded by the mu opioid receptor gene (Oprm). Confocal laser microscopy revealed that fibers containing EM-2-like immunoreactivity (-LI) were distributed in close apposition to fibers showing MOR-1-LI (exon 4-LI) and to MOR-1C-LI (exons 7/8/9-LI) in the superficial laminae of the lumbar spinal cord. We also observed colocalization of EM-2-LI and MOR-1-LI in a few fibers of lamina II, and colocalization of EM-2-LI and MOR-1C-LI in laminae I-II, and V-VI. To assess the functional relevance of the MOR-1 variants in endomorphin analgesia, we examined the effects of antisense treatments that targeted individual exons within the Oprm1 gene on EM-1 and EM-2 analgesia in the tail flick test. This antisense mapping study implied mu opioid receptor mechanisms for the endomorphins are distinct from those of morphine or morphine-6beta-glucuronide (M6G).


Subject(s)
Alternative Splicing/genetics , Oligopeptides/metabolism , Pain/metabolism , Posterior Horn Cells/metabolism , Receptors, Opioid, mu/metabolism , Amino Acid Sequence/genetics , Animals , Base Sequence/genetics , Exons/genetics , Immunohistochemistry , Male , Mice , Mice, Inbred Strains , Oligopeptides/genetics , Oligopeptides/pharmacology , Pain/drug therapy , Pain/physiopathology , Posterior Horn Cells/cytology , Posterior Horn Cells/drug effects , Protein Structure, Tertiary/genetics , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/genetics
8.
Neuroscience ; 106(4): 833-42, 2001.
Article in English | MEDLINE | ID: mdl-11682168

ABSTRACT

Opioids inhibit nociceptive transmission at the level of the spinal cord, possibly through inhibition of neurotransmitter release by presynaptic mu opioid receptors (MORs) thus preventing the activation of ascending pathways and the perception of pain. Most nociceptive primary afferents are unmyelinated fibers containing peptides such as substance P and/or calcitonin gene-related peptide. However, few terminals contain both substance P and MOR. Recently, we identified new carboxy-terminal MOR splice variants that are localized in the superficial laminae of the dorsal horn. We now report the precise cellular distribution of two of these MOR-1 variants, MOR-1C (exon 7/8/9 epitope) and MOR-1D (exon 8/9 epitope), at the ultrastructural level. In the superficial laminae of the dorsal horn, the majority of the labeling of MOR-1C and MOR-1D was found in unmyelinated axons. This distribution contrasts with that of MOR-1 (exon 4 epitope), in which labeling is equally found in dendrites and soma, as well as in axons. The presence of dense core vesicles in many of the MOR-1C-like immunoreactive terminals implies that this splice variant might be involved in presynaptic inhibition of transmitter release from peptide-containing afferents to the dorsal horn. Consistent with this finding, confocal microscopy analyses showed that many MOR-1C profiles in laminae I-II also contained calcitonin gene-related peptide, whereas fewer MOR-1 profiles contained either substance P or calcitonin gene-related peptide in this same region. From these findings we suggest that there are differential distributions of MOR-1 splice variants as well as distinct peptide colocalizations in the dorsal horn.


Subject(s)
Afferent Pathways/metabolism , Alternative Splicing/physiology , Nociceptors/metabolism , Posterior Horn Cells/metabolism , Presynaptic Terminals/metabolism , Receptors, Opioid, mu/metabolism , Synaptic Transmission/physiology , Afferent Pathways/ultrastructure , Animals , Calcitonin Gene-Related Peptide/metabolism , Epitopes/genetics , Epitopes/metabolism , Immunohistochemistry , Male , Microscopy, Electron , Nerve Fibers/metabolism , Nerve Fibers/ultrastructure , Nociceptors/ultrastructure , Pain/metabolism , Pain/physiopathology , Posterior Horn Cells/ultrastructure , Presynaptic Terminals/ultrastructure , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary/genetics , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/genetics , Substance P/metabolism
9.
Free Radic Biol Med ; 31(8): 943-53, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11595379

ABSTRACT

We have previously shown that overexpressing cRel, a transcription factor of the Rel/NF-kappa B family, concomitantly inhibits proliferation of HeLa cells and makes them resistant against TNF alpha-induced apoptosis. Both effects rely on the upregulation of the manganese superoxide dismutase (MnSOD), a mitochondrial enzyme that converts O(2)(*-) in H(2)O(2). Here we describe additional alterations induced by cRel, namely mitochondrial clustering and accumulation of dense dark granules near the nucleus. These changes preferentially occur in cells that display a sustained cRel expression in the nucleus and that are cell-cycle arrested. As the cell-cycle arrest, these changes are reproduced by directly overexpressing MnSOD or by treating cells with H(2)O(2), suggesting they are due to MnSOD induction and ensuing H(2)O(2) accumulation. We propose that mitochondria cluster because they are damaged by the H(2)O(2) they overproduce. They would then be autophagocytosed and degraded in secondary lysosomes. In support of this scenario, we documented the occurrence of oxidative damage and the presence of lysosomes in the area of mitochondrial clustering. In addition, we identified the dense dark granules as lipofuscin, based on their autofluorescence. Lipofuscin could directly originate from the mitochondrial degradation products that would aggregate and become indigestible because of the presence of H(2)O(2) in the secondary lysosomes. Altogether, our findings show that cRel overexpression in HeLa cells creates, via the induction of MnSOD, an oxidative injury that culminates in mitochondrial degeneration, proliferation blockage, and resistance against TNF alpha-induced apoptosis.


Subject(s)
Cell Nucleus/physiology , HeLa Cells/cytology , Hydrogen Peroxide/metabolism , Mitochondria/physiology , Proto-Oncogene Proteins c-rel/metabolism , Superoxide Dismutase/metabolism , Up-Regulation/genetics , Cell Cycle/physiology , Cell Division/physiology , Cell Survival/physiology , HeLa Cells/metabolism , Humans , Lipofuscin/metabolism , Oxidation-Reduction , Proto-Oncogene Proteins c-rel/genetics , Superoxide Dismutase/genetics , Up-Regulation/physiology
10.
Neuroreport ; 12(14): 3069-72, 2001 Oct 08.
Article in English | MEDLINE | ID: mdl-11568638

ABSTRACT

The mu opioid receptor MOR-1 is internalized by many mu agonists, but not morphine. To see whether differences in the intracellular carboxy terminus influences internalization, we examined internalization of a splice variant of the mu opioid receptor, MOR-1C, in the lateral septum of the mouse in vivo. Following intracerebroventricular (i.c.v.) saline treatment, MOR-1C-like immunoreactivity (LI) within neurons in naive mice was found predominantly in clusters close to the plasma membrane. Following either intracerebroventricular [d-Ala2, MePhe4,Gly(ol)5]enkephalin (DAMGO) or morphine, MOR-1C-LI clustered into endosomes in the cytoplasm. This effect was suppressed by prior administration of the opioid antagonist naloxone. In contrast, only DAMGO, and not morphine, internalized MOR-1-LI. These results illustrate differences in internalization between two MOR-1 variants that have alternative splicing at the COOH terminus.


Subject(s)
Alternative Splicing/physiology , Endocytosis/physiology , Morphine/pharmacology , Neurons/metabolism , Protein Transport/physiology , Receptors, Opioid, mu/genetics , Septal Nuclei/metabolism , Alternative Splicing/drug effects , Animals , Endocytosis/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Immunohistochemistry , Male , Mice , Naloxone/pharmacology , Neurons/cytology , Neurons/drug effects , Protein Isoforms/drug effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary/genetics , Protein Transport/drug effects , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/metabolism , Septal Nuclei/cytology , Septal Nuclei/drug effects
11.
J Biol Chem ; 276(29): 27322-8, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11350953

ABSTRACT

Rel/nuclear factor (NF)-kappaB transcription factors play a major role in the regulation of programmed cell death. A few anti-apoptotic Rel/NF-kappaB target genes have been characterized; they act either downstream in the apoptotic pathway or upstream, for example at the tumor necrosis factor (TNF) receptor level. We found using DNA arrays, reverse transcription-polymerase chain reaction, and immunofluorescence that Rel/NF-kappaB factors up-regulate DcR1, a receptor for TNF-related apoptosis-inducing ligand (TRAIL), a cytokine of the TNF family that induces apoptosis in tumor cells. Four related receptors bind TRAIL, two death receptors (DR4 and DR5) that signal apoptosis and two decoy receptors (DcR1 and DcR2) that act as dominant negative inhibitors of TRAIL-mediated apoptosis. DcR1 is devoid of an intracellular domain and is anchored at the cell surface membrane by a glycophospholipid. Our results indicate that overexpression of cRel or activation of endogenous Rel/NF-kappaB factors by TNFalpha in HeLa cells up-regulates DcR1 without changing the expression of DcR2, DR4, and DR5 and makes cells resistant against TRAIL-induced apoptosis. This resistance is a consequence of DcR1 up-regulation, because it was abolished when DcR1 was removed from the cell surface by a phosphatidylinositol phospholipase C. Therefore, Rel/NF-kappaB transcription factors could regulate the sensitivity of cells to TRAIL, by controlling the ratio of TRAIL-decoy to -death receptors.


Subject(s)
Apoptosis/physiology , Membrane Glycoproteins/physiology , NF-kappa B/physiology , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/physiology , Up-Regulation/physiology , Apoptosis Regulatory Proteins , Base Sequence , DNA Primers , Fluorescent Antibody Technique , GPI-Linked Proteins , HeLa Cells , Humans , Receptors, Tumor Necrosis Factor, Member 10c , Reverse Transcriptase Polymerase Chain Reaction , TNF-Related Apoptosis-Inducing Ligand , Transcription Factor RelA , Tumor Necrosis Factor Decoy Receptors
12.
J Neurosci ; 21(10): 3656-64, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11331395

ABSTRACT

Although the tachykinins substance P (SP) and neurokinin A (NKA) are coreleased from primary afferent nociceptors and act via neurokinin (NK) receptors, their differential effects in vivo are not known. Despite pharmacological evidence that NKA preferentially binds NK-2 receptors, this receptor is not found in spinal cord neurons. Thus, in the present studies, we compared the extent to which SP and NKA contribute to spinal nociceptive processing via the NK-1 receptor. We found that SP and NKA induce NK-1 receptor internalization with identical dose dependence and induce increases in intracellular calcium at the same concentrations, suggesting that SP and NKA equally activate the NK-1 receptor. We found, however, that the selective NK-1 receptor antagonist GR 205171 blocked NKA but not SP-induced NK-1 receptor internalization in the rat spinal cord in vivo and in embryonic day 19 rat spinal neurons in vitro. Using this selectivity of GR 205171 for NKA-induced NK-1 receptor activation, we examined the relative contribution of SP and NKA to noxious stimulus-induced activation of spinal NK-1 receptors. We estimate that NKA contributes to at least 50% of the NK-1 receptor activation in lamina I. Under inflammatory conditions, all noxious stimulus-induced NK-1 receptor internalization in deep dorsal horn neurons was blocked by GR 205171, suggesting that it is entirely NKA-mediated. Substance P-mediated NK-1 receptor internalization was focused at the site of termination of stimulated nociceptors but NKA also activated NK-1 receptors at more distant sites. We conclude that NKA not only targets the NK-1 receptor but may be a predominant pronociceptive primary afferent neurotransmitter.


Subject(s)
Neurokinin A/metabolism , Receptors, Neurokinin-1/metabolism , Signal Transduction/physiology , Spinal Cord/metabolism , Substance P/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Intracellular Fluid/metabolism , Male , Neurokinin A/pharmacology , Neurokinin-1 Receptor Antagonists , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Pain Measurement/drug effects , Piperidines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Spinal Cord/cytology , Spinal Cord/drug effects , Substance P/pharmacology , Tetrazoles/pharmacology
13.
Cancer Res ; 61(6): 2656-64, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11289144

ABSTRACT

Rel/nuclear factor kappaB transcription factors were shown to have either pro- or antiapoptotic as well as pro- or antiproliferative functions, and it is often assumed that the outcome of their activation depends on the cell type or cellular context. Inconsistent with this assumption, we show here that cRel is able in one cell type to inhibit proliferation, protect against apoptosis induced by tumor necrosis factor alpha (TNF-alpha) + cycloheximide (CHX), and increase the basal rate of apoptosis. Both the effects of proliferation inhibition and protection against TNF-alpha + CHX-induced apoptosis are massive and occur in the same cells. Using reverse transcription-PCR, Western blot and immunofluorescence, and transactivation assays, we found that the manganese superoxide dismutase (MnSOD), an enzyme that converts O2*- in H2O2, is up-regulated by cRel through a kappaB site in intron 2. Inhibition of MnSOD induction by antisense oligonucleotides and overexpression of MnSOD respectively reverts and mimics both the antiproliferative and antiapoptotic effects of cRel, suggesting that they both occur via the induction of this gene. On one hand, MnSOD could improve the efficiency of cRel-overexpressing cells in eliminating toxic O2*- produced on TNF-alpha treatment, explaining why they escape TNF-alpha-induced apoptosis. On the other hand, cRel-overexpressing cells should accumulate H2O2. We present evidence linking this H2O2 accumulation to the proliferation arrest induced by cRel. Therefore, different effects on proliferation and apoptosis could arise from the induction of MnSOD and thus coexist in cRel-overexpressing cells.


Subject(s)
Apoptosis/physiology , Proto-Oncogene Proteins c-rel/physiology , Superoxide Dismutase/biosynthesis , Apoptosis/drug effects , Base Sequence , Cell Cycle/physiology , Cell Division/drug effects , Cell Division/physiology , Cycloheximide/toxicity , Gene Expression Regulation, Enzymologic , Genetic Vectors , HeLa Cells , Humans , Hydrogen Peroxide/metabolism , Introns/genetics , Mitochondria/enzymology , NF-kappa B/genetics , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Oxidation-Reduction , Protein Synthesis Inhibitors/toxicity , Proto-Oncogene Proteins c-rel/biosynthesis , Proto-Oncogene Proteins c-rel/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transfection , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/physiology
14.
Neuroscience ; 100(1): 141-53, 2000.
Article in English | MEDLINE | ID: mdl-10996465

ABSTRACT

The present study examined immunohistochemically the CNS distributions of a splice variant of the mu-opioid receptor, MOR-1D, in both rats and mice. In MOR-1D, exon 4 of MOR-1 is replaced by two additional exons that code for seven amino acids. Using rabbit antisera, we compared immunohistochemically the regional distribution of a C-terminal epitope of MOR-1D to that of a C-terminal epitope from MOR-1 and a C-terminal epitope from another splice variant, MOR-1C. The general distribution of MOR-1D-like immunoreactivity was similar in both mouse and rat. MOR-1D-like immunoreactivity was seen in the dentate gyrus and in the mossy fibers of the hippocampal formation, the nucleus of the solitary tract and the area postrema, the inferior olivary nucleus, the nucleus ambiguous, the spinal trigeminal nucleus and the spinal cord. MOR-1D-like immunoreactivity was not observed in some regions containing dense MOR-1-like immunoreactivity, such as the striatum or the locus coeruleus. In regions containing MOR-1, MOR-1C and MOR-1D, the pattern of each variant was unique.MOR-1D and MOR-1C are splice variants of the cloned mu-opioid receptor MOR-1. Although they differ only at the tip of the carboxy terminus, they show marked differences in their regional distributions, as determined immunohistochemically by epitopes in their unique carboxy termini. Since the splice variants are derived from the same gene, these differences in regional distribution imply region-specific messenger RNA processing.


Subject(s)
Central Nervous System/metabolism , DNA, Recombinant , Epitopes/metabolism , Mice/metabolism , Rats/metabolism , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/immunology , Animals , Cell Line , DNA, Complementary/physiology , Humans , Immunohistochemistry , Peptide Fragments/immunology , Receptors, Opioid, mu/metabolism , Tissue Distribution , Trans-Activators , Transfection
15.
Neuroreport ; 11(9): 1953-7, 2000 Jun 26.
Article in English | MEDLINE | ID: mdl-10884050

ABSTRACT

The present study examined the distribution in the human spinal cord of a unique carboxy terminus sequence contained within MOR-1C, one of the recently described splice variants of the cloned mu opioid receptor gene MOR-1. Although MOR-1-like immunoreactivity (LI) and delta opioid receptor-like immunoreactivity were observed only in the superficial laminae, MOR-1C-LI was abundant in the superficial laminae of the dorsal horn and around the central canal. In the substantia gelatinosa, MOR-1C-LI was found in small diameter axonal elements, the cytoplasm and the plasmalemma of small spinal neurons and dendrites in inner lamina II and in some fibers within Lissauer's tract. These studies imply the presence of MOR-1C in human spinal cord and its distribution suggests that it plays a role in the control of pain processing.


Subject(s)
DNA, Recombinant , Peptide Fragments/metabolism , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/genetics , Spinal Cord/metabolism , Animals , Female , Genetic Variation , Humans , Immunohistochemistry , Male , Middle Aged , Neurons/metabolism , Neurons/ultrastructure , Protein Isoforms/metabolism , Rats , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Spinal Cord/cytology , Tissue Distribution
16.
J Comp Neurol ; 419(2): 244-56, 2000 Apr 03.
Article in English | MEDLINE | ID: mdl-10723002

ABSTRACT

The present study examined immunohistochemically the regional distribution of the mu opioid receptor splice variant MOR-1C by using a rabbit antisera generated against the C-terminal peptide sequences and compared it with MOR-1. Overall, the distribution of MOR-1C-like immunoreactivity (-LI) differed from MOR-1-LI. Both MOR-1C-LI and MOR-1-LI were prominent in a few central nervous system regions, including the lateral parabrachial nucleus, the periaqueductal gray, and laminae I-II of the spinal trigeminal nuclei and the spinal cord. In the striatum, hippocampal formation, presubiculum and parasubiculum, amygdaloid nuclei, thalamic nuclei, locus coeruleus, and nucleus ambiguous MOR-1-LI predominated, whereas MOR-1C-LI was absent or sparse. Conversely, MOR-1C-LI exceeded MOR-1-LI in the lateral septum, the deep laminae of the spinal cord, and most hypothalamic nuclei such as the median eminence, periventricular, suprachiasmatic, supraoptic, arcuate, paraventricular, ventromedial, and dorsomedial nuclei. Double-labeling studies showed colocalization of the two receptors in neurons of the lateral septum, but not in the median eminence or in the arcuate nucleus, even though both MOR-1 isoforms were expressed. Because both MOR-1 and MOR-1C are derived from the same gene, these differences in regional distribution represent region-specific mRNA processing. The regional distributions reported in this study involve the epitope seen by the combinations of exons 7, 8, and 9. However, if other MOR-1 variants containing exons 7, 8, and 9 exist, the antisera would not distinguish between them and MOR-1C.


Subject(s)
Brain/metabolism , Rats/metabolism , Receptors, Opioid, mu/metabolism , Alternative Splicing , Amino Acid Sequence/genetics , Animals , CHO Cells , Central Nervous System/metabolism , Cricetinae , DNA, Recombinant , Genetic Variation , Immunoblotting , Immunohistochemistry , Male , Molecular Sequence Data , Peptide Fragments/genetics , Protein Processing, Post-Translational , Rabbits , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/genetics , Tissue Distribution
17.
J Neurosci ; 20(23): 8578-84, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11102461

ABSTRACT

Although both pre- and postsynaptic mechanisms have been implicated in the analgesia produced by mu-opioids at the spinal cord, it is not known under what conditions these different controls come into play. Because the mu-opioid receptor (MOR) can be visualized in individual lamina II excitatory interneurons and internalizes into endosomes on ligand binding, we tested whether MOR internalization could be monitored and used to measure postsynaptic MOR signaling. To test whether endogenous opioids modulate these lamina II interneurons during noxious stimulation, we next assessed the magnitude of postsynaptic MOR internalization under a variety of nociceptive conditions. As observed in other systems, we show that MOR internalization in dorsal horn interneurons is demonstrated readily in response to opioid ligands. The MOR internalization is dose-dependent, with a similar dose-response to that observed for opioid-induced increases in potassium conductance. We demonstrate that MOR internalization in lamina II neurons correlates precisely with the extent of analgesia produced by intrathecal DAMGO. These results suggest that MOR internalization provides a good marker of MOR signaling in the spinal cord and that postsynaptic MORs on lamina II interneurons likely participate in the analgesia that is produced by exogenous opioids. We found, however, that noxious stimuli, under normal or inflammatory conditions, did not induce MOR internalization. Thus, endogenous enkephalins and endomorphins, thought to be released during noxious peripheral stimuli, do not modulate nociceptive messages via postsynaptic MORs on lamina II interneurons. We suggest that any endogenous opioids that are released by noxious stimuli target presynaptic MORs or delta-opioid receptors.


Subject(s)
Interneurons/metabolism , Narcotics/administration & dosage , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Receptors, Opioid, mu/metabolism , Synapses/metabolism , Analgesics, Opioid/pharmacology , Animals , Dose-Response Relationship, Drug , Drug Implants , Electric Stimulation , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , In Vitro Techniques , Inflammation/chemically induced , Inflammation/metabolism , Injections, Spinal , Injections, Subcutaneous , Interneurons/cytology , Interneurons/drug effects , Ligands , Morphine/administration & dosage , Oligopeptides/administration & dosage , Pain Measurement , Piperidines/administration & dosage , Posterior Horn Cells/cytology , Potassium/metabolism , Rats , Receptors, Opioid, mu/agonists , Remifentanil , Signal Transduction/drug effects , Substantia Gelatinosa/cytology , Substantia Gelatinosa/drug effects , Substantia Gelatinosa/metabolism
18.
J Neurosci ; 19(21): 9642-53, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10531466

ABSTRACT

Although opioids can reduce stimulus-evoked efflux of Substance P (SP) from nociceptive primary afferents, the consequences of this reduction on spinal cord nociceptive processing has not been studied. Rather than assaying SP release, in the present study we examined the effect of opioids on two postsynaptic measures of SP release, Fos expression and neurokinin-1 (NK-1) receptor internalization, in the rat. The functional significance of the latter was first established in in vitro studies that showed that SP-induced Ca(2+) mobilization is highly correlated with the magnitude of SP-induced NK-1 receptor internalization in dorsal horn neurons. Using an in vivo analysis, we found that morphine had little effect on noxious stimulus-evoked internalization of the NK-1 receptor in lamina I neurons. However, internalization was reduced when we coadministered morphine with a dose of an NK-1 receptor antagonist that by itself was without effect. Thus, although opioids may modulate SP release, the residual release is sufficient to exert maximal effects on the target NK-1 receptors. Morphine significantly reduced noxious stimulus-induced Fos expression in lamina I, but the Fos inhibition was less pronounced in neurons that expressed the NK-1 receptor. Taken together, these results suggest that opioid analgesia predominantly involves postsynaptic inhibitory mechanisms and/or presynaptic control of non-SP-containing primary afferent nociceptors.


Subject(s)
Analgesia, Epidural , Analgesics, Opioid/pharmacology , Morphine/pharmacology , Pain/physiopathology , Posterior Horn Cells/physiology , Receptors, Neurokinin-1/physiology , Signal Transduction/physiology , Spinal Cord/physiology , Substance P/physiology , Analgesics, Opioid/administration & dosage , Animals , Cells, Cultured , Embryo, Mammalian , Male , Neurokinin-1 Receptor Antagonists , Piperidines/pharmacology , Posterior Horn Cells/drug effects , Proto-Oncogene Proteins c-fos/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tetrazoles/pharmacology
19.
Mol Pharmacol ; 56(2): 396-403, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10419560

ABSTRACT

We have identified four new mu-opiod receptor (MOR)-1 exons, indicating that the gene now contains at least nine exons spanning more than 200 kilobases. Replacement of exon 4 by combinations of the new exons yields three new receptors. When expressed in Chinese hamster ovary cells, all three variants displayed high affinity for mu-opioid ligands, but kappa and delta drugs were inactive. However, there were subtle, but significant, differences in the binding profiles of the three variants among themselves and from MOR-1. Immunohistochemically, the major variant, MOR-1C, displayed a regional distribution quite distinct from that of MOR-1. Region-specific processing also was seen at the mRNA level. Antisense mapping revealed that the four new exons were all involved in morphine analgesia. Together with two other variants generated from alternative splicing of exon 4, there are now six distinct MOR-1 receptors.


Subject(s)
Alternative Splicing , Protein Isoforms/genetics , Receptors, Opioid, mu/genetics , Animals , Brain/metabolism , Cloning, Molecular , Cricetinae , Exons/genetics , Male , Mice , Mice, Inbred ICR , Protein Isoforms/biosynthesis , Protein Isoforms/isolation & purification , RNA, Messenger/biosynthesis , RNA, Messenger/metabolism , Receptors, Opioid, mu/biosynthesis , Receptors, Opioid, mu/isolation & purification
20.
Proc Natl Acad Sci U S A ; 96(1): 260-5, 1999 Jan 05.
Article in English | MEDLINE | ID: mdl-9874806

ABSTRACT

It is common to think of gray matter as the site of integration in neural circuits and white matter as the wires that connect different groups of neurons. The dorsal column (DC) white matter, for example, is the spinal cord axonal pathway through which a topographic map of the body is conveyed to the somatosensory cortex. We now describe a network of neurons located along the midline of the DCs. The neurons are present in several mammals, including primates and birds, and have a profuse dendritic arbor that expresses both the neuron-specific marker, microtubule-associated protein-2, and the neurokinin-1 receptor, a target of the neuropeptide, substance P. Electron microscopy and double immunostaining for synaptophysin and a marker of gamma-aminobutyric acid-ergic terminals documented a rich synaptic input to these neurons. Finally, injection of a gamma-aminobutyric acid type A receptor antagonist or of substance P into the cerebrospinal fluid of the rat spinal cord induced Fos expression and internalization of the neurokinin-1 receptor in these neurons, respectively, indicating that the DC neurons are under tonic inhibitory control and can respond to neurotransmitters that circulate in the cerebrospinal fluid.


Subject(s)
Nerve Net/cytology , Neurons/cytology , Neuropil/cytology , Spinal Cord/cytology , Animals , Antigens, Differentiation , Cats , Dendrites/ultrastructure , Immunohistochemistry , Macaca mulatta , Male , Mice , Microtubule-Associated Proteins/isolation & purification , Nerve Net/ultrastructure , Neurons/ultrastructure , Neuropil/ultrastructure , Neurotransmitter Agents/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/isolation & purification , Songbirds , Species Specificity , Spinal Cord/ultrastructure , Synapses
SELECTION OF CITATIONS
SEARCH DETAIL
...