Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proteomics ; 18(7): e1700408, 2018 04.
Article in English | MEDLINE | ID: mdl-29406625

ABSTRACT

Adverse life experiences increase the lifetime risk to several stress-related psychopathologies, such as anxiety or depressive-like symptoms following stress in adulthood. However, the neurochemical modulations triggered by stress have not been fully characterized. Neuropeptides play an important role as signaling molecules that contribute to physiological regulation and have been linked to neurological and psychiatric diseases. However, little is known about the influence of stress on neuropeptide regulation in the brain. Here, we have performed an exploratory study of how neuropeptide expression at adulthood is modulated by experiencing a period of multiple stressful experiences. We have targeted hippocampus and prefrontal cortex (PFC) brain areas, which have previously been shown to be modulated by stressors, employing a targeted liquid chromatography-mass spectrometry (LC-MS) based approach that permits broad peptide coverage with high sensitivity. We found that in the hippocampus, Met-enkephalin, Met-enkephalin-Arg-Phe, and Met-enkephalin-Arg-Gly-Leu were upregulated, while Leu-enkephalin and Little SAAS were downregulated after stress. In the PFC area, Met-enkephalin-Arg-Phe, Met-enkephalin-Arg-Gly-Leu, peptide PHI-27, somatostatin-28 (AA1-12), and Little SAAS were all downregulated. This systematic evaluation of neuropeptide alterations in the hippocampus and PFC suggests that stressors impact neuropeptides and that neuropeptide regulation is brain-area specific. These findings suggest several potential peptide candidates, which warrant further investigations in terms of correlation with depression-associated behaviors.


Subject(s)
Gene Expression Regulation , Hippocampus/metabolism , Neuropeptides/genetics , Prefrontal Cortex/metabolism , Stress, Psychological/metabolism , Animals , Chromatography, Liquid , Enkephalin, Methionine/genetics , Hippocampus/physiology , Male , Mass Spectrometry , Prefrontal Cortex/physiology , Proteomics , Rats , Somatostatin-28/genetics , Stress, Psychological/genetics
2.
Dev Neurosci ; 35(4): 326-37, 2013.
Article in English | MEDLINE | ID: mdl-23838073

ABSTRACT

Approximately 500,000 infants are born prematurely each year in the United States. These infants typically require an extensive stay in the neonatal intensive care unit (NICU), where they experience on average 14 painful and invasive procedures each day. These procedures, including repeated heel lance, insertion of intravenous lines, and respiratory and gastric suctioning, typically result in an inflammatory response, inducing pain and stress in the newborn. Remarkably, the majority of these procedures are performed in the complete absence of pre- or post-emptive analgesics. Recent clinical studies report that former NICU patients have increased thresholds for pain and stress later in life as compared with term-born infants. However, to date, the mechanisms whereby early-life inflammation alters later-life response to stress and pain are not known. The present studies were conducted to determine if neonatal injury impairs adult responses to anxiety- and stress-provoking stimuli. As we have previously reported that early-life pain results in a significant increase in opioid peptide expression within the midbrain periaqueductal gray, the role of endogenous opioids in our behavioral studies was also examined. Male and female rats received an intraplantar injection of the inflammatory agent carrageenan (1%) on the day of birth. In adulthood, animals were assessed for changes in response to anxiety- and stress-provoking stimuli using the open field and forced swim tests, respectively. Injury-induced changes in sucrose preference and stress-induced analgesia were also assessed. As adults, neonatally injured animals displayed a blunted response to both anxiety- and stress-provoking stimuli, as indicated by significantly more time spent in the inner area of the open field and a 2-fold increase in latency to immobility in the forced swim test as compared to controls. No change in sucrose preference was observed. Using in situ hybridization and immunohistochemistry, we observed a 2-fold increase in enkephalin mRNA and protein expression, respectively, in stress-related brain regions including the central amygdala and lateral septum. Administration of the opioid receptor antagonist naloxone reversed the attenuated responses to forced swim stress and stress-induced analgesia, suggesting the changes in stress-related behavior were opioid-dependent. Together, these data contribute to mounting evidence that neonatal injury in the absence of analgesics has adverse effects that are both long-term and polysystemic.


Subject(s)
Animals, Newborn/physiology , Stress, Psychological/psychology , Amygdala/physiology , Analgesia , Anhedonia , Animals , Anxiety/psychology , Behavior, Animal/drug effects , Densitometry , Enkephalin, Methionine/biosynthesis , Enkephalin, Methionine/genetics , Enkephalins/biosynthesis , Enkephalins/genetics , Female , Food Preferences/drug effects , Immunohistochemistry , In Situ Hybridization , Opioid Peptides/physiology , Periaqueductal Gray/physiology , Pregnancy , Protein Precursors/biosynthesis , Protein Precursors/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Septum of Brain/physiology , Stress, Psychological/physiopathology , Sucrose/pharmacology , Swimming/psychology
3.
Int Immunopharmacol ; 99: 107996, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34311187

ABSTRACT

The antitumor effects of methionine enkephalin (MENK), also known as opioid growth factor (OGF), including its inhibitory effects on cutaneous squamous cell carcinoma (CSCC), have been established. In this study, we determined the precise mechanism by which MENK suppresses CSCC cell growth. In particular, MENK induced G0/G1 cell cycle arrest and promoted apoptosis in CSCC cells via the Bcl-2/Bax/Caspase-3 signaling pathway. Moreover, MENK reduced immunosuppression by downregulating the number of myeloid-derived suppressor cells (MDSCs) and regulating the polarization of tumor-associated macrophages from M2 to M1 in vivo. Furthermore, JAK2/STAT3, an important tumor-promotion and immunosuppression signaling pathway that is involved in MDSC expansion in tumors and macrophage polarization, was inhibited. These findings highlight the potential of the JAK2/STAT3 signaling pathway as a therapeutic target and suggest the clinical application of MENK for CSCC.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Enkephalin, Methionine/metabolism , Enkephalin, Methionine/pharmacology , Macrophages/drug effects , Myeloid-Derived Suppressor Cells/drug effects , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Polarity/drug effects , Cell Proliferation/drug effects , Cytokines/metabolism , Enkephalin, Methionine/genetics , Female , Humans , Immunosuppression Therapy , Janus Kinase 2/metabolism , Mice, Inbred BALB C , Mice, Nude , RNA, Small Interfering , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
4.
Int Immunopharmacol ; 99: 107999, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34315116

ABSTRACT

This study examined the antitumor effect of methionine enkephalin (MENK) against lung cancer in vivo and in vitro and explored the underlying mechanisms. Changes in the immune status of the tumor microenvironment (TME) in response to MENK administration were examined in mice. MENK significantly inhibited the proliferation of lung cancer cells in vivo and in vitro by regulating the Wnt/ß-catenin pathway and causing cell cycle arrest at the G0/G1 phase. Knockdown of opioid growth factor receptor abolished the effect of MENK on lung cancer cells. The immune status of the TME of mice differed between the MENK and control groups. MENK increased the infiltration of M1-type macrophages, natural killer cells, CD8+ T cells, CD4+ T cells, and dendritic cells into the TME, and decreased the proportion of myeloid inhibitory cells and M2-type macrophages. Immunohistochemical analysis of the expression of cytokines in the TME showed that MENK upregulated IL-15, IL-21, IFN-γ, and granzyme B and downregulated IL-10 and TGF-ß1 in mice. Taken together, these finding indicate that MENK may be a potential agent for lung cancer treatment in the future, especially for overcoming immune escape and immune resistance.


Subject(s)
Enkephalin, Methionine/metabolism , Enkephalin, Methionine/pharmacology , Lung Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/drug effects , Dendritic Cells/drug effects , Enkephalin, Methionine/genetics , Female , Humans , Lung Neoplasms/metabolism , Macrophages/drug effects , Mice , Mice, Inbred C57BL , RNA, Small Interfering , T-Lymphocytes/drug effects , Wnt Signaling Pathway/drug effects
5.
Science ; 245(4916): 415-7, 1989 Jul 28.
Article in English | MEDLINE | ID: mdl-2569236

ABSTRACT

Astrocytes have many neuronal characteristics, such as neurotransmitter receptors, ion channels, and neurotransmitter uptake systems. Cultured astrocytes were shown to express certain neuropeptide genes, with specificity for both the gene expressed and the brain region from which the cells were prepared. Somatostatin messenger RNA and peptides were detected only in cerebellar astrocytes, whereas proenkephalin messenger RNA and enkephalin peptides were present in astrocytes of cortex, cerebellum, and striatum. Cholecystokinin was not expressed in any of the cells. These results support the hypothesis that peptides synthesized in astrocytes may play a role in the development of the central nervous system.


Subject(s)
Astrocytes/metabolism , Gene Expression Regulation , Neuropeptides/genetics , Animals , Animals, Newborn , Blotting, Northern , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Corpus Striatum/cytology , Corpus Striatum/metabolism , Enkephalin, Methionine/biosynthesis , Enkephalin, Methionine/genetics , Enkephalins/biosynthesis , Enkephalins/genetics , Neuropeptides/biosynthesis , Protein Precursors/biosynthesis , Protein Precursors/genetics , RNA, Messenger/analysis , Radioimmunoassay , Rats , Somatostatin/biosynthesis , Somatostatin/genetics
6.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 40(6): 1053-5, 2009 Nov.
Article in Zh | MEDLINE | ID: mdl-20067118

ABSTRACT

OBJECTIVE: To investigate the Methionine Enkephalin (M-Enk) expression in lamina II of cat spinal cord and the atypical complex terminal (ACT) after complete dorsal rhizotomy. METHODS: 10 male adult cats were divided into 2 groups after carrying out the complete dorsal rhizotomy: acute group, which survived 2 weeks; chronic group, which survived 2 months. At maturity, all cats were perfused after anaesthesia, the L6 segments of the spinal cord were removed and prepared for light and electron microscopy of M-Enk immunohistochemistry and for observing of ACT. RESULTS: M-Enk immuno-reactivity can be seen in laminal II lateral sides both in operation and control sides in acute group. They were both decreased in chronic group on the photos by naked eyes, and little intensive in operation side on the photos. The immuno-positive ACTs have been founded in the Lamina II of chronic operation side beside some M-Enk immunopositive simple terminals, they are round or ellipse in shape and usually form flat or convex two synapse with two post-compounds. ACTs contained both clear vesicles and dense core vesicles. CONCLUSION: M-Enk express in operation side after complete dorsal rhizotomy is mainly on the lateral side in lamina II. The M-Enk immuno-positive ACT may imply the mechanism of spinal cord plasticity.


Subject(s)
Enkephalin, Methionine/metabolism , Ganglia, Spinal/metabolism , Neuronal Plasticity/physiology , Spinal Cord/physiology , Animals , Cats , Enkephalin, Methionine/genetics , Ganglia, Spinal/surgery , Male , Rhizotomy , Spinal Cord/metabolism
7.
Peptides ; 29(12): 2266-75, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18930087

ABSTRACT

Chimeric peptide of Met-enkephalin and FMRFa (YGGFMKKKFMRFa-YFa), a kappa-opioid receptor specific peptide, did not induce tolerance and cross-tolerance effects to its analgesic action on day 5 after pretreatment with either YFa or morphine for 4 days. However, pretreatment with YFa for 4 days led to the development of cross-tolerance to the analgesic effects of morphine and also 4 days of pretreatment of morphine resulted in the expression of tolerance to its own analgesic effects. Similar expression of tolerance and cross-tolerance were also observed when YFa was compared with the kappa receptor agonist peptide dynorphin A(1-13) [DynA(1-13)]. Cross-tolerance effects between YFa and DynA(1-13) analgesia were also not observed on day 5. Interestingly, when YFa and DynA(1-13) were tested for their analgesic effects for 5 days, reduction in analgesia on day 3 was observed in case of DynA(1-13) whereas YFa maintained its analgesia for 5 days. Thus, chimeric peptide YFa may serve as a useful probe to understand pain modulation and expression of tolerance and cross-tolerance behavior with other opioids.


Subject(s)
Analgesics/pharmacology , Drug Tolerance , Enkephalin, Methionine/genetics , Morphine/pharmacology , Oligopeptides/genetics , Recombinant Fusion Proteins/pharmacology , Analgesia/methods , Analgesics/administration & dosage , Animals , Dynorphins/administration & dosage , Dynorphins/pharmacology , Male , Mice , Morphine/administration & dosage , Pain Measurement/methods , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, kappa/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics
8.
Cell Transplant ; 15(3): 225-30, 2006.
Article in English | MEDLINE | ID: mdl-16719057

ABSTRACT

We have reported that transplantation of adrenal medullary chromaffin cells that release endogenous opioid peptides into pain modulatory regions in the CNS produce significant antinociceptive effects in patients with terminal cancer pain. However, the usefulness of this procedure is minimal because the availability of human adrenal tissue is very limited. Alternative xenogeneic materials, such as porcine and bovine adrenal chromaffin cells present problems of immune rejection and possible pathogenic contamination. In an attempt to develop opioid peptide-producing cells of autologous origin, we have transfected human mesenchymal stem cells (hMeSCs) with a mammalian expression vector containing a fusion gene of green fluorescent protein (GFP) and human preproenkephalin (hPPE), a precursor protein for enkephalin opioid peptides. Enkephalins are major neurotransmitters that play an important role in analgesia by activating peripheral opioid receptors. Following the establishment of stable transfection of hMeSCs, the expressions of hPPE and GFP were confirmed and the production of methionine enkephalin (Met-enkephalin) was significantly increased compared to control naive hMeSCs (p < 0.05). Our in vitro data demonstrated that genetically engineered hMeSCs with transfected hPPE gene can constitutively produce opioid peptide Met-enkephalin at an augmented high level. hMeSCs are relatively easy to isolate from a patient's bone marrow aspirates and expand in culture by repeated passages. Autologous hMeSCs would not require immunosuppression when transplanted back into the same patient. Through targeted gene manipulation such as hPPE gene transfection, this may offer a virtually unlimited safe cell supply for the treatment of opioid-sensitive pain in humans.


Subject(s)
Analgesics/metabolism , Enkephalin, Methionine/genetics , Enkephalin, Methionine/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Analgesics/therapeutic use , Analgesics, Opioid/therapeutic use , Cell Proliferation , Cells, Cultured , DNA/genetics , Enkephalins/genetics , Gene Expression Regulation/genetics , Gene Fusion , Genetic Engineering , Genetic Vectors , Green Fluorescent Proteins/genetics , Humans , Mesenchymal Stem Cells/cytology , Pain/drug therapy , Protein Precursors/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
9.
Sci Rep ; 6: 32799, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27605249

ABSTRACT

Neuropathic pain often results from peripheral nerve damage, which can involve immune response. Local leukocyte-derived opioid peptides or exogenous opioid agonists inhibit neuropathy-induced mechanical hypersensitivity in animal models. Since neuropathic pain can also be augmented by heat, in this study we investigated the role of opioids in the modulation of neuropathy-evoked heat hypersensitivity. We used a chronic constriction injury of the sciatic nerve in wild-type and opioid peptide-knockout mice, and tested opioid effects in heat and mechanical hypersensitivity using Hargreaves and von Frey tests, respectively. We found that although perineural exogenous opioid agonists, including peptidergic ligands, were effective, the endogenous opioid peptides ß-endorphin, Met-enkephalin and dynorphin A did not alleviate heat hypersensitivity. Specifically, corticotropin-releasing factor, an agent triggering opioid peptide secretion from leukocytes, applied perineurally did not attenuate heat hypersensitivity in wild-type mice. Exogenous opioids, also shown to release opioid peptides via activation of leukocyte opioid receptors, were equally analgesic in wild-type and opioid peptide-knockout mice, indicating that endogenous opioids do not contribute to exogenous opioid analgesia in heat hypersensitivity. Furthermore, exogenously applied opioid peptides were ineffective as well. Conversely, opioid peptides relieved mechanical hypersensitivity. Thus, both opioid type and sensory modality may determine the outcome of neuropathic pain treatment.


Subject(s)
Analgesics, Opioid/pharmacology , Neuralgia/drug therapy , Neuralgia/etiology , Opioid Peptides/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Animals , Corticotropin-Releasing Hormone/pharmacology , Dynorphins/genetics , Dynorphins/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Enkephalin, Methionine/genetics , Enkephalin, Methionine/metabolism , Enkephalin, Methionine/pharmacology , Hot Temperature/adverse effects , Male , Mice, Inbred C57BL , Mice, Knockout , Neuralgia/genetics , Opioid Peptides/genetics , Peripheral Nervous System/metabolism , Receptors, Opioid/agonists , beta-Endorphin/genetics , beta-Endorphin/metabolism , beta-Endorphin/pharmacology
10.
Mol Endocrinol ; 4(9): 1408-15, 1990 Sep.
Article in English | MEDLINE | ID: mdl-2233751

ABSTRACT

Previous results have shown that the relative abundance of proenkephalin mRNA in the rat heart is comparable to the levels found in the brain; however, the extractable enkephalin-containing peptide levels are much lower in the heart. This lack of correspondence between the levels of transcript and peptide could arise from either the inefficient translation of proenkephalin transcripts or the translation of proenkephalin transcripts into peptides that are rapidly secreted or degraded. To distinguish between these possibilities, the translational status of proenkephalin mRNA in the rat heart was established by Northern blot analysis of sucrose density gradient-sedimented polysomal fractions and compared to the striatum, which is known to efficiently translate proenkephalin transcripts. In both tissues, we detected 1.5-kilobase transcripts, but an additional larger transcript of approximately 3.6 kilobases was detected in the heart. Both transcripts were associated primarily with polyribosomes, suggesting active translation of proenkephalin mRNA in the rat heart. RIA of the culture media and extracts from primary cultures of neonatal rat cardiomyocytes indicated the presence of immunoreactive Met-enkephalin-Arg6-Phe7, which was stimulated by 8-(4-chlorophenylthio)cAMP. These results suggest that proenkephalin transcripts are translated in the heart and that detectable levels of immunoreactive Met-enkephalin-Arg6-Phe7 are present in the media and cell extracts of primary cultures of neonatal rat cardiomyocytes.


Subject(s)
Enkephalins/genetics , Myocardium/metabolism , Polyribosomes/metabolism , Protein Precursors/genetics , Transcription, Genetic , Animals , Animals, Newborn , Cells, Cultured , Centrifugation, Density Gradient , Corpus Striatum/metabolism , Enkephalin, Methionine/analogs & derivatives , Enkephalin, Methionine/analysis , Enkephalin, Methionine/genetics , Male , Nucleic Acid Hybridization , Protein Biosynthesis , RNA, Messenger/genetics , Rats , Rats, Inbred Strains , Ribonucleases/antagonists & inhibitors
11.
Zhongguo Zhen Jiu ; 35(9): 917-21, 2015 Sep.
Article in Zh | MEDLINE | ID: mdl-26721148

ABSTRACT

OBJECTIVE: To observe the analgesic effect of electroacupuncture (EA) on collagen-induced arthritis (CIA) rats and its regulating effect on inflammation reaction and the endogenous opioid system of synovial tissues. Methods A total of 30 healthy male Wistar rats were randomly divided into a control group, a model group and an EA group, 10 rats in each one. The chronic pain model of CIA rats was made by cattle type-II collagen in the model group and EA group. Rats in the EA group were treated with EA at "Zusanli" (ST 36) and "Kunlun" (BL 60) for 30 min from 16th day after model establishment, once a day for consecutive 10 days. Rats in the control group did not receive any treatment. Rats in the model group were treated with fixation as the EA group. Threshold of pain, arthritis index, paw swelling were measured before model establishment and 16 d, 20 d, 23 d and 25 d after model establishment. The levels of beta-endorphin (ß-END), met-enkephalin (met-ENK), dynorphin A (Dyn A) were measured by radioimmunoassay; the mRNA expressions of mu opioid receptor (MOR), kappa opioid receptor (KOR) and delta opioid receptor (DOR) in synovial tissues of CIA rats were detected by I quantitative polymerase chain reaction (qPCR). RESULTS: Compared with the control group, threshold of pain was reduced (all P<0. 01), arthritis index was increased (all P<0. 01) and paw swelling was increased (all P<0. 01) in the model group on the 16th day, 20th day, 23rd day, 25th day after model establishment. Compared with the model group, the threshold of pain was increased in the EA group (all P<0. 01), arthritis index and paw swelling were reduced (all P<0. 01) on the 23rd day and 25th day after model establishment. Compared with the control group, the level of Dyn A in synovial tissues of CIA rats was increased in the model group (P<0. 01); the mRNA expressions of MOR, KOR and DOR were down-regulated lower than 0. 5 fold of normal level. Compared with the model group, the level of ß-END in synovial tissues of the knee joint was increased in the EA group (P<0. 05), and the mRNA expressions of MOR, KOR and DOR in synovial tissues of CIA rats were up-regulated more than 2 folds of normal level. CONCLUSION: The intervention of EA on chronic pain of CIA rats is superior, which is likely to be related with effects of EA on anti-inflammation and up-regulation of synovial tissue ß-END and MOR, KOR, DOR.


Subject(s)
Analgesics, Opioid/immunology , Arthritis, Rheumatoid/therapy , Chronic Pain/therapy , Electroacupuncture , Synovial Fluid/immunology , Acupuncture Analgesia , Acupuncture Points , Animals , Arthritis, Rheumatoid/immunology , Cattle , Chronic Pain/immunology , Dynorphins/genetics , Dynorphins/immunology , Enkephalin, Methionine/genetics , Enkephalin, Methionine/immunology , Humans , Male , Rats , Rats, Wistar , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/immunology , beta-Endorphin/genetics , beta-Endorphin/immunology
12.
Gene ; 39(2-3): 269-74, 1985.
Article in English | MEDLINE | ID: mdl-3937764

ABSTRACT

Enkephalins are pentapeptides with opioid activity that have been found in brain and other neural tissues. They are released by proteolytic processing of the proenkephalin, which contains several enkephalin sequences each flanked by pairs of basic amino acid (aa) residues. We have constructed an artificial variant of the proenkephalin gene by concatenation of synthetic oligodeoxynucleotides (oligo) coding for Met-enkephalin preceded by two arginines. One of the resulting structures, containing eleven enkephalin sequences separated by pairs of arginine codons, was cloned in the expression vector pRE31. The biological activity of enkephalin was detected after the digestion of the isolated plasmid-coded protein with trypsin and carboxypeptidase B. The product of the synthetic gene may thus serve as a defined simplified substrate for the study of the not yet fully understood enzymatic mechanisms of proenkephalin processing.


Subject(s)
Enkephalins/genetics , Protein Precursors/genetics , Animals , Antigens, Viral, Tumor/genetics , Cloning, Molecular , DNA, Recombinant , Enkephalin, Methionine/genetics , Escherichia coli/genetics , Gene Expression Regulation , Molecular Weight , Oligodeoxyribonucleotides/chemical synthesis , Protein Biosynthesis , beta-Galactosidase/genetics
13.
FEBS Lett ; 237(1-2): 183-6, 1988 Sep 12.
Article in English | MEDLINE | ID: mdl-3139451

ABSTRACT

Searching for somatic cells expressing the preproenkephalin (A) gene in the testis, we have isolated Sertoli cells from the testes of 20-day-old rats. Cultured Sertoli cells contained a single species (about 1.5 kb) of preproenkephalin mRNA, and follicle-stimulating hormone (FSH) transiently increased the mRNA abundance to a maximum (about 30 molecules per cell) at 12 h. Various compounds that activate the cyclic AMP system in Sertoli cells similarly increased the abundance of preproenkephalin mRNA. Moreover, FSH increased intracellular Met-enkephalin immunoreactive peptides in Sertoli cells. Thus, the preproenkephalin gene expression in Sertoli cells is positively regulated by FSH through the cyclic AMP system.


Subject(s)
Enkephalins/genetics , Follicle Stimulating Hormone/pharmacology , Gene Expression Regulation/drug effects , Genes/drug effects , Protein Precursors/genetics , Sertoli Cells/metabolism , Transcription, Genetic/drug effects , Actins/genetics , Animals , Cells, Cultured , Enkephalin, Methionine/biosynthesis , Enkephalin, Methionine/genetics , Kinetics , Male , RNA, Messenger/genetics , Rats , Reference Values , Sertoli Cells/drug effects
14.
Neuroscience ; 55(4): 1067-74, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8232897

ABSTRACT

The time-course effects of one month of estrogen upon calcitonin gene-related peptide - and methionine-enkephalin-immunoreactivity in the periventricular preoptic nucleus and medial preoptic nucleus were semi-quantitatively investigated with a computer-based image analysis system. Female Wistar rats were ovariectomized and implanted subcutaneously with a 10-mm-long silastic capsule containing estradiol-17 beta, or with a blank capsule, as a control. Estradiol-17 beta-treated rats were killed at days 1, 4, 7, 10, 14 and 28 after the implantation of estradiol-17 beta. To investigate the details of changes in calcitonin gene-related peptide- and methionine-enkephalin-immunoreactive fibers in the periventricular preoptic nucleus and medial preoptic nucleus, a grid, made up of 8 x 16 squares (one square corresponding to 50 x 50 microns in the true section), was set on the wall of the third ventricle, and immunoreactivity within each square was measured with an image analyser. In the control rats, calcitonin gene-related peptide- and methionine-enkephalin-immunoreactive fibers were distributed in the periventricular preoptic nucleus and medial preoptic nucleus. In the estradiol-17 beta-treated rats, calcitonin gene-related peptide-immunoreactive fibers increased prominently at day 1, day 7 and day 10 in the periventricular preoptic nucleus, whereas methionine-enkephalin-immunoreactive fibers increased at day 1, day 14 and day 28 in the periventricular preoptic nucleus and medial preoptic nucleus. These findings suggest that the mechanism underlying the increases in these calcitonin gene-related peptide- and methionine-enkephalin-immunoreactive fibers after estrogen treatment might be different.


Subject(s)
Calcitonin Gene-Related Peptide/biosynthesis , Enkephalin, Methionine/biosynthesis , Estradiol/pharmacology , Gene Expression Regulation/drug effects , Preoptic Area/drug effects , Animals , Calcitonin Gene-Related Peptide/genetics , Enkephalin, Methionine/genetics , Female , Image Processing, Computer-Assisted , Nerve Fibers/chemistry , Nerve Fibers/ultrastructure , Ovariectomy , Preoptic Area/metabolism , Preoptic Area/ultrastructure , Rats , Rats, Wistar , Time Factors
15.
Mol Cell Endocrinol ; 113(2): 155-63, 1995 Sep 22.
Article in English | MEDLINE | ID: mdl-8674823

ABSTRACT

Mouse neuroblastoma Neuro-2a cells were examined for the expression of pro-enkephalin mRNA, protein, and Met-enkephalin ([Met]-Enk) peptide. Reverse transcriptase/polymerase chain reaction (RT/PCR) and in situ hybridization demonstrated the presence of pro-enkephalin mRNA in these cells. Immunocytochemistry using an antibody which recognizes pro-enkephalin and high pressure liquid chromatography (HPLC) followed by radioimmunoassay indicated that pro-enkephalin was synthesized in these cells and processed to yield the bioactive pentapeptide, [Met]-Enk. Furthermore, release studies showed that the [Met]-Enk was secreted from these cells with high K+ stimulation. Using double labeling, in situ hybridization combined with immunocytochemistry, we demonstrated that prohormone convertase 2 (PC2) mRNA is colocalized with pro-enkephalin in the same Neuro-2a cells, suggesting that this enzyme may be responsible for processing this precursor. we also showed the presence of vasopressin mRNA and arginine-vasopressin peptide in these cells using in situ hybridization and immunocytochemistry, respectively. Thus, the Neuro-2a cells are a multiple neuropeptide-producing cell line and an excellent model for studying the mechanisms involved in the synthesis, intracellular targeting and processing of endogenous pro-enkephalin and pro-vasopressin, as well as other transfected neuropeptide precursors.


Subject(s)
Enkephalin, Methionine/genetics , Gene Expression , Neuroblastoma/metabolism , RNA, Messenger/metabolism , Vasopressins/genetics , Animals , Arginine Vasopressin/analysis , Chromatography, High Pressure Liquid , Enkephalin, Methionine/metabolism , Enkephalins/analysis , Enkephalins/genetics , Immunohistochemistry , In Situ Hybridization , Mice , Polymerase Chain Reaction , Proprotein Convertase 2 , Protein Precursors/analysis , Protein Precursors/genetics , RNA, Messenger/analysis , RNA-Directed DNA Polymerase , Subtilisins/genetics , Tumor Cells, Cultured , Vasopressins/analysis
16.
J Mol Neurosci ; 11(1): 43-56, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9826785

ABSTRACT

Pituitary adenylate cyclase activating polypeptide-27 (PACAP-27) caused a dose-dependent increase in met-enkephalin secretion and increased production of met-enkephalin peptide and proenkephalin A (PEnk) mRNA in bovine chromaffin cells, at concentrations as low as 300 pM. PACAP-38 was less potent than PACAP-27, but had similar effects. Vasoactive intestinal polypeptide (VIP) (1-100 nM) was without appreciable effect on either enkephalin secretion or biosynthesis, implicating PACAP type I receptors in PACAP-stimulated enkephalin secretion and synthesis. PACAP type I receptors can activate adenylate cyclase and stimulate phospholipase C through heterotrimeric G protein interactions, leading to increased intracellular cyclic AMP (cAMP), inositol triphosphate (IP3)-mediated calcium mobilization, and calcium- and diacylglycerol (DAG)-mediated protein kinase C (PKC) activation. Enkephalin secretion evoked by 10-100 nM PACAP-27 was not inhibited by 1 microM (-)-202-791, an L-type specific dihydropyridine calcium channel blocker, but was inhibited 65-80% by the arylalkylamine calcium channel blocker D600. Forty mM potassium-evoked secretion was inhibited > 90% by both D600 and (-)-202-791, 25 microM forskolin-induced secretion was blocked < 50% by D600 and was unaffected by (-)-202-791, and 100 nM phorbol myristate acetate (PMA)-induced secretion was unaffected by either D600 or (-)-202-791. Enkephalin biosynthesis was increased by 10 nM PACAP-27, as measured by increased met-enkephalin pentapeptide content and PEnk A mRNA levels. PACAP-, forskolin-, and PMA-stimulated enkephalin synthesis were not blocked by D600 or (-)-202-791. Elevated potassium-induced enkephalin biosynthesis upregulation was completely blocked by either D600 or (-)-202-791 at the same concentrations. PACAP acting through type I PACAP receptors couples calcium influx-dependent enkephalin secretion and calcium influx-independent enkephalin biosynthesis in chromaffin cells. Restriction of the effects of enhanced calcium influx to stimulation of secretion, but not of biosynthesis, is unique to PACAP. By contrast, potassium-induced enkephalin biosynthesis upregulation is completely calcium influx dependent, specifically via calcium influx through L-type calcium channels. We propose that subpopulations of voltage-dependent calcium channels are differentially linked to intracellular signal transduction pathways that control neuropeptide gene expression and secretion in chromaffin cells.


Subject(s)
Calcium Signaling/drug effects , Chromaffin Cells/drug effects , Enkephalin, Methionine/metabolism , Neuropeptides/pharmacology , Adrenal Glands/cytology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Calcium Channels, L-Type , Cattle , Chromaffin Cells/metabolism , Colforsin/pharmacology , Cyclic AMP/physiology , Enkephalin, Methionine/biosynthesis , Enkephalin, Methionine/genetics , Enkephalins/genetics , Gene Expression Regulation/drug effects , Male , Models, Biological , Pituitary Adenylate Cyclase-Activating Polypeptide , Potassium/pharmacology , Protein Precursors/genetics , Radioimmunoassay , Tetradecanoylphorbol Acetate/pharmacology , Vasoactive Intestinal Peptide/pharmacology
17.
Brain Res Mol Brain Res ; 10(3): 241-50, 1991 Jun.
Article in English | MEDLINE | ID: mdl-1653391

ABSTRACT

The distribution and characteristics of preproenkephalin (PPenk) mRNA and enkephalin-containing (EC) peptides are compared in CNS and adrenal tissues from Syrian hamsters and Sprague-Dawley rats. Total cellular RNA extracts from both rat and hamster tissues produce a single hybridization band of PPenk mRNA of approximately 1500 bases when analyzed by Northern blot hybridization. Quantitation by solution hybridization reveals that in the hamster the highest levels of PPenk mRNA are found in adrenal (16.3 +/- 1.4 pg equivalents/micrograms RNA (mean +/- S.E.M.)) and striatum (13.3 +/- 0.7), followed by hypothalamus (0.8 +/- 0.2), and hippocampus (0.4 +/- 0.2). In the rat the highest levels of PPenk mRNA are in the striatum (35 +/- 2 pg/micrograms RNA) followed by the hypothalamus (3.0 +/- 0.5), hippocampus (0.3 +/- 0.1) and adrenal (0.18 +/- 0.04). Thus, the rank order of abundance of PPenk mRNA is similar in these CNS tissues for rat and hamster. The hamster adrenal levels are more than 90-fold greater than those of the rat. The abundance of EC peptides in both hamster and rat tissues mirror the rank order found with PPenk mRNA. Hamster adrenal contains the highest level of EC peptides (441 +/- 37 pmol/mg protein (mean +/- S.E.M.)) which is more than 400-fold greater than that of the rat adrenal and 8- to 12-fold greater than that found in rat and hamster striatum or hypothalamus. Both size exclusion chromatography and Western blot analysis indicate that EC peptides in hamster adrenal are predominantly large proenkephalin-like peptides with approximately 6 copies of Met- and 1 copy of Leu-enkephalin and that included in their number is a prominent EC peptide with a molecular weight of 34 kDa. Unilateral denervation of the hamster adrenal results in a time-dependent ipsilateral decrease in EC peptide and PPenk mRNA levels. Thus, by day 8 postsurgery, PPenk mRNA levels have declined by an average of 80% while EC peptides are reduced by 68% when compared to the innervated contralateral adrenal. These results demonstrate the great abundance of PPenk mRNA and EC peptides in the hamster adrenal. They also demonstrate the apparent need for transsynaptic impulse activity to maintain the high steady-state levels of PPenk and EC peptides. These characteristics of the hamster adrenal system provide opportunities for physiological and pharmacological investigations of the regulation of proenkephalin gene expression.


Subject(s)
Adrenal Glands/physiology , Brain/physiology , Denervation , Enkephalin, Leucine/analysis , Enkephalin, Methionine/analysis , Enkephalins/genetics , Protein Precursors/genetics , RNA, Messenger/analysis , Adrenal Glands/innervation , Adrenal Medulla/physiology , Animals , Cricetinae , Enkephalin, Leucine/genetics , Enkephalin, Methionine/genetics , Epinephrine/metabolism , Liver/physiology , Mesocricetus , Norepinephrine/metabolism , Organ Specificity , RNA, Messenger/genetics , Rats , Rats, Inbred Strains , Reference Values
18.
Brain Res Mol Brain Res ; 33(1): 111-20, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8774952

ABSTRACT

[Met5]-enkephalin, encoded by the preproenkephalin (PPE) gene, serves as a growth factor during brain development in addition to its role as a neurotransmitter. This study examined the relationship of gene and peptide expression in the developing (postnatal day 6) rat brain by disrupting peptide-receptor interaction with either a brief (4-6 h) or continuous opioid receptor blockade using a single injection of 1 or 50 mg/kg naltrexone (NTX), respectively; such perturbations result in growth inhibition or acceleration, respectively. In the caudate putamen, an area that has completed neurogenesis by postnatal day 6 and has an abundance of PPE mRNA and enkephalins in adulthood, NTX did not influence PPE mRNA in either NTX group, or the enkephalin levels in the 1 mg/kg NTX group. [Met5]-enkephalin values in the neostriatum, however, were 67-183% greater than controls in rats given 50 mg/kg NTX, beginning 5 min after drug injection. In the cerebellum, PPE mRNA expression was depressed from 5 min to 4 h in the 1 mg/kg NTX group, and was normal thereafter; mRNA levels in the 50 mg/kg NTX group were markedly subnormal for 24 h. Enkephalin levels were significantly depressed within 5 min of drug injection and remained so for 4 h in the 1 mg/kg NTX group, but were elevated to approximately 135% of control values at 8, 16, and 24 h. Enkephalin levels were not changed in the cerebellum of the 50 mg/kg NTX group, or in the plasma of either NTX group. These data suggest that a single exposure to NTX can affect transcriptional and translational mechanisms related to PPE mRNA and opioid peptide expression in a rapid and sustained manner, and that this treatment elicits a specific pattern of alterations dependent upon the brain region sampled, drug dosage, and/or the duration of opioid receptor blockade. Additionally, our results indicate that the decreased DNA synthesis in external germinal cells occurring after opioid receptor blockade as recorded earlier may be related to an increase in the potent opioid growth factor, [Met5]-enkephalin.


Subject(s)
Enkephalin, Methionine/genetics , Enkephalins/genetics , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Protein Precursors/genetics , RNA, Messenger/biosynthesis , Animals , Caudate Nucleus/metabolism , Cerebellum/metabolism , Enkephalin, Methionine/antagonists & inhibitors , Enkephalins/antagonists & inhibitors , Female , Male , Protein Precursors/antagonists & inhibitors , Putamen/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Time Factors
19.
Brain Res Mol Brain Res ; 60(1): 133-9, 1998 Sep 18.
Article in English | MEDLINE | ID: mdl-9748541

ABSTRACT

Opioids have been implicated in sexual differentiation of the brain and in the regulation of reproductive behavior and endocrinology of mammals. Previous studies have indicated that estrogen administration in adults regulates preproenkephalin MRNA levels in several hypothalamic brain nuclei. We have determined preproenkephalin mRNA levels in estrogen-treated juvenile male and female rats to investigate the developmental pattern of estrogenic regulation of enkephalinergic neurons in the medial preoptic area. Rats were treated with estradiol benzoate (20 microgram/kg/day) or oil from day 21 to 23. Sections of the medial preoptic area (mPOA) were studied by in situ hybridization histochemistry at the single cell level and quantified with the assistance of an image analysis system. Our data indicate that males contain higher levels of preproenkephalin mRNA per neuron than females. In addition, our results indicate that estrogen causes an upward shift in the amount of mRNA expressed per cell, females demonstrating a greater response to estrogen than males. An increase in soma cell area following estrogen treatment was observed only in female mPOA enkephalinergic neurons. Taken together, these results indicate a sex difference in total preproenkephalin levels and in estrogenic regulation of preproenkephalin mRNA in the POA of juvenile rats. These results are discussed in relation to the differential role opioids may play in male and female reproductive physiology.


Subject(s)
Enkephalins/genetics , Estrogens/physiology , Preoptic Area/chemistry , Protein Precursors/genetics , Sex Characteristics , Sexual Maturation , Animals , Cell Size/physiology , Enkephalin, Methionine/genetics , Female , Gene Expression Regulation, Developmental , In Situ Hybridization , Male , Neurons/chemistry , Neurons/cytology , Neurons/physiology , Preoptic Area/cytology , Preoptic Area/growth & development , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
20.
Brain Res Mol Brain Res ; 60(2): 160-7, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9757025

ABSTRACT

[Met5]-enkephalin, encoded by the preproenkephalin (PPE) gene, serves as a growth factor (opioid growth factor, OGF) during cardiac development in addition to its role as a neuroregulator. This study examined the ontogeny and relationship of gene and peptide expression in the mammalian heart during late embryonic, preweaning, and postweaning periods. Values for PPE mRNA of hearts in rats from embryonic day 16 (E16) to postnatal day 1 were 33 to 50% of levels found in adults. Adult values for the mature heart were comparable to those in the caudate, an area of the rat brain rich in PPE mRNA. Message gradually decreased during the first postnatal week to 10% of adult values and remained so until weaning. PPE mRNA on days 35 and 50 were three- and sevenfold, respectively, higher than at 21 days, and in adults was more than 50% greater than at day 50. Message for PPE in neonatal heart was regulated rapidly and in a sustained fashion by excess opioid agonist (OGF) or blockade of opioid-receptor interaction. [Met5]-enkephalin levels increased sevenfold between E18 and E20, and another 1.6-fold until birth. Having reached a zenith in the neonate, values for enkephalin-like peptide decreased gradually through the 2nd postnatal week, and were extremely low in adulthood. Indeed, a 43-fold difference in peptide levels was detected between neonatal and adult rat heart. These data provide evidence for the expression of a tightly regulated and distinct growth factor (OGF) during the crucial periods of cell proliferation and differentiation in the mammalian heart, and reveal that the source of OGF is autocrine and/or paracrine.


Subject(s)
Embryonic and Fetal Development , Enkephalin, Methionine/genetics , Enkephalins/genetics , Fetal Heart/metabolism , Gene Expression Regulation, Developmental , Heart/growth & development , Myocardium/metabolism , Protein Precursors/genetics , Aging , Animals , Caudate Nucleus/metabolism , DNA Probes , Enkephalin, Methionine/biosynthesis , Female , Gestational Age , Heart/embryology , Liver/metabolism , Male , Pro-Opiomelanocortin/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL