Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Endocr Regul ; 57(1): 152-161, 2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37561834

RESUMO

Objective. Modified levels of pro- (caspase3, Bax) and anti-apoptotic (Bcl-2) regulatory proteins have been detected in certain brain areas of schizophrenic patients indicating a possible dysregulation of apoptosis. In the present study, effects of antipsychotics, haloperidol (HAL) and olanzapine (OLA), on the gene expression of caspase3 (casp3), Bax and Bcl-2 were studied in vitro in mouse hippocampal mHippoE-2 cell line and in vivo in the hippocampus of MK-801 animal schizophrenia model with the aim to provide evidence that antipsychotics may affect the activity of apoptosis-related markers. Methods. mHippoE-2 cells were incubated with MK-801 (20 µM), HAL (10 µM), and OLA (10 µM) alone or combined, MK-801+HAL/OLA, for 24, 48, and 72 h. Male Sprague Dawley rats were injected with saline or MK-801 (0.5 mg/kg) for 6 days and since the 7th day, they were treated with vehicle (VEH), HAL (1 mg/kg) or OLA (2 mg/kg) for the next 7 days. The casp3, Bax and Bcl-2 gene expression in mHippoE-2 cells and rat hippocampus was measured by RT-PCR. Results. In mHippoE-2 cells, casp3 gene expression was increased by MK-801 and OLA treatments alone for 48 h, HAL treatment alone for 24 and 72 h, and co-treatment with MK-801+OLA for 24 and 72 h compared to controls. HAL and OLA suppressed the stimulatory effect of MK-801 on casp3 mRNA levels in cells after 48 h of incubation. Bax mRNA levels in mHippoE-2 cells were decreased after HAL treatment for 24 and 48 h, and also after co-treatment with MK-801+HAL for 72 h. In vivo, MK-801 decreased mRNA levels of both pro-apoptotic markers, casp3 and Bax, in hippocampus of VEH-treated rats and Bax mRNA levels in hippocampus of HAL-treated animals. OLA reversed the inhibitory effect of MK-801 on casp3 expression in the VEH-treated animals. Neither MK-801 nor antipsychotics induced changes in the gene expression of anti-apoptotic marker Bcl-2 in mHippoE-2 cells as well as hippocampus of rats. Conclusions. The results of the present study demonstrate that antipsychotics, HAL and OLA, may affect mRNA levels of pro-apoptotic markers in hippocampal cells in vitro, but not in vivo. The obtained data do not clearly support the assumed potentiating role of MK-801 in inducing apoptosis in specific brain areas and a possible protective role of antipsychotics against induction of apoptosis. The obtained data may contribute to a deeper insight into the neurodevelopmental changes connected with schizophrenia.


Assuntos
Antipsicóticos , Ratos , Masculino , Camundongos , Animais , Antipsicóticos/farmacologia , Haloperidol/farmacologia , Olanzapina/farmacologia , Caspase 3/farmacologia , Maleato de Dizocilpina/farmacologia , Proteína X Associada a bcl-2/genética , Benzodiazepinas/farmacologia , Ratos Sprague-Dawley , Apoptose , Hipocampo
2.
Int J Mol Sci ; 23(14)2022 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-35887056

RESUMO

Aberrant neurogenesis in the subventricular zone (SVZ) and hippocampus (HIP) contributes to schizophrenia pathogenesis. Haloperidol (HAL) and olanzapine (OLA), commonly prescribed antipsychotics for schizophrenia treatment, affect neurogenesis too. The effect of HAL and OLA on an mHippoE-2 cell line was studied in vitro where we measured the cell number and projection length. In vivo, we studied the gene expression of DCX, Sox2, BDNF, and NeuN in the SVZ and HIP in an MK-801-induced animal schizophrenia model. Cells were incubated with HAL, OLA, and MK-801 for 24, 48, and 72 h. Animals were injected for 6 days with saline or MK801 (0.5 mg/kg), and from the 7th day with either vehicle HAL (1 mg/kg) or OLA (2 mg/kg), for the next 7 days. In vitro, HAL and OLA dose/time-dependently suppressed cells' proliferation and shortened their projection length. HAL/OLA co-treatment with MK-801 for 24 h reversed HAL's/OLA's inhibitory effect. In vivo, HAL and OLA suppressed DCX and NeuN genes' expression in the HIP and SVZ. MK-801 decreased DCX and NeuN genes' expression in the HIP and OLA prevented this effect. The data suggest that subchronic HAL/OLA treatment can inhibit DCX and NeuN expression. In an MK-801 schizophrenia model, OLA reversed the MK-801 inhibitory effect on DCX and NeuN and HAL reversed the effect on DCX expression; however, only in the HIP.


Assuntos
Antipsicóticos , Esquizofrenia , Animais , Antipsicóticos/uso terapêutico , Benzodiazepinas , Proliferação de Células , Modelos Animais de Doenças , Maleato de Dizocilpina/farmacologia , Maleato de Dizocilpina/uso terapêutico , Haloperidol/farmacologia , Haloperidol/uso terapêutico , Hipocampo , Olanzapina/farmacologia , Esquizofrenia/tratamento farmacológico , Esquizofrenia/genética
3.
Neurochem Res ; 46(2): 159-164, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33170479

RESUMO

The corticotropin-releasing hormone family of peptides is involved in regulating the neuroendocrine stress response. Also, the vagus nerve plays an important role in the transmission of immune system-related signals to brain structures, thereby orchestrating the neuroendocrine stress response. Therefore, we investigated gene expression of urocortin 2 (Ucn2) and c-fos, a markers of neuronal activity, within the hypothalamic paraventricular nucleus (PVN), a brain structure involved in neuroendocrine and neuroimmune responses, as well as in the adrenal medulla and spleen in vagotomized rats exposed to immune challenge. In addition, markers of neuroendocrine stress response activity were investigated in the adrenal medulla, spleen, and plasma. Intraperitoneal administration of lipopolysaccharide (LPS) induced a significant increase of c-fos and Ucn2 gene expression in the PVN, and adrenal medulla as well as increases of plasma corticosterone levels. In addition, LPS administration induced a significant increase in the gene expression of tyrosine hydroxylase (TH) and phenylethanolamine N-methyltransferase (PNMT) in the adrenal medulla. In the spleen, LPS administration increased gene expression of c-fos, while gene expression of TH and PNMT was significantly reduced, and gene expression of Ucn2 was not affected. Subdiaphragmatic vagotomy significantly attenuated the LPS-induced increases of gene expression of c-fos and Ucn2 in the PVN and Ucn2 in the adrenal medulla. Our data has shown that Ucn2 may be involved in regulation of the HPA axis in response to immune challenge. In addition, our findings indicate that the effect of immune challenge on gene expression of Ucn2 is mediated by vagal pathways.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Urocortinas/metabolismo , Medula Suprarrenal/efeitos dos fármacos , Animais , Hormônio Liberador da Corticotropina/genética , Masculino , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos Sprague-Dawley , Baço/efeitos dos fármacos , Urocortinas/genética , Vagotomia , Nervo Vago/cirurgia
4.
Anticancer Drugs ; 32(10): 1011-1018, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34145181

RESUMO

The sympathetic nervous system participates in the development and progression of several cancer types and this effect is mediated mainly via ß-adrenergic signaling. However, the potential of ß-adrenergic signaling blockade to prevent cancer development after exposure to carcinogens has not been investigated, yet. Therefore, in our study, we determined the effect of the ß-blocker propranolol on the development and progression of mammary cancer induced in female rats by administration of the chemical carcinogen N-methyl-N-nitrosourea (MNU). The propranolol treatment (20 mg/kg body weight) started 12 days after MNU administration and lasted 10 weeks. We found that both saline and propranolol treatment significantly increased gene expression of the catecholamine-synthesizing enzyme tyrosine hydroxylase, indicating that repeated injection of saline or propranolol-induced stress in these two groups. However, compared to the vehicle-treated group, propranolol slightly delayed the development and moderately reduced the incidence of mammary carcinoma in animals. To evaluate the mechanisms mediating the effect of propranolol on the development of MNU-induced cancer, we investigated several parameters of the tumor microenvironment and found that propranolol increased gene expression of Casp3. Our data indicate that propranolol treatment that starts after exposure to carcinogens might represent a new, useful approach for preventing the development of cancer, especially in stressed individuals. However, the potential efficiency of propranolol treatment for preventing cancer development and progression in individuals exposed to carcinogens needs further investigation.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Propranolol/farmacologia , Animais , Caspase 3/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Metilnitrosoureia/farmacologia , Feniletanolamina N-Metiltransferase/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos beta/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Tirosina 3-Mono-Oxigenase/efeitos dos fármacos
5.
Endocr Regul ; 55(3): 153-162, 2021 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-34523299

RESUMO

Objective. Changes in the brain derived neurotrophic factor (BDNF) and glucocorticoid receptor (GR) expression in the prefrontal cortex (PFC) and hippocampus (HIP) are associated with psychiatric diseases and stress response. Chronic mild stress (CMS) may alter BDNF as well as GR levels in both the PFC and the HIP. The aim of the present study was to find out whether chronic treatment with a typical antipsychotic haloperidol (HAL) and an atypical antipsychotic aripiprazole (ARI) may modify the CMS effect on the BDNF and GR expression in the above-mentioned structures. Methods. The rats were exposed to CMS for 3 weeks and from the 7th day of CMS injected with vehicle (VEH), HAL (1 mg/kg) or ARI (10 mg/kg) for 4 weeks. BDNF and GR mRNA levels were established in the PFC and the HIP by Real Time PCR, whereas, PFC and HIP samples were obtained by punching them from 500 µm thick frozen sections. C-Fos immunoreactivity was analyzed in the PFC and the HIP on 30 µm thick paraformaldehyde fixed sections. Weight gain and corticosterone (CORT) levels were also measured. Results. The CMS and HAL suppressed the BDNF and GR mRNA levels in the PFC. In the HIP, CMS elevated BDNF mRNA levels that were suppressed by HAL and ARI treatments. The CMS decreased the c-Fos immunoreactivity in the PFC in both HAL- and ARI-treated animals. In the HIP, HAL increased the c-Fos immunoreactivity that was again diminished in animals exposed to CMS. Stressed animals gained markedly less weight until the 7th day of CMS, however, later their weight gain did not differ from the unstressed ones or was even higher in CMS+HAL group. Un-stressed HAL and ARI animals gained less weight than the VEH ones. Neither CMS nor HAL/ARI affected the plasma CORT levels. Conclusion. The present data indicate that HAL and ARI in the doses 1 mg/kg or 10 mg/kg, respectively, does not modify the effect of the CMS preconditioning on the BDNF and GR mRNA levels in the PFC or the HIP. However, HAL seems to modify the CMS effect on the HIP activation.


Assuntos
Antipsicóticos , Haloperidol , Animais , Antipsicóticos/farmacologia , Aripiprazol/farmacologia , Fator Neurotrófico Derivado do Encéfalo/genética , Hipocampo , Córtex Pré-Frontal , Ratos , Receptores de Glucocorticoides/genética
6.
Eur J Neurosci ; 51(12): 2376-2393, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31883212

RESUMO

The brain exerts complex effects on the initiation and progression of cancer in the body. However, the influence of cancer localized in peripheral tissues on the brain has been only partially described. Therefore, we investigated gene expression in brain structures that participate in transmitting viscerosensory signals, regulating autonomic functions and food intake, as well as cognition in C57Bl/6J mice with B16-F10 melanoma. In addition, we investigated the relationship between peripheral inflammation and neuroinflammation. We found increased neuronal activity in the nucleus of the solitary tract of tumor-bearing mice, whereas neuronal activity in the A1/C1 catecholaminergic cell group, parabrachial nucleus, lateral hypothalamic area, ventromedial nucleus of the hypothalamus, paraventricular nucleus of the hypothalamus, and hippocampus was decreased. In the majority of investigated brain structures, we found increased gene expression of IL-1ß, whereas gene expression of IL-6 and NF-κB was reduced or unchanged compared with controls. Melanoma-bearing mice also showed increased gene expression of tyrosine hydroxylase in the A1/C1 catecholaminergic cell group, nucleus of the solitary tract, and locus coeruleus, as well as reduced mRNA levels of hypocretin neuropeptide precursor protein in the lateral hypothalamic area, and proopiomelanocortin in the arcuate nucleus. In addition, we found reduced mRNA levels of Bcl-2, brain-derived neurotrophic factor, and doublecortin in the hippocampus. Our data indicate that skin melanoma induces complex changes in the brain, and these changes are most probably caused by cancer-related signals mediated by pro-inflammatory cytokines.


Assuntos
Hipotálamo , Melanoma , Animais , Encéfalo , Cognição , Ingestão de Alimentos , Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Sensação
7.
Stress ; 19(5): 528-34, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27416924

RESUMO

Accumulated evidence indicates that sympathetic nerves may potentiate tumor growth, including melanoma. To elucidate possible mechanisms for this effect, we performed chemical sympathectomy by intraperitoneal (i.p.) injection of the neurotoxin 6-hydroxydopamine hydrobromide (100 mg/kg of body weight); in nine adult male C57BL/6J mice; nine control mice received i.p. vehicle (VEH). Seven days later, all mice were injected subcutaneously with 3 × 10(3) B16-F10 melanoma cells. Mice were euthanized 20 d after injection of melanoma cells, for measurement of tumor weight and expression of genes related to sympathetic signaling, apoptosis, hypoxia and angiogenesis in tumor tissue. To assess potential involvement of the hypothalamo-pituitary-adrenocortical axis in the effect of sympathectomy on melanoma growth, concentrations of plasma corticosterone and level of glucocorticoid receptor mRNA in tumor tissue were determined. We found that sympathectomy significantly attenuated melanoma growth (tumor weight 0.29 ± 0.16 g versus 1.02 ± 0.30 g in controls; p < 0.05). In tumor tissue from sympathectomized mice, we found significantly increased gene expression (measured by real-time PCR), relative to VEH-injected controls, of tyrosine hydroxylase, neuropeptide Y and glucocorticoid receptor (all p < 0.05), and alpha1, beta1 and beta3 adrenergic receptors (all p < 0.025), and factors related to apoptosis (Bcl-2 and caspase-3; p < 0.05) and hypoxia (hypoxia inducible factor 1 alpha) (p = 0.005). Plasma corticosterone concentrations were significantly elevated (p < 0.05) in these mice. Our findings indicate that sympathectomy induces complex changes in the tumor microenvironment reducing melanoma growth. Such complex changes should be considered in the prediction of responses of cancer patients to interventions affecting sympathetic signaling in tumor tissue and its environment.


Assuntos
Regulação Neoplásica da Expressão Gênica , Melanoma Experimental/cirurgia , Sistema Nervoso Simpático/cirurgia , Animais , Apoptose/fisiologia , Caspase 3/metabolismo , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeo Y/metabolismo , Oxidopamina/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Simpatectomia Química , Carga Tumoral , Microambiente Tumoral , Tirosina 3-Mono-Oxigenase/metabolismo
8.
Am J Physiol Regul Integr Comp Physiol ; 308(6): R517-29, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25589013

RESUMO

The involvement of the nonclassical renin-angiotensin system (RAS) in the adrenomedullary response to stress is unclear. Therefore, we examined basal and immobilization stress (IMO)-triggered changes in gene expression of the classical and nonclassical RAS receptors in the rat adrenal medulla, specifically the angiotensin II type 2 (AT2) and type 4 (AT4) receptors, (pro)renin receptor [(P)RR], and Mas receptor (MasR). All RAS receptors were identified, with AT2 receptor mRNA levels being the most abundant, followed by the (P)RR, AT1A receptor, AT4 receptor, and MasR. Following a single IMO, AT2 and AT4 receptor mRNA levels decreased by 90 and 50%, respectively. Their mRNA levels were also transiently decreased by repeated IMO. MasR mRNA levels displayed a 75% transient decrease as well. Conversely, (P)RR mRNA levels were increased by 50% following single or repeated IMO. Because of its abundance, the function of the (P)RR was explored in PC-12 cells. Prorenin activation of the (P)RR increased phosphorylation of extracellular signal-regulated kinase 1/2 and tyrosine hydroxylase at Ser(31), likely increasing its enzymatic activity and catecholamine biosynthesis. Together, the broad and dynamic changes in gene expression of the nonclassical RAS receptors implicate their role in the intricate response of the adrenomedullary catecholaminergic system to stress.


Assuntos
Medula Suprarrenal/metabolismo , Imobilização , Sistema Renina-Angiotensina , Estresse Psicológico/metabolismo , Medula Suprarrenal/fisiopatologia , Animais , Catecolaminas/biossíntese , Modelos Animais de Doenças , Regulação da Expressão Gênica , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Células PC12 , Fosforilação , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , ATPases Translocadoras de Prótons/genética , ATPases Translocadoras de Prótons/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Tipo 2 de Angiotensina/genética , Receptor Tipo 2 de Angiotensina/metabolismo , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Sistema Renina-Angiotensina/genética , Serina , Estresse Psicológico/genética , Estresse Psicológico/fisiopatologia , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/metabolismo , ATPases Vacuolares Próton-Translocadoras
9.
Gen Physiol Biophys ; 33(3): 357-64, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24968410

RESUMO

The left and right ventricles fulfill different role in heart function. Here we compare chamber specific changes in local catecholamine concentrations; gene expression and the receptor protein amount of all three ß-adrenoceptors (ß-AR) in rat right heart ventricles exposed to acute (1 session) and repeated (7 sessions) immobilization stress (IMMO) vs. previously observed changes in left ventricles. Density of muscarinic receptors as main cardio-inhibitive receptors was also measured. In the right ventricles, noradrenaline and adrenaline were increased. No ß1-AR changes were observed, in spite of the increased sympathetic activity. On the other hand, we have found a decrease of ß2-AR gene expression (reduction to 30%) after 7 IMMO and protein (to 59%) after 1 IMMO. ß3-AR gene expression was increased after 7 IMMO. Muscarinic receptor density was not changed. When comparing correlation in left and right ventricles, there was strong correlation between adrenaline and ß2-AR gene expression, protein and ß3-AR gene expression in the left ventricles while only correlation between adrenaline and ß2-AR mRNA and protein in the right ventricles was found. Our results show that maintenance of cardiac homeostasis under stress conditions are to a great extent achieved by a balance between different receptors and also by a balanced receptor changes in left vs. right ventricles. Taken together, decrease of cardio-stimulating ß2-AR represents a new important mechanism by which ß2-AR contributes to the heart physiology.


Assuntos
Ventrículos do Coração/metabolismo , Ventrículos do Coração/fisiopatologia , Receptores Adrenérgicos beta/metabolismo , Receptores Muscarínicos/metabolismo , Estresse Fisiológico , Animais , Sítios de Ligação , Catecolaminas/biossíntese , Epinefrina/biossíntese , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Masculino , Norepinefrina/biossíntese , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Restrição Física
10.
J Neurochem ; 125(2): 185-92, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23320836

RESUMO

The corticotropin-releasing hormone (CRH) family regulates the endocrine stress response. Here, we examined the effect of immobilization stress (IMO) on gene expression of adrenomedullary CRH family members. Urocortin 2 (Ucn2) has the highest basal gene expression and is increased by > 30-fold in response to single IMO and about 10-fold after six daily repeated IMO. IMO also induced a smaller rise in CRH (six-fold) and CRH receptor type 1 (CRHR1; two-fold with single IMO). The influence of glucocorticoids was examined. Dexamethasone (DEX) or corticosterone greatly increased Ucn2 mRNA levels in PC12 cells in a dose-dependent manner. The DEX elicited rise in Ucn2 was abolished by actinomycin D pre-treatment, indicating a transcriptionally mediated response. DEX also triggered a rise in CRHR1 and lowered CRH mRNA levels. In CRH-knockout mice, where the IMO-induced rise in corticosterone was attenuated, the response of IMO on Ucn2, as well as CRHR2 mRNAs was absent. Overall, the results suggest that the stress-triggered rise in glucocorticoids is involved in the large induction of Ucn2 mRNA levels by IMO, which may allow Ucn2 to act in an autocrine/paracrine fashion to modulate adrenomedullary function, or act as an endocrine hormone.


Assuntos
Medula Suprarrenal/metabolismo , Hormônio Liberador da Corticotropina/biossíntese , Glucocorticoides/metabolismo , Estresse Psicológico/metabolismo , Urocortinas/biossíntese , Animais , Regulação da Expressão Gênica , Masculino , Camundongos Knockout , Peptídeos , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Hormônio Liberador da Corticotropina/biossíntese , Restrição Física
11.
J Neuroimmunol ; 376: 578033, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36738563

RESUMO

Accumulated data indicate that inflammation affecting brain structures participates in the development of cancer-related cachexia. However, the mechanisms responsible for the induction and progression of cancer-related neuroinflammation are still not fully understood. Therefore, we studied the time-course of neuroinflammation in selected brain structures and cachexia development in tumor-bearing rats. After tumor cells inoculation, specifically on the 7th, 14th, 21st, and 28th day of tumor growth, we assessed the presence of cancer-associated cachexia in rats. Changes in gene expression of inflammatory factors were studied in selected regions of the hypothalamus, brain stem, and circumventricular organs. We showed that the initial stages of cancer growth (7th and 14th day after tumor cells inoculation), are not associated with cachexia, or increased expression of inflammatory molecules in the brain. Even when we did not detect cachexia in tumor-bearing rats by the 21st day of the experiment, the inflammatory brain reaction had already started, as we found elevated levels of interleukin 1 beta, interleukin 6, tumor necrosis factor alpha, and glial fibrillary acidic protein mRNA levels in the nucleus of the solitary tract. Furthermore, we found increased interleukin 1 beta expression in the locus coeruleus and higher allograft inflammatory factor 1 expression in the vascular organ of lamina terminalis. Ultimately, the most pronounced manifestations of tumor growth were present on the 28th day post-inoculation of tumor cells. In these animals, we detected cancer-related cachexia and significant increases in interleukin 1 beta expression in all brain areas studied. We also observed significantly decreased expression of the glial cell activation markers allograft inflammatory factor 1 and glial fibrillary acidic protein in most brain areas of cachectic rats. In addition, we showed increased expression of cluster of differentiation 163 and cyclooxygenase 2 mRNA in the hypothalamic paraventricular nucleus, A1/C1 neurons, and area postrema of cachectic rats. Our data indicate that cancer-related cachexia is associated with complex neuroinflammatory changes in the brain. These changes can be found in both hypothalamic as well as extrahypothalamic structures, while their extent and character depend on the stage of tumor growth.


Assuntos
Caquexia , Fibrossarcoma , Ratos , Animais , Ratos Wistar , Caquexia/metabolismo , Interleucina-1beta/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Doenças Neuroinflamatórias , Encéfalo/metabolismo , Fibrossarcoma/metabolismo , Inflamação/metabolismo , RNA Mensageiro
12.
Cell Mol Neurobiol ; 32(5): 795-800, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22198558

RESUMO

With acute stress, the release of adrenomedullary catecholamines is important for handling the emergency situation. However, when chronic or repeated, stress alters the allostatic load and leads to a hyperadrenergic state, resulting in the development or worsening of a wide range of diseases. To help elucidate the mechanism, we examined the effects of single and repeated immobilization stress on gene expression of components of neurosecretory vesicles in the adrenal medulla. Male Sprague-Dawley rats were exposed to immobilization stress once for 2 h (1× IMO) or daily for six consecutive days (6× IMO). Compared to unstressed controls, 1× IMO elevated gene expression of vesicular monoamine transporter 2 (VMAT2). In response to 6× IMO, not only was VMAT2 mRNA still elevated, but chromogranin A (CgA) and chromogranin B (CgB) mRNAs were also increased two to three-fold above basal levels. To investigate the possible role of the hypothalamic-pituitary-adrenal axis in the induction of VMAT2, PC12 cells were treated with the synthetic glucocorticoid dexamethasone, which was found to elevate VMAT2 mRNA expression. The findings suggest that following repeated stress, elevations of various components of neurosecretory vesicles in the adrenal can facilitate more efficient utilization of the well-characterized heightened catecholamine levels.


Assuntos
Medula Suprarrenal/metabolismo , Regulação da Expressão Gênica , Sistemas Neurossecretores/metabolismo , Vesículas Secretórias/metabolismo , Estresse Fisiológico/genética , Medula Suprarrenal/efeitos dos fármacos , Animais , Dexametasona/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/metabolismo , Masculino , Sistemas Neurossecretores/efeitos dos fármacos , Células PC12 , Ratos , Ratos Sprague-Dawley , Vesículas Secretórias/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos , Proteínas Vesiculares de Transporte de Monoamina/genética , Proteínas Vesiculares de Transporte de Monoamina/metabolismo
13.
Neurochem Int ; 152: 105224, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34798194

RESUMO

CRH system integrates responses to stress challenges, whereas antipsychotics may impinge on this process. Effect of haloperidol (HAL) and aripiprazole (ARI) on chronic mild stress (CMS) induced neurobehavioral and CRH/CRHR1 system changes was studied in functionally interconnected rat brain areas including prefrontal cortex (PFC), bed nucleus of the stria terminalis (BNST), hypothalamic paraventricular nucleus (PVN), hippocampus (HIP), and amygdala (AMY). Animals were exposed to CMS for 3-weeks and since the 7th day of CMS injected with vehicle (VEH), HAL (1 mg/kg) or ARI (10 mg/kg) for 4-weeks. Expression levels of CRH, CRHR1, and c-fos genes and anxiety-like and anhedonia behavioural patterns were evaluated. CMS in VEH animals suppressed CRH gene expression in the PFC and BNST, c-fos expression in all areas, except HIP, and increased CRHR1 gene expression in the HIP. Antipsychotics decreased CRH gene expression in all areas, except HIP and by CMS elevated CRHR1 expression in the HIP (ARI also in AMY). CMS and antipsychotics decreased the sucrose preference. Aripiprazole prevented CRH expression decrease in the BNST and sucrose preference induced by CMS. Haloperidol increased time spent in the EPM open arms. These data indicate that HAL and ARI selectively influenced behavioural parameters and CRH/CRHR1 gene expression levels in CMS animals.


Assuntos
Aripiprazol/farmacologia , Comportamento Animal/efeitos dos fármacos , Hormônio Liberador da Corticotropina/efeitos dos fármacos , Haloperidol/farmacologia , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Antipsicóticos/farmacologia , Ansiedade/induzido quimicamente , Ansiedade/tratamento farmacológico , Hormônio Liberador da Corticotropina/metabolismo , Hormônio Liberador da Corticotropina/farmacologia , Haloperidol/metabolismo , Masculino , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo
14.
J Biol Chem ; 285(52): 41172-86, 2010 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-20978123

RESUMO

Hypertension is a serious risk factor for cardiovascular disease, and the angiotensinogen (AGT) gene locus is associated with human essential hypertension. The human AGT (hAGT) gene has an A/G polymorphism at -6, and the -6A allele is associated with increased blood pressure. However, transgenic mice containing 1.2 kb of the promoter with -6A of the hAGT gene show neither increased plasma AGT level nor increased blood pressure compared with -6G. We have found that the hAGT gene has three additional SNPs (A/G at -1670, C/G at -1562, and T/G at -1561). Variants -1670A, -1562C, and -1561T almost always occur with -6A, and variants -1670G, -1562G, and -1561G almost always occur with -6G. Therefore, the hAGT gene may be subdivided into either -6A or -6G haplotypes. We show that these polymorphisms affect the binding of HNF-1α and glucocorticoid receptor to the promoter, and a reporter construct containing a 1.8-kb hAGT gene promoter with -6A haplotype has 4-fold increased glucocorticoid-induced promoter activity as compared with -6G haplotype. In order to understand the physiological significance of these haplotypes in an in vivo situation, we have generated double transgenic mice containing either the -6A or -6G haplotype of the hAGT gene and the human renin gene. Our ChIP assay shows that HNF-1α and glucocorticoid receptor have stronger affinity for the chromatin obtained from the liver of transgenic mice containing -6A haplotype. Our studies also show that transgenic mice containing -6A haplotype have increased plasma AGT level and increased blood pressure as compared with -6G haplotype. Our studies explain the molecular mechanism involved in association of the -6A allele of the hAGT gene with hypertension.


Assuntos
Alelos , Angiotensinogênio/sangue , Pressão Sanguínea/genética , Haplótipos , Hipertensão/sangue , Polimorfismo de Nucleotídeo Único , Angiotensinogênio/genética , Animais , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Humanos , Hipertensão/genética , Fígado/metabolismo , Camundongos , Camundongos Transgênicos , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Renina/genética , Renina/metabolismo
15.
Cell Mol Neurobiol ; 30(8): 1459-65, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21046458

RESUMO

Vesicular monoamine transporters (VMATs) mediate transmitter uptake into neurosecretory vesicles. There are two VMAT isoforms, VMAT1 and VMAT2, encoded by separate genes and displaying different cellular distributions and pharmacological properties. We examined the effect of immobilization stress (IMO) on expression of VMATs in the rat adrenal medulla. Under basal conditions, VMAT1 is widely expressed in all adrenal chromaffin cells, while VMAT2 is co-localized with tyrosine hydroxylase (TH) but not phenylethanolamine N-methyltransferase (PNMT), indicating its expression in norepinephrine (NE)-, but not epinephrine (Epi)-synthesizing chromaffin cells. After exposure to IMO, there was no change in levels of VMAT1 mRNA. However, VMAT2 mRNA was elevated after exposure of rats to 2 h IMO once (1× IMO) or daily for 6 days (6× IMO). The changes in VMAT2 mRNA were reflected by increased VMAT2 protein after the repeated IMO. Immunofluorescence revealed an increased number of cells expressing VMAT2 following repeated IMO and its colocalization with PNMT in many chromaffin cells. The findings suggest an adaptive mechanism in chromaffin cells whereby enhanced catecholamine storage capacity facilitates more efficient utilization of the well-characterized heightened catecholamine biosynthesis with repeated IMO stress.


Assuntos
Medula Suprarrenal/citologia , Células Cromafins/metabolismo , Epinefrina/biossíntese , Estresse Fisiológico , Proteínas Vesiculares de Transporte de Monoamina/genética , Proteínas Vesiculares de Transporte de Monoamina/metabolismo , Animais , Células Cromafins/enzimologia , Regulação da Expressão Gênica , Masculino , Feniletanolamina N-Metiltransferase/metabolismo , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Restrição Física
16.
J Neuroimmunol ; 336: 577030, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31473519

RESUMO

The mechanisms underlying stress-related modulation of immune function via urocortin 1 and urocortin 2 have been only vaguely described. Therefore, we investigated the effect of LPS injection or immobilization stress on gene expression of urocortin 1 and urocortin 2 in the rat spleen, along with the potential involvement of glucocorticoids. Our data showed: a) different regulation of urocortin 1 and urocortin 2 gene expression in the rat spleen under different stressful conditions (LPS vs. immobilization stress) and b) diverse effects of stress-induced adrenal glucocorticoids on this process. Our findings indicate a specific, rather than general regulation of splenic immune function by urocortins during stressful conditions.


Assuntos
Hormônio Liberador da Corticotropina/biossíntese , Glucocorticoides/fisiologia , Baço/metabolismo , Estresse Psicológico/metabolismo , Urocortinas/biossíntese , Animais , Corticosterona/sangue , Hormônio Liberador da Corticotropina/genética , Expressão Gênica , Masculino , Ratos , Ratos Sprague-Dawley , Estresse Psicológico/genética , Estresse Psicológico/psicologia , Urocortinas/genética
17.
Neurochem Int ; 125: 47-56, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30738079

RESUMO

The mechanisms responsible for the anti-inflammatory effects of antidepressants are only partially understood. Published data indicate that the vagal anti-inflammatory pathway could be involved in mediating this effect. Therefore, we investigated the influence of subdiaphragmatic vagotomy on the anti-inflammatory effect of fluoxetine in rats injected with lipopolysaccharide (LPS) to induce an inflammatory response. The extent of this response was determined by measurement of TNF-α, IL-1ß, and IL-6 plasma levels, along with gene expression of TNF-α, IL-1ß, and IL-6 in the spleen and selected structures of the brain. To evaluate possible central mechanisms, c-fos mRNA levels were determined in the nucleus of the solitary tract, dorsal motor nucleus of the vagus, paraventricular hypothalamic nucleus, basolateral amygdala, central nucleus of the amygdala, hippocampus, and frontal cortex. We found that pretreatment with fluoxetine substantially prevented LPS-induced increases of pro-inflammatory cytokines in plasma and gene expression in the spleen and brain in animals with an intact vagus nerve. However, in vagotomized animals, fluoxetine pretreatment only partially attenuated the LPS-induced increase in these markers of peripheral inflammation. Our data has shown that fluoxetine exerts potent anti-inflammatory effects in both the periphery and brain. Moreover, we found that the peripheral anti-inflammatory action of fluoxetine is mediated, at least partially, by activation of a vagal anti-inflammatory pathway. The role of the vagus nerve in mediating the anti-inflammatory effects of antidepressants has been marginally explored and our findings highlight its potential contribution to this mechanism of action of antidepressants.


Assuntos
Anti-Inflamatórios/farmacologia , Antidepressivos/farmacologia , Fluoxetina/farmacologia , Mediadores da Inflamação/metabolismo , Nervo Vago/metabolismo , Animais , Vias Eferentes/efeitos dos fármacos , Vias Eferentes/metabolismo , Mediadores da Inflamação/antagonistas & inibidores , Masculino , Ratos , Ratos Sprague-Dawley , Vagotomia/tendências , Nervo Vago/efeitos dos fármacos , Nervo Vago/cirurgia
18.
J Neuroimmunol ; 337: 577068, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31606594

RESUMO

Recent data indicate that peripheral, as well as hypothalamic pro-inflammatory cytokines play an important role in the development of cancer cachexia. However, there are only a few studies simultaneously investigating the expression of inflammatory molecules in both the periphery and hypothalamic structures in animal models of cancer cachexia. Therefore, using the Yoshida ascites hepatoma rat's model of cancer cachexia we investigated the gene expression of inflammatory markers in the spleen along with the paraventricular and arcuate nuclei, two hypothalamic structures that are involved in regulating energy balance. In addition, we investigated the effect of intracerebroventricular administration of PS-1145 dihydrochloride (an Ikß inhibitor) on the expression of selected inflammatory molecules in these hypothalamic nuclei and spleen. We observed significantly reduced food intake in tumor-bearing rats. Moreover, we found significantly decreased expression of IL-6 in the spleen as well as decreased NF-κB in the paraventricular nucleus of rats with Yoshida ascites hepatoma. Similarly, expression of TNF-α, IL-1ß, NF-κB, and COX-2 in the arcuate nucleus was significantly reduced in tumor-bearing rats. Administration of PS-1145 dihydrochloride reduced only the gene expression of COX-2 in the hypothalamus. Based on our findings, we suggest that the growing Yoshida ascites hepatoma decreased food intake by mechanical compression of the gut and therefore this model is not suitable for investigation of the inflammation-related mechanisms of cancer cachexia development.


Assuntos
Ascite/metabolismo , Encéfalo/metabolismo , Caquexia/metabolismo , Carcinoma Hepatocelular/metabolismo , Mediadores da Inflamação/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Baço/metabolismo , Animais , Ascite/complicações , Ascite/imunologia , Encéfalo/imunologia , Caquexia/etiologia , Caquexia/imunologia , Carcinoma Hepatocelular/complicações , Carcinoma Hepatocelular/imunologia , Linhagem Celular Tumoral , Inflamação/etiologia , Inflamação/imunologia , Inflamação/metabolismo , Neoplasias Hepáticas Experimentais/complicações , Neoplasias Hepáticas Experimentais/imunologia , Masculino , Ratos , Ratos Wistar , Baço/imunologia
19.
J Neuroendocrinol ; : e12595, 2018 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-29604138

RESUMO

Corticotropin-releasing factor is well known activator of the hypothalamic-pituitary-adrenocortical axis, that represents crucial system participating on stress response of the organism. Urocortins are members of the corticotropin-releasing factor family of peptides with proposed effects on neuroendocrine and behavioral stress response mechanisms. Urocortin 2, one of three known urocortins, is present in central and peripheral stress response system and its expression can be augmented by glucocorticoids. In the present study we have examined how glucocorticoid withdrawal affects urocortin 2 gene expression after acute immobilization in the adrenal medulla and selected brain areas in rats. We used pharmacological adrenalectomy to block synthesis of corticosterone. Our results show that the immobilization-induced rise in urocortin 2 mRNA levels in rat adrenal medulla was not inhibited by glucocorticoid withdrawal. On the other hand, observed changes in the brain indicate that the effect of stress and pharmacological adrenalectomy on urocortin 2 gene expression is site-specific. While in the paraventricular nucleus and locus coeruleus the immobilization induced rise of urocortin 2 was not inhibited by pharmacological adrenalectomy in the arcuate nucleus and central amygdala it was. Moreover, we have seen a significant depletion of urocortin 2 plasma levels after immobilization. The immobilization induced rise of urocortin 2 gene expression in rat adrenal medulla and brain areas regulating stress response pathways and preservation of its induction after adrenalectomy suggests a role of urocortin 2 in the neuroendocrine stress response of an organism. This article is protected by copyright. All rights reserved.

20.
Neuroendocrinology ; 86(2): 69-76, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17556846

RESUMO

Subcutaneous injection of diluted formalin (0.2 ml of 4% solution/100 g BW) can influence the increase of plasma epinephrine levels in rats exposed to exteroceptive (handling, immobilization), as well as to interoceptive stressors (insulin-induced hypoglycemia), without having any effect on norepinephrine release. In the present studies, the effect of the above-mentioned stressors has been investigated on formalin-induced prolactin (PRL) and corticosterone secretion. Administrations of formalin via chronically implanted subcutaneous cannula into the hind limb without handling induce an immediate increase in both plasma PRL and corticosterone levels. While PRL concentration reaches its peak value within 5 min then returns to the basal level by the end of the 30th min, corticosterone level also starts to rise immediately after formalin administration reaching its highest concentration within 15-30 min, but it remains at this high level during the next 60 min, then it declines and returns to the pre-injection level. Application of formalin to animals exposed to different heterotypic stressors (like handling or insulin-induced hypoglycemia) produces an attenuated PRL response, while plasma corticosterone levels induced by the same nociceptive component remained unchanged. Combinations of formalin injection with immobilization also show an attenuated PRL response. The present data indicate that plasma PRL response to formalin is related to its acute nociceptive phase, and application of different stressors prior to formalin injection significantly attenuate plasma PRL levels, while it does not influence corticosterone responses.


Assuntos
Nociceptores/fisiologia , Dor/sangue , Prolactina/sangue , Estresse Fisiológico/sangue , Doença Aguda , Animais , Corticosterona/sangue , Formaldeído/farmacologia , Manobra Psicológica , Hipoglicemia/sangue , Hipoglicemia/induzido quimicamente , Hipoglicemiantes , Injeções Subcutâneas , Insulina , Masculino , Dor/fisiopatologia , Medição da Dor , Ratos , Ratos Sprague-Dawley , Restrição Física , Estresse Fisiológico/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa