Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Hormones (Athens) ; 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38740711

RESUMO

PURPOSE: Corticotropin-releasing hormone (CRH) plays an important role in relief of pain by releasing analgesia-associated molecules in several inflammatory states. During inflammation, peripheral CRH acts on cells of the immune system to stimulate the local expression of proopiomelanocortin (POMC) and the production of ß-endorphin, which in turn binds to opioid receptors on sensory neurons to produce antinociception. In the present study, we further investigated the role of endogenous CRH in inflammatory pain by determining the effects of Crh-deficiency on this process. METHODS: For this purpose, we used Crh-deficient (Crh-/-) mice and their wildtype (Crh + / +) littermates in the CFA (Complete Freund's Adjuvant)-induced inflammatory pain model. Pain thresholds were evaluated with the Hargreaves apparatus. RESULTS: Our experiments showed that Crh deficiency led to increased pain response, which was associated with decreased POMC mRNA levels in locally inflamed paws of these mice. Furthermore, Crh-/- mice had higher paw edema than Crh + / + mice. Histological evaluation of inflamed paw tissues revealed increased inflammatory response in Crh-/- mice. Protein levels of proinflammatory cytokines, such as IL-6, TNF-α, and IL-1ß, were higher in inflamed tissue of Crh-/- mice compared to wildtype mice. Corticosterone replacement increased the pain threshold of Crh-/- mice, restored their paw volume to the levels of wildtype mice, and significantly reduced their proinflammatory cytokine levels. Furthermore, glucocorticoid administration significantly increased POMC mRNA expression in the inflamed paw. CONCLUSION: Our data suggest that genetic deficiency of CRH is associated with increased pain. This effect is likely attributable to the accompanying glucocorticoid insufficiency and is in part mediated by opioids expressed locally.

2.
Biomedicines ; 9(9)2021 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-34572370

RESUMO

Dehydroepiandrosterone (DHEA), an adrenal and neurosteroid hormone with strong neuroprotective and immunomodulatory properties, and ligand for all high-affinity neurotrophin tyrosine kinase receptors (Trk), also exerts important effects on hyperalgesia. Its synthetic, 17-spiro-epoxy analogue, BNN27, cannot be converted to estrogen or androgen as DHEA; it is a specific agonist of TrkA, the receptor of pain regulator Nerve Growth Factor (NGF), and it conserves the immunomodulatory properties of DHEA. Our study aimed to evaluate the anti-nociceptive and anti-inflammatory properties of BNN27 during Complete Freund's Adjuvant (CFA)-induced inflammatory hyperalgesia in mice. Hyperalgesia was evaluated using the Hargreaves test. Inflammatory markers such as cytokines, NGF and opioids were measured, additionally to corticosterone and the protein kinase B (AKT) signaling pathway. We showed for the first time that treatment with BNN27 reversed hyperalgesia produced by CFA. The effect of BNN27 involved the inhibition of NGF in the dorsal root ganglia (DRG) and the increased synthesis of opioid peptides and their receptors in the inflamed paw. We also found alterations in the cytokine levels as well as in the phosphorylation of AKT2. Our findings strongly support that BNN27 represents a lead molecule for the development of analgesic and anti-inflammatory compounds with potential therapeutic applications in inflammatory hyperalgesia.

3.
Curr Mol Pharmacol ; 11(1): 72-80, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-27784217

RESUMO

BACKGROUND: Hypothalamic corticotrophin-releasing hormone (CRH) has a key role in coordinating and controlling complex responses to stress, both systemically, by stimulating the expression of the pituitary POMC gene, and thus, resulting in increased production of ACTH and adrenal glucocorticoid release, and locally since CRH has been identified in several peripheral tissues. CRH seems to exert its effects through interaction with two known so far receptors, CRF1R and CRF2R. The mRNA and protein of CRH family of peptides and their receptors are expressed at several peripheral tissues including rodent and human skin. In addition to CRH, skin expresses POMC and its products, including ACTH while recent studies have shown the presence of glucocorticoids also in skin. OBJECTIVE: This review aims to summarize the role of CRH in the physiology and pathophysiology of human and rodent skin. RESULTS: It is clear that a) locally produced CRH is involved in the inflammatory process, b) CRH has been shown to stimulate angiogenesis in vivo and chemotaxis of endothelial cells in vitro, and c) CRH mRNA and peptide have been identified in skin. CONCLUSION: Based on the above we hypothesize that CRH plays a crucial role in several inflammatory pathologies of the skin as well as in cutaneous wound healing, which are all discussed in the present review.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Pele/metabolismo , Animais , Humanos , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Pele/patologia , Dermatopatias/metabolismo , Dermatopatias/patologia
4.
PLoS One ; 6(7): e21654, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21765902

RESUMO

Corticotropin-releasing hormone, or factor, (CRH or CRF) exerts important biological effects in multiple peripheral tissues via paracrine/autocrine actions. The aim of our study was to assess the effects of endogenous CRH in the biology of mouse and human skin fibroblasts, the primary cell type involved in wound healing. We show expression of CRH and its receptors in primary fibroblasts, and we demonstrate the functionality of fibroblast CRH receptors by induction of cAMP. Fibroblasts genetically deficient in Crh (Crh-/-) had higher proliferation and migration rates and compromised production of IL-6 and TGF-ß1 compared to the wildtype (Crh+/+) cells. Human primary cultures of foreskin fibroblasts exposed to the CRF(1) antagonist antalarmin recapitulated the findings in the Crh-/- cells, exhibiting altered proliferative and migratory behavior and suppressed production of IL-6. In conclusion, our findings show an important role of fibroblast-expressed CRH in the proliferation, migration, and cytokine production of these cells, processes associated with the skin response to injury. Our data suggest that the immunomodulatory effects of CRH may include an important, albeit not explored yet, role in epidermal tissue remodeling and regeneration and maintenance of tissue homeostasis.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Derme/citologia , Fibroblastos/metabolismo , Animais , Anticorpos Neutralizantes/imunologia , Apoptose , Movimento Celular , Proliferação de Células , Hormônio Liberador da Corticotropina/deficiência , Fibroblastos/citologia , Humanos , Interleucina-6/biossíntese , Interleucina-6/imunologia , Camundongos , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Fator de Crescimento Transformador beta1/biossíntese , Fator de Crescimento Transformador beta1/imunologia
5.
Mol Cancer ; 9: 261, 2010 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-20875132

RESUMO

INTRODUCTION: Stress has been shown to be a tumor promoting factor. Both clinical and laboratory studies have shown that chronic stress is associated with tumor growth in several types of cancer. Corticotropin Releasing Factor (CRF) is the major hypothalamic mediator of stress, but is also expressed in peripheral tissues. Earlier studies have shown that peripheral CRF affects breast cancer cell proliferation and motility. The aim of the present study was to assess the significance of peripheral CRF on tumor growth as a mediator of the response to stress in vivo. METHODS: For this purpose we used the 4T1 breast cancer cell line in cell culture and in vivo. Cells were treated with CRF in culture and gene specific arrays were performed to identify genes directly affected by CRF and involved in breast cancer cell growth. To assess the impact of peripheral CRF as a stress mediator in tumor growth, Balb/c mice were orthotopically injected with 4T1 cells in the mammary fat pad to induce breast tumors. Mice were subjected to repetitive immobilization stress as a model of chronic stress. To inhibit the action of CRF, the CRF antagonist antalarmin was injected intraperitoneally. Breast tissue samples were histologically analyzed and assessed for neoangiogenesis. RESULTS: Array analysis revealed among other genes that CRF induced the expression of SMAD2 and ß-catenin, genes involved in breast cancer cell proliferation and cytoskeletal changes associated with metastasis. Cell transfection and luciferase assays confirmed the role of CRF in WNT- ß-catenin signaling. CRF induced 4T1 cell proliferation and augmented the TGF-ß action on proliferation confirming its impact on TGFß/SMAD2 signaling. In addition, CRF promoted actin reorganization and cell migration, suggesting a direct tumor-promoting action. Chronic stress augmented tumor growth in 4T1 breast tumor bearing mice and peripheral administration of the CRF antagonist antalarmin suppressed this effect. Moreover, antalarmin suppressed neoangiogenesis in 4T1 tumors in vivo. CONCLUSION: This is the first report demonstrating that peripheral CRF, at least in part, mediates the tumor-promoting effects of stress and implicates CRF in SMAD2 and ß-catenin expression.


Assuntos
Neoplasias da Mama/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Estresse Fisiológico/fisiologia , Animais , Western Blotting , Neoplasias da Mama/etiologia , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Hormônio Liberador da Corticotropina/antagonistas & inibidores , Hormônio Liberador da Corticotropina/genética , Feminino , Imunofluorescência , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos BALB C , Pirimidinas/farmacologia , Pirróis/farmacologia , Radioimunoensaio , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estresse Fisiológico/genética
6.
Endocrinology ; 151(7): 3214-24, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20463057

RESUMO

Scavenger receptor class B type I (SR-BI) facilitates the reverse transport of excess cholesterol from peripheral tissues to the liver via high-density lipoproteins. In steroidogenic tissues, SR-BI supplies cholesterol for steroid hormone production. We show here that the transcription of the human SR-BI gene is subject to feedback inhibition by glucocorticoid in adrenal and ovarian cells. SR-BI mRNA levels were increased in adrenals from corticosterone-insufficient Crh(-/-) mice, whereas corticosterone replacement by oral administration inhibited SR-BI gene expression in these mice. SR-BI mRNA levels were increased in adrenals from wild-type mice treated with metyrapone, a drug that blocks corticosterone synthesis. Experiments in adrenocortical H295R and ovarian SKOV-3 cells using cycloheximide and siRNA-mediated gene silencing revealed that glucocorticoid-mediated inhibition of SR-BI gene transcription requires de novo protein synthesis and the glucocorticoid receptor (GR). No direct binding of GR to the SR-BI promoter could be demonstrated in vitro and in vivo, suggesting an indirect mechanism of repression of SR-BI gene transcription by GR in adrenal cells. Deletion analysis established that the region of the human SR-BI promoter between nucleotides -201 and -62 is sufficient to mediate repression by glucocorticoid. This region contains putative binding sites for transcriptional repressors that could play a role in SR-BI gene regulation in response to glucocorticoid. In summary, this is the first report showing that glucocorticoid suppress SR-BI expression suggesting that steroidogenic tissues maintain steroid hormone homeostasis by prohibiting SR-BI-mediated high-density lipoprotein cholesterol uptake when the endogenous levels of glucocorticoid are elevated.


Assuntos
Glândulas Suprarrenais/citologia , Glucocorticoides/farmacologia , Ovário/citologia , Receptores Depuradores Classe B/metabolismo , Animais , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Eletroforese em Gel de Poliacrilamida , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas/genética , RNA Interferente Pequeno , Receptores de Glucocorticoides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores Depuradores Classe B/genética
7.
Mol Cancer ; 8: 30, 2009 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-19490624

RESUMO

INTRODUCTION: Cancer cells secrete bioactive peptides that act in an autocrine or paracrine fashion affecting tumor growth and metastasis. Corticotropin-releasing factor (CRF), a hypothalamic neuropeptide that controls the response to stress, has been detected in breast cancer tissues and cell lines. CRF can affect breast cancer cells in an autocrine or paracrine manner via its production from innervating sympathetic neurons or immune cells. METHODS: In the present study we report our findings regarding the impact of CRF on breast cancer cell motility and invasiveness. For this purpose we used the MCF7 breast cancer cell line and evaluated the effect of CRF on motility and invasiveness using the wound-healing and boyden-chamber assays. In addition, we measured the effect of CRF on molecules that mediate motility by western blot, immunofluorescence, ELISA and RT-PCR. RESULTS: Our findings show that: 1. CRF transiently inhibited the apoptosis of MCF7 cells. 2. CRF enhanced MCF7 cell motility in a wound healing assay and their invasiveness through extracellular matrix. 3. CRF increased actin polymerization, phosphorylation of Focal Adhesion Kinase (FAK), providing a potential mechanism for the observed induction of MCF7 motility. 4. CRF induced the expression of Cox-1 but not Cox-2 in MCF7 cells as well as the production of prostaglandins, factors known to promote invasiveness and metastasis. CONCLUSION: Overall, our data suggest that CRF stimulates cell motility and invasiveness of MCF7 cells most probably via induction of FAK phosphorylation and actin filament reorganization and production of prostaglandins via Cox1. Based on these findings we postulate that the stress neuropeptide CRF present in the vicinity of tumors (either produced locally by the tumor cells themselves or by nearby normal cells or secreted from the innervations of surrounding tissues) may play an important role on breast tumor growth and metastatic capacity, providing a potential link between stress and tumor progression.


Assuntos
Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Hormônio Liberador da Corticotropina/fisiologia , Actinas/metabolismo , Análise de Variância , Apoptose , Western Blotting , Linhagem Celular Tumoral , Movimento Celular , Hormônio Liberador da Corticotropina/farmacologia , Ciclo-Oxigenase 2/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Imunofluorescência , Quinase 1 de Adesão Focal/metabolismo , Humanos , Invasividade Neoplásica , Fosforilação , Prostaglandinas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Mol Pharmacol ; 75(4): 793-800, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19124613

RESUMO

Upon binding of the corticotropin-releasing factor (CRF) analog sauvagine to the type 1 CRF receptor (CRF(1)), the amino-terminal portion of the peptide has been shown to lie near Lys257 in the receptor's second extracellular loop (EL2). To test the hypothesis that EL2 residues play a role in the binding of sauvagine to CRF(1) we carried out an alanine-scanning mutagenesis study to determine the functional role of EL2 residues (Leu251 to Val266). Only the W259A, F260A, and W259A/F260A mutations reduced the binding affinity and potency of sauvagine. In contrast, these mutations did not seem to significantly alter the overall receptor conformation, in that they left unchanged the affinities of the ligands astressin and antalarmin that have been suggested to bind to different regions of CRF(1). The W259A, F260A, and W259A/F260A mutations also decreased the affinity of the endogenous ligand, CRF, implying that these residues may play a common important role in the binding of different peptides belonging to CRF family. Parallel amino acid deletions of the two peptides produced ligands with various affinities for wild-type CRF(1) compared with the W259A, F260A, and W259A/F260A mutants, supporting the interaction between the amino-terminal residues 8 to 10 of sauvagine and the corresponding region in CRF with EL2 of CRF(1). This is the first time that a specific region of CRF(1) has been implicated in detailed interactions between the receptor and the amino-terminal portion of peptides belonging to the CRF family.


Assuntos
Alanina/genética , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Sequência de Aminoácidos , Proteínas de Anfíbios/química , Proteínas de Anfíbios/genética , Proteínas de Anfíbios/metabolismo , Ligação Competitiva/genética , Linhagem Celular , Humanos , Dados de Sequência Molecular , Família Multigênica , Fragmentos de Peptídeos/química , Hormônios Peptídicos/química , Hormônios Peptídicos/genética , Hormônios Peptídicos/metabolismo , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética , Receptores de Hormônio Liberador da Corticotropina/química , Receptores de Hormônio Liberador da Corticotropina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA