Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cells ; 12(18)2023 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-37759478

RESUMO

Osteoarthritis (OA) is the most prevalent joint disease associated with chronic pain. OA pain is often accompanied by mood disorders. We addressed the role of the Prokineticin (PK) system in pain and mood alterations in a mice OA model induced with monosodium iodoacetate (MIA). The effect of a PK antagonist (PC1) was compared to that of diclofenac. C57BL/6J male mice injected with MIA in the knee joint were characterized by allodynia, motor deficits, and fatigue. Twenty-eight days after MIA, in the knee joint, we measured high mRNA of PK2 and its receptor PKR1, pro-inflammatory cytokines, and MMP13. At the same time, in the sciatic nerve and spinal cord, we found increased levels of PK2, PKR1, IL-1ß, and IL-6. These changes were in the presence of high GFAP and CD11b mRNA in the sciatic nerve and GFAP in the spinal cord. OA mice were also characterized by anxiety, depression, and neuroinflammation in the prefrontal cortex and hippocampus. In both stations, we found increased pro-inflammatory cytokines. In addition, PK upregulation and reactive astrogliosis in the hippocampus and microglia reactivity in the prefrontal cortex were detected. PC1 reduced joint inflammation and neuroinflammation in PNS and CNS and counteracted OA pain and emotional disturbances.

2.
J Clin Med ; 12(13)2023 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-37445367

RESUMO

Knee osteoarthritis (OA) is a chronic degenerative inflammatory-based condition caused by a cascade of different intra-articular molecules including several cytokines. Among the cytokines, prokineticins (PKs) have recently been identified as important mediators of inflammation and pain. This observational study examined the potential involvement of PK2 in degenerative or traumatic knee disease. Fifteen patients presenting knee osteoarthritis (OA group) and 15 patients presenting a traumatic meniscal tear (TM group) were studied. Synovial fluid samples from affected knees were assessed for PK2, IL-10, and TNF-α using the ELISA method. At a long-term follow-up (minimum 5 years, mean = 6.1 years), patients in the TM group underwent clinical re-evaluation with PROMs (Tegner Activity Scale, IKDC, Lysholm, SKV); in addition, X-ray visualization was used to assess the presence of secondary OA. PK2 was detected in synovial fluids of both TM and OA patients and the levels were comparable between the two groups, while IL-10 levels were significantly greater in the OA group than those in TM patients. PK2 levels correlated with those of IL-10. PK2 levels were greater in blood effusions compared to clear samples, did not differ significantly between sexes, nor were they related to differences in weight, height, or injury (meniscal laterality, time since dosing). No correlation was found between PROMs and radiological classifications in patients in the TM group at final follow-up. These data are the first observations of PK2 in synovial fluid following traumatic meniscus injury. These findings suggest possible further prognostic indices and therapeutic targets to limit the development of secondary OA.

3.
Int J Mol Sci ; 23(17)2022 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-36077245

RESUMO

MRAP2 is a small simple transmembrane protein arranged in a double antiparallel topology on the plasma membrane. It is expressed in the paraventricular nucleus of the hypothalamus, where it interacts with various G protein-coupled receptors, such as the prokineticin receptors, and regulates energy expenditure and appetite. The aim of this work was to analyze the functional role of the specific arginine residue at position 125 of MRAP2, which affects protein conformation, dimer formation, and PKR2 binding. Results obtained with the MRAP2 mutants R125H and R125C, which are found in human patients with extreme obesity, and mouse MRAP2, in which arginine 125 is normally replaced by histidine, were compared with those obtained with human MRAP2. Understanding the mechanism by which MRAP2 regulates G protein-coupled receptors helps in elucidating the metabolic pathways involved in metabolic dysfunction and in developing new drugs as specific targets of the MRAP2-PKR2 complex.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Arginina , Animais , Arginina/metabolismo , Humanos , Hipotálamo/metabolismo , Camundongos , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
4.
Biomolecules ; 12(3)2022 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-35327666

RESUMO

Melanocortin Receptor Accessory Protein 2 (MRAP2) modulates the trafficking and signal transduction of several G-protein-coupled receptors (GPCRs) involved in the control of energy homeostasis, such as Prokineticin receptors (PKRs). They bind the endogenous ligand prokineticin 2 (PK2), a novel adipokine that has an anorexic effect and modulates thermoregulation and energy homeostasis. In the present work, we used biochemical techniques to analyze the mechanism of interaction of MRAP2 with PKR2 and we identified the specific amino acid regions involved in the complex formation. Our results indicate that MRAP2 likely binds to the N-terminal region of PKR2, preventing glycosylation and consequently the correct receptor localization. We also identified a C-terminal region of MRAP2 that is critical for the interaction with PKR2. Consequently, we analyzed the role of the prokineticin transduction system in the regulation of MRAP2 expression in tissues involved in the control of food intake: at the central level, in hypothalamic explants, and at the peripheral level, in adipocytes. We demonstrated the modulation of MRAP2 expression by the prokineticin transduction system.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Melanocortinas , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Transporte/metabolismo , Melanocortinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Melanocortina/metabolismo , Transdução de Sinais
5.
Life (Basel) ; 12(2)2022 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-35207461

RESUMO

Prokineticins are a new class of chemokine-like peptides that bind their G protein-coupled receptors, PKR1 and PKR2, and promote chemotaxis and the production of pro-inflammatory cytokines following tissue injury or infection. This review summarizes the major cellular and biochemical mechanisms of prokineticins pathway regulation that, like other chemokines, include: genetic polymorphisms; mRNA splice modulation; expression regulation at transcriptional and post-transcriptional levels; prokineticins interactions with cell-surface glycosaminoglycans; PKRs degradation, localization, post-translational modifications and oligomerization; alternative signaling responses; binding to pharmacological inhibitors. Understanding these mechanisms, which together exert substantial biochemical control and greatly enhance the complexity of the prokineticin-receptor network, leads to novel opportunities for therapeutic intervention. In this way, besides targeting prokineticins or their receptors directly, it could be possible to indirectly influence their activity by modulating their expression and localization or blocking the downstream signaling pathways.

6.
Biomedicines ; 9(11)2021 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-34829877

RESUMO

Prokineticins are a new class of chemokine-like peptides involved in a wide range of biological and pathological activities. In particular, prokineticin 2 (PK2), prokineticin receptor 1 (PKR1) and prokineticin receptor 2 (PKR2) play a central role in modulating neuroinflammatory processes. PK2 and PKRs, which are physiologically expressed at very low levels, are strongly upregulated during inflammation and regulate neuronal-glial interaction. PKR2 is mainly overexpressed in neurons, whereas PKR1 and PK2 are mainly overexpressed in astrocytes. Once PK2 is released in inflamed tissue, it is involved in both innate and adaptive responses: it triggers macrophage recruitment, production of pro-inflammatory cytokines, and reduction of anti-inflammatory cytokines. Moreover, it modulates the function of T cells through the activation of PKR1 and directs them towards a pro-inflammatory Th1 phenotype. Since the prokineticin system appears to be upregulated following a series of pathological insults leading to neuroinflammation, we will focus here on the involvement of PK2 and PKRs in those pathologies that have a strong underlying inflammatory component, such as: inflammatory and neuropathic pain, Alzheimer's disease, Parkinson's disease, multiple sclerosis, stroke, obesity, diabetes, and gastrointestinal inflammation.

7.
Int J Mol Sci ; 22(21)2021 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-34769347

RESUMO

Chemotherapy-induced neuropathy (CIN) is a major adverse effect associated with many chemotherapeutics, including bortezomib (BTZ). Several mechanisms are involved in CIN, and recently a role has been proposed for prokineticins (PKs), a chemokine family that induces proinflammatory/pro-algogen mediator release and drives the epigenetic control of genes involved in cellular differentiation. The present study evaluated the relationships between epigenetic mechanisms and PKs in a mice model of BTZ-induced painful neuropathy. To this end, spinal cord alterations of histone demethylase KDM6A, nuclear receptors PPARα/PPARγ, PK2, and pro-inflammatory cytokines IL-6 and IL-1ß were assessed in neuropathic mice treated with the PK receptors (PKRs) antagonist PC1. BTZ treatment promoted a precocious upregulation of KDM6A, PPARs, and IL-6, and a delayed increase of PK2 and IL-1ß. PC1 counteracted allodynia and prevented the increase of PK2 and of IL-1ß in BTZ neuropathic mice. The blockade of PKRs signaling also opposed to KDM6A increase and induced an upregulation of PPAR gene transcription. These data showed the involvement of epigenetic modulatory enzymes in spinal tissue phenomena associated with BTZ painful neuropathy and underline a role of PKs in sustaining the increase of proinflammatory cytokines and in exerting an inhibitory control on the expression of PPARs through the regulation of KDM6A gene expression in the spinal cord.


Assuntos
Bortezomib/toxicidade , Hormônios Gastrointestinais/metabolismo , Histona Desmetilases/metabolismo , Hiperalgesia/patologia , Neuropeptídeos/metabolismo , Dor/patologia , Doenças do Sistema Nervoso Periférico/patologia , Medula Espinal/patologia , Animais , Antineoplásicos/toxicidade , Citocinas/metabolismo , Hormônios Gastrointestinais/genética , Histona Desmetilases/genética , Hiperalgesia/induzido quimicamente , Hiperalgesia/genética , Hiperalgesia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeos/genética , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Dor/induzido quimicamente , Dor/genética , Dor/metabolismo , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/metabolismo , Medula Espinal/metabolismo
8.
Cell Signal ; 83: 110000, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33811988

RESUMO

Prokineticin 1 (pk1) and prokineticin 2 (pk2) interact with two structurally related G-protein coupled receptors, prokineticin receptor 1 (PKR1) and prokineticin receptor 2 (PKR2). Cellular signalling studies show that the activated receptors can evoke Ca2+-mobilization, pertussis toxin-sensitive ERK phosphorylation, and intracellular cAMP accumulation, which suggests the partecipation of several G protein subtypes, such as Gq/11, Gi/o and Gs. However, direct interactions with these transduction proteins have not been studied yet. Here we measured by bioluminescence resonance energy transfer (BRET) the association of PKR1 and PKR2 with different heterotrimeric Gα proteins in response to pk1 and pk2 activation. Using host-cell lines carrying gene deletions of Gαq/11 or Gαs, and pertussis toxin treatment to abolish the receptor interactions with Gαi/o, we determined that both receptors could couple with comparable efficiency to Gq/11 and Gi/o, but far less efficiently to Gs or other pertussis toxin-insensitive G proteins. We also used BRET methodology to assess the association of prokineticin receptors with ß-arrestin isoforms. Fluorescent versions of the isoforms were transfected both in HEK293 cells and in double KO ß-arrestin 1/2 mouse fibroblasts, to study receptor interaction with the reconstituted individual ß-arrestins without background expression of the endogenous genes. Both receptors formed stable BRET-emitting complexes with ß-arrestin 2 but not with ß-arrestin 1, indicating strong selectivity for the former. In all the studied transducer interactions and in both receptors, pk2 was more potent than pk1 in promoting receptor binding to transduction proteins.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Sistemas do Segundo Mensageiro , beta-Arrestina 2/metabolismo , AMP Cíclico/genética , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/genética , beta-Arrestina 2/genética
9.
Front Physiol ; 11: 591850, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33281622

RESUMO

Vasculogenesis and angiogenesis are key processes of placental development, which occur throughout pregnancy. Placental vasculogenesis occurs during the first trimester of pregnancy culminating in the formation of hemangioblasts from intra-villous stem cells. Placental angiogenesis occurs subsequently, forming new blood vessels from existing ones. Angiogenesis also takes place at the fetomaternal interface, allowing essential spiral arteriole remodeling to establish the fetomaternal circulation. Vasculogenesis and angiogenesis in animal models and in humans have been studied in a wide variety of in vitro, physiological and pathological conditions, with a focus on the pro- and anti-angiogenic factors that control these processes. Recent studies revealed roles for new families of proteins, including direct participants such as the prokineticin family, and regulators of these processes such as the homeobox genes. This review summarizes recent advances in understanding the molecular mechanisms of actions of these families of proteins. Over the past decade, evidence suggests increased production of placental anti-angiogenic factors, as well as angiogenic factors are associated with fetal growth restriction (FGR) and preeclampsia (PE): the most threatening pathologies of human pregnancy with systemic vascular dysfunction. This review also reports novel clinical strategies targeting members of these family of proteins to treat PE and its consequent effects on the maternal vascular system.

10.
Front Immunol ; 11: 2119, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33072073

RESUMO

Neurotoxicity is a common side effect of chemotherapeutics that often leads to the development of chemotherapy-induced peripheral neuropathy (CIPN). The peptide Prokineticin 2 (PK2) has a key role in experimental models of CIPN and can be considered an insult-inducible endangering mediator. Since primary afferent sensory neurons are highly sensitive to anticancer drugs, giving rise to dysesthesias, the aim of our study was to evaluate the alterations induced by vincristine (VCR) and bortezomib (BTZ) exposure in sensory neuron cultures and the possible preventive effect of blocking PK2 signaling. Both VCR and BTZ induced a concentration-dependent reduction of total neurite length that was prevented by the PK receptor antagonist PC1. Antagonizing the PK system also reduced the upregulation of PK2, PK-R1, TLR4, IL-6, and IL-10 expression induced by chemotherapeutic drugs. In conclusion, inhibition of PK signaling with PC1 prevented the neurotoxic effects of chemotherapeutics, suggesting a promising strategy for neuroprotective therapies against the sensory neuron damage induced by exposure to these drugs.


Assuntos
Antineoplásicos/toxicidade , Bortezomib/toxicidade , Hormônios Gastrointestinais/antagonistas & inibidores , Proteínas do Tecido Nervoso/antagonistas & inibidores , Neuropeptídeos/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Síndromes Neurotóxicas/prevenção & controle , Células Receptoras Sensoriais/efeitos dos fármacos , Triazinas/farmacologia , Vincristina/toxicidade , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação para Baixo , Avaliação Pré-Clínica de Medicamentos , Hormônios Gastrointestinais/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/fisiologia , Neuritos/efeitos dos fármacos , Neuritos/ultraestrutura , Neuroimunomodulação/efeitos dos fármacos , Neuropeptídeos/fisiologia , Fármacos Neuroprotetores/uso terapêutico , RNA Mensageiro/biossíntese , Células Receptoras Sensoriais/fisiologia , Células Receptoras Sensoriais/ultraestrutura , Triazinas/uso terapêutico
11.
Cells ; 8(11)2019 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-31766244

RESUMO

Among mediators of inflammation, chemokines play a pivotal role in the neuroinflammatory process related to Alzheimer's disease (AD). The chemokine Bv8/prokineticin 2 (PROK2) is a critical player in inflammatory and neuroinflammatory diseases and has been demonstrated to be involved in Aß toxicity. The aim of the present study was to extend the research to rats chronically intracerebroventricularly (i.c.v.) injected with Aß, to an AD transgenic mouse model, and subsequently to AD patients, mainly with the aim of detecting a potential biomarker. Real-time PCR and immunofluorescence analysis were used to evaluate Prokineticin-2 (PROK2) mRNA and the corresponding protein levels in both animal and human AD brain extracts, and the ELISA test was used to measure the amount of PROK2 in the serum of AD patients. We demonstrated a significant upregulation of PROK2 levels in brain tissues of Aß1-42 i.c.v. injected rats, transgenic AD mice (Tg2576), and in the hippocampus of AD patients. Additionally, through a pilot study, an approximate twofold increase of PROK2 levels has been proved in the serum of AD patients, compared to the control subjects, identifying a potential blood-based biomarker of the disease.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Suscetibilidade a Doenças , Hormônios Gastrointestinais/genética , Hormônios Gastrointestinais/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Biomarcadores , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Hormônios Gastrointestinais/sangue , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Neuropeptídeos/sangue , Agregados Proteicos , Agregação Patológica de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos
12.
Brain Behav Immun ; 82: 422-431, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31525509

RESUMO

Vincristine (VCR) treatment is often associated to painful neuropathy. Its development is independent from antitumoral mechanism and involves neuroinflammation. We investigated the role of the chemokine prokineticin (PK)2 in a mouse model of VCR induced neuropathy using a PK-receptors (PK-R) antagonist to counteract its development. We also evaluated emotional like deficits in VCR mice. VCR (0,1 mg/kg) was i.p. injected in C57BL/6J male mice once a day for 14 consecutive days. Pain, anxiety and depressive like behaviors were assessed in animals. PK2, PK-Rs, cytokines, neuroinflammatory markers (CD68, CD11b, GFAP, TLR4) and ATF3 were evaluated in DRG, spinal cord, prefrontal cortex and hippocampus. The PK-Rs antagonist PC1, was s.c. injected (150 µg/kg) twice a day from day 7 (hypersensitivity state) until day 14. Its effect on pain and neuroinflammation was evaluated. VCR mice developed neuropathic pain but not mood alterations. After 7 days of VCR treatment we observed a neuroinflammatory condition in DRG with high levels of PK-Rs, TLR4, CD68, ATF3 and IL-1ß without relevant alterations in spinal cord. At day 14, an upregulation of PK system and a marked neuroinflammation was evident also in spinal cord. Moreover, at the same time, we observed initial alterations in supraspinal brain areas. PC1 treatment significantly counteracted neuropathic pain and blunted neuroinflammation.


Assuntos
Hormônios Gastrointestinais/metabolismo , Neuralgia/induzido quimicamente , Neuralgia/metabolismo , Neuropeptídeos/metabolismo , Vincristina/toxicidade , Animais , Ansiedade/induzido quimicamente , Ansiedade/metabolismo , Comportamento Animal/efeitos dos fármacos , Citocinas/metabolismo , Depressão/induzido quimicamente , Depressão/metabolismo , Modelos Animais de Doenças , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroimunomodulação/efeitos dos fármacos , Distribuição Aleatória , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
13.
Front Pharmacol ; 10: 622, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31231219

RESUMO

Alzheimer's disease is the most common neurodegenerative disorder characterized by the presence of ß-amyloid aggregates deposited as senile plaques and by the presence of neurofibrillary tangles of tau protein. To date, there is a broad consensus on the idea that neuroinflammation is one of the most important component in Alzheimer's disease pathogenesis. Chemokines and their receptors, beside the well-known role in the immune system, are widely expressed in the nervous system, where they play a significant role in the neuroinflammatory processes. Prokineticins are a new family of chemokine-like molecules involved in numerous physiological and pathological processes including immunity, pain, inflammation, and neuroinflammation. Prokineticin 2 (PROK2) and its receptors PKR1 and PKR2 are widely expressed in the central nervous system in both neuronal and glial cells. In Alzheimer's disease, PROK2 sustains the neuroinflammatory condition and contributes to neurotoxicity, since its expression is strongly upregulated by amyloid-ß peptide and reversed by the PKR antagonist PC1. This review aims to summarize the current knowledge on the neurotoxic and/or neuroprotective function of chemokines in Alzheimer's disease, focusing on the prokineticin system: it represents a new field of investigation that can stimulate the research of innovative pharmacotherapeutic strategies.

14.
J Neuroinflammation ; 16(1): 89, 2019 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-30995914

RESUMO

BACKGROUND: Neuropathy is a dose-limiting side effect of many chemotherapeutics, including bortezomib. The mechanisms underlying this condition are not fully elucidated even if a contribution of neuroinflammation was suggested. Here, we investigated the role of a chemokine family, the prokineticins (PKs), in the development of bortezomib-induced peripheral neuropathy (BIPN), and we used a PK receptor antagonist to counteract the development and progression of the pathology. METHODS: Neuropathy was induced in male C57BL/6J mice by using a protocol capable to induce a detectable neuropathic phenotype limiting systemic side effects. The presence of allodynia (both mechanical and thermal) and thermal hyperalgesia was monitored over time. Mice were sacrificed at two different time points: 14 and 28 days after the first bortezomib (BTZ) injection. At these times, PK system activation (PK2 and PK-Rs), macrophage and glial activation markers, and cytokine production were evaluated in the main station involved in pain transmission (sciatic nerve, DRG, and spinal cord), and the effect of a PK receptors antagonist (PC1) on the same behavioral and biochemical parameters was assessed. Structural damage of DRG during BTZ treatment and an eventual protective effect of PC1 were also evaluated. RESULTS: BTZ induces in mice a dose-related allodynia and hyperalgesia and a progressive structural damage to the DRG. We observed a precocious increase of macrophage activation markers and unbalance of pro- and anti-inflammatory cytokines in sciatic nerve and DRG together with an upregulation of GFAP in the spinal cord. At higher BTZ cumulative dose PK2 and PK receptors are upregulated in the PNS and in the spinal cord. The therapeutic treatment with the PK-R antagonist PC1 counteracts the development of allodynia and hyperalgesia, ameliorates the structural damage in the PNS, decreases the levels of activated macrophage markers, and prevents full neuroimmune activation in the spinal cord. CONCLUSIONS: PK system may be a strategical pharmacological target to counteract BTZ-induced peripheral neuropathy. Blocking PK2 activity reduces progressive BTZ toxicity in the DRG, reducing neuroinflammation and structural damage to DRG, and it may prevent spinal cord sensitization.


Assuntos
Antineoplásicos/toxicidade , Bortezomib/toxicidade , Hormônios Gastrointestinais/metabolismo , Neuropeptídeos/metabolismo , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/metabolismo , Animais , Modelos Animais de Doenças , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo
15.
Prog Mol Biol Transl Sci ; 161: 149-179, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30711026

RESUMO

Prokineticins are two conserved small proteins (~8kDa), prokineticin 1 (PROK1; also called EG-VEGF) and prokineticin 2 (PROK2; also called Bv8), with an N-terminal AVITGA sequence and 10 cysteines forming 5 disulfide bridges. PROK1 and PROK2 bind to two highly related G protein-coupled receptors (GPCRs), prokineticin receptor 1 (PROKR1) and prokineticin receptor 2 (PROKR2). Prokineticins and their receptors are widely expressed. PROK1 is predominantly expressed in peripheral tissues, especially steroidogenic organs, whereas PROK2 is mainly expressed in the central nervous system and nonsteroidogenic cells of the testes. Prokineticins signaling has been implicated in several important physiological functions, including gastrointestinal smooth muscle contraction, circadian rhythm regulation, neurogenesis, angiogenesis, pain perception, mood regulation, and reproduction. Dysregulation of prokineticins signaling has been observed in a variety of diseases, such as cancer, ischemia, and neurodegeneration, in which prokineticins signaling seems to be a promising therapeutic target. Based on the phenotypes of knockout mice, PROKR2 and PROK2 have recently been identified as causative genes for idiopathic hypogonadotropic hypogonadism, a developmental disorder characterized by impaired development of gonadotropin-releasing hormone neurons and infertility. In vitro functional studies with these disease-associated PROKR2 mutations uncovered some novel features for this receptor, such as biased signaling, which may be used to understand GPCR signaling regulation in general.


Assuntos
Doença , Saúde , Receptores Acoplados a Proteínas G/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Sequência de Aminoácidos , Animais , Humanos , Mutação/genética , Nociceptividade , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/química , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética
16.
Neurol Sci ; 38(Suppl 1): 27-30, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28527062

RESUMO

Prokineticins (PK) 1 and 2 belong to a new family of chemokines capable to interact with two different G coupled receptors: Prokineticin receptor (PKR)1 and 2. Both prokineticins and their receptors are widely distributed in different tissues and regulate several biological functions. In particular, a role of the PK system in inflammation and nociception has been established. PKRs are expressed in regions of the nervous system associated with pain and in primary sensitive neurons they colocalize with transient potential receptor vanilloid-TRPV1 providing an anatomical interaction in nociceptor sensitization. Moreover, PKs are strongly upregulated in immune and glial cells and sustain a proinflammatory loop in inflamed tissues. Recent evidences indicate that the block of the PK system represents a promising strategy to contrast inflammation and pain.


Assuntos
Inflamação/metabolismo , Dor/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Células Receptoras Sensoriais/metabolismo , Animais , Humanos , Inflamação/diagnóstico , Inflamação/epidemiologia , Dor/diagnóstico , Dor/epidemiologia , Células Receptoras Sensoriais/patologia
17.
Neuropharmacology ; 108: 39-48, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27140692

RESUMO

Bv8/prokineticin 2 (PK2) is a member of a bioactive family of peptides that regulate multiple functions in the CNS including hyperalgesia, neurogenesis, neuronal survival and inflammation. Recent studies have associated PK2 and prokineticin receptors (PKR) with human diseases, but because their role in neuropathology is still debated we examined whether prokineticins exert a protective or deleterious role in models of cerebral ischemia and ischemic tolerance in vitro. In order to mimic cerebral ischemia, we exposed primary murine cortical cell cultures or rat organotypic hippocampal slices to appropriate periods of oxygen-glucose deprivation (OGD), which leads to neuronal damage 24 h later. Ischemic tolerance was induced by exposing hippocampal slices to a preconditioning subtoxic pharmacological stimulus (3 µM NMDA for 1 h) 24 h before the exposure to OGD. Bv8 (10-100 nM) attenuated OGD injury in cortical cultures and hippocampal slices, and the effect was prevented by the PKR antagonist PC7. The development of OGD tolerance was associated with an increase in the expression of PK2, PKR1 and PKR2 mRNA and proteins and was prevented by addition of the antagonist PC7 into the medium during preconditioning. Both Bv8 at protective concentrations and the NMDA preconditioning stimulus promoted the phosphorylation of ERK1/2 and Akt. These findings indicate that the prokineticin system can be up-regulated by a defensive preconditioning subtoxic NMDA stimulus and that PK2 may act as an endogenous neuroprotective factor through the activation of the ERK1/2 and Akt transduction pathways.


Assuntos
Isquemia Encefálica/metabolismo , Córtex Cerebral/metabolismo , Hormônios Gastrointestinais/biossíntese , Hipocampo/metabolismo , Neuropeptídeos/biossíntese , Fármacos Neuroprotetores/metabolismo , Animais , Anuros , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Isquemia Encefálica/prevenção & controle , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Relação Dose-Resposta a Droga , Hormônios Gastrointestinais/farmacologia , Hormônios Gastrointestinais/uso terapêutico , Hipocampo/efeitos dos fármacos , Camundongos , Neuropeptídeos/farmacologia , Neuropeptídeos/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Técnicas de Cultura de Órgãos , Ratos
18.
Am J Physiol Endocrinol Metab ; 307(6): E515-26, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25074983

RESUMO

Early studies have reported a phase-shifting effect of growth hormone secretagogues (GHSs). This study aimed to determine the mechanism of action of GHSs. We examined the response of the hypothalamic suprachiasmatic nuclei (SCN) to growth hormone releasing peptide-6 (GHRP-6) by assessing effects on the phase of locomotor activity rhythms, SCN neuronal discharges, and the potential signaling pathways involved in the drug action on circadian rhythms. The results showed that bolus administration of GHRP-6 (100 µg/kg ip) at the beginning of subjective night (CT12) induced a phase delay of the free-running rhythms in male C57BL/6J mice under constant darkness, but did not elicit phase shift at other checked circadian time (CT) points. The phase-delay effect of GHRP-6 was abolished by d-(+)-Lys-GHRP-6 (GHS receptor antagonist), KN-93 [calcium/calmodulin-dependent protein kinase II (CaMK) II inhibitor], or anti-phosphorylated (p)-cAMP response element-binding protein (CREB) antibody. Further analyses demonstrated that GHRP-6 at CT12 induced higher calcium mobilization and neuronal discharge in the SCN compared with that at CT6, decreased the levels of glutamate and γ-aminobutyric acid, increased the levels of p-CaMKII, p-CREB, and period 1, and delayed the circadian expressions of circadian locomotor output cycles kaput, Bmal1, and prokineticin 2 in the SCN; these signaling changes resulted in behavioral phase delay. Collectively, GHRP-6 induces a CT-dependent phase delay via activating GHS receptor and the downstream signaling, which is partially similar to the signaling cascade of light-induced phase delay at early night. These novel observations may help to better understand the role of GHSs in circadian physiology.


Assuntos
Ritmo Circadiano/efeitos dos fármacos , Receptores de Grelina/agonistas , Animais , Sinalização do Cálcio/efeitos dos fármacos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cromatografia Líquida de Alta Pressão , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Interpretação Estatística de Dados , Citometria de Fluxo , Ácido Glutâmico/metabolismo , Imuno-Histoquímica , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Proteínas Circadianas Period/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Núcleo Supraquiasmático/efeitos dos fármacos , Núcleo Supraquiasmático/fisiologia , Ácido gama-Aminobutírico/metabolismo
19.
Curr Neuropharmacol ; 4(3): 207-15, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18615143

RESUMO

Bv8 is a small protein secreted by frog skin. Mammalian homologues of Bv8, the prokineticins PK1 and PK2, and their G-protein coupled receptors PKR1 and PKR2 have been identified and linked to several biological effects. Bv8 elicits a dose-dependent reduction in nociceptive threshold to thermal and mechanical stimuli applied to the skin of tail and paw of rats and mice and increases the sensitivity to nociceptive mediators as capsaicin and prostaglandins. The receptors for Bv8/PKs are present in a fraction of peptidergic population of C-fibre neurons, and in a fraction of A myelinated-fibre neurons. In mouse and rat dorsal root ganglia, PKR-expressing neurons also express TRPV1 and the activation of PKRs sensitises TPRV1 to the action of capsaicin. Mice lacking PKR1 gene exhibit impaired Bv8-induced hyperalgesia, develop deficient responses to noxious heat, capsaicin and protons and show reduced thermal and mechanical hypersensitivity to paw inflammation, indicating a requirement for PKR1 signalling associated with activation and sensitisation of primary afferent fibres. PKs are highly expressed by neutrophils and other inflammatory cells and must be considered as new pronociceptive mediators in inflammatory tissues. Bv8-like hyperalgesic activity was demonstrated in extracts of rat inflammatory granulocytes. Bv8 stimulate macrophage and T lymphocyte to differentiate between an inflammatory and Th1 profile indicating that Bv8/PK proteins play a role in immuno-inflammatory responses. Blockade of PKRs may represent a novel therapeutic strategy in acute and inflammatory pain conditions.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA