Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Med Life ; 16(9): 1428-1432, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38107718

RESUMO

This study examined the reno-protective potential of Compound 21 during renal ischemia-reperfusion injury by regulating the PI3K expression. 20 adult male Swiss-albino mice, aged 8-12 weeks and weighing 20-30g, were randomly assigned to four equal groups: sham, control, vehicle, and Compound 21. Serum urea, creatinine, inflammatory mediators, tissue 8-isoprostane, and myeloperoxidase were quantified using ELISA. Compared to the sham group, blood levels of urea, creatinine, TNF-α, IL-6, and IL-10 were significantly higher in the ischemia-reperfusion group than in the sham group (p<0.05). However, these indicators were significantly lower in the Compound 21 group (p<0.05). Histological analysis revealed significant renal tissue damage in the ischemia-reperfusion group (p<0.05), which was significantly reduced in the Compound 21 group (p<0.05). PCR results showed that PI3K expression was significantly lower (p<0.05) in the control group compared to the sham group but significantly higher in the Compound 21 group (p<0.05). Furthermore, P-AKT expression levels in the control group were considerably lower than in the sham group (p<0.05). On the other hand, the level of P-AKT expression in the Compound 21 group was significantly upregulated compared to the control group (p<0.05). The findings revealed that Compound 21 could mitigate renal dysfunction induced by ischemia-reperfusion injury in male mice through modulation of the PI3K/AKT signaling pathway, resulting in decreased levels of pro-inflammatory cytokines and renal oxidative stress markers.


Assuntos
Receptor Tipo 2 de Angiotensina , Traumatismo por Reperfusão , Animais , Masculino , Camundongos , Creatinina , Isquemia , Rim , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Tipo 2 de Angiotensina/agonistas , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/prevenção & controle , Traumatismo por Reperfusão/patologia , Ureia/sangue
2.
J Med Life ; 16(8): 1188-1193, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38024827

RESUMO

Trastuzumab is a successful treatment option for HER2-positive breast cancer, but a decline in left ventricular ejection fraction (LVEF) and an increase in inflammatory and cardiac enzyme biomarkers can lead to cessation and termination of therapy. This study aimed to investigate the ability of Coenzyme Q10 (Coq10) to avoid these adverse effects. The study included 100 female patients with HER2+ (HER2+3 or amplified gene) breast cancer. All patients underwent standard adjuvant chemotherapy regimens, which involved a four-cycle treatment of Adriamycin, Cyclophosphamide, Docetaxel, and an initial 8 mg/kg loading dose of trastuzumab, followed by a year of 6 mg/kg maintenance doses every three weeks. One group of 50 patients received trastuzumab and a placebo, while the other 50 were given trastuzumab and CoQ10 for a full year. The CoQ10-treated group exhibited a statistically significant decrease in levels of monocyte chemoattractant protein-1 (MCP-1), interleukin-6 (IL6), soluble toll-like receptor 4 (sTLR4), and cardiac troponin I (cTnI) compared to the control group (p<0.05). However, there was no significant difference in the mean F2-isoprostane levels between the treated and the control groups at any data collection point. Furthermore, the CoQ10-treated group experienced a significant reduction in the decline of EF levels compared to the control group at all stages except for baseline. According to our findings, Coenzyme Q10 protected patients with HER2+3 breast cancer from the cardiotoxicity of trastuzumab by increasing ejection fraction and decreasing inflammatory biomarkers and cardiac enzyme levels.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Trastuzumab/efeitos adversos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Cardiotoxicidade/etiologia , Cardiotoxicidade/prevenção & controle , Cardiotoxicidade/tratamento farmacológico , Volume Sistólico , Receptor ErbB-2 , Anticorpos Monoclonais Humanizados/efeitos adversos , Função Ventricular Esquerda , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos
3.
J Med Life ; 16(6): 941-947, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37675176

RESUMO

Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. This study aimed to investigate the potential protective effect of the lungs in sepsis by modulating inflammatory and oxidative stress markers. Twenty-four adult male Swiss-albino mice, aged 8-12 weeks and weighing 20-30 g, were divided into four equal groups (n=6): sham (laparotomy only), CLP (laparotomy plus cecal ligation and puncture), vehicle (DMSO administered one hour before CLP), and Ticagrelor (50 mg/kg IP administered one hour before CLP). Tissue levels of pro-inflammatory and oxidative stress markers in the lung were assessed using ELISA. F2 isoprostane levels were significantly higher in the sepsis group (p<0.05) compared to the sham group, while Ticagrelor significantly decreased the inflammatory and oxidative stress markers compared to the sepsis group. All mice in the sepsis group had considerable (p=0.05) lung tissue damage, but Ticagrelor considerably decreased lung tissue injury (p=0.05). Furthermore, Ticagrelor was found to reduce tissue cytokine levels of the lung (IL-1, TNF a, IL-6, F2 isoprostane, GPR 17, MIF) in male mice during CLP-induced polymicrobial sepsis by modulation of pro-inflammatory and oxidative stress cascade signaling pathways.


Assuntos
Endotoxemia , Sepse , Masculino , Animais , Camundongos , Endotoxemia/tratamento farmacológico , F2-Isoprostanos , Ticagrelor/farmacologia , Ticagrelor/uso terapêutico , Pulmão
4.
J Med Life ; 16(6): 915-919, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37675177

RESUMO

Ischemia-reperfusion injury (IRI) is a major contributor to acute and chronic kidney failure, heart failure, and ischemic stroke. This study aimed to investigate the therapeutic potential of Iberin, known for its anti-inflammatory, antioxidant, and antiapoptotic properties, in a rat model of renal IRI. Twenty-four adult male rats were randomly divided into four groups: Group I (Sham group) underwent laparotomy without IRI induction; Group II (Control group) underwent laparotomy followed by renal artery clamping for 30 minutes to induce ischemia, followed by 2 hours of reperfusion; Group III (Iberin treatment group) received a pre-injection of Iberin (15 mg/kg) and underwent 30 minutes of ischemia followed by 2 hours of reperfusion; and Group IV (Vehicle-treated group) received the vehicle (ethanol) 1 hour prior to ischemia and reperfusion induction. Iberin was diluted with ethanol. Biomarkers associated with inflammation, oxidative stress, and apoptosis were measured using enzyme-linked immunosorbent assay. Iberin treatment significantly reduced levels of inflammatory cytokines interleukin-1ß (IL-1ß) and IL-6, Bcl-2-associated X protein (BAX), tumor necrosis factor α (TNF-α), nuclear factor kappa p56, high mobility group B1, and neutrophil gelatinase-associated lipocalin. Moreover, Iberin increased levels of heat shock protein and Bcl2 compared to the control and vehicle groups. Iberin treatment prolonged the ischemic tolerance of renal tissue, potentially preventing or delaying irreversible injuries. These findings highlight the potential of Iberin as a promising candidate for mitigating renal injury caused by ischemia-reperfusion, due to its ability to modulate inflammatory markers.


Assuntos
Falência Renal Crônica , Traumatismo por Reperfusão , Masculino , Animais , Ratos , Traumatismo por Reperfusão/tratamento farmacológico , Rim , Isotiocianatos
5.
J Med Life ; 16(5): 759-765, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37520489

RESUMO

Evolocumab, a PCSK-9 inhibitor, is known for its ability to reduce low-density lipoprotein cholesterol (LDL-C). This study aimed to investigate the effects of evolocumab, alone or in combination with atorvastatin, on the progression of atherosclerosis. Fifty male domestic rabbits were randomly assigned to five groups: control, high cholesterol diet, evolocumab vehicle (dimethyl sulfoxide, DMSO), evolocumab alone, and evolocumab plus atorvastatin. Serum levels of interleukin 10 (IL-10), IL-17, IL-1ß, intracellular adhesion molecule (ICAM), and vascular adhesion molecule (VCAM) were measured. Toll-like receptor (TLR) expression on monocytes was evaluated using flow cytometry. Histopathological examination and measurement of intimal thickness (IT) were also conducted. The results revealed that the evolocumab produced a statistically significant (p<0.05) reduction in lipid profile at 5 weeks, with the peak effect occurring at 10 weeks. Furthermore, the inhibitor reduced TLRs at 10 weeks to 10.83±1.8 and intimal thickness to 160.66±9.45. IL-17, IL-1ß, ICAM, and VCAM were significantly reduced by evolocumab treatment, with the improvement of the histopathological changes in the aortic wall. The combination of evolocumab and atorvastatin caused a more statistically significant reduction in TLRs at 10 weeks to 5.08±1.2 and intimal thickness to 121.79±5.3. IL-17, IL-1ß, ICAM, and VCAM were significantly (p<0.05) reduced by the combination, and the histopathological changes in the aortic wall were significantly improved. In conclusion, evolocumab delays the progression of atherosclerosis by modulating inflammatory pathways.


Assuntos
Aterosclerose , Interleucina-17 , Animais , Masculino , Coelhos , Atorvastatina/farmacologia , Atorvastatina/uso terapêutico , Aterosclerose/tratamento farmacológico , LDL-Colesterol , Resultado do Tratamento
6.
J Med Life ; 16(2): 317-324, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36937464

RESUMO

Ischemia and reperfusion injury (I/R) is a serious condition leading to organ failure, characterized by poor blood supply followed by rapid resuscitation of blood flow and reoxygenation. Renal failure caused by renal ischemia has high mortality and morbidity. This study aimed to explore the potential role of Semaglutide as a novel and effective therapeutic strategy for acute renal failure. Additionally, we aimed to assess the possible protective effect of Semaglutide on kidney I/R injury in mice through modulation of the inflammatory and oxidative pathways via phosphatidylinositol 3-kinase/adenosine triphosphate (PI3K/AKT) activation. We employed twenty-eight albino mice to induce the I/R injury model by clamping the renal artery for 30 min followed by a period of reperfusion for 2 hours. The control group was exposed to I/R injury, while the Semaglutide-treated group was pretreated with the drug 12 hours before induction of ischemia at a dose of 100 nmol/L/kg via the intraperitoneal route (i.p). In addition, the DMSO-treated group was subjected to similar conditions to the Semaglutide-treated group. At the end of the experiments, kidneys and blood samples were collected for investigation. Semaglutide could act as a protective agent against acute kidney injury by reducing inflammatory molecules such as tumor necrosis factor-alpha (TNF-α) and its cognate receptor, TNF-α R, interleukine-6 (IL-6). Furthermore, Semaglutide reduced F8 isoprostane levels, increased PI3K and AKT levels in renal tissues, and mitigated renal damage. Semaglutide had renoprotective effects via modulation of the inflammatory response and oxidative pathway by targeting the PI3K/AKT signaling pathway.


Assuntos
Injúria Renal Aguda , Traumatismo por Reperfusão , Animais , Camundongos , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Apoptose , Rim , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia
7.
Wiad Lek ; 76(1): 122-130, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36883500

RESUMO

OBJECTIVE: The aim: This study was set out to assess the potential protective impact of MK0752 (a gamma secretase inhibitor) on sepsis-induced renal injury through modulation of inflammatory and oxidative stress pathways. PATIENTS AND METHODS: Materials and methods: Twenty-four Swiss-albino mice aged between eight and twelve week and weighted twenty to thirty-seven grams were randomly allocated into four groups (n=6 in each group). Sham group (laparotomy without cecal ligation and puncture (CLP), sepsis group (laparotomy with CLP), vehicle-treated group (equivalent volume of DMSO before the CLP), MK0752 treated group (5 mg/kg) single daily dose for three days before the CLP. Blood samples were used to assess the serum levels of urea and creatinine. The kidneys were used to assess tissue levels of the TNF-α, IL-10, IL-6, TNFR1, VEGF, notch1, jagged1 and tissue damage by histopathological analysis. RESULTS: Results: The current study shows that pretreatment with MK0752 ameliorates the renal damage by significantly reducing the proinflammatory cytokines and notch1 signaling. CONCLUSION: Conclusions: Taken together, these results suggest that MK0752 could be protective against the renal injury induced by sepsis through its ameliorative impact on renal architecture and modulating cytokines and Notch1 singling pathway. Further studies regarding the role of Notch signaling pathways would be worthwhile.


Assuntos
Secretases da Proteína Precursora do Amiloide , Sepse , Camundongos , Animais , Modelos Animais de Doenças , Sepse/complicações , Sepse/tratamento farmacológico , Citocinas , Rim
8.
J Med Life ; 16(12): 1852-1856, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38585526

RESUMO

Renal ischemia-reperfusion injury (IRI) is a critical health concern that aggravates the pathophysiology of acute kidney injury (AKI), leading to high mortality rates in intensive care units. Cardamonin is a natural compound with anti-inflammatory and antioxidant properties. The current study aimed to evaluate the renoprotective impact of cardamonin against AKI induced by renal IRI. Male rats (n=5 per group) were divided into four groups: the sham group underwent anesthesia and abdominal incision only; the control group experienced bilateral renal artery clamping for 30 minutes followed by 2 hours of reperfusion; the vehicle group received the cardamonin vehicle 30 minutes before ischemia induction; and the cardamonin group was administered 5 mg/kg of cardamonin 30 minutes before ischemia. Blood urea nitrogen (BUN) and creatinine were measured to assess the renal function. Tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1ß), interleukin-6 (IL-6), caspase 3, and F2-isoprostane were assessed in renal tissues. Kidney injury was examined using the hematoxylin and eosin stain method. Compared to the sham group, the control group exhibited significantly higher levels of BUN, creatinine, TNF-α, IL-1ß, IL-6, F2-isoprostane, and caspase 3 in renal tissues, along with severe kidney injury as evidenced by histological analysis. Compared to the control group, pretreatment with cardamonin resulted in a significant reduction in these biomarkers and alleviated renal damage. Cardamonin had renoprotective effects against renal ischemia and reperfusion injury via modulating inflammation, oxidative stress, and apoptosis pathways.


Assuntos
Injúria Renal Aguda , Chalconas , Traumatismo por Reperfusão , Ratos , Masculino , Animais , Caspase 3/metabolismo , Caspase 3/farmacologia , Fator de Necrose Tumoral alfa , Interleucina-6 , Creatinina , F2-Isoprostanos/metabolismo , F2-Isoprostanos/farmacologia , Ratos Sprague-Dawley , Rim , Isquemia/tratamento farmacológico , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/prevenção & controle , Apoptose , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/prevenção & controle , Estresse Oxidativo , Reperfusão , Inflamação/tratamento farmacológico , Inflamação/patologia
9.
J Med Life ; 15(2): 241-251, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35419091

RESUMO

Ischemia-reperfusion injury (IRI) can be defined as changes in the functions and structures of the tissues resulting from the restoration of blood after a period of ischemia. This study aimed to assess the potential protective effect of Fimasartan (angiotensin receptor antagonist) in the bilateral renal IRI in male rats through its potential effect on renal functions, modulation of the inflammatory cascade, oxidative stress, and apoptotic effect. The animals were equally assigned into four groups. The sham (negative control) group was exposed to surgical conditions without induction of IRI. The control group was exposed to ischemia by occluding the renal pedicles by clamps for 30 min, followed by restoration of blood for 2h. The vehicle-treated group received dimethyl sulfoxide (DMSO) by intraperitoneal injection (IP) 30 minutes before clamping. Fimasartan-treated group: rats pretreated with Fimasartan a dose of 3 mg/kg IP; this was half hour before occluding the renal pedicles. Animals were then exposed to 30 min ischemia (clamping the renal pedicles) followed by 2h reperfusion by releasing the clamps. Blood samples were collected to examine the levels of serum urea and creatinine. Renal tissue was used to measure the levels of cytokines (TNFα, IL-6) and total antioxidant capacity (TAC). Immunohistochemistry was used to assess the levels of Bax, caspase 3, and Bcl-2. Histopathological analyses were performed to detect the parenchymal injury. The present study shows that pretreatment with Fimasartan improves kidney function through its effects on oxidative stress, cytokines, and apoptotic markers.


Assuntos
Injúria Renal Aguda , Traumatismo por Reperfusão , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Apoptose , Compostos de Bifenilo/metabolismo , Compostos de Bifenilo/farmacologia , Compostos de Bifenilo/uso terapêutico , Feminino , Humanos , Rim , Masculino , Estresse Oxidativo , Pirimidinas , Ratos , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Tetrazóis
10.
Wiad Lek ; 75(11 pt 2): 2791-2803, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36591770

RESUMO

OBJECTIVE: The aim: To investigate the Nephroprotective potential of Olmesartan in RIRI via modulation of the Nrf2/OH-1 signaling pathway. PATIENTS AND METHODS: Materials and methods: Thirty male rats were equally divided into four groups. The sham group was exposed to surgical conditions without induction of RIRI. The control group was exposed to ischemia by clamping the renal pedicles for 30 min, followed by 2h of blood restoration. The vehicle-treated group was received dimethyl sulfoxide (DMSO) by intraperitoneal injection (IP) 30 min before clamping. RESULTS: Results: Olmesartan-treated group was pretreated with Olmesartan a dose of 10 mg/kg IP; 30 min prior to induction of ischemia. Following 30 min of ischemia, the clamps were released and allowed to the reperfusion for 2 h. Blood samples were collected to examine the levels of serum urea and creatinine. Kidney tissue was used to measure the levels of cytokines (TNFα, IL6, MCP, BAX, BCL2 and isoprostane F2. Immunohistochemistry was used to assess the levels of Nrf2 and HO-1. Histological analyses were used to detect the tubular damage in the kidney. CONCLUSION: Conclusions: The results showed that Olmesartan alleviates renal tissue damage through activating the antioxidant effect mediated by Nrf2 signaling.


Assuntos
Fator 2 Relacionado a NF-E2 , Traumatismo por Reperfusão , Masculino , Humanos , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/farmacologia , Fator 2 Relacionado a NF-E2/uso terapêutico , Estresse Oxidativo , Rim/patologia , Transdução de Sinais , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/prevenção & controle , Traumatismo por Reperfusão/metabolismo
11.
J Med Life ; 15(12): 1553-1562, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36762326

RESUMO

Cyclosporine A (CsA), a well-known immunosuppressive drug, has been prescribed after organ transplantation and in a variety of disorders with an immunological origin. Nephrotoxicity is one of the most frequently stated problems associated with CsA, and therefore the treatment with CsA remains a big challenge. This study sets out to assess the ameliorative influences of Candesartan Cilexetil (CC) on oxidative stress and the nephrotoxic effect of CsA in a rat model. Twenty-four Wister Albino rats, 7-8-week-old, weighing 150-250g, were randomly categorized into three groups (eight animals in each group). These groups were the (1) CsA-treated group, (2) vehicle-treated group, and (3) CC-treated group. Bodyweights were assessed at the start and end of experiments. Renal function test and levels of glutathione peroxidase 1 catalase -CAT (Gpx1), catalase (CAT), superoxide dismutase (SOD), interleukin -2 (IL-2), and malondialdehyde (MDA) were investigated in renal tissues. Histological changes in kidneys were also evaluated. Data showed that levels of urea and creatinine in serum and levels of IL-2 and MDA in renal tissues were elevated in the CsA-treated group, with severe histological changes compared with the control group. Furthermore, tissue levels of Gpx1, CAT, and SOD were significantly decreased in CsA-treated in comparison with the control group. Treatment with CC for the rats subjected to CSA resulted in a marked reduction in levels of serum urea and creatinine and tissue levels of IL-2 and MDA. Levels of Gpx1, CAT, and SOD in renal tissues were greater in the CC-treatment group compared with the CsA-treated group. CC treatment reduced the deterioration of renal morphology compared with CsA treatment. The findings of this study suggest that CC could prevent CSA-induced nephrotoxicity through its anti-inflammatory and antioxidant influences. Considerably more work needs to be done to determine the mechanistic insight behind the ameliorative effect of CC.


Assuntos
Nefropatias , Insuficiência Renal , Animais , Ratos , Ciclosporina/efeitos adversos , Catalase , Interleucina-2/metabolismo , Interleucina-2/farmacologia , Interleucina-2/uso terapêutico , Nefropatias/induzido quimicamente , Nefropatias/tratamento farmacológico , Nefropatias/prevenção & controle , Creatinina , Ratos Wistar , Imunossupressores/efeitos adversos , Rim , Estresse Oxidativo , Ureia/metabolismo , Ureia/farmacologia , Ureia/uso terapêutico , Superóxido Dismutase/metabolismo
12.
Mol Pharmacol ; 94(1): 674-688, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29724789

RESUMO

The structurally related, but distinct neuropeptides, neuromedin U (NmU) and neuromedin S (NmS) are ligands of two G protein-coupled NmU receptors (NMU1 and NMU2). Hypothalamic NMU2 regulates feeding behavior and energy expenditure and has therapeutic potential as an anti-obesity target, making an understanding of its signaling and regulation of particular interest. NMU2 binds both NmU and NmS with high affinity, resulting in receptor-ligand co-internalization. We have investigated whether receptor trafficking events post-internalization are biased by the ligand bound and can therefore influence signaling function. Using recombinant cell lines expressing human NMU2, we demonstrate that acute Ca2+ signaling responses to NmU or NmS are indistinguishable and that restoration of responsiveness (resensitization) requires receptor internalization and endosomal acidification. The rate of NMU2 resensitization is faster following NmU compared with NmS exposure, but is similar if endothelin-converting enzyme-1 activity is inhibited or knocked down. Although acute activation of extracellular signal-regulated kinase (ERK) is also similar, activation by NMU2 is longer lasting if NmS is the ligand. Furthermore, when cells are briefly challenged before removal of free, but not receptor-bound ligand, activation of ERK and p38 mitogen-activated protein kinase by NmS is more sustained. However, only NmU responses are potentiated and extended by endothelin-converting enzyme-1 inhibition. These data indicate that differential intracellular ligand processing produces different signaling and receptor resensitization profiles and add to the findings of other studies demonstrating that intracellular ligand processing can shape receptor behavior and signal transduction.


Assuntos
Receptores de Neurotransmissores/metabolismo , Transdução de Sinais/fisiologia , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Linhagem Celular , Enzimas Conversoras de Endotelina/metabolismo , Metabolismo Energético , Células HEK293 , Humanos , Ligantes , Sistema de Sinalização das MAP Quinases/fisiologia , Neuropeptídeos/metabolismo , Obesidade/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Neurotox Res ; 34(2): 263-272, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29589276

RESUMO

Alzheimer's disease is a neurodegenerative disease that affects 44 million people worldwide, costing the world $605 billion to care for those affected not taking into account the physical and psychological costs for those who care for Alzheimer's patients. Dipentylammonium is a simple amine, which is structurally similar to a number of other identified sigma-1 receptor ligands with high affinities such as (2R-trans)-2butyl-5-heptylpyrrolidine, stearylamine and dodecylamine. This study investigates whether dipentylammonium is able to provide neuroprotective effects similar to those of sigma-1 receptor agonists such as PRE-084. Here we identify dipentylammonium as a sigma-1 receptor ligand with nanomolar affinity. We have found that micromolar concentrations of dipentylammonium protect from glutamate toxicity and prevent NFκB activation in HT-22 cells. Micromolar concentrations of dipentylammonium also protect stably expressing amyloid precursor protein Swedish mutant (APP/Swe) Neuro2A cells from toxicity induced by 150 µM dopamine, suggesting that dipentylammonium may be useful for the treatment of Parkinsonian symptoms in Alzheimer's patients which are often associated with a more rapid deterioration of cognitive and physical ability. Finally, we found that low micromolar concentrations of dipentylammonium could out preform known sigma-1 receptor agonist PRE-084 in potentiating neurite outgrowth in Neuro2A cells, further suggesting that dipentylammonium has a potential use in the treatment of neurodegenerative diseases and could be acting through the sigma-1 receptor.


Assuntos
Ácidos Borônicos/farmacologia , Dopamina/farmacologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Ácido Glutâmico/farmacologia , Imidazóis/farmacologia , Crescimento Neuronal/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Receptores sigma/metabolismo , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Humanos , L-Lactato Desidrogenase/metabolismo , NF-kappa B/metabolismo , Neuroblastoma/patologia , Pentazocina/farmacologia , Transporte Proteico/efeitos dos fármacos , Ensaio Radioligante , Trítio/farmacocinética , Receptor Sigma-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA