Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Life Sci ; 351: 122808, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38852796

ABSTRACT

As a precursor of protoporphyrin IX (PpIX), an endogenous pro-apoptotic and fluorescent molecule, 5-Aminolevulinic acid (5-ALA) has gained substantial attention for its potential in fluorescence-guided surgery as well as photodynamic therapy (PDT). Moreover, 5-ALA-PDT has been suggested as a promising chemo-radio sensitization therapy for various cancers. However, insufficient 5-ALA-induced PpIX fluorescence and the induction of multiple resistance mechanisms may hinder the 5-ALA-PDT clinical outcome. Reduced efficacy and resistance to 5-ALA-PDT can result from genomic alterations, tumor heterogeneity, hypoxia, activation of pathways related to cell surveillance, production of nitric oxide, and most importantly, deregulated 5-ALA transporter proteins and heme biosynthesis enzymes. Understanding the resistance regulatory mechanisms of 5-ALA-PDT may allow the development of effective personalized cancer therapy. Here, we described the mechanisms underlying resistance to 5-ALA-PTD across various tumor types and explored potential strategies to overcome this resistance. Furthermore, we discussed future approaches that may enhance the efficacy of treatments using 5-ALA-PDT.


Subject(s)
Aminolevulinic Acid , Drug Resistance, Neoplasm , Neoplasms , Photochemotherapy , Photosensitizing Agents , Aminolevulinic Acid/pharmacology , Humans , Photochemotherapy/methods , Neoplasms/drug therapy , Neoplasms/pathology , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Animals , Protoporphyrins/pharmacology , Protoporphyrins/metabolism
2.
Med Oncol ; 41(3): 70, 2024 Feb 10.
Article in English | MEDLINE | ID: mdl-38340190

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is one of the world's largest health concerns with growing global incidence and mortality. The potential value of the neurokinin-1 receptor as a therapeutic target has been reported in several tumor types, including CRC. Here we examined the potential anti-tumor effects of a clinically approved neurokinin-1 receptor antagonist, aprepitant, alone and its combination with 5-Fluorouracil (5-FU) as a first choice CRC chemotherapeutic drug, in both in vitro and in vivo models of CRC. METHODS: MTT assay was employed for assessing cell proliferation. mRNA expression levels were determined by quantitative real-time PCR (qRT-PCR). Flow cytometric analysis of apoptosis was performed using an Annexin-V/propidium iodide assay kit. We finally conducted an in vivo experiment in a mouse model of CRC to confirm the in vitro antiproliferative activity of aprepitant and 5-FU. RESULTS: We found that aprepitant and 5-FU significantly reduced CRC cell viability. The combination of drugs exhibited potent synergistic growth inhibitory effects on CRC cells. Moreover, aprepitant and 5-FU induced apoptosis and altered the levels of apoptotic genes (up-regulation of Bax, and p53 along with downregulation of Bcl-2). Importantly, the aprepitant and 5-FU combination showed a more pronounced impact on apoptosis and associated genes than either of the agents alone. Furthermore, aprepitant reduced tumor growth in vivo and led to significantly longer survival time, and this effect was more prominent when using the aprepitant and 5-FU combination. CONCLUSIONS: Collectively, combinatory treatment with aprepitant and 5-FU potentially exerts synergistic growth inhibition and apoptosis induction in CRC, deserving further consideration as a novel strategy for CRC patients.


Subject(s)
Colorectal Neoplasms , Fluorouracil , Animals , Mice , Humans , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Aprepitant/pharmacology , Colorectal Neoplasms/pathology , Xenograft Model Antitumor Assays , Drug Synergism , Apoptosis , Cell Proliferation , Cell Line, Tumor
3.
Int. j. cardiovasc. sci. (Impr.) ; 36: e20230016, jun.2023. tab, graf
Article in English | LILACS-Express | LILACS | ID: biblio-1528761

ABSTRACT

Abstract Background: Coronary artery disease (CAD) is the most common form of cardiac disease with high morbidity and mortality rates. Objectives: In this study, we evaluated the expression of miR-27a and miR-27b as biomarkers in peripheral blood mononuclear cells (PBMCs) of patients with CAD and investigated its correlation with cholesterol-efflux transporter, ATP-binding cassette transporter A1 (ABCA1). Method: This study was performed on 54 men with CAD and 51 healthy, sex- and age-matched control participants. The expression of miR-27a/b and ABCA1 genes in PBMCs were measured by quantitative real-time polymerase chain reaction (qRT-PCR). The protein expression of ABCA1 was assessed by Western blotting. Concurrently, the specificity and sensitivity of miR-27a/b was evaluated through receiver operating characteristic (ROC) curve. The significance level adopted in the statistical analysis was 5%. Results: We found that miR-27a and miR-27b expression were significantly increased, while both mRNA and protein expression of ABCA1 were markedly reduced in the PBMCs of CAD patients in comparison to non-CAD controls. miR-27a/27b expression was also shown to be inversely correlated with ABCA1. ROC analysis showed that the miR-27a had an area under the ROC curve (AUC) of about 92.6 (sensitivity 83.3٪ and specificity 86.6٪) and miR-27b had an AUC of about 93.0 (sensitivity 86.6٪ and specificity 80.0 (%, suggesting the diagnostic potential of miR-27a/b in CAD patients. Conclusions: Our data suggested a possible role of miR-27a/b in CAD pathogenesis. Additionally, we proposed that miR-27a/b expression in PBMCs may have potential clinical implications in the diagnosis of CAD patients, but further validations in large cohorts are required.

4.
Biofactors ; 49(4): 900-911, 2023.
Article in English | MEDLINE | ID: mdl-37092793

ABSTRACT

Glioblastoma multiforme (GBM) is the most malignant type of cerebral neoplasm in adults with a poor prognosis. Currently, combination therapy with different anti-cancer agents is at the forefront of GBM research. Hence, this study aims to evaluate the potential anti-cancer synergy of a clinically approved neurokinin-1 receptor antagonist, aprepitant, and 5-aminolevulinic acid (5-ALA), a prodrug that elicits fluorescent porphyrins in gliomas on U-87 human GBM cells. We found that aprepitant and 5-ALA effectively inhibited GBM cell viability. The combinatorial treatment of these drugs exerted potent synergistic growth inhibitory effects on GBM cells. Moreover, aprepitant and 5-ALA induced apoptosis and altered the levels of apoptotic genes (up-regulation of Bax and P53 along with downregulation of Bcl-2). Furthermore, aprepitant and 5-ALA increased the accumulation of protoporphyrin IX, a highly pro-apoptotic and fluorescent photosensitizer. Aprepitant and 5-ALA significantly inhibited GBM cell migration and reduced matrix metalloproteinases (MMP-2 and MMP-9) activities. Importantly, all these effects were more prominent following aprepitant-5-ALA combination treatment than either drug alone. Collectively, the combination of aprepitant and 5-ALA leads to considerable synergistic anti-proliferative, pro-apoptotic, and anti-migratory effects on GBM cells and provides a firm basis for further evaluation of this combination as a novel therapeutic approach for GBM.


Subject(s)
Aminolevulinic Acid , Glioblastoma , Adult , Humans , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Aprepitant/pharmacology , Aprepitant/therapeutic use , Neurokinin-1 Receptor Antagonists/pharmacology , Neurokinin-1 Receptor Antagonists/therapeutic use , Cell Line, Tumor
5.
Appl Biochem Biotechnol ; 195(12): 7796-7807, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37093533

ABSTRACT

Prostate cancer is one of the main global health threats for men which is in close association with chronic inflammation. Neuropeptide substance P (SP), acting through neurokinin receptor (NK-1R), induces various pro-inflammatory responses which are strongly involved in the pathogenesis of several diseases as well as cancer. Therefore, we aimed to investigate the pro-inflammatory functions of the SP/NK1R complex in prostate cancer and the therapeutic effects of its inhibition by NK-1R antagonist, aprepitant, in vitro. MTT assay was conducted for the cytotoxicity assessment of aprepitant in prostate cancer cells. The protein expression levels were evaluated by Western blot assay. Quantitative real-time PCR (qRT-PCR) was applied to measure mRNA expression levels of pro-inflammatory cytokines. Concurrently, the protein concentrations of pro-inflammatory cytokines were also analyzed by enzyme-linked immunosorbent assay. We observed that SP increased the levels of pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α), while treatment with aprepitant reduced the effects of SP. We also indicated that SP increased the protein levels of nuclear factor-kappa B (NF-κB), as the main regulator of inflammatory processes, and also an NF-κB target gene, cyclooxygenase 2 (COX-2) in prostate cancer cells, while treatment with aprepitant reversed these effects. Taken together, our findings highlight the importance of the SP/NK1R system in the modulation of pro-inflammatory responses in prostate cancer cells and suggest that aprepitant may be developed as a novel anti-inflammatory agent for the management of cancer-associated inflammation.


Subject(s)
NF-kappa B , Prostatic Neoplasms , Male , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Substance P/metabolism , Substance P/pharmacology , Substance P/therapeutic use , Signal Transduction , Aprepitant/pharmacology , Aprepitant/therapeutic use , Inflammation/drug therapy , Inflammation/metabolism , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Interleukin-1beta/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics
6.
Biofactors ; 49(3): 534-559, 2023.
Article in English | MEDLINE | ID: mdl-36651605

ABSTRACT

Diabetes is a significant public health issue known as the world's fastest-growing disease condition. It is characterized by persistent hyperglycemia and subsequent chronic complications leading to organ dysfunction and, ultimately, the failure of target organs. Substance P (SP) is an undecapeptide that belongs to the family of tachykinin (TK) peptides. The SP-mediated activation of the neurokinin 1 receptor (NK1R) regulates many pathophysiological processes in the body. There is also a relation between the SP/NK1R system and diabetic processes. Importantly, deregulated expression of SP has been reported in diabetes and diabetes-associated chronic complications. SP can induce both diabetogenic and antidiabetogenic effects and thus affect the pathology of diabetes destructively or protectively. Here, we review the current knowledge of the functional relevance of the SP/NK1R system in diabetes pathogenesis and its exploitation for diabetes therapy. A comprehensive understanding of the role of the SP/NK1R system in diabetes is expected to shed further light on developing new therapeutic possibilities for diabetes and its associated chronic conditions.


Subject(s)
Diabetes Mellitus , Substance P , Humans , Substance P/genetics , Substance P/pharmacology , Receptors, Neurokinin-1/genetics , Receptors, Neurokinin-1/metabolism , Diabetes Mellitus/drug therapy , Diabetes Mellitus/genetics
7.
Cell Biochem Biophys ; 80(4): 819-827, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36197642

ABSTRACT

Ovarian cancer is the seventh most common cancer globally, and the second most common cancer among women with significant mortality. Toward this end, it is shown that substance P (SP) is involved in tumor initiation and progression through the neurokinin-1 receptor (NK1R). However, the exact molecular mechanism of the SP/NK1R system in ovarian cancer is not yet fully clarified. In this in vitro study, we decided to investigate the effect of the SP/NK1R system and blockage of NK1R by its specific antagonist (Aprepitant) on the proliferation of ovarian cancer cells as well as the alteration of inflammatory pathways. Our results revealed that Aprepitant stimulated apoptotic cell death and attenuated inflammation of ovarian cancer cells through the NF-kB and P53 signaling pathways. After treatment with Aprepitant, the expression of downstream anti-apoptotic genes related to the NF-kB pathway (survivine and bcl2) was decreased. However, we indicated the positive effect of SP on the proliferation of ovarian cancer cells by inducing the expression of NF-kB protein and NF-kB anti-apoptotic target genes. Moreover, Pro-apoptotic p53 target genes (P21 and Bax) were increased through aprepitant treatment, while SP attenuated these genes' expression. Besides, ROS generation in ovarian cancer cells after treatment with SP induced, while blocking of NK1R with Aprepitant reduced the level of ROS generation. Given this, our data suggest that this NK1R might be used as an important therapeutic target in ovarian cancer and Aprepitant could be considered a new drug in ovarian cancer therapy.


Subject(s)
Ovarian Neoplasms , Receptors, Neurokinin-1 , Apoptosis , Aprepitant/pharmacology , Cell Line, Tumor , Cell Proliferation , Female , Humans , NF-kappa B/metabolism , Ovarian Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Receptors, Neurokinin-1/genetics , Receptors, Neurokinin-1/metabolism , Substance P/metabolism , Substance P/pharmacology , Tumor Suppressor Protein p53 , bcl-2-Associated X Protein/metabolism
8.
Biomed Res Int ; 2022: 8082608, 2022.
Article in English | MEDLINE | ID: mdl-36177059

ABSTRACT

Background: Osteosarcoma, the most frequent osteogenic malignancy, has become a serious public health challenge due to its high morbidity rates and metastatic potential. Recently, the neurokinin-1 receptor (NK-1R) is proved to be a promising target in cancer therapy. This study is aimed at determining the effect of aprepitant, a safe and Food and Drug Administration (FDA) approved NK-1R antagonist, on osteosarcoma cell migration and metastasis, and to explore its underlying mechanism of action. Methods: Colorimetric MTT assay was employed to assess cell viability and cytotoxicity. A wound-healing assay was used to examine migration ability. The desired genes' protein and mRNA expression levels were measured by western blot assay and quantitative real-time PCR (qRT-PCR), respectively. Gelatinase activity was also measured by zymography. Results: We found that aprepitant inhibited MG-63 osteosarcoma cell viability in a dose-dependent manner. We also observed that aprepitant inhibited the migrative phenotype of osteosarcoma cells and reduced the expression levels and activities of matrix metalloproteinases (MMP-2 and MMP-9). Aprepitant also reduced the expression of an angiogenic factor, VEGF protein, and NF-κB as an important transcriptional regulator of metastasis-related genes. Conclusion: Collectively, our observations indicate that aprepitant modulates the metastatic behavior of human osteosarcoma cells, which may be applied to an effective therapeutic approach for patients with metastatic osteosarcoma.


Subject(s)
Bone Neoplasms , Osteosarcoma , Angiogenesis Inducing Agents/pharmacology , Aprepitant/pharmacology , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , NF-kappa B , Neurokinin-1 Receptor Antagonists/pharmacology , Neurokinin-1 Receptor Antagonists/therapeutic use , Osteosarcoma/pathology , RNA, Messenger/genetics , Receptors, Neurokinin-1/genetics , Receptors, Neurokinin-1/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/pharmacology
9.
Toxicol Appl Pharmacol ; 440: 115951, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35235860

ABSTRACT

BACKGROUND: Overexpression of the angiotensin-II receptor and renin-angiotensin system (RAS) has been reported in several malignancies, including colorectal-cancer (CRC), indicating its potential value as a therapeutic target. Here we explored the impact of targeting the RAS using an angiotensin II receptor blocker, valsartan, alone and its combination with Fluorouracil (5-FU) in in vitro and in vivo models of CRC. METHODS: Anti-proliferative activity of valsartan was evaluated in 2-/3-dimensional in vitro and in vivo CRC mouse models. The anti-migratory effects of this agent was assessed by wound-healing assay, while apoptosis was studied using 4',6-diamidino-2-phenylindole or DAPI staining, and staining with Annexin-V-fluorescein isothiocyanate with analysis using FACS. Gene-expression was determined at mRNA and protein levels. We further evaluated the anti-inflammatory properties of valsartan by histological analysis and the measurement of oxidative/antioxidant markers. Gelatin zymography was used to measure matrix metalloproteinase-2 and -9 activity (MMP-2 and 9). RESULTS: Valsartan suppressed CRC cell-growth and synergistically enhanced the anti-tumor-activities of 5-FU by induction of apoptosis, BAX, BCL2, P53 and modulation of the cell cycle. Valsartan inhibited the cell migration by perturbation of MMP2/9. Furthermore, valsartan inhibited tumor-growth, and this was more pronounced when using the valsartan/5-FU combination. The plausible mechanism for this is via the induction of ROS and down-regulation of SOD, thiol/catalase as well as VEGF. Valsartan may protect cells against intestinal fibrosis by modulation of pro-fibrotic and pro-inflammatory factors including interleukins and Col1A1 expression. CONCLUSIONS: Our findings demonstrated that targeting RAS pathway using Valsartan interferes with cell-proliferation, induces apoptosis, reduces migration and synergistically interacts with 5-FU, supporting further studies on this new therapeutic approach for colorectal cancer.


Subject(s)
Colorectal Neoplasms , Matrix Metalloproteinase 2 , Angiotensins/therapeutic use , Animals , Colorectal Neoplasms/metabolism , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Mice , Valsartan/pharmacology , Valsartan/therapeutic use
10.
Life Sci ; 296: 120448, 2022 May 01.
Article in English | MEDLINE | ID: mdl-35247438

ABSTRACT

Oxidative stress which refers to redox imbalance with increased generation of reactive oxygen species (ROS) has been associated with the pathophysiology of diverse disease conditions. Recently, a close, yet not fully understood, relation between oxidative stress and neuropeptides, in particular, substance P (SP), has been reported in certain conditions. SP has been shown to affect the cellular redox environment through activation of neurokinin-1receptor (NK1R). It seems that SP/NK1R system and oxidative stress can act either synergistically or antagonistically in a context-dependent manner, thereby, influencing the pathology of various clinical disorders either destructively or protectively. Importantly, the interactions between oxidative stress and SP/NK1R system can be pharmacologically targeted. Therefore, a better understanding of the redox modulatory properties of SP/NK1R signaling will pave the way for identifying new therapeutic possibilities for attenuating oxidative stress-mediated damage. Towards this end, we performed a comprehensive search through PubMed/Medline and Scopus databases and discussed all related existing literature regarding the interplay between oxidative stress and SP/NK1R system as well as their implication in various clinical disorders, to provide a clear view and hence better management of oxidative damage.


Subject(s)
Neurokinin-1 Receptor Antagonists/pharmacology , Oxidative Stress/physiology , Receptors, Neurokinin-1/metabolism , Stress, Psychological/metabolism , Substance P/metabolism , Animals , Humans , Magnesium Deficiency/metabolism , Neurokinin-1 Receptor Antagonists/therapeutic use , Oxidation-Reduction , Signal Transduction/drug effects
11.
Biochim Biophys Acta Mol Cell Res ; 1869(5): 119221, 2022 05.
Article in English | MEDLINE | ID: mdl-35134443

ABSTRACT

AIMS: Prostate cancer continues to be one of the main global health issues in men. Neuropeptide substance P (SP) acting via neurokinin-1receptor (NK1R) promotes tumorigenicity in many human malignant tumors. However, its pro-tumorigenic functions and the therapeutic effects of its inhibition in prostate cancer remain unclear. METHODS: MTT assay was employed for measuring cellular proliferation and cytotoxicity. mRNAs and proteins expression levels were evaluated by qRT-PCR and western blot assay, respectively. Gelatinase activity was assessed by zymography. The migration ability was defined using wound-healing assay. Flow cytometry was employed to evaluate the cell cycle distribution. We also performed an in vivo experiment in a mouse model of prostate cancer to confirm the in vitro therapeutic effect of targeting the SP/NK1R system. RESULTS: We found a noticeable increase in the expression of the truncated isoform of NK1R as an oncogenic NK1R splice variant in tumor cells. We also demonstrated that SP promotes both proliferative and migrative phenotypes of prostate cancer through modifying cell cycle-related proteins (c-Myc, cyclin D1, cyclin B1, p21), and apoptosis-related genes (Bcl-2 and Bax), promoting cell migration and increasing MMP-2 and MMP-9 expression and activity, while aprepitant administration could remarkably reverse these effects. SP also stimulated tumor growth in vivo, which was correlated with shorter survival times, while aprepitant reversed this effect and led to significantly longer survival time. SIGNIFICANCE: Our findings suggest that SP/NK1R system may serve as a novel therapeutic target in prostate cancer and support the possible candidacy of aprepitant in future prostate cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Aprepitant/pharmacology , Cell Proliferation/drug effects , Receptors, Neurokinin-1/metabolism , Substance P/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Aprepitant/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Male , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Nude , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Neurokinin-1/genetics , Substance P/therapeutic use , Survival Rate , Transplantation, Heterologous
12.
Mol Biol Rep ; 49(2): 1067-1076, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34766230

ABSTRACT

BACKGROUND: Cervical cancer, an aggressive gynecological cancer, seriously threatens women's health worldwide. It is recently reported that neuropeptide substance P (SP) regulates many tumor-associated processes through neurokinin-1 receptor (NK1R). Therefore, we used cervical cancer cell line (HeLa) to investigate the functional relevance of the SP/NK1R system in cervical cancer pathogenesis. METHODS: Cellular proliferation and cytotoxicity were analyzed by colorimetric MTT assay. Quantitative real-time PCR (qRT-PCR) was used to measure mRNA expression levels of desired genes. Cell cycle distribution and apoptosis were evaluated by flow cytometry. A wound-healing assay was employed to assess migration ability. RESULTS: We found that the truncated isoform of NK1R(NK1R-Tr) is the dominantly expressed form of the receptor in Hela cells. We also indicated that that SP increased HeLa cell proliferation while treatment with NK1R antagonist, aprepitant, inhibited HeLa cell viability in a dose and time-dependent manner. SP also alters the levels of cell cycle regulators (up-regulation of cyclin B1 along with downregulation of p21) and apoptosis-related genes (up-regulation of Bcl-2 along with downregulation of Bax) while aprepitant reversed these effects. Aprepitant also induced arrest within the G2 phase of the cell cycle and subsequent apoptosis. Furthermore, SP promoted the migrative phenotype of HeLa cells and increased MMP-2 and MMP-9 expression while aprepitant exposure significantly reversed these effects. CONCLUSION: Collectively, our results indicate the importance of the SP / NK1R system in promoting both proliferative and migrative phenotypes of cervical cancer cells and suggest that aprepitant may be developed as a novel treatment for combating cervical cancer.


Subject(s)
Receptors, Neurokinin-1/metabolism , Substance P/metabolism , Uterine Cervical Neoplasms/genetics , Apoptosis/drug effects , Aprepitant/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Neurokinin-1 Receptor Antagonists/pharmacology , Receptors, Neurokinin-1/genetics , Signal Transduction/drug effects , Uterine Cervical Neoplasms/therapy
13.
Neurol Res Int ; 2021: 9966000, 2021.
Article in English | MEDLINE | ID: mdl-34917417

ABSTRACT

Altered redox balance is among the main contributing factors developing glioblastoma multiforme (GBM), a highly aggressive grade IV brain tumor. Neuropeptide substance P (SP) plays a key role in modifying the cellular redox environment by activating the neurokinin-1 receptor (NK1R). In this study, we aimed to investigate the redox-modulating properties of both SP and a commercially available NK1R antagonist, aprepitant in GBM cells. To detect the effect of aprepitant on the viability of U87 glioblastoma cells, resazurin assay was applied. The level of intracellular ROS was assessed using 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA) assay. The expression of glutaredoxin, a well-known redox-active protein, was measured by quantitative real-time polymerase chain reaction (qRT-PCR). Concurrently, the activity of glutaredoxin was also analyzed by a commercial kit (ZellBio GmbH). We found that SP increased the intracellular levels of reactive oxygen species (ROS) in U87 GBM cells, and aprepitant remarkably decreased this effect. We also explored the effects of SP/NK1R signaling on the glutaredoxin system as a major cellular redox buffer in GBM cells. SP reduced both expression and enzymatic activity of glutaredoxin, and these effects were significantly decreased by aprepitant. In conclusion, our results suggest a possible involvement of SP/NK1R signaling in GBM pathogenesis through oxidative stress and offering new insight for the application of aprepitant as a redox-modulating strategy in GBM patients.

14.
Iran J Basic Med Sci ; 24(4): 499-505, 2021 Apr.
Article in English | MEDLINE | ID: mdl-34094032

ABSTRACT

OBJECTIVES: Glioblastoma multiforme (GBM), a highly aggressive Grade IV brain tumor, is a significant public health issue due to its poor prognosis and incurability. Neuropeptide substance P (SP) plays a critical role in GBM tumor growth and development via activation of neurokinin-1receptor (NK1R). Moreover, SP is a pro-oxidant factor contributing to oxidative stress in various cell types. However, the link between SP and oxidative stress in cancer cells is not fully investigated. Here, we aimed to identify the effects of SP and NK1R antagonist, aprepitant, on the redox status of GBM cells. MATERIALS AND METHODS: Resazurin assay was employed to determine the effect of aprepitant on viability of U87 glioblastoma cells. 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA) assay was employed to measure the levels of intracellular reactive oxygen species (ROS). A quantitative real-time polymerase chain reaction was applied to measure the expression of proteins of the thioredoxin system. Commercial kits (ZellBio GmbH) were also used to measure the enzymatic activity of these proteins. RESULTS: We found that SP increased ROS level in U87 GBM cells, and aprepitant significantly reduced this effect. Furthermore, we found that SP could also affect the thioredoxin system, a central antioxidant enzyme defense system. SP reduced both expression and enzymatic activity of the thioredoxin system's proteins, Trx and thioredoxin reductase (TrxR) and these effects were significantly reduced by aprepitant. CONCLUSION: Our results indicated that SP activation of NK1R represented a link between oxidative stress and GBM and highlighted the need for further validations in future studies.

15.
Curr Mol Pharmacol ; 14(4): 559-569, 2021 10 25.
Article in English | MEDLINE | ID: mdl-33357209

ABSTRACT

Prostate cancer (PCa) is the second most prevalent cancer and the fifth leading cause of cancer-related deaths among men. Androgen deprivation therapy (ADT) is the most frequently used therapeutic strategy in PCa; however, the development of resistance to ADT, known as castration- resistant prostate cancer (CRPC), continues to be a major obstacle against the successful treatment of PCa. The abnormal activation of the androgen receptor (AR) signaling pathway has been found as one of the main contributing factors to the development of resistance in CRPC. Therefore, AR regulatory strategies are urgently required to combat resistance. Recently, microRNAs (miRNAs) have been found as major AR regulatory factors affecting ADT resistance. MiRNAs can target AR itself, AR-related genes, AR splice variants, AR-related signaling pathways as well as cancer stem cells (CSCs), and play critical roles in regulating ADT resistance. Due to their capability to affect various genes and signaling pathways, miRNAs are now being studied for their potential role as a new therapeutic target in CRPC. It has been recommended that combination therapies, including miRNAs and existing drugs, can synergistically decrease castration resistance. miRNAs also have prognostic values for ADT, and their expression profiling in CRPC patients before therapeutic scheduling may enable the physician to diagnose patients who are ADT-resistant. Overall, extant evidence obviously supports the predictive and therapeutic potential of miRNAs in CRPC patients. This review summarizes the available information about the microRNA-mediated AR controlling mechanisms involved in ADT resistance.


Subject(s)
MicroRNAs , Prostatic Neoplasms, Castration-Resistant , Androgen Antagonists/therapeutic use , Humans , Male , MicroRNAs/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, Androgen/therapeutic use , Signal Transduction
16.
Clin Genet ; 98(4): 322-330, 2020 10.
Article in English | MEDLINE | ID: mdl-32266968

ABSTRACT

The neuropeptide substance P (SP) triggers a variety of tumor-promoting signaling pathways through the activation of neurokinin-1receptor (NK1R), a class of neurokinin G protein-coupled receptors superfamily. Recent researches in our and other laboratories have shown the overexpression of both SP and NK1R in breast cancer (BC) patients. SP/NK1R signaling is strongly implicated in the pathogenesis of BC through affecting cell proliferation, migration, metastasis, angiogenesis, and resistance. Therefore, SP/NK1R signaling responses must be rigorously regulated; otherwise, they would contribute to a more aggressive BC phenotype. Recently, microRNAs (miRNAs) as a specific class of epigenetic regulators have been shown to regulate NK1R and thus, controlling SP/NK1R signaling responses in BC. This review summarizes the current knowledge of the role of SP/NK1R signaling and its therapeutic potentials in BC. We also provide an overview regarding the effects of miRNA-mediated NK1R regulatory mechanisms in controlling BC tumorigenesis to gain a clearer view and thus better management of cancer.


Subject(s)
Breast Neoplasms/genetics , MicroRNAs/genetics , Receptors, Neurokinin-1/genetics , Substance P/genetics , Breast Neoplasms/epidemiology , Breast Neoplasms/pathology , Cell Proliferation/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Phosphorylation/genetics , Signal Transduction/genetics
17.
Crit Rev Eukaryot Gene Expr ; 29(4): 333-342, 2019.
Article in English | MEDLINE | ID: mdl-31679294

ABSTRACT

Acute myocardial infarction (AMI), or heart attack, is a major public health problem, responsible for 3 to 4 million deaths each year. Despite great improvements in diagnostic and therapeutic strategies, it remains one of the most lethal types of heart disease. Therefore, the identification of molecular mechanisms involved in AMI pathogenesis might help us to develop new therapeutic and diagnostic approaches. MicroRNAs (21- to 24-nucleotide noncoding RNAs) have been shown to play important roles in AMI pathogenesis by affecting multiple cellular processes, including cardiac cell proliferation, apoptosis, survival, regeneration, and autophagy. Thus, targeting microRNAs might have great clinical significance for the treatment of AMI patients. Moreover, aberrant miRNA expression patterns can serve as an ideal diagnostic and prognostic biomarker for AMI patients. This review aims to give an overview of recent studies that have addressed the therapeutic potency of microRNAs in AMI. We also summarize the potential use of microRNAs as diagnostic and prognostic biomarkers for AMI.


Subject(s)
Biomarkers/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Myocardial Infarction/genetics , Apoptosis/genetics , Biomarkers/blood , Cell Proliferation/genetics , Cell Survival/genetics , Humans , MicroRNAs/blood , Myocardial Infarction/diagnosis , Myocardial Infarction/therapy , Myocardium/metabolism , Myocardium/pathology , Prognosis
18.
Psychiatry Res ; 272: 467-473, 2019 02.
Article in English | MEDLINE | ID: mdl-30611966

ABSTRACT

There is growing interest on the impact of diet on depressive disorders. However, there are limited data on the association between dietary patterns and depression symptoms among Iranian adolescents. The aim of this study was to evaluate the association between dietary patterns and depression score among Iranian adolescent girls. Data were obtained from 750 adolescent girls, who were recruited from several schools using a random cluster sampling. Dietary data were collected using a valid and reliable food frequency questionnaire (FFQ). Beck's depression inventory (BDI-II) was used to assess depression severity score. The prevalence of girls diagnosed with depression was 29.1%. Three major dietary patterns were identified in the study based on factor analysis: "Healthy", "Traditional" and "Western". The multivariate-adjusted odds ratio of having depressive symptoms was 0.55 (95% confidence intervals, 0.34-0.89) for the highest versus lowest quartile of the healthy dietary pattern score. However, there were no significant associations between the Traditional and Western dietary patterns and depression symptoms. Our results indicate that adherence to a Healthy dietary pattern characterized by high intakes of fruits, vegetables, fish and dairy products is associated with a lower probability of having depressive symptoms among Iranian adolescent girls.


Subject(s)
Depressive Disorder/epidemiology , Depressive Disorder/psychology , Diet, Healthy/psychology , Adolescent , Child , Cross-Sectional Studies , Depressive Disorder/diet therapy , Diet, Healthy/trends , Diet, Western/adverse effects , Diet, Western/psychology , Female , Humans , Iran/epidemiology , Prevalence , Psychiatric Status Rating Scales , Surveys and Questionnaires
19.
Cell Oncol (Dordr) ; 42(2): 131-141, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30645730

ABSTRACT

BACKGROUND: Chemotherapy and radiation therapy are the most common types of cancer therapy. The development of chemo/radio-resistance remains, however, a major obstacle. Altered redox balances are among of the main factors mediating therapy resistance. Therefore, redox regulatory strategies are urgently needed to overcome this problem. Recently, microRNAs have been found to act as major redox regulatory factors affecting chemo/radio-resistance. MicroRNAs play critical roles in regulating therapeutic resistance through the regulation of antioxidant enzymes, redox-sensitive signaling pathways, cancer stem cells, DNA repair mechanisms and autophagy. CONCLUSIONS: Here, we summarize current knowledge on microRNA-mediated redox regulatory mechanisms underlying chemo/radio-resistance. This knowledge may form a basis for a better clinical management of cancer patients.


Subject(s)
Drug Resistance, Neoplasm/genetics , MicroRNAs/metabolism , Neoplasms/genetics , Animals , Antioxidants/metabolism , Humans , MicroRNAs/genetics , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Oxidation-Reduction
20.
J Cell Biochem ; 120(3): 2801-2809, 2019 03.
Article in English | MEDLINE | ID: mdl-30260014

ABSTRACT

Breast cancer is the most common cause of cancer death in women and presents a serious therapeutic challenge worldwide. Traditional treatments are less successful at targeting cancer tumors, leading to recurrent treatment-resistant secondary malignancies. Oncolytic virotherapy (OV) is a novel anticancer strategy with therapeutic implications at targeting cancer cells by using mechanisms that differ from conventional therapies. Administration of OVs either alone or in combination with standard therapies provide new insights regarding the effectiveness and improvement of treatment responses for breast cancer patients. This review summarizes cellular, animal and clinical studies investigating therapeutic potency of oncolytic virotherapy in breast cancer treatment for a better understanding and hence a better management of this disease.


Subject(s)
Breast Neoplasms/therapy , Oncolytic Virotherapy/trends , Animals , Breast Neoplasms/pathology , Female , Humans , Neoplastic Stem Cells/pathology , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , Receptors, Virus/metabolism , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...