Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
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
2.
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
3.
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
4.
J Cell Biochem ; 120(5): 6868-6877, 2019 May.
Article in English | MEDLINE | ID: mdl-30426530

ABSTRACT

Cervical cancer (CC) is one of the most common cancers among females, and it is most notable in developing countries. The exact etiology of CC is poorly understood; but, smoking, oral contraceptives, immunosuppression, and infection with human papillomavirus (HPV) may increase the risk of CC. There is also an association between CC and oxidative stress. Oxidative stress is caused by a disturbed oxidant-antioxidant balance in favor of the former, leading to an excessive generation of free radicals, particularly reactive oxygen species (ROS), and subsequently to biological damages. Thus, redox enzymatic and nonenzymatic regulators are required to maintain the redox homeostasis. Dysregulated antioxidants system and the pathogenic role of oxidative stress in CC have been investigated in several clinical and preclinical studies. In this study, we reviewed studies that have addressed the cross-talk between oxidative stress and CC pathogenesis and resistance to therapy.

5.
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
6.
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
7.
J Cell Biochem ; 119(10): 7905-7912, 2018 11.
Article in English | MEDLINE | ID: mdl-30011093

ABSTRACT

The concentrations of adenosine may increase under ischemic conditions in the tumor microenvironment, and then it enters the systemic circulation. Adenosine controls cancer progression and responses to therapy by regulating angiogenesis, cell survival, apoptosis, cell proliferation, and metastases in tumors. Hence, adenosine metabolism, adenosine-generating enzymes, and adenosine signaling are potentially novel therapeutic targets in a wide range of pathological conditions, including cerebral and cardiac ischemic diseases, inflammatory disorders, immunomodulatory disorders, and, of special interest in this review, cancer. This review summarizes the role of adenosine in the pathogenesis of head and neck cancer for a better understanding of how this may be applied to treating this type of cancer.


Subject(s)
Adenosine/metabolism , Head and Neck Neoplasms/metabolism , Animals , Humans , Neovascularization, Pathologic/metabolism , Signal Transduction/physiology
8.
J Cell Physiol ; 232(8): 1966-1970, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27925197

ABSTRACT

The coagulation protease Factor Xa (FXa) triggers a variety of signaling pathways through activation of protease-activated receptors (PARs) and non-PAR receptors. FXa-mediated signaling is strongly implicated in the pathogenesis of several inflammatory diseases including fibrosis, cardiovascular diseases, and cancer. Thus, targeting of FXa can have great clinical significance in terms of the treatment of these disorders. This review summarizes the current knowledge about the mechanism of FXa signaling in cellular and animal systems under (patho) physiological conditions for a better understanding and hence a better management of FXa-induced disorders. J. Cell. Physiol. 232: 1966-1970, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Atherosclerosis/metabolism , Factor Xa/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Neoplasms/metabolism , Signal Transduction , Animals , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/pharmacology , Atherosclerosis/blood , Atherosclerosis/drug therapy , Factor Xa Inhibitors/therapeutic use , Humans , Inflammation/blood , Inflammation/drug therapy , Inflammation Mediators/antagonists & inhibitors , Neoplasms/blood , Neoplasms/drug therapy , Signal Transduction/drug effects
9.
J Cell Physiol ; 232(3): 482-485, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27458694

ABSTRACT

Thrombin initiates proinflammatory signaling responses through activation of protease-activated receptors (PARs) in in vitro and in vivo systems. Proinflammatory signaling function of thrombin increases secretion of proinflammatory cytokines and chemokines, triggers vascular permeability, promotes leukocyte migration, and induces adhesion molecule expression. Thrombin as a potent signaling molecule is strongly implicated in a number of proinflammatory disorders including severe sepsis, cancer, cardiovascular disease, and of special interest in this review neurodegenerative disorders. This review summarizes the role of thrombin in the pathogenesis of central nervous system (CNS) inflammatory diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD), promoting greater understanding and clinical management of these diseases. J. Cell. Physiol. 232: 482-485, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Central Nervous System Diseases/metabolism , Central Nervous System Diseases/pathology , Inflammation/metabolism , Inflammation/pathology , Thrombin/metabolism , Animals , Central Nervous System/metabolism , Central Nervous System/pathology , Humans , Signal Transduction
10.
J Cell Physiol ; 232(9): 2323-2329, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28004386

ABSTRACT

Thrombin-induced activation of protease-activated receptors (PARs) represents a link between inflammation and cancer. Proinflammatory signaling functions of thrombin are associated with several inflammatory diseases including neurodegenerative, cardiovascular, and of special interest in this review cancer. Thrombin-induced inflammatory responses up-regulates expression of cytokines, adhesion molecules, angiogenic factors, and matrix-degrading proteases that facilitate tumor cells proliferation, angiogenesis, invasion, and metastasis. This review summarizes the current knowledge about the mechanisms of thrombin-mediated proinflammatory responses in cancer pathology for a better understanding and hence a better management of this disease.


Subject(s)
Inflammation Mediators/metabolism , Inflammation/metabolism , Neoplasms/metabolism , Signal Transduction , Thrombin/metabolism , Angiogenic Proteins/metabolism , Animals , Cell Adhesion Molecules , Cell Movement , Cell Proliferation , Cytokines/metabolism , Humans , Inflammation/pathology , Neoplasm Metastasis , Neoplasms/pathology , Neovascularization, Pathologic , Peptide Hydrolases , Receptors, Proteinase-Activated/metabolism
11.
J Cell Physiol ; 232(12): 3221-3225, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28092101

ABSTRACT

WNT/B-CATENIN signaling pathway is one of the key dysregulated pathways in different tumor types, which regulate the expression of several genes involved in cell proliferation, differentiation, and survival. This pathway is being modulated by sex-determining region Y-box (SOX) family genes. The functions of these genes are suggested as tumor suppressor or oncogene. SOX genes transcribe a group of transcription factors that play important roles in embryonic development and carcinogenesis. Among them, SOX15 is recently been identified as a novel tumor suppressor in pancreatic and esophagus cancers with a potential role in modulating Wnt/b-catenin signaling. This report summarizes the current knowledge about Wnt/b-catenin signaling pathway and its cross talk with SOX15 with particular emphasis on the value of SOX gene expression as prognostic or predictive biomarker in cancer.


Subject(s)
Signal Transduction , Animals , Gene Expression Regulation , Humans , Neoplasms/metabolism , SOX Transcription Factors/metabolism , Wnt Signaling Pathway
12.
J Cell Biochem ; 118(8): 1994-1999, 2017 08.
Article in English | MEDLINE | ID: mdl-28135008

ABSTRACT

Interferons (INFs) elicit antiviral responses in tumor cells upon binding to cell surface receptors. Oncolytic virotherapy (OV) is an effective antitumor therapeutic approach which in combination with standard radiotherapy or chemotherapy regimens potentiates treatment responses in cancer patients. However, oncolytic viruses are susceptible to the IFN-induced antiviral state in the tumor microenvironment. A number of studies have, therefore, investigated the effects of combined therapy of IFN signaling pharmacological inhibitors with oncolytic viruses, which result in improved virus replication and oncolysis. This review summarizes the current knowledge of the mechanisms of interferon-mediated tumor resistance to oncolytic virotherapy and provides new insights regarding the effectiveness of combinatorial treatment strategies to attenuate INF-induced OV resistance for greater clinical significance in the treatment of cancer patients. J. Cell. Biochem. 118: 1994-1999, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Gene Expression Regulation, Neoplastic/immunology , Hematopoietic Stem Cells/immunology , Interferons/immunology , Neoplasms/immunology , Oncolytic Virotherapy , Stem Cell Niche/immunology , Bone Marrow/immunology , Bone Marrow/pathology , Chemokine CXCL12/genetics , Chemokine CXCL12/immunology , Dinoprostone/immunology , Dinoprostone/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/immunology , Extracellular Matrix/pathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/immunology , Gene Expression Regulation, Neoplastic/genetics , Hematopoietic Stem Cells/pathology , Humans , Integrins/genetics , Integrins/immunology , Interferons/genetics , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Oncolytic Viruses/growth & development , Oncolytic Viruses/immunology , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , Signal Transduction , Stem Cell Niche/genetics
13.
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
14.
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
15.
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
16.
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
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL