Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Iran J Allergy Asthma Immunol ; 22(2): 183-189, 2023 Apr 30.
Article in English | MEDLINE | ID: mdl-37496411

ABSTRACT

The therapeutic and immunomodulatory potential of mesenchymal stem cells (MSCs) in rheumatoid arthritis (RA) has attracted considerable scientific attention in recent decades. This study aimed to evaluate the expression of genes encoding interleukin (IL)4 and IL10, as well as interferon-gamma (IFNG) and transforming growth factor beta (TGFB1) in refractory RA patients following intravenous injection of autologous bone marrow-derived MSCs (BM-MSCs). This study was registered in Iranian Registry of Clinical Trials (IRCT) (2015102824760N1) and ClinicalTrials.gov (identifier: NCT03333681). Blood samples were taken from 13 patients before and 1 and 6 months after the MSC injection to evaluate the clinical manifestations, paraclinical factors, and expression of IL4, IL10, IFNG, and TGFB1 genes employing the SYBR Green real-time reverse-transcriptase polymerase chain reaction (RT-PCR) technique. There was a significant increase in the expression of TGFB1 at 1 and 6 months after the MSC injection compared to that in the baseline, while the expression of IL4 and IL10 did not change significantly. On the other hand, the expression of IFNG increased significantly after 1 month but decreased significantly at 6 months compared to 1 month after the intervention. Nevertheless, it showed no significant decrease compared to the baseline. A significant decrease was observed for the expression of IFNG 6 months after the injection compared to that after 1 month, which was in concordance with the rise in the expression of the TGFB1 gene. A significant change in the gene expression of TGFB1 and IFNG in our study was consistent with the amelioration of clinical manifestations, suggesting a mechanism of action for MSCs in the treatment of RA.


Subject(s)
Arthritis, Rheumatoid , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/therapy , Arthritis, Rheumatoid/metabolism , Gene Expression , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-4/metabolism , Iran , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Transforming Growth Factor beta/metabolism
2.
Prog Biophys Mol Biol ; 180-181: 19-27, 2023.
Article in English | MEDLINE | ID: mdl-37080435

ABSTRACT

Natural substances are increasingly being used as cancer treatments. Scutellarin, as a flavonoid, recently has been identified in a Chinese herbal extract called Erigeron breviscapus (Vant.). Scutellarin is being researched for its potential benefits due to the discovery that it possesses a variety of biological effects, such as neuroprotective, anti-bacterial, and anti-viral properties. In addition to these biological functions, scutellarin has also been found to have anti-tumor properties. The underlying mechanisms of scutellarin's anticancer activity involve its ability to inhibit various signaling pathways, such as Jak/STAT, ERK/AMPK, and Wnt/ß-catenin. Additionally, scutellarin activates intrinsic and extrinsic apoptotic pathways, which causes the death of tumor cells, interrupts the cell cycle, and promotes its arrest. By limiting metastasis, angiogenesis, drug resistance, and other tumorigenic processes, scutellarin also reduces the aggressiveness of tumors. Despite its promising anticancer activity, scutellarin faces several challenges in its clinical development, including poor solubility, bioavailability, and pharmacokinetic properties. Therefore, it has been suggested that certain modifications can enhance the pharmacogenetic capabilities of scutellarin to decrease its limited water solubility. In conclusion, scutellarin represents a potential candidate for cancer treatment and further studies are needed to explore its clinical utility and optimize its therapeutic potential.


Subject(s)
Neoplasms , Plant Extracts , Signal Transduction , Apigenin/pharmacology , Apigenin/therapeutic use , Medicine, Traditional , Neoplasms/drug therapy
3.
Cell Biol Int ; 46(10): 1557-1570, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35811438

ABSTRACT

A crucial component of the immune system are chemokiness. Chemokine's dysregulation has been linked to a number of pathological diseases. Recently, CXCL17, a chemokine belonging to the CXC subfamily, was identified. With regard to a number of physiological conditions and disorders, CXCL17 either has homeostatic or pathogenic effects. Some research suggests that CXCL17 is an orphan ligand, despite the fact that G protein-coupled receptor (GPR) 35 has been suggested as a possible receptor for CXCL17. Since CXCL17 is primarily secreted by mucosal epithelia, such as those in the digestive and respiratory tracts, under physiological circumstances, this chemokine is referred to as a mucosal chemokine. Macrophages and monocytes are the cells that express GPR35 and hence react to CXCL17. In homeostatic conditions, this chemokine has anti-inflammatory, antibacterial, and chemotactic properties. CXCL17 promotes angiogenesis, metastasis, and cell proliferation in pathologic circumstances like malignancies. However, other studies suggest that CXCL17 may have anti-tumor properties. Additionally, studies have shown that CXCL17 may have a role in conditions such as idiopathic pulmonary fibrosis, multiple sclerosis, asthma, and systemic sclerosis. Additionally, deregulation of CXCL17 in some diseases may serve as a biomarker for diagnosis and prognosis. Clarifying the underlying mechanism of CXCL17's activity in homeostatic and pathological situations may thus increase our understanding of its role and hold promise for the development of novel treatment strategies.


Subject(s)
Chemokines, CXC , Infections , Inflammation , Neoplasms , Chemokines , Chemokines, CXC/physiology , Humans , Infections/immunology , Inflammation/immunology , Neoplasms/immunology , Receptors, G-Protein-Coupled
4.
Front Immunol ; 13: 861931, 2022.
Article in English | MEDLINE | ID: mdl-35677043

ABSTRACT

Chemokines exert crucial roles in inducing immune responses through ligation to their canonical receptors. Besides these receptors, there are other atypical chemokine receptors (ACKR1-4) that can bind to a wide range of chemokines and carry out various functions in the body. ACKR2, due to its ability to bind various CC chemokines, has attracted much attention during the past few years. ACKR2 has been shown to be expressed in different cells, including trophoblasts, myeloid cells, and especially lymphoid endothelial cells. In terms of molecular functions, ACKR2 scavenges various inflammatory chemokines and affects inflammatory microenvironments. In the period of pregnancy and fetal development, ACKR2 plays a pivotal role in maintaining the fetus from inflammatory reactions and inhibiting subsequent abortion. In adults, ACKR2 is thought to be a resolving agent in the body because it scavenges chemokines. This leads to the alleviation of inflammation in different situations, including cardiovascular diseases, autoimmune diseases, neurological disorders, and infections. In cancer, ACKR2 exerts conflicting roles, either tumor-promoting or tumor-suppressing. On the one hand, ACKR2 inhibits the recruitment of tumor-promoting cells and suppresses tumor-promoting inflammation to blockade inflammatory responses that are favorable for tumor growth. In contrast, scavenging chemokines in the tumor microenvironment might lead to disruption in NK cell recruitment to the tumor microenvironment. Other than its involvement in diseases, analyzing the expression of ACKR2 in body fluids and tissues can be used as a biomarker for diseases. In conclusion, this review study has tried to shed more light on the various effects of ACKR2 on different inflammatory conditions.


Subject(s)
Endothelial Cells , Receptors, Chemokine , Chemokines , Endothelial Cells/metabolism , Female , Humans , Inflammation/pathology , Pregnancy , Receptors, CCR10 , Receptors, Chemokine/metabolism
5.
Clin Exp Hypertens ; 44(6): 539-547, 2022 Aug 18.
Article in English | MEDLINE | ID: mdl-35722928

ABSTRACT

Hypothyroidism disturbs redox homeostasis and takes part in cardiovascular system dysfunction. Considering antioxidant and cardio-protective effects of PPAR-γ agonists including pioglitazone (POG) and rosiglitazone (RSG), the present study was aimed to determine the effect of POG or RSG on oxidants and antioxidants indexes in the heart and aorta tissues of Propylthiouracil (PTU)-induced hypothyroid rats. MATERIALS AND METHODS: The animals were divided into six groups: (1) Control; (2) propylthiouracil (PTU), (3) PTU-POG 10, (4) PTU-POG 20, (5) PTU-RSG 2, and (6) PTU-RSG 4. Hypothyroidism was induced in rats by giving 0.05% propylthiouracil (PTU) in drinking water for 42 days. The rats of PTU-POG 10 and PTU-POG 20 groups received 10 and 20 mg/kg POG, respectively, besides PTU, and the rats of PTU-RSG 2 and PTU-RSG 4 groups received 2 and 4 mg/kg RSG, respectively, besides PTU. The animals were sacrificed, and the serum of the rats was collected to measure thyroxine level. The heart and aorta tissues were also removed for the measurement of biochemical oxidative stress markers. RESULTS: Hypothyroidism was induced by PTU administration, which was indicated by lower serum thyroxine levels. Hypothyroidism also was accompanied by a decrease of catalase (CAT), superoxide dismutase (SOD) activities, and thiol concentration in the heart and aorta tissues while increased level of malondialdehyde (MDA). Interestingly, administration of POG or RSG dramatically reduced oxidative damage in the heart and aorta, as reflected by a decrease in MDA and increased activities of SOD, CAT, and thiol content. CONCLUSION: The results of this study showed that administration of POG or RSG decreased oxidative damage in the heart and aorta tissues induced by hypothyroidism in rats.


Subject(s)
Hypothyroidism , Thyroxine , Animals , Antioxidants/pharmacology , Heart , Hypothyroidism/chemically induced , Hypothyroidism/complications , Hypothyroidism/drug therapy , Oxidative Stress , PPAR gamma , Pioglitazone/pharmacology , Propylthiouracil/adverse effects , Rats , Rats, Wistar , Sulfhydryl Compounds , Superoxide Dismutase/metabolism , Thyroxine/adverse effects
6.
Rep Biochem Mol Biol ; 11(1): 10-19, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35765531

ABSTRACT

Background: inflammatory chemokines such as CCL2 and CCL5 are involved in the progress of osteoarthritis. Crocin with antioxidant and anti-inflammatory properties can reduce the symptoms of osteoarthritis (OA). This study was performed investigate the effect of Krocina™, on the gene expressions and plasma levels of CCL2 and CCL5 in OA patients. Methods: The study included 35 patients that were randomized in the Krocina™ and placebo groups. The intervention was Krocina™ 15mg daily for four months. Clinical and paraclinical parameters were measured. CCL2 and CCL5 genes expression and plasma levels were determined using the SYBR Green Real-Time RT-PCR and Enzyme-linked Immunosorbent Assay (ELISA) techniques. Results: The C-reactive protein (CRP) value in the Krocina™ group and the visual analogue scale (VAS) value in the Krocina™ and placebo groups decreased significantly after the intervention. The gene expression of CCL2 in the Krocina™ and placebo groups decreased significantly. On the contrary, the gene expression of CCL5 in the Krocina™ and placebo groups increased significantly. Moreover, the plasma levels of CCL2 in the Krocina™ and placebo groups decreased meaningfully. There was no difference regarding the plasma levels of CCL5 within the Krocina™ and placebo groups before and after the intervention in either of the groups. Conclusion: Administration of Krocina™ reduced the clinical signs of inflammation and CRP and VAS value. Also, Krocina™ significantly decreased the plasma levels and gene expression of CCL2 in osteoarthritis patients.

7.
Physiol Rep ; 10(7): e15243, 2022 04.
Article in English | MEDLINE | ID: mdl-35373925

ABSTRACT

Viral infections are linked to the progression of inflammatory reactions and oxidative stress that play pivotal roles in systemic diseases. To confirm this phenomenon, in the present study, TNF-α level and oxidative stress markers were examined in the liver, kidney, and pancreas of HTLV1-infected male BALB/c mice. To this end, twenty BALB/c mice were divided into HTLV1-infected mice that were inoculated with 1-million HTLV1-infected cells (MT-2), and the control groups. Two months after inoculation, the peripheral blood, mesenteric lymph nodes, liver, kidney, and pancreas were collected after deep anesthetization of mice (ketamine, 30 mg/kg). The extracted DNA of mesenteric lymph nodes was obtained to quantify proviral load (PVL) using quantitative real-time polymerase chain reaction (qRT-PCR). The levels of lipid peroxidation, total thiol (SH), nitric oxide (NO), TNF-α, catalase (CAT), and superoxide dismutase (SOD) activities were examined in the liver, kidney, and pancreases. Furthermore, histopathological changes in the liver and kidney were evaluated. In liver tissue, the levels of MDA, TNF-α, and blood cell infiltration were significantly increased, and the levels of CAT and SOD were significantly decreased. In the kidney, a reduction in SOD, CAT, and total SH and an increase in MDA and NO were observed. In the pancreas, CAT activity, total SH, and SOD were decreased, and the levels of MDA and NO were enhanced. In terms of TNF-α production, it has been shown that the level of this inflammatory cytokine was increased in the liver, kidney, and pancreas. The HTLV1 may have a role in inducing inflammatory reactions and oxidative stress pathways in the tissues.


Subject(s)
Pancrelipase , Superoxide Dismutase , Animals , Kidney/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Oxidative Stress , Pancreas/metabolism , Pancrelipase/metabolism , Pancrelipase/pharmacology , Superoxide Dismutase/metabolism
8.
Clin Exp Immunol ; 207(2): 164-175, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35020885

ABSTRACT

Cancer is considered a life-threatening disease, and several factors are involved in its development. Chemokines are small proteins that physiologically exert pivotal roles in lymphoid and non-lymphoid tissues. The imbalance or dysregulation of chemokines has contributed to the development of several diseases, especially cancer. CCL19 is one of the homeostatic chemokines that is abundantly expressed in the thymus and lymph nodes. This chemokine, which primarily regulates immune cell trafficking, is involved in cancer development. Through the induction of anti-tumor immune responses and inhibition of angiogenesis, CCL19 exerts tumor-suppressive functions. In contrast, CCL19 also acts as a tumor-supportive factor by inducing inflammation, cell growth, and metastasis. Moreover, CCL19 dysregulation in several cancers, including colorectal, breast, pancreatic, and lung cancers, has been considered a tumor biomarker for diagnosis and prognosis. Using CCL19-based therapeutic approaches has also been proposed to overcome cancer development. This review will shed more light on the multifarious function of CCL19 in cancer and elucidate its application in diagnosis, prognosis, and even therapy. It is expected that the study of CCL19 in cancer might be promising to broaden our knowledge of cancer development and might introduce novel approaches in cancer management.


Subject(s)
Lung Neoplasms , Lymph Nodes , Chemokine CCL19/metabolism , Chemokines/metabolism , Humans , Lung Neoplasms/metabolism , Neovascularization, Pathologic , Prognosis , Receptors, CCR7/metabolism
9.
Biotechnol Appl Biochem ; 69(1): 61-69, 2022 Feb.
Article in English | MEDLINE | ID: mdl-33289168

ABSTRACT

The immune checkpoint molecules are involved in the regulation of T cells in order to prevent them from attacking to sell tissues and play a role in the immune response homeostasis. Application of the immune checkpoint inhibitors (ICIs) has provided a promising therapeutic approach in pathologies where the immune system is suppressed. The extended utilization of ICIs in several cancers has caused immune-related side effects in the cardiovascular system like cardiomyopathy and myocarditis. Cardiac toxicity, one of the main side effects of the ICIs-based therapeutic approach, has less been concerned; however, during the last years, many cases of fatal heart failure and myocarditis have been reported in patients treated with ICIs. In this review article, we attempted to discuss the cardiac adverse effects of inhibiting different immune checkpoint molecules.


Subject(s)
Myocarditis , Neoplasms , Cardiotoxicity/etiology , Humans , Immune Checkpoint Inhibitors , Immunotherapy/adverse effects , Myocarditis/chemically induced , Neoplasms/drug therapy
10.
Immunol Invest ; 51(4): 1060-1086, 2022 May.
Article in English | MEDLINE | ID: mdl-33522348

ABSTRACT

Interleukin (IL)-33, a member of IL-1 cytokine family, is produced by various immune cells and acts as an alarm to alert the immune system after epithelial or endothelial cell damage during cell necrosis, infection, stress, and trauma. The biological functions of IL-33 largely depend on its ligation to the corresponding receptor, suppression of tumorigenicity 2 (ST2). The pathogenic roles of this cytokine have been implicated in several disorders, including allergic disease, cardiovascular disease, autoimmune disease, infectious disease, and cancers. However, alerted levels of IL-33 may result in either disease amelioration or progression. Genetic variations of IL33 gene may confer protective or susceptibility risk in the onset of autoimmune diseases. The purpose of this review is to discuss the involvement of IL-33 and ST2 in the pathogenesis of a variety of autoimmune disorders, such as autoimmune rheumatic, neurodegenerative, and endocrine diseases.


Subject(s)
Autoimmune Diseases , Interleukin-33 , Autoimmune Diseases/genetics , Autoimmunity/genetics , Cytokines , Humans , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/genetics , Signal Transduction
11.
Immunology ; 165(1): 44-60, 2022 01.
Article in English | MEDLINE | ID: mdl-34716913

ABSTRACT

Cytokines are considered vital mediators of the immune system. Down- or upregulation of these mediators is linked to several inflammatory and pathologic situations. IL-26 is referred to as an identified member of the IL-10 family and IL-20 subfamily. Due to having a unique cationic structure, IL-26 exerts diverse functions in several diseases. Since IL-26 is mainly secreted from Th17, it is primarily considered a pro-inflammatory cytokine. Upon binding to its receptor complex (IL-10R1/IL-20R2), IL-26 activates multiple signalling mediators, especially STAT1/STAT3. In cancer, IL-26 induces IL-22-producing cells, which consequently decrease cytotoxic T-cell functions and promote tumour growth through activating anti-apoptotic proteins. In hypersensitivity conditions such as rheumatoid arthritis, multiple sclerosis, psoriasis and allergic disease, this cytokine functions primarily as the disease-promoting mediator and might be considered a biomarker for disease prognosis. Although IL-26 exerts antimicrobial function in infections such as hepatitis, tuberculosis and leprosy, it has also been shown that IL-26 might be involved in the pathogenesis and exacerbation of sepsis. Besides, the involvement of IL-26 has been confirmed in other conditions, including graft-versus-host disease and chronic obstructive pulmonary disease. Therefore, due to the multifarious function of this cytokine, it is proposed that the underlying mechanism regarding IL-26 function should be elucidated. Collectively, it is hoped that the examination of IL-26 in several contexts might be promising in predicting disease prognosis and might introduce novel approaches in the treatment of various diseases.


Subject(s)
Disease Susceptibility , Interleukins/genetics , Interleukins/metabolism , Animals , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation , Humans , Infections/etiology , Infections/metabolism , Infections/pathology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Interleukins/chemistry , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Organ Specificity/genetics , Organ Specificity/immunology , Protein Transport , Signal Transduction , Structure-Activity Relationship
12.
J Complement Integr Med ; 19(2): 311-321, 2022 Jun 01.
Article in English | MEDLINE | ID: mdl-34506695

ABSTRACT

OBJECTIVES: Many diabetes-related complications are caused by oxidative stress. In the current study, the protective effect of Cinnamomum cassia against diabetes-induced liver and kidney oxidative stress was evaluated. METHODS: The male Wistar rats (n=48) were randomly divided into six groups including; control group received 500 µL normal saline orally for 42 days. Diabetes groups received intraperitoneally (i.p.) streptozotocin (STZ) as single-dose (60 mg/kg, i.p.). Cinnamon extract (100, 200, 400 mg/kg) and metformin (300 mg/kg) were orally administered to diabetic rats for 42 days. After the experiment period, the animals were anesthetized and the liver and kidney tissues were quickly removed and restored for oxidative stress evaluation. The levels of malondialdehyde (MDA), total thiol content, glutathione (GSH), nitric oxide (NO) metabolites, as well as, superoxide dismutase (SOD) and catalase (CAT) activities were measured in kidney and liver tissue. RESULTS: The level of MDA, SOD, and CAT activities increased significantly, while the total thiol content, and NO production were significantly reduced in diabetic animals compared to the control group (from p<0.05 to p<0.001). Treatment with cinnamon extract significantly decreased the MDA level, as well as, SOD and CAT activities in the liver and kidney of diabetic rats (from p<0.05 to p<0.001). In the liver and kidney of cinnamon treated groups, GSH and total thiol contents and NO production were significantly higher than diabetic group (from p<0.05 to p<0.001). CONCLUSIONS: Cinnamon extract due to its potent antioxidant property could be effective in decrease of diabetes-induced oxidative stress that plays a major role in renal and hepatic complications.


Subject(s)
Cinnamomum aromaticum , Diabetes Mellitus, Experimental , Animals , Antioxidants/metabolism , Cinnamomum aromaticum/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Glutathione/metabolism , Kidney , Lipid Peroxidation , Liver , Male , Nitric Oxide/metabolism , Oxidative Stress , Plant Extracts/metabolism , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Rats , Rats, Wistar , Streptozocin/metabolism , Streptozocin/pharmacology , Streptozocin/therapeutic use , Sulfhydryl Compounds/metabolism , Sulfhydryl Compounds/pharmacology , Sulfhydryl Compounds/therapeutic use , Superoxide Dismutase/metabolism
13.
Cancer Med ; 10(15): 5191-5202, 2021 08.
Article in English | MEDLINE | ID: mdl-34128588

ABSTRACT

Cancer is a leading cause of death which imposes a substantial financial burden. Among the several mechanisms involved in cancer progression, imbalance of immune cell-derived factors such as cytokines and chemokines plays a central role. IL-25, as a member of the IL-17 cytokine subfamily, exerts a paradoxical role in cancer, including tumor supportive and tumor suppressive. Hence, we have tried to clarify the role of IL-25 and its receptor in tumor progression and cancer prognosis. It has been confirmed that IL-25 exerts a tumor-suppressive role through inducing infiltration of eosinophils and B cells into the tumor microenvironment and activating the apoptotic pathways. In contrast, the tumor-supportive function has been implemented by activating inflammatory cascades, promoting cell cycle, and inducing type-2 immune responses. Since IL-25 has been dysregulated in tumor tissues and this dysregulation is involved in cancer development, its examination can be used as a tumor diagnostic and prognostic biomarker. Moreover, IL-25-based therapeutic approaches have shown promising results in cancer inhibition. In cancers in which IL-25 has a tumor-suppressive function, employing IL-25-enhancing approaches, such as Virulizin® and dihydrobenzofuran administration, has potentially inhibited tumor cell growth. On the other hand, in the case of IL-25-dependent tumor progression, using IL-25 blocking methods, including anti-IL-25 antibodies, might be a complementary approach to the other anticancer agent. Collectively, it is hoped, IL-25 might be a promising target in cancer treatment.


Subject(s)
Interleukin-17/physiology , Neoplasms , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/physiology , Benzofurans/therapeutic use , Bile , Biomarkers, Tumor/physiology , Breast Neoplasms/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Cycle/physiology , Cell Proliferation/drug effects , Disease Progression , Female , Humans , Immunity, Cellular , Interleukin-17/antagonists & inhibitors , Interleukin-17/metabolism , Liver Neoplasms/metabolism , Male , Mice , Neoplasms/etiology , Neoplasms/immunology , Neoplasms/mortality , Neoplasms/therapy , Prognosis , Prostatic Neoplasms/metabolism , Receptors, Interleukin-17/metabolism , Receptors, Interleukin-17/physiology , Signal Transduction/physiology , Tissue Extracts/therapeutic use , Tumor Microenvironment/immunology
14.
Int Immunopharmacol ; 97: 107681, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33932697

ABSTRACT

Cancer is a leading cause of death worldwide and imposes a substantial financial burden. Therefore, it is essential to develop cost-effective approaches to inhibit tumor growth and development. The imbalance of cytokines and chemokines play an important role among different mechanisms involved in cancer development. One of the strongly conserved chemokines that is constitutively expressed in skin epithelia is the chemokine CXCL14. As a member of the CXC subfamily of chemokines, CXCL14 is responsible for the infiltration of immune cells, maturation of dendritic cells, upregulation of major histocompatibility complex (MHC)-I expression, and cell mobilization. Moreover, dysregulation of CXCL14 in several cancers has been identified by several studies. Depending on the type or origin of the tumor and components of the tumor microenvironment, CXCL14 plays a conflicting role in cancer. Although fibroblast-derived CXCL14 has a tumor-supportive role, epithelial-derived CXCL14 mainly inhibits tumor progression. Hence, this review will elucidate what is known on the mechanisms of CXCL14 and its therapeutic approaches in tumor treatment. CXCL14 is a promising approach for cancer immunotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Chemokines, CXC/metabolism , Neoplasms/immunology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/agonists , Biomarkers, Tumor/analysis , Biomarkers, Tumor/antagonists & inhibitors , Chemokines, CXC/agonists , Chemokines, CXC/analysis , Chemokines, CXC/antagonists & inhibitors , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/genetics , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Up-Regulation
15.
Expert Rev Mol Med ; 23: e4, 2021 04 21.
Article in English | MEDLINE | ID: mdl-33880989

ABSTRACT

Today, cancer is one of the leading causes of death worldwide. Lately, cytokine and chemokine imbalances have gained attention amongst different involved pathways in cancer development and attracted much consideration in cancer research. CXCL16, as a member of the CXC subgroup of chemokines, has been attributed to be responsible for immune cell infiltration into the tumour microenvironment. The aberrant expression of CXCL16 has been observed in various cancers. This chemokine has been shown to play a conflicting role in tumour development through inducing pro-inflammatory conditions. The infiltration of various immune and non-immune cells such as lymphocytes, cancer-associated fibroblasts and myeloid-derived suppressor cells by CXCL16 into the tumour microenvironment has complicated the tumour fate. Given this diverse role of CXCL16 in cancer, a better understanding of its function might build-up our knowledge about tumour biology. Hence, this study aimed to review the impact of CXCL16 in cancer and explored its therapeutic application. Consideration of these findings might provide opportunities to achieve novel approaches in cancer treatment and its prognosis.


Subject(s)
Chemokines, CXC , Neoplasms , Animals , Chemokine CXCL16 , Chemokines, CXC/genetics , Humans , Neoplasms/genetics , Receptors, CXCR6 , Receptors, Chemokine , Receptors, Scavenger , Receptors, Virus , Tumor Microenvironment
16.
Prog Biophys Mol Biol ; 164: 63-71, 2021 09.
Article in English | MEDLINE | ID: mdl-33894206

ABSTRACT

Despite significant signs of progress in cancer treatment over the past decade, either cancer prevalence or mortality continuously grow worldwide. Current anti-cancer agents show insignificant effectiveness, followed by serious side effects. It is important to find new, highly efficient pharmacological agents to increase cancer patients' clinical outcomes. Curcumin, a polyphenolic compound, has gained growing attention because of its anti-cancer properties. Curcumin can hinder the development, migration, and metastasis of cancer cells. The anti-cancer effects of curcumin are principally attributed to the regulation of several cellular signaling pathways, including MAPK/PI3K/Akt, Wnt/ß-catenin, JAK/STAT, and NF-ĸB signaling pathways. Furthermore, curcumin can affect the expression and function of tumor-suppressive and oncogenic long non-coding RNAs (lncRNAs). In this study, we briefly reviewed the modulatory effect of curcumin on dysregulated tumor-supportive and tumor-suppressive lncRNAs in several cancers. It is hoped that a better understanding of curcumin's anti-cancer properties would pave the way for the development of a therapeutic approach in cancer.


Subject(s)
Antineoplastic Agents , Curcumin , Neoplasms , RNA, Long Noncoding , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Curcumin/pharmacology , Curcumin/therapeutic use , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/therapeutic use , RNA, Long Noncoding/genetics , RNA, Long Noncoding/therapeutic use
17.
Int J Rheum Dis ; 24(4): 567-581, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33550689

ABSTRACT

BACKGROUND: Genetic polymorphisms in the endoplasmic reticulum aminopeptidase gene ERAP2 has been attributed with the etiopathogenesis of ankylosing spondylitis (AS). Here we assessed the association of ERAP2 gene single nucleotide polymorphisms (SNPs) with AS predisposition in Iranian patients and determined their effect on the inflammatory state of the patients. METHODS: For genotyping of rs2548538, rs2287988, and rs17408150 SNPs using a real-time allelic discrimination approach, DNA was extracted from the whole blood of 250 AS patients and 250 healthy individuals. RNA of the peripheral blood mononuclear cells was separated, cDNA was synthesized, and transcriptional levels of cytokines, including interleukin (IL)-17A, IL-23, IL-10, and transforming growth factor-ß, were measured. Enzyme-linked immunosorbent assay was used to measure the serum concentration on the cytokines. RESULTS: Three ERAP2 gene SNPs were not associated significantly with AS risk. Nonetheless, rs2287988 and rs17408150 SNPs showed statistically significant association with susceptibility to the disease in those AS patients who were positive for human leukocyte antigen (HLA)-B27. Transcriptional level and serum concentration of IL-17A and IL-23 were higher, but those of IL-10 were lower in both AS patients and the HLA-B27-positive patient group relative to the control group. Nevertheless, ERAP2 gene SNPs in the HLA-B27-positive AS patients did not affect the transcription level and serum concentration of cytokines. CONCLUSIONS: ERAP2 gene rs2287988 and rs17408150 SNPs are associated with susceptibility to AS, but they are probably not determining the levels of IL-17A, IL-23, and IL-10 in this disease.


Subject(s)
Aminopeptidases/genetics , Polymorphism, Single Nucleotide , Spondylitis, Ankylosing/genetics , Adult , Biomarkers/blood , Case-Control Studies , Cytokines/blood , Cytokines/genetics , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Inflammation Mediators/blood , Iran , Male , Middle Aged , Phenotype , Risk Assessment , Risk Factors , Spondylitis, Ankylosing/diagnosis , Spondylitis, Ankylosing/immunology
18.
IUBMB Life ; 73(4): 659-669, 2021 04.
Article in English | MEDLINE | ID: mdl-33625758

ABSTRACT

One of the most common malignant tumors is hepatocellular carcinoma (HCC). Progression of HCC mainly results from highly complex molecular and pathological pathways. Midkine (MDK) is a growth factor that impacts viability, migration, and other cell activities. Since MDK has been involved in the inflammatory responses, it has been claimed that MDK has a crucial role in HCC. MDK acts as an anti-apoptotic factor, which mediates tumor cell viability. In addition, MDK blocks anoikis to promote metastasis. There is also evidence that MDK is involved in angiogenesis. It has been shown that the application of anti-MDK approaches might be promising in the treatment of HCC. Besides, due to the elevated expression in HCC, MDK has been proposed as a biomarker in the prognosis and diagnosis of HCC. In this review, we will discuss the role of MDK in HCC. It is hoped that the development of new strategies concerning MDK-based therapies will be promising in HCC management.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Midkine/physiology , Animals , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/pathology , Genetic Therapy/methods , Humans , Immunotherapy/methods , Liver/blood supply , Liver/pathology , Liver Neoplasms/diagnosis , Liver Neoplasms/pathology , Midkine/blood , Midkine/chemistry , Neovascularization, Pathologic/metabolism , RNA Interference
19.
Gene ; 732: 144336, 2020 Mar 30.
Article in English | MEDLINE | ID: mdl-31935514

ABSTRACT

In the present study, we aimed to evaluate effects of autologous mesenchymal stem cells (MSCs) intravenous administration on the response of B cells, BAFF, APRIL, and their receptors on the surface of B cells at 1, 6, and 12 month follow-up periods in refractory rheumatoid arthritis (RA) patients. Thirteen patients with refractory RA received autologous MSCs. Plasma levels of BAFF and APRIL were measured employing ELISA method, followed by estimating B cell population and BAFFRs evaluation by flow cytometry technique. Gene expression of BAFF, APRIL, and their receptors on B cell surface in PBMCs was evaluated by SYBR Green real-time PCR technique. Plasma concentration of BAFF significantly decreased 1 and 6 months after the MSCT (MSCs Transplantation). Plasma concentration of APRIL significantly decreased 1 month after the MSCT. Percentages of CD19 + B cells in the PBMC population significantly decreased 12 months after the MSCT. Percentages of BR3 + CD19 + B cells and BCMA + CD19 + B cells significantly decreased at the 12th month after the MSCT. The gene expression of BAFF in the PBMC population significantly decreased during 6, and 12 months after the MSCT. The gene expression of APRIL significantly decreased on month 6 after the MSCT. The gene expression of BR3 significantly decreased during 1, 6, and 12 months after the MSCT. The MSCT seems to decrease B cells response because of the reduced production of BAFF and APRIL cytokines and decrease the expression of their receptors on the surface of B cells.


Subject(s)
Arthritis, Rheumatoid/therapy , B-Cell Activating Factor/metabolism , B-Cell Activation Factor Receptor/metabolism , Down-Regulation , Mesenchymal Stem Cell Transplantation/methods , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Administration, Intravenous , Adult , Antigens, CD19/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , B-Cell Activating Factor/genetics , B-Cell Activation Factor Receptor/genetics , B-Lymphocytes/immunology , Female , Gene Expression Profiling , Humans , Middle Aged , Treatment Outcome , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics
20.
Rep Biochem Mol Biol ; 8(1): 9-14, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31334281

ABSTRACT

BACKGROUND: Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder characterized by altered bowel habits and abdominal pain in the absence of a recognizable structural anomaly. The pathogenesis of IBS has been associated with inflammation and the expression of pro-inflammatory chemokines, such as CCL2 and CCL16. Our study aimed to investigate the relationship between the serum levels of CCL2 and CCL16 and IBS. Additionally, we examined how serum levels of these chemokines relate to IBS subtypes. METHODS: Patients with IBS diagnosed according to the Rome III criteria participated in this study (n= 96). Healthy individuals with no history of allergic, autoimmune, chronic or active gastrointestinal infectious diseases were used as controls (n= 44). The serum levels of CCL2 and CCL16 was measured via enzyme-linked immunosorbent assay (ELISA). RESULTS: A significant decrease in the serum levels of CCL16 and CCL2 was observed in the patients with IBS. Additionally, the serum levels of CCL16 in IBS patients with diarrhea (D-IBS) was significantly higher than those with the mixed IBS (M-IBS) subtype. CONCLUSION: The significant increase in the serum levels of CCL-16 in patients with D-IBS compared to patients with M-IBS suggests that CCL-16 may be used as an immunological biomarker to differentiate between these two subtypes.

SELECTION OF CITATIONS
SEARCH DETAIL
...