Moureq R. Alotaibi
King Saud University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Moureq R. Alotaibi.
Molecular Immunology | 2017
Sheikh F. Ahmad; Ahmed Nadeem; Mushtaq Ahmad Ansari; Saleh A. Bakheet; Sabry M. Attia; Khairy M.A. Zoheir; Laila Al-Ayadhi; Mohammad Zeed Alzahrani; Abdulaziz M.S. Alsaad; Moureq R. Alotaibi; Adel R.A. Abd-Allah
HighlightsEnhanced pro‐inflammatory cytokine production (IL‐21/IL‐22) on PBMC in AU.Decreased anti‐inflammatory molecules (IL‐27/CTLA‐4) on PBMC in AU.Induction of IL‐21/IL‐22 protein and mRNA expression in AU children.Decreased IL‐27 mRNA expression in children with AU. ABSTRACT Accumulating evidence suggests an association between immune dysfunction and autism disorders in a significant subset of children. In addition, an imbalance between pro‐ and anti‐inflammatory pathways has been proposed to play an important role in the pathogenesis of several neurodevelopmental disorders including autism; however, the role of anti‐inflammatory molecules IL‐27 and CTLA‐4 and pro‐inflammatory cytokines IL‐21 and IL‐22 has not previously been explored in autistic children. In the current study, we investigated the expression of IL‐21, IL‐22, IL‐27, and CD152 (CTLA‐4) following an in‐vitro immunological challenge of peripheral blood mononuclear cells (PBMCs) from children with autism (AU) or typically‐developing children (TD) with phorbol‐12‐myristate 13‐acetate (PMA) and ionomycin. In our study, cells from children with AU had increased IL‐21 and IL‐22 and decreased CTLA‐4 expression on CD4+ T cells as compared with cells from the TD control. Similarly, AU cells showed decreased IL‐27 production by CD14+ cells compared to that of TD control cells. These results were confirmed by real‐time PCR and western blot analyses. Our study shows dysregulation of the immune balance in cells from autistic children as depicted by enhanced pro‐inflammatory cytokines, ‘IL‐21/IL‐22’ and decreased anti‐inflammatory molecules, ‘IL‐27/CTLA‐4’. Thus, further study of this immune imbalance in autistic children is warranted in order to facilitate development of biomarkers and therapeutics.
Immunobiology | 2017
Naif O. Al-Harbi; Ahmed Nadeem; Mohammed M. Al-Harbi; Khairy M.A. Zoheir; Mushtaq Ahmad Ansari; Ahmed M. El-Sherbeeny; Khalid Mashay Al-Anazi; Moureq R. Alotaibi; Sheikh F. Ahmad
Psoriatic inflammation has been shown to be associated with cardiovascular dysfunction and systemic inflammation. Recently, psoriasis has also been linked to hepatic disorders, however underlying mechanism connecting the two are unknown. IL-17A being a central pro-inflammatory cytokine in the pathogenesis of psoriasis may be involved in hepatic inflammation through its receptor and downward signaling; however so far no study has investigated IL-17A related signaling in the liver during psoriasis in a murine model. Therefore, this study explored psoriasis-induced hepatic inflammation and concurrent metabolic changes. Mice were applied topically imiquimod (IMQ) to develop psoriatic inflammation. Additionally mice were also treated either with IL-17A or anti-IL17A antibody to explore the role of IL-17 related signaling in liver. Mice were then assessed for hepatic inflammation through assessment of inflammatory/oxidative stress markers (IL-17RC, NFκB, IL-6, MCP-1, IL-1β, GM-CSF, ICAM-1, iNOS, lipid peroxides and myeloperoxidase activity) as well as hepatic injury (alanine aminotransferase, aspartate aminotransferase and alkaline phosphatase) and protein/lipid metabolic biomarkers (total proteins, albumin, total bilirubin, triglycerides, HDL cholesterol, and total cholesterol). IMQ treatment led to hepatic inflammation as evidenced by increased pro-inflammatory cytokines and oxidative stress with concomitant dysregulation in hepatic protein/lipid metabolism. Treatment with IL-17A further aggravated, whereas treatment with anti-IL17A antibody ameliorated IMQ-induced changes in hepatic injury/inflammation and protein/lipid metabolism. Our study shows for the first time that psoriatic inflammation leads to hepatic inflammation which results in dysregulated protein/lipid metabolism through IL-17RC/NFκB signaling. This could result in increased risk of cardiovascular dysfunction in patients with psoriasis.
International Immunopharmacology | 2017
Naif O. Al-Harbi; Ahmed Nadeem; Mushtaq Ahmad Ansari; Mohammed M. Al-Harbi; Moureq R. Alotaibi; Abdulaziz M.S. Alsaad; Sheikh F. Ahmad
Abstract Psoriatic patients have systemic inflammation as well as oxidative stress, which are associated with cardiovascular disorders such as atherosclerosis, hypertension myocardial infarction, and stroke. Psoriasis has also been shown to be associated with kidney disease in several studies. Both disorders also have strong component of oxidative stress which usually emanates from NADPH oxidases (NOXs) and inducible nitric oxide synthase (iNOS). However, whether psoriatic inflammation leads to renal oxidative stress and dysfunction remains unexplored. Therefore, this study investigated the effect of imiquimod (IMQ)‐induced psoriatic inflammation on kidney function and inflammation in a murine model. Mice were topically applied IMQ followed by various analyses in kidney/blood related to inflammation and kidney function. Psoriatic inflammation in mice was associated with kidney dysfunction as reflected by increased serum creatinine and blood urea nitrogen. Kidney dysfunction was paralleled by upregulation of ROS generating enzymes such as NOX2, NOX4 and iNOS with concomitant oxidative stress. Treatment either with general antioxidant, N‐acetyl cysteine or NOX/iNOS inhibitors led to improvement of IMQ‐induced renal dysfunction and oxidative stress. On the contrary, buthionine sulfoximine, oxidant inducer further aggravated IMQ‐induced renal impairment and oxidant‐antioxidant imbalance. Our data suggest that psoriatic inflammation causes kidney dysfunction where NOXs and iNOS play important roles. Treatment with antioxidants may be considered as adjunct therapy in psoriatic patients with kidney disease. HighlightsPsoriatic inflammation causes kidney dysfunction in mice.Psoriatic inflammation causes renal oxidative inflammation.Diphenyliodonium and 1400W attenuate psoriasis related kidney dysfunction.N‐acetyl cysteine attenuates psoriasis related kidney dysfunction.Buthionine sulfoximine aggravates psoriasis related kidney dysfunction.
Asian Pacific Journal of Cancer Prevention | 2016
Othman A. Al-Shabanah; Moureq R. Alotaibi; Salim S. Al Rejaie; Ali Alhoshani; Mashal M. Almutairi; Musaad A. Alshammari; Mohamed M. Hafez
Objective: Breast cancer is global female health problem worldwide. Most of the currently used agents for breast cancer treatment have toxic side-effects. Ginseng root, an oriental medicine, has many health benefits and may exhibit direct anti-cancer properties. This study was performed to assess the effects of ginseng on breast cancer cell lines. Materials and Methods: Cytotoxicity of ginseng extract was measured by MTT assay after exposure of MDA-MB-231, MCF-10A and MCF-7 breast cancer cells to concentrations of 0.25, 0.5, 1, 1.5, 2 and 2.5 mg/well. Expression levels of p21WAF, p16INK4A, Bcl-2, Bax and P53 genes were analyzed by quantitative real time PCR. Results: The treatment resulted in inhibition of cell proliferation in a dose-and time-dependent manner. p53, p21WAF1and p16INK4A expression levels were up-regulated in ginseng treated MDA-MB-231 and MCF-7 cancer cells compared to untreated controls and in MCF-10A cells. The expression levels of Bcl2 in the MDA-MB-231 and MCF-7 cells were down-regulated. In contrast, that of Bax was significantly up-regulated. Conclusion: The results of this study revealed that ginseng may inhibit breast cancer cell growth by activation of the apoptotic pathway.
Toxicology and Applied Pharmacology | 2018
Sabry M. Attia; M.A. Al-Hamamah; Moureq R. Alotaibi; Gamaleldin I. Harisa; Mohammed M. Attia; Sheikh F. Ahmad; Mushtaq Ahmad Ansari; Ahmed Nadeem; Saleh A. Bakheet
ABSTRACT Histone deacetylases (HDACs), which regulate transcription and specific functions such as tumor suppression by p53, are frequently altered in tumors and have a contentious role in carcinogenesis. HDAC inhibitors, which have a long history of use in psychiatry and neurology, have recently been tested as possible treatments for tumors. Belinostat received regulatory approval in the USA on July 3, 2014, for use against peripheral T‐cell lymphoma. However, the unavailability of information on belinostat genotoxicity in normal cells and the molecular mechanisms involved in the genetic instability after exposure to belinostat encouraged us to conduct this study. Our data showed that the exposure of mice to belinostat at the recommended human doses induced chromosome breakage, whole‐chromosome lagging, and oxidative DNA damage in bone marrow cells in a dose‐dependent manner. The expression levels of 84 genes involved in the DNA damage signaling pathway were evaluated by using an RT2 Profiler PCR array. Belinostat exposure altered the expression of 25 genes, with statistically significant changes observed in 17 genes. The array results were supported by RT‐PCR and western blotting experiments. Collectively, our results showed that belinostat exposure caused oxidative DNA damage and downregulated the expression of genes involved in DNA damage repair, which may be responsible for belinostat‐induced genomic instability. Thus, the clinical usage of this drug should be weighed against the hazards of carcinogenesis, and the observed genotoxicity profile of belinostat may support further development of efficient HDAC inhibitors with weaker genotoxicity. HighlightsBelinostat was a genotoxic at the recommended human doses.Belinostat‐induced oxidative genomic damageBelinostat downregulated the expression of genes involved in DNA damage pathway.Its clinical usage should be weighed against the hazards of carcinogenesis.
Radiation Research | 2018
Jingwen Xu; Nipa Patel; Tareq Saleh; Emmanuel K. Cudjoe; Moureq R. Alotaibi; Yingliang Wu; Santiago Lima; Adam M. Hawkridge; David A. Gewirtz
Studies of radiation interaction with tumor cells often focus on apoptosis as an end point; however, clinically relevant doses of radiation also promote autophagy and senescence. Moreover, functional p53 has frequently been implicated in contributing to radiation sensitivity through the facilitation of apoptosis. To address the involvement of apoptosis, autophagy, senescence and p53 status in the response to radiation, the current studies utilized isogenic H460 non-small cell lung cancer cells that were either p53-wild type (H460wt) or null (H460crp53). As anticipated, radiosensitivity was higher in the H460wt cells than in the H460crp53 cell line; however, this differential radiation sensitivity did not appear to be a consequence of apoptosis. Furthermore, radiosensitivity did not appear to be reduced in association with the promotion of autophagy, as autophagy was markedly higher in the H460wt cells. Despite radiosensitization by chloroquine in the H460wt cells, the radiation-induced autophagy proved to be essentially nonprotective, as inhibition of autophagy via 3-methyl adenine (3-MA), bafilomycin A1 or ATG5 silencing failed to alter radiation sensitivity or promote apoptosis in either the H460wt or H460crp53 cells. Radiosensitivity appeared to be most closely associated with senescence, which occurred earlier and to a greater extent in the H460wt cells. This finding is consistent with the in-depth proteomics analysis on the secretomes from the H460wt and H460crp53 cells (with or without radiation exposure) that showed no significant association with radioresistance-related proteins, whereas several senescence-associated secretory phenotype (SASP) factors were upregulated in H460wt cells relative to H460crp53 cells. Taken together, these findings indicate that senescence, rather than apoptosis, plays a central role in determination of radiosensitivity; furthermore, autophagy is likely to have minimal influence on radiosensitivity under conditions where autophagy takes the nonprotective form.
Pharmacological Reports | 2018
Faisal Imam; Naif O. Al-Harbi; Mohammad M. Al-Harbi; Mushtaq Ahmad Ansari; Abdullah F. Alasmari; Mohd Nazam Ansari; Wael A. Alanazi; Saleh A. Bahashwan; Mashal M. Almutairi; Musaad A. Alshammari; Mohammad Rashid Khan; Abdulaziz M.S. Alsaad; Moureq R. Alotaibi
BACKGROUND Doxorubicin is an effective, potent and commonly used anthracycline-related anticancer drug; however, cardiotoxicity compromises its therapeutic potential. Apremilast, a novel phosphodiesterase type 4-inhibitor, reported to have anti-inflammatory effects and modulating many inflammatory mediators. METHODS The present study investigated the influence of apremilast against doxorubicin-induced cardiotoxicity in male Wistar rats. A total, 24 animals were divided into four groups of six animal each. Group 1, served as control and received normal saline. Group 2 animals, received doxorubicin (20 mg kg-1, ip). Group 3 and 4, treatment group, received doxorubicin (20 mg kg-1, ip) with the same schedule as group-2, plus apremilast (10 and 20 mg kg-1 day-1, po) respectively. Oxidative stress, caspase-3 enzyme activity, gene expression and protein expression were tested. RESULTS The results of the present study demonstrated that administration of apremilast reversed doxorubicin-induced cardiotoxicity. CONCLUSION These findings suggested that apremilast can attenuate doxorubicin-induced cardiotoxicity via inhibition of oxidative stress mediated activation of nuclear factor-kappa B signaling pathways.BACKGROUND Doxorubicin is an effective, potent and commonly used anthracycline-related anticancer drug; however, cardiotoxicity compromises its therapeutic potential. Apremilast, a novel phosphodiesterase type 4-inhibitor, reported to have anti-inflammatory effects and modulating many inflammatory mediators. METHODS The present study investigated the influence of apremilast against doxorubicin-induced cardiotoxicity in male Wistar rats. A total, 24 animals were divided into four groups of six animal each. Group 1, served as control and received normal saline. Group 2 animals, received doxorubicin (20mgkg-1, ip). Group 3 and 4, treatment group, received doxorubicin (20mgkg-1, ip) with the same schedule as group-2, plus apremilast (10 and 20mgkg-1day-1, po) respectively. Oxidative stress, caspase-3 enzyme activity, gene expression and protein expression were tested. RESULTS The results of the present study demonstrated that administration of apremilast reversed doxorubicin-induced cardiotoxicity. CONCLUSION These findings suggested that apremilast can attenuate doxorubicin-induced cardiotoxicity via inhibition of oxidative stress mediated activation of nuclear factor-kappa B signaling pathways.
International Immunopharmacology | 2018
Naif O. Al-Harbi; Ahmed Nadeem; Sheikh F. Ahmad; Moureq R. Alotaibi; Abdullah F. Alasmari; Wael A. Alanazi; Mohammad M. Al-Harbi; Ahmad M. El-Sherbeeny; Khalid E. Ibrahim
&NA; Sepsis affects millions of people worldwide and is associated with acute kidney injury (AKI). Innate and adaptive immune cells have been shown to play an important role in AKI through release of various inflammatory mediators which include reactive oxidant species (ROS). Acetate, a short chain fatty acid produced by gut bacteria has anti‐inflammatory properties and has also been shown to modulate oxidative stress in different immune cells. Effects of acetate have been shown to be both GPR43 dependent and independent in different cells/tissues. However, the role of acetate on T cell NADPH oxidase (NOX2)/ROS signaling remains unexplored during sepsis‐induced AKI. Therefore, the current study investigated the effect of acetate on sepsis‐induced AKI parameters and T cell oxidant‐antioxidant balance. Our results show that acetate ameliorates sepsis‐induced AKI as reflected by a decrease in serum, creatinine/blood urea nitrogen and renal myeloperoxidase activity/lipid peroxides and restoration of kidney tubular structure. Moreover, acetate administration was associated with correction of oxidant‐antioxidant imbalance in T cells during sepsis‐induced AKI. Acetate produced its inhibitory effects on NOX2/ROS signaling via attenuation of histone deacetylase activity in T cells which was induced during AKI. Overall, the data suggest that acetate might be beneficial during sepsis‐induced AKI by restoration of oxidant‐antioxidant balance in T cells.
International Immunopharmacology | 2018
Sheikh F. Ahmad; Mushtaq Ahmad Ansari; Ahmed Nadeem; Saleh A. Bakheet; Laila Al-Ayadhi; Moureq R. Alotaibi; Ali Alhoshani; Khaled A. Al-Hosaini; Sabry M. Attia
&NA; Autism spectrum disorder (ASD) is a heterogeneous disorder diagnosed based on the severity of abnormalities in social skills. Several studies have acknowledged the presence of abnormal immune functions among individuals diagnosed with ASD. HLA‐DR (human leukocyte antigen‐antigen D related) has been shown to play a significant role in several inflammatory and neurological disorders; however, the role of HLA‐DR signaling in ASD has not yet been fully clarified. In this study, we investigated the role of HLA‐DR signaling in children with ASD. Flow cytometric analysis, using peripheral blood mononuclear cells (PBMCs), revealed the numbers of CD4+, CD8+, CD28+, CXCR4+, and CCR7+ expressing HLA‐DR cells in typically developing (TD) controls and children with ASD. We also determined the numbers of IFN‐&ggr;+, IL‐21+, and Foxp3+ expressing HLA‐DR cells in TD controls and in children with ASD using PBMCs. We observed mRNA and protein expression levels of HLA‐DR by RT‐PCR and western blotting analysis. Our results revealed that children with ASD had significantly increased numbers of HLA‐DR+CD4+, HLA‐DR+CD8+, CD28+HLA‐DR+, HLA‐DR+CXCR4+, HLA‐DR+CCR7+ cells compared with TD controls. We found that children with ASD showed increased HLA‐DR+IFN‐&ggr;+ and HLA‐DR+IL‐21+ and decreased HLA‐DR+Foxp3+ expression levels compared with TD controls. Furthermore, children with ASD showed higher HLA‐DR mRNA and protein expression levels compared with TD controls. These results indicated that HLA‐DR could play an essential role in the immune abnormalities associated with ASD.
Asian Pacific Journal of Cancer Prevention | 2018
Moureq R. Alotaibi; Zeinab K. Hassan; Salim S. Al-Rejaie; Musaad A. Alshammari; Mashal M. Almutairi; Ali Alhoshani; Wael A. Alanazi; Mohamed H Hafez; Othman A. Al-Shabanah
Background Breast cancer is affected by the immune system in that different cytokines play roles in its initiation and progression. Interleukin-10 (IL-10), an anti-inflammatory cytokine, is an immunosuppressive factor involved in tumorigenesis. The present study was conducted to investigate the gene silencing effect of a small interference RNA (siRNA) targeting IL-10 on the apoptotic pathway in breast cancer cell line. Methods The siRNA targeting IL-10 and a glyceraldehyde 3-phosphate dehydrogenase (GAPDH) clone were introduced into MDA-MB-231 cells. Real-time PCR assays were used to determine IL-10 and GAPDH gene expression levels, in addition to those for protein kinase B (AKT), phosphoinositide 3-kinase (PI3K), B-cell lymphoma 2 (Bcl2), caspase-3 and caspase-9 genes related to apoptosis. Results Inhibition of IL-10 by the siRNA accelerated apoptosis and was accompanied by significant increase in caspase-3 and caspase-9 and a significant decrease in PI3K, AKT and Bcl2 expression levels compared to the non-transfected case. Conclusions In conclusion, the production of IL-10 may represent a new escape mechanism by breast cancer cells to evade destruction by the immune system. IL-10 gene silencing causes down regulation of both PI3K/AKT and Bcl2 gene expression and also increases the Bbc3, BAX caspase3, and caspase 3 cleavage expression levels. IL–10 might represent a promising new target for therapeutic strategies.