57 results on '"Moureq R. Alotaibi"'
Search Results
2. Treatment with the anti-CD20 monoclonal antibody rituximab mitigates gonadal disruptions in the collagen-induced arthritis in male DBA/1 J mouse model
- Author
-
Mohammed A. Al-Hamamah, Moureq R. Alotaibi, Sheikh F. Ahmad, Ahmed Nadeem, Mohamed S.M. Attia, Mushtaq A. Ansari, Saleh A. Bakheet, Mohammed M. Alanazi, and Sabry M. Attia
- Subjects
Male ,Health, Toxicology and Mutagenesis ,Antibodies, Monoclonal ,Antineoplastic Agents ,Arthritis, Experimental ,Arthritis, Rheumatoid ,Mice ,Mice, Inbred DBA ,Semen ,Antirheumatic Agents ,Genetics ,Animals ,Rituximab ,Molecular Biology - Abstract
Rheumatoid arthritis (RA), which is driven by persistent activation of the immune system, primarily affects the joints. Several reports have estimated the risk of gonadal disruptions in arthritic patients, with potential attributable risk factors such as treatments with the disease-modifying antirheumatic drugs and the influence of the disease itself. The FDA approved rituximab, a therapy for non-Hodgkin's lymphoma, for management of RA in February 2006. However, the influence of repeated treatment with rituximab on gonadal function in RA has not been reported yet. Thus, the aim of the presents study is to evaluate whether repeated treatment with the clinically relevant dose of rituximab may change the gonadal disruptions in collagen-induced arthritis in male DBA/1 J mouse, a model of RA. Testicular disruptions, as determined by the sperm DNA strand breaks, spermatocyte chromosomal analysis and spermiogram examination have been conducted by the use of standard techniques. Additionally, we aimed to test whether the anti-rheumatic effect of rituximab also decreases the cellular oxidant-antioxidant imbalance in arthritic male DBA/1 J mice. Repeated treatment of naïve control DBA/1 J mice with rituximab did not exhibit any significant deleterious effects. Moreover, repeated administration of rituximab to the arthritic DBA/1 J mice suppressed disease severity and decreased testicular disruptions. Rituximab treatment also diminished gonadal oxidative stress, through decreasing reactive oxygen species generation and restoring the reduced glutathione level in arthritic DBA/1 J mice. In conclusion, rituximab is a safe therapeutic agent and can mitigate gonadal disruptions induced by arthritis, which insinuates the importance for arthritic patients especially at reproductive age.
- Published
- 2022
3. Aneugenic and clastogenic alterations in the DBA/IJ mouse model of rheumatoid arthritis treated with rituximab, an anti-CD20 antibody
- Author
-
Sabry M. Attia, Mohammed A. Al-Hamamah, Moureq R. Alotaibi, Abdullah F. Alasmari, Mohamed S.M. Attia, Sheikh F. Ahmad, Mohamed A. Mahmoud, Ahmed Nadeem, Mushtaq A. Ansari, and Saleh A. Bakheet
- Subjects
Health, Toxicology and Mutagenesis ,Genetics - Published
- 2023
4. In vitro and in vivo antitumor studies of potential anticancer agents of platinum(II) complexes of dicyclopentadiene and dithiocarbamates§
- Author
-
Adam A A Sulaiman, Homood M As Sobeai, Eman Aldawood, Ahmad Abogosh, Khalid Alhazzani, Moureq R Alotaibi, Saeed Ahmad, Ali Alhoshani, and Anvarhusein A Isab
- Subjects
Metals and Alloys ,Biophysics ,Antineoplastic Agents ,Apoptosis ,Biochemistry ,Biomaterials ,Mice ,Indenes ,Chemistry (miscellaneous) ,Coordination Complexes ,Cell Line, Tumor ,Animals ,Humans ,Dimethyl Sulfoxide ,Cisplatin ,Drug Screening Assays, Antitumor ,Melanoma ,Platinum - Abstract
Three platinum(II) complexes of dicyclopentadiene (DCP) and dithiocarbamates (DTCs), namely, [Pt(η4-DCP)(Me2DTC)]PF6 (1), [Pt(η4-DCP)(Et2DTC)]PF6 (2), and [Pt(η4-DCP)(Bz2DTC)]PF6 (3) [Me2DTC = dimethyldithiocarbamate, Et2DTC = diethyldithiocarbamate, and Bz2DTC = dibenzyldithiocarbamate] were prepared and characterized by elemental analysis, IR, 1H, and 13C Nuclear Magnetic Resonance spectroscopy. The spectroscopic data indicated the coordination of both DCP and DTC ligands to platinum(II). The solution chemistry of complex 1 revealed that the complexes are stable in both dimethyl sulfoxide (DMSO) and 1:1 mixture of DMSO:H2O. In vitro cytotoxicity of the complexes relative to cisplatin was tested using MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay, against CHL-1 (human melanoma cancer cells), MDA-MB-231 (breast cancer cells), A549 (lung cancer cells), and B16 (murine melanoma cancer cells). The antiproliferative effect of all three prepared complexes was found to be significantly higher than cisplatin. Furthermore, flow cytometric analysis of complex 1 showed that the complex induced apoptosis, oxidative stress, mitochondrial potential depolarization and cell cycle arrest in a concentration-dependent pattern in the CHL-1 cells. Confirmation of apoptosis via gene expression analysis demonstrated down-regulation of anti-apoptotic genes and up-regulation of pro-apoptotic genes in the CHL-1 cells. Wound-healing assays also lent support to the strong cytotoxicity of the complexes. In vivo studies showed a significant reduction of tumor volume at the end of the experiment. In addition, the drug did not change the weight of the mice. In conclusion, complex 1 inhibited cell proliferation in vitro and reduced tumor growth in vivo.
- Published
- 2022
5. Angiotensin II type 1 receptor blockade attenuates gefitinib-induced cardiac hypertrophy via adjusting angiotensin II-mediated oxidative stress and JNK/P38 MAPK pathway in a rat model
- Author
-
Wael A. Alanazi, Hussain N. Alhamami, Metab Alharbi, Khalid Alhazzani, Abdulrahman S. Alanazi, Sary Alsanea, Nemat Ali, Abdullah F. Alasmari, Ahmed Z. Alanazi, Moureq R. Alotaibi, and Mohammed Alswayyed
- Subjects
Pharmacology ,Pharmaceutical Science - Abstract
Gefitinib is a tyrosine kinase inhibitor (TKI) of the epidermal growth factor receptor (EGFR), used for the treatment of advanced or metastatic non-small cell lung cancer. Recently, studies proved that Gefitinib-induced cardiotoxicity through induction of oxidative stress leads to cardiac hypertrophy. The current study was conducted to understand the mechanisms underlying gefitinib-induced cardiac hypertrophy through studying the roles of angiotensin II (AngII), oxidative stress, and mitogen-activated protein kinase (MAPK) pathway. Male Wistar albino rats were treated with valsartan, gefitinib, or both for four weeks. Blood samples were collected for AngII and cardiac markers measurement, and hearts were harvested for histological study and biochemical analysis. Gefitinib caused histological changes in the cardiac tissues and increased levels of cardiac hypertrophy markers, AngII and its receptors. Blocking of AngII type 1 receptor (AT1R) via valsartan protected hearts and normalized cardiac markers, AngII levels, and the expression of its receptors during gefitinib treatment. valsartan attenuated gefitinib-induced NADPH oxidase and oxidative stress leading to down-regulation of JNK/p38-MAPK pathway. Collectively, AT1R blockade adjusted AngII-induced NADPH oxidase and JNK/p38-MAPK leading to attenuation of gefitinib-induced cardiac hypertrophy. This study found a pivotal role of AngII/AT1R signaling in gefitinib-induced cardiac hypertrophy, which may provide novel approaches in the management of EGFRIs-induced cardiotoxicity.
- Published
- 2022
6. Clearance of therapy‐induced senescent tumor cells by the senolytic ABT‐263 via interference with BCL‐X L –BAX interaction
- Author
-
Liliya Tyutyunyk-Massey, Tareq Saleh, Jason Reed, Valerie J. Carpenter, Andrew J. Souers, Anthony C. Faber, Joel D. Leverson, Graeme F. Murray, Hisashi Harada, David A. Gewirtz, and Moureq R. Alotaibi
- Subjects
BCL‐XL ,0301 basic medicine ,Senescence ,Cancer Research ,senescence ,medicine.medical_treatment ,Bcl-xL ,chemotherapy ,lcsh:RC254-282 ,senolytic ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,ABT‐263 ,Genetics ,medicine ,Doxorubicin ,Senolytic ,Etoposide ,Chemotherapy ,Navitoclax ,biology ,business.industry ,Cancer ,General Medicine ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,radiation ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Molecular Medicine ,business ,medicine.drug - Abstract
Tumor cells undergo senescence in response to both conventional and targeted cancer therapies. The induction of senescence in response to cancer therapy can contribute to unfavorable patient outcomes, potentially including disease relapse. This possibiliy is supported by our findings that tumor cells induced into senescence by doxorubicin or etoposide can give rise to viable tumors in vivo. We further demonstrate sensitivity of these senescent tumor cells to the senolytic ABT‐263 (navitoclax), therefore providing a “two‐hit” approach to eliminate senescent tumor cells that persist after exposure to chemotherapy or radiation. The sequential combination of therapy‐induced senescence and ABT‐263 could shift the response to therapy toward apoptosis by interfering with the interaction between BCL‐XL and BAX. The administration of ABT‐263 after either etoposide or doxorubicin also resulted in marked, prolonged tumor suppression in tumor‐bearing animals. These findings support the premise that senolytic therapy following conventional cancer therapy may improve therapeutic outcomes and delay disease recurrence.
- Published
- 2020
7. EGFR Inhibitor Gefitinib Induces Cardiotoxicity through the Modulation of Cardiac PTEN/Akt/FoxO3a Pathway and Reactive Metabolites Formation: In Vivo and in Vitro Rat Studies
- Author
-
Mohamed W. Attwa, Ali Alhoshani, Moureq R. Alotaibi, Abdullah Al-Dhfyan, Shireen Hourani, Hesham M. Korashy, Abdelali Agouni, Adnan A. Kadi, Sabah Akhtar, Fawaz E. Alanazi, and Asad Zeidan
- Subjects
PTEN ,animal structures ,H9c2 rat cardiomyocyte cells ,CYP1A1 ,FoxO3a ,010501 environmental sciences ,Toxicology ,01 natural sciences ,03 medical and health sciences ,Gefitinib ,medicine ,Epidermal growth factor receptor ,Protein kinase B ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,0105 earth and related environmental sciences ,EGFR inhibitors ,0303 health sciences ,Cardiotoxicity ,biology ,Chemistry ,General Medicine ,Caediotoxicity ,Apoptosis ,biology.protein ,Cancer research ,medicine.drug - Abstract
Gefitinib (GEF) is a selective inhibitor of the epidermal growth factor receptor (EGFR) used to treat non-small cell lung cancer. Yet, few cases of cardiotoxicity have been reported. However, the role of the PTEN/Akt/FoxO3a pathway, which mediates GEF anticancer activity, in GEF cardiotoxicity remains unclear. For this purpose, in vitro H9c2 cells and in vivo rat cardiomyocytes were utilized as study models. Treatment of H9c2 cells and Sprague-Dawley rats with GEF significantly induced the expression of hypertrophic and apoptotic markers at mRNA and protein levels with an increased plasma level of troponin. This was accompanied by induction of autophagy and mitochondrial dysfunction in H9c2 cells. Inhibition of cardiac EGFR activity and Akt cellular content of in vitro and in vivo rat cardiomyocytes by GEF increased PTEN and FoxO3a gene expression and cellular content. Importantly, treatment of H9c2 cells with PI3K/Akt inhibitor increased PTEN and FoxO3a mRNA expression associated with potentiation of GEF cardiotoxicity. In addition, by using LC-MS/MS, we showed that GEF is metabolized in the rat heart microsomes into one cyanide- and two methoxylamine-adduct reactive metabolites, where their formation was entirely blocked by CYP1A1 inhibitor, α-naphthoflavone. The current study concludes that GEF induces cardiotoxicity through modulating the expression and function of the cardiac PTEN/AKT/FoxO3a pathway and the formation of CYP1A1-mediated reactive metabolites.
- Published
- 2020
8. A newly synthesized platinum-based compound (PBC-II) increases chemosensitivity of HeLa ovarian cancer cells via inhibition of autophagy
- Author
-
Adam A. A. Sulaiman, Faten Abdullah Alaqil, Homood M. As Sobeai, Moureq R. Alotaibi, Khalid Alhazzani, Nasser Hadal Alotaibi, Mashal M. Almutairi, and Anvarhusein A. Isab
- Subjects
0301 basic medicine ,Pharmaceutical Science ,Article ,HeLa ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Autophagy ,medicine ,Cytotoxicity ,PI3K/AKT/mTOR pathway ,Platinum ,Pharmacology ,chemistry.chemical_classification ,Cisplatin ,Reactive oxygen species ,biology ,lcsh:RM1-950 ,Acridine orange ,biology.organism_classification ,digestive system diseases ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,Ovarian ,chemistry ,Doxorubicin ,Apoptosis ,030220 oncology & carcinogenesis ,IL-10 ,Cancer research ,medicine.drug - Abstract
There are many mechanisms of resistance, chemoresistance of HeLa cells to anti-cancer agents seems to be autophagy-mediated. While using very effective anti-cancers such as Doxorubicin and cisplatin, cells overcome the cytotoxicity of these drugs through promotion of what so-called cytoprotective autophagy. Here in this study, we sought to introduce a novel platinum-based compound PBC-II that possesses anti-cancer activity. Our data showed that PBC-II is able to induce apoptosis at relatively low concentrations, with no detectable reactive oxygen species (ROS). However, further experiments demonstrated that exposure of HeLa cells to PBC-II did not promote autophagy; rather, it resulted in accumulation of p62 and decrease in LC3-II levels. Autophagy was then promoted in HeLa cells pharmacologically by Doxorubicin and genetically by siRNA IL-10. In order to confirm promotion of autophagy in our model, we performed acridine orange staining to assess for autophagy under microscope as well as via flow cytometry. We then measured protein level of autophagy markers p62 and LC3 by western blot. Our data indicated that PBC-II interferes with therapy-induced autophagy. We also determined PI3K activity while co-incubation of PBC-II with autophagy inducers. It was clear that PI3K activation decreased when PBC-II was co-administered with autophagy inducers. Collectively, PBC-II exerts unique anti-proliferative effects associated with inhibition of autophagy, which indicates that PBC-II is potentially a promising agent to be used in resistant ovarian tumors. Keywords: Ovarian, Autophagy, Doxorubicin, IL-10, Platinum, HeLa
- Published
- 2019
9. Improving the understanding of originator and biosimilar biologics among healthcare providers in Saudi Arabia
- Author
-
Mahmoud Mosli, Doaa A. Bintaleb, Turki Al-Thunian, Moureq R. Alotaibi, Wajed A. Alshammari, Hanan Al Rayes, Najah K. Alharbi, Ghada A Aldrees, Ali Mohammed Asiri, Rana Almadany, Mohammed A. Omair, Atheer T. Alotaibi, Ahmed Z. Alotaibi, Maryam A. Alharaibi, Munira Alwaihibi, Wesam M. Asser, Abdulrazaq Al Jazaeri, Nouf A. Alanazi, and Tariq M. Alhawassi
- Subjects
030203 arthritis & rheumatology ,Pharmacology ,Medical education ,business.industry ,lcsh:RM1-950 ,Pharmaceutical Science ,Biosimilar ,Article ,Clinical Practice ,03 medical and health sciences ,Pharmacoeconomics ,0302 clinical medicine ,lcsh:Therapeutics. Pharmacology ,030220 oncology & carcinogenesis ,Pharmacovigilance ,Medicine ,Patentability ,business ,Healthcare providers - Abstract
The loss of patentability of many originator biologics has led to the rapid introduction of biosimilar agents. The anticipated economic benefit of introducing such agent has been accompanied by vagueness surrounding their biotechnology, approval requirements, positioning in treatment paradigms and potential for adverse events. The Second Symposium on Biologics and Biosimilars “Beyond Clinical Practice” was held on 24th-26th January 2020 aiming at improving the understanding of these new agents in a diverse interactive conference and to guide stakeholders how to introduce biosimilars into clinical practice. The symposium consisted of 4 tracks and 3 workshops. A total of 217 participants attended the meeting. The majority were pharmacists (78.8%) followed by physicians (18.9%) and other healthcare providers (2.3%). The workshops covered the following topics: basics of pharmacoeconomics, pharmacovigilance and patients’ perspective toward biosimilar biologics. While, the 4 main tracks included: Introduction to biosimilars, challenges in clinical practice, regulatory and pharmacoeconomic aspects and Challenges in biosimilar pharmacovigilance.
- Published
- 2020
10. Protective effect of valsartan against doxorubicin‐induced cardiotoxicity: Histopathology and metabolomics in vivo study
- Author
-
Faisal N Alotaibi, Mohammed Alswayyed, Ahmed Z Alanazi, Wael A. Alanazi, Khaldoon Aljerian, Homood M. As Sobeai, Ali Alhoshani, Moureq R. Alotaibi, and Khalid Alhazzani
- Subjects
Male ,Cardiotonic Agents ,Health, Toxicology and Mutagenesis ,Inflammation ,macromolecular substances ,Pharmacology ,Toxicology ,Biochemistry ,Rats, Sprague-Dawley ,In vivo ,Fibrosis ,polycyclic compounds ,medicine ,Animals ,Metabolomics ,Doxorubicin ,Molecular Biology ,Cardiotoxicity ,biology ,Chemistry ,organic chemicals ,technology, industry, and agriculture ,General Medicine ,medicine.disease ,Rats ,carbohydrates (lipids) ,Gene Expression Regulation ,Valsartan ,Apoptosis ,biology.protein ,Molecular Medicine ,Creatine kinase ,medicine.symptom ,medicine.drug - Abstract
Doxorubicin (DOX) treatment has been associated with cardiotoxicity. Therefore, it is crucial to search for a therapeutic that can effectively mitigate DOX-induced cardiotoxicity. This study was conducted to investigate the protective effects of valsartan (VAL) against DOX-induced cardiotoxicity. Sprague-Dawley rats were divided into four treatment groups: Group I: Control, Group II: VAL (30 mg/kg, ip), Group III: DOX (15 mg/kg, ip), and Group IV: VAL + DOX (30 + 15 mg/kg, ip). All groups were treated every other day for 14 days. Blood was isolated for biochemical and metabolomics studies, and sections of the heart were also analyzed for histopathological and immunohistochemical alterations to detect changes in P53, BAX, BCL-2, and P62 expression. The combination of VAL + DOX resulted in a marked decrease in cardiac biomarker enzymes (aminotransferase and creatine phosphokinase) compared to DOX monotherapy. In addition, the histopathological examination of the VAL + DOX combination revealed a low percentage of fibrosis and inflammation. Immunohistochemical expression of p53 and BAX was significantly reduced, whereas BCL-2 expression was significantly increased in the VAL + DOX treatment group compared to DOX monotherapy. Also, the combination of VAL + DOX reverses the negative effect of DOX on nuclear p62 expression. Analysis of serum metabolites showed that DOX monotherapy reduced the number of several amino acids, whereas the combination of VAL + DOX restored these metabolic pathways. This study revealed the potential cardioprotective effect of VAL, which may provide novel and promising approaches for managing cardiotoxicity induced by DOX.
- Published
- 2021
11. Anticancer Activity and Apoptosis Induction of Gold(III) Complexes Containing 2,2′-Bipyridine-3,3′-dicarboxylic Acid and Dithiocarbamates
- Author
-
Saeed Ahmad, Wajhul Qamar, Ali Alhoshani, Adam A. A. Sulaiman, Muhammad Monim-ul-Mehboob, Moureq R. Alotaibi, Khalid Alhazzani, Anvarhusein A. Isab, and Homood M. As Sobeai
- Subjects
Programmed cell death ,dithiocarbamates ,Pyridines ,gold(III) ,Pharmaceutical Science ,Antineoplastic Agents ,010402 general chemistry ,01 natural sciences ,Medicinal chemistry ,2,2'-Bipyridine ,Article ,2,2′-bipyridine-3,3′-dicarboxylic acid ,Analytical Chemistry ,Dimethyldithiocarbamate ,HeLa ,chemistry.chemical_compound ,QD241-441 ,Coordination Complexes ,Thiocarbamates ,Drug Discovery ,medicine ,Humans ,Physical and Theoretical Chemistry ,Dithiocarbamate ,chemistry.chemical_classification ,Cisplatin ,biology ,010405 organic chemistry ,Organic Chemistry ,apoptosis ,biology.organism_classification ,0104 chemical sciences ,Dicarboxylic acid ,chemistry ,Chemistry (miscellaneous) ,Apoptosis ,A549 Cells ,anti-cancer activity ,MCF-7 Cells ,Molecular Medicine ,Gold ,medicine.drug ,HeLa Cells - Abstract
Three novel gold(III) complexes (1–3) of general composition [Au(Bipydc)(S2CNR2)]Cl2 (Bipydc = 2,2′-bipyridine-3,3′-dicarboxylic acid and R = methyl for dimethyldithiocarbamate (DMDTC), ethyl for diethyldithiocarbamate (DEDTC), and benzyl for dibenzyldithiocarbamate (DBDTC)) have been synthesized and characterized by elemental analysis, FTIR and NMR spectroscopic techniques. The spectral results confirmed the presence of both the Bipydc and dithiocarbamate ligands in the complexes. The in vitro cytotoxic studies demonstrated that compounds 1–3 were highly cytotoxic to A549, HeLa, MDA-231, and MCF-7 cancer cells with activities much higher (about 25-fold) than cisplatin. In order to know the possible mode of cell death complex 2, [Au(Bipydc)(DEDTC)]Cl2 was further tested for induction of apoptosis towards the MCF-7 cells. The results indicated that complex 2 induces cell death through apoptosis.
- Published
- 2021
12. Assessment of DNA repair efficiency in the inbred BTBR T+tf/J autism spectrum disorder mouse model exposed to gamma rays and treated with JNJ7777120
- Author
-
Sabry M. Attia, Haneen A. Al-Mazroua, S. A. Bakheet, M.S.A. Attia, Moureq R. Alotaibi, Abdulaziz M.S. Alsaad, H.A. Alomar, Sheikh F. Ahmad, and Ahmed Nadeem
- Subjects
Pharmacology ,medicine.medical_specialty ,DNA damage ,Chemistry ,DNA repair ,Glutathione ,Ligand (biochemistry) ,medicine.disease ,medicine.disease_cause ,030227 psychiatry ,Comet assay ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Endocrinology ,medicine.anatomical_structure ,Autism spectrum disorder ,Internal medicine ,medicine ,Bone marrow ,Biological Psychiatry ,Oxidative stress - Abstract
Information regarding DNA repair in autism is limited to a few studies, which have reported inconsistent results. Therefore, we designed a study to determine whether DNA repair efficiency is altered in autism and to investigate whether the H4 ligand JNJ7777120 can enhance DNA repair efficiency in BTBR T+tf/J (BTBR) mice; we also attempted to elucidate the mechanism(s) underlying this amelioration. Evaluation of DNA damage using the comet assay on bone marrow cells showed increased levels of DNA damage in BTBR mice compared with age-matched control C57BL/6J mice. Conversely, BTBR animals pretreated with 20 mg/kg JNJ7777120 for five days exhibited significant decreases in DNA damage compared with that of control BTBR mice. Our results also indicated higher sensitivity of BTBR mice exposed to gamma rays to DNA damage generation. A marked difference was observed between BTBR and C57BL/6J mice at different sampling times after irradiation, with BTBR mice showing a higher percentage of DNA damage and slower repair rate than that of C57BL/6J mice. JNJ7777120 led to enhanced repair of the DNA damage induced by radiation when administered to BTBR mice five days prior to radiation. Additionally, oxidative stress in BTBR mice was significantly elevated with a reduced GSH/GSSG ratio; significant amelioration was subsequently observed in JNJ7777120-pretreated BTBR mice. Furthermore, repetitive behaviors were also attenuated in BTBR mice by JNJ7777120 treatment without altering locomotor activity. Our results suggest that JNJ7777120 can be developed for use as a therapeutic agent to enhance DNA repair efficiency in autism spectrum disorder.
- Published
- 2019
13. DAPTA, a C-C chemokine receptor 5 (CCR5) antagonist attenuates immune aberrations by downregulating Th9/Th17 immune responses in BTBR T+ Itpr3tf/J mice
- Author
-
Musaad A. Alshammari, Moureq R. Alotaibi, Saleh A. Bakheet, Mushtaq A. Ansari, Sheikh F. Ahmad, Sabry M. Attia, Abdullah F. Alasmari, Ahmed Nadeem, and Haneen A. Al-Mazroua
- Subjects
0301 basic medicine ,Pharmacology ,Chemistry ,viruses ,Antagonist ,virus diseases ,FOXP3 ,CCR5 receptor antagonist ,Marble burying ,03 medical and health sciences ,Chemokine receptor ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Downregulation and upregulation ,RAR-related orphan receptor gamma ,030217 neurology & neurosurgery - Abstract
Autism spectrum disorder (ASD) is a prevalent neurodevelopmental disorder characterized by deficits in social interaction, communication, and repetitive behaviors. BTBR T+ Itpr3tf/J (BTBR) mice, a preclinical autistic model featuring ASD symptoms as defined by social relations, was used in this study. We evaluated the potentially protective effect of D -Ala-peptide T-amide (DAPTA), a selective C-C chemokine receptor 5 (CCR5) antagonist, in BTBR mice. CCR5 is considered a potential therapeutic target in different neurodegenerative disorders. BTBR and C57 mice were intraperitoneally (i.p) treated with the DAPTA (0.01 mg/kg, i.p, once daily) for 7 days. We examined the effect of DAPTA by evaluating marble burying and administering repetitive behavior tests. We employed flow cytometry to assess the effect of DAPTA on CCR5+, CD4+CCR5+, CCR5+IL-6+, CCR5+IL-9+, CCR5+IL-17A+, CCR5+RORγT+, CCR5+IL-10+, and CCR5+Foxp3+ in spleen cells. We further explored the effects of DAPTA on IL-6, IL-9, IL-17A, RORγT, IL-10, and Foxp3 protein and mRNA expression levels in the brain tissues. DAPTA administration significantly decreased marble burying and repetitive behavior in BTBR mice. Additionally, DAPTA treatment inhibited CCR5+, CD4+CCR5+, CCR5+IL-6+, CCR5+IL-9+, CCR5+IL-17A+, CCR5+RORγT+, and upregulated CCR5+IL-10+, and CCR5+Foxp3+ production. We further observed that DAPTA downregulated IL-6, IL-9, IL-17A, and RORγT, and increased IL-10 and Foxp3 protein and mRNA expression. Therefore, our results suggest that DAPTA administration represents a potential treatment strategy for patients with ASD.
- Published
- 2019
14. Inhibition of spleen tyrosine kinase attenuates psoriasis-like inflammation in mice through blockade of dendritic cell-Th17 inflammation axis
- Author
-
Khalid E. Ibrahim, Mohammed M. Al-Harbi, Musaad A. Alshammari, Ali Alhoshani, Sheikh F. Ahmad, Ahmad M. El-Sherbeeny, Moureq R. Alotaibi, Ahmed Nadeem, Khalid S. Alzahrani, and Naif O. Al-Harbi
- Subjects
Male ,0301 basic medicine ,Syk ,Inflammation ,chemical and pharmacologic phenomena ,RM1-950 ,Proinflammatory cytokine ,Mice ,Random Allocation ,03 medical and health sciences ,0302 clinical medicine ,Psoriasis ,medicine ,Animals ,Protein Kinase Inhibitors ,Pharmacology ,Mice, Inbred BALB C ,CD40 ,Innate immune system ,biology ,business.industry ,hemic and immune systems ,Dendritic Cells ,General Medicine ,Dendritic cell ,TLR7 ,Spleen tyrosine kinase ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Th17 Cells ,Th17 ,Therapeutics. Pharmacology ,Inflammation Mediators ,medicine.symptom ,business ,Spleen - Abstract
Psoriasis is a debilitating autoimmune disease of the skin characterized by acanthosis and hyperkeratosis resulting from excessive growth of keratinocytes in the epidermis and inflammatory infiltrates in the dermis. Innate immune cells such as dendritic cells (DCs), perform a critical role in the pathophysiology of psoriasis by presenting inflammatory/costimulatory signals for differentiation of Th17 cells. Recent studies point to the involvement of spleen tyrosine kinase (SYK) in inflammatory signaling cascade of DCs. However, it is yet to be determined whether SYK inhibition in DCs would lead to diminishment of psoriatic inflammation. Therefore, our study evaluated the effects of SYK inhibitor, R406 on imiquimod (IMQ)-induced psoriasis-like inflammation, expression of costimulatory/inflammatory molecules in DCs and their relationship with Th17/Treg cells. Our data show that R406 causes attenuation of IMQ-induced dermal inflammation as shown by reduction in ear/back skin thickness, acanthosis and myeloperoxidase activity. This was concurrent with reduction in inflammatory cytokines and co-stimulatory molecules in CD11c + DCs such as IL-6, IL-23, MHCII, and CD40. This favoured the suppression of Th17 cells and upregulation of Treg cells in R406-treated mice with psoriasis-like inflammation. Direct activation of TLR7 by IMQ in splenocytic cultures led to increased SYK expression in CD11c + DCs and release of IL-23/IL-6. IMQ-induced IL-6/IL-23 levels were significantly diminished by SYK inhibitor, R406 in splenocytic cultures. In essence, our study shows that SYK inhibition supresses psoriasis-like inflammation by modifying DC function in mice. Further, it implies that SYK inhibition could be a prospective therapeutic approach for the treatment of psoriasis-like inflammation.
- Published
- 2019
15. Dysregulation of T cell immunoglobulin and mucin domain 3 (TIM-3) signaling in peripheral immune cells is associated with immune dysfunction in autistic children
- Author
-
Musaad A. Alshammari, Ali Alhoshani, Saleh A. Bakheet, Moureq R. Alotaibi, Sabry M. Attia, Sheikh F. Ahmad, Mushtaq A. Ansari, Ahmed Nadeem, and Laila Y. Al-Ayadhi
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,T cell ,CD14 ,Immunology ,CD11a ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigens, CD ,Internal medicine ,medicine ,Humans ,Autistic Disorder ,Child ,Hepatitis A Virus Cellular Receptor 2 ,Molecular Biology ,biology ,Chemistry ,FOXP3 ,Immune dysregulation ,Cross-Sectional Studies ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Gene Expression Regulation ,Child, Preschool ,Leukocytes, Mononuclear ,biology.protein ,Cytokines ,Female ,Antibody ,CD8 ,Signal Transduction ,030215 immunology - Abstract
Evidence suggests that immune dysregulation is associated with autism spectrum disorder (ASD). T cell immunoglobulin and mucin domain-3 (TIM-3) has a critical role in several inflammatory disorders; however, the role of TIM-3 signaling has not been demonstrated in ASD. In the present study, we assessed the role of TIM-3 signaling in children with ASD. We expected that increased numbers of TIM-3+ cells could alter immune function in children with ASD. We revealed production of TIM-3 on CD3+, CD4+, CD8+, CD11a+,b+, CD14+, CD62P+, and CXCR5+ PBMCs in children with ASD and typically developing (TD) controls using immunofluorescent staining. We further demonstrated the production of IL-1β, IFN-γ, IL-17 A, and Foxp3 in TIM-3+ PBMCs of TD controls and individuals with ASD. We also observed the mRNA expression levels of TIM-3, CD11a,b, CD14, IL-1β and IFN-γ using RT-PCR. We further assessed the protein levels of TIM-3, IL-1β, CXCR5, and IFN-γ using western blotting. The results showed that children with ASD had increased numbers of CD3+TIM-3+, CD4+TIM-3+, CD8+TIM-3+, CD11a,b+TIM-3+, CD14+TIM-3+, CD62P+TIM-3+ and CXCR5+TIM-3+ cells compared with TD controls. Our results further showed that children with ASD had increased IL-1β+TIM-3+, IFN-γ+TIM-3+, and IL-17+TIM-3+, and decreased Foxp3+TIM-3+ production compared with that in TD controls. Our results indicated that children with ASD significantly induced TIM-3, CD11a,b, CD14, CXCR5, IL-1β and IFN-γ mRNA and protein expression levels compared with TD controls. The results suggested that detection of TIM-3 signaling could contribute to the early diagnoses of ASD.
- Published
- 2019
16. In vivo Assessment of Combined Effects of Glibenclamide and Losartan in Diabetic Rats
- Author
-
Moureq R. Alotaibi, Ahmed T Almnaizel, Salim S. Al-Rejaie, Amal J. Fatani, Mohammed M. Ahmed, and Hatem M. Abuohashish
- Subjects
Blood Glucose ,0301 basic medicine ,020205 medical informatics ,medicine.medical_treatment ,02 engineering and technology ,Pharmacology ,medicine.disease_cause ,Losartan ,Diabetes Mellitus, Experimental ,Glibenclamide ,Lipid peroxidation ,03 medical and health sciences ,chemistry.chemical_compound ,Diabetes mellitus ,Glyburide ,0202 electrical engineering, electronic engineering, information engineering ,medicine ,Animals ,Hypoglycemic Agents ,Prospective Studies ,chemistry.chemical_classification ,Glutathione Peroxidase ,Original Paper ,Kidney ,Superoxide Dismutase ,business.industry ,Glutathione peroxidase ,Insulin ,General Medicine ,medicine.disease ,Glutathione ,Rats ,Oxidative Stress ,Glutathione Reductase ,medicine.anatomical_structure ,chemistry ,030101 anatomy & morphology ,business ,Oxidative stress ,medicine.drug - Abstract
Objective: Diabetic complications involve multiple pathological pathways, including hyperglycemia-induced oxidative stress and inflammation. Combination therapy is usually employed to improve treatment outcomes and to lower potential adverse effects. In this study, we evaluated the effects of antidiabetic and antihypertensive agents, glibenclamide (GLI) and losartan (LT), on diabetes mellitus (DM)-associated metabolic changes in rats. Materials and Methods: Streptozotocin-induced diabetic animals were orally treated with GLI 5 mg/kg and/or LT 25 mg/kg for 4 weeks. Blood glucose, insulin, aspartate aminotransferase, alanine aminotransferase, urinary creatinine, and urea levels were measured. Serum, liver, and kidney values of inflammatory markers, such as interleukin-1β, tumor necrosis factor alpha, and interleukin-6 were assessed, along with lipid peroxidation products (e.g., thiobarbituric acid reactive substances), endogenous antioxidants (e.g., glutathione), as well as antioxidant enzyme activities (e.g., catalase, superoxide dismutase, and glutathione peroxidase). Finally, histological changes in liver and kidney tissues were evaluated. Results: DM markedly induced systemic, hepatic, and renal inflammation and lowered antioxidant defense mechanisms. Treatment of diabetic rats with either GLI or LT significantly improved liver and kidney functions and histological structure. Moreover, both medications reduced signs of oxidative stress and inflammation in blood, liver, and kidney samples. Combining GLI and LT showed similar protective potential against systemic, hepatic, and renal oxidative stress and inflammation. Conclusion: Adding LT to GLI therapy revealed prospective antioxidant and anti-inflammatory action, while no synergistic or additive effects were observed.
- Published
- 2018
17. Dysregulation of the expression of HLA-DR, costimulatory molecule, and chemokine receptors on immune cells in children with autism
- Author
-
Ali Alhoshani, Moureq R. Alotaibi, Saleh A. Bakheet, Laila Y. Al-Ayadhi, Sabry M. Attia, Khaled A. Al-Hosaini, Sheikh F. Ahmad, Mushtaq A. Ansari, and Ahmed Nadeem
- Subjects
0301 basic medicine ,Autism Spectrum Disorder ,T-Lymphocytes ,Immunology ,C-C chemokine receptor type 7 ,Cell Separation ,Lymphocyte Activation ,Peripheral blood mononuclear cell ,Immunophenotyping ,Interferon-gamma ,03 medical and health sciences ,Chemokine receptor ,0302 clinical medicine ,Immune system ,Costimulatory and Inhibitory T-Cell Receptors ,Antigens, CD ,mental disorders ,HLA-DR ,Humans ,Immunology and Allergy ,Medicine ,Child ,Cells, Cultured ,Pharmacology ,business.industry ,Interleukins ,CD28 ,FOXP3 ,Forkhead Transcription Factors ,HLA-DR Antigens ,Flow Cytometry ,030104 developmental biology ,Child, Preschool ,Receptors, Chemokine ,business ,030217 neurology & neurosurgery ,CD8 ,Signal Transduction - Abstract
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-γ+, 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-γ+ 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.
- Published
- 2018
18. In vivo and in vitro studies evaluating the chemopreventive effect of metformin on the aryl hydrocarbon receptor-mediated breast carcinogenesis
- Author
-
Homood M. As Sobeai, Fawaz E. Alanazi, Hesham M. Korashy, Naif O. Al-Harbi, Abdullah Al-Dhfyan, Ali Alhoshani, Moureq R. Alotaibi, and Khalid Alhazzani
- Subjects
QH301-705.5 ,CYP1B1 ,DMBA ,In vivo rat ,Apoptosis ,medicine.disease_cause ,Breast cancer ,medicine ,polycyclic compounds ,Biology (General) ,skin and connective tissue diseases ,biology ,Breast carcinogenesis ,Chemistry ,AhR ,Cancer ,Cell cycle ,medicine.disease ,Aryl hydrocarbon receptor ,Metformin ,Cancer research ,biology.protein ,Original Article ,General Agricultural and Biological Sciences ,Carcinogenesis ,Mammosphere - Abstract
Metformin (MET) is a clinically used anti-hyperglycemic agent that shows activities against chemically-induced animal models of cancer. A study from our laboratory showed that MET protectes against 7, 12-dimethylbenz[a]anthracene (DMBA)-induced carcinogenesis in vitro human non-cancerous epithelial breast cells (MCF10A) via activation of the aryl hydrocarbon receptor (AhR). However, it is unclear whether MET can prevent the initiation of breast carcinogenesis in an in vivo rat model of AhR-induced breast carcinogenesis. Therefore, the main aims of this study are to examine the effect of MET on protecting against rat breast carcinogenesis induced by DMBA and to explore whether this effect is medicated through the AhR pathway. In this study, treatment of female rats with DMBA initiated breast carcinogenesis though inhibiting apoptosis and tumor suppressor genes while inducing oxidative DNA damage and cell cycle proliferative markers. This effect was associated with activation of AhR and its downstream target genes; cytochrome P4501A1 (CYP1A1) and CYP1B1. Importantly, MET treatment protected against DMBA-induced breast carcinogenesis by restoring DMBA effects on apoptosis, tumor suppressor genes, DNA damage, and cell proliferation. Mechanistically using in vitro human breast cancer MCF-7 cells, MET inhibited breast cancer stem cells spheroids formation and development by DMBA, which was accompanied by a proportional inhibition in CYP1A1 gene expression. In conclusion, the study reports evidence that MET is an effective chemopreventive therapy for breast cancer by inhibiting the activation of CYP1A1/CYP1B1 pathway in vivo rat model.
- Published
- 2021
19. Clearance of therapy-induced senescent tumor cells by the senolytic ABT-263 via interference with BCL-X
- Author
-
Tareq, Saleh, Valerie J, Carpenter, Liliya, Tyutyunyk-Massey, Graeme, Murray, Joel D, Leverson, Andrew J, Souers, Moureq R, Alotaibi, Anthony C, Faber, Jason, Reed, Hisashi, Harada, and David A, Gewirtz
- Subjects
Male ,BCL‐XL ,senescence ,Carcinogenesis ,Topoisomerase Inhibitors ,bcl-X Protein ,Apoptosis ,chemotherapy ,Models, Biological ,senolytic ,ABT‐263 ,Cell Line, Tumor ,Humans ,Cellular Senescence ,Research Articles ,Etoposide ,bcl-2-Associated X Protein ,Sulfonamides ,Aniline Compounds ,Radiation ,Cell Death ,Tumor Burden ,radiation ,HEK293 Cells ,Doxorubicin ,Protein Binding ,Research Article - Abstract
Senescent tumor cells can be selectively eliminated by the BH3 mimetic, ABT‐263 (navitoclax), via senolysis. We show that ABT‐263 inhibits the interaction between anti‐apoptotic BCL‐2 family member, BCL‐XL, and pro‐apoptotic effector, BAX. This results in senescent tumor cell death in vitro and reduced tumor volume in vivo. This work highlights the utilization of senolytic agents to enhance efficacy of anticancer therapy., Tumor cells undergo senescence in response to both conventional and targeted cancer therapies. The induction of senescence in response to cancer therapy can contribute to unfavorable patient outcomes, potentially including disease relapse. This possibiliy is supported by our findings that tumor cells induced into senescence by doxorubicin or etoposide can give rise to viable tumors in vivo. We further demonstrate sensitivity of these senescent tumor cells to the senolytic ABT‐263 (navitoclax), therefore providing a “two‐hit” approach to eliminate senescent tumor cells that persist after exposure to chemotherapy or radiation. The sequential combination of therapy‐induced senescence and ABT‐263 could shift the response to therapy toward apoptosis by interfering with the interaction between BCL‐XL and BAX. The administration of ABT‐263 after either etoposide or doxorubicin also resulted in marked, prolonged tumor suppression in tumor‐bearing animals. These findings support the premise that senolytic therapy following conventional cancer therapy may improve therapeutic outcomes and delay disease recurrence.
- Published
- 2020
20. EGFR Inhibitor Gefitinib Induces Cardiotoxicity through the Modulation of Cardiac PTEN/Akt/FoxO3a Pathway and Reactive Metabolites Formation
- Author
-
Ali, Alhoshani, Fawaz E, Alanazi, Moureq R, Alotaibi, Mohamed W, Attwa, Adnan A, Kadi, Abdullah, Aldhfyan, Sabah, Akhtar, Shireen, Hourani, Abdelali, Agouni, Asad, Zeidan, and Hesham M, Korashy
- Subjects
Male ,Membrane Potential, Mitochondrial ,Myocardium ,Forkhead Box Protein O3 ,PTEN Phosphohydrolase ,Antineoplastic Agents ,Gefitinib ,Cardiotoxicity ,Cell Line ,ErbB Receptors ,Rats, Sprague-Dawley ,Microsomes ,Animals ,Protein Kinase Inhibitors ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Gefitinib (GEF) is a selective inhibitor of the epidermal growth factor receptor (EGFR) used to treat non-small cell lung cancer. Yet, few cases of cardiotoxicity have been reported. However, the role of the PTEN/Akt/FoxO3a pathway, which mediates GEF anticancer activity, in GEF cardiotoxicity remains unclear. For this purpose
- Published
- 2020
21. Vorinostat is genotoxic and epigenotoxic in the mouse bone marrow cells at the human equivalent doses
- Author
-
Moureq R. Alotaibi, Saleh A. Bakheet, Sabry M. Attia, Mushtaq A. Ansari, Mohamed S.M. Attia, Sheikh F. Ahmad, Mohammed A. Al-Hamamah, Mohamed K. Al-Khalifa, and Ahmed Nadeem
- Subjects
0301 basic medicine ,Male ,DNA Repair ,DNA repair ,medicine.drug_class ,DNA damage ,Down-Regulation ,Antineoplastic Agents ,Apoptosis ,Bone Marrow Cells ,Biology ,Toxicology ,medicine.disease_cause ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,Epigenetics ,Vorinostat ,Chromosome Aberrations ,Dose-Response Relationship, Drug ,Histone deacetylase inhibitor ,DNA Methylation ,Comet assay ,Oxidative Stress ,030104 developmental biology ,DNA methylation ,Cancer research ,Comet Assay ,Carcinogenesis ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Vorinostat was approved as the first histone deacetylase inhibitor for the management of cutaneous T cell lymphoma. However, it's in vivo genetic and epigenetic effects on non-cancerous cells remain poorly understood. As genetic and epigenetic changes play a critical role in the pathogenesis of carcinogenesis, we investigated whether vorinostat induces genetic and epigenetic alterations in mouse bone marrow cells. Bone marrow cells were isolated 24 h following the last oral administration of vorinostat at the doses of 25, 50, or 100 mg/kg/day for five days (approximately equal to the recommended human doses). The cells were then used to assess clastogenicity and aneugenicity by the micronucleus test complemented by fluorescence in situ hybridization assay; DNA strand breaks, oxidative DNA strand breaks, and DNA methylation by the modified comet assay; apoptosis by annexin V/PI staining analysis and the occurrence of the hypodiploid DNA content; and DNA damage/repair gene expression by polymerase chain reaction (PCR) Array. The expression of the mRNA transcripts were also confirmed by real-time PCR and western blot analysis. Vorinostat caused structural chromosomal damage, numerical chromosomal abnormalities, DNA strand breaks, oxidative DNA strand breaks, DNA hypomethylation, and programed cell death in a dose-dependent manner. Furthermore, the expression of numerous genes implicated in DNA damage/repair were altered after vorinostat treatment. Accordingly, the genetic/epigenetic mechanism(s) of action of vorinostat may play a role in its carcinogenicity and support the continued study and development of new compounds with lower toxicity.
- Published
- 2020
22. Therapeutic treatment with Ibrutinib attenuates imiquimod-induced psoriasis-like inflammation in mice through downregulation of oxidative and inflammatory mediators in neutrophils and dendritic cells
- Author
-
Moureq R. Alotaibi, Khalid E. Ibrahim, Khalid S. Alzahrani, Sheikh F. Ahmad, Ahmed Nadeem, Sabry M. Attia, Hafiz Majid Mahmood, Ahmad M. El-Sherbeeny, Faleh Alqahtani, Naif O. Al-Harbi, Mohammed M. Al-Harbi, and Saleh A. Bakheet
- Subjects
0301 basic medicine ,Male ,BALB 3T3 Cells ,Neutrophils ,Down-Regulation ,Inflammation ,medicine.disease_cause ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Immune system ,Piperidines ,Psoriasis ,medicine ,Bruton's tyrosine kinase ,Animals ,Peroxidase ,Skin ,Pharmacology ,Innate immune system ,Imiquimod ,biology ,business.industry ,Nitrotyrosine ,Adenine ,Dendritic Cells ,medicine.disease ,Oxidative Stress ,030104 developmental biology ,chemistry ,Ibrutinib ,Cancer research ,biology.protein ,medicine.symptom ,Inflammation Mediators ,business ,Oxidation-Reduction ,030217 neurology & neurosurgery ,Oxidative stress ,Signal Transduction - Abstract
Psoriasis is clinically characterized by well-demarcated silvery plaques which may appear on the extremities, scalp, and sacral area. The multidimensional interactions among innate immune cells [neutrophils and dendritic cells (DCs)], adaptive immune cells and skin resident cells result in characteristic features of psoriatic inflammation such as acanthosis, hyperkeratosis, and parakeratosis. Tec family kinases are involved in the pathogenesis of several inflammatory diseases. One of them is Bruton’s tyrosine kinase (BTK) which is reported to carry out inflammatory and oxidative signaling in neutrophils and DCs. Effect of BTK inhibitor with regard to psoriatic inflammation has not been explored previously especially in a therapeutic setting. In the current investigation, effect of BTK inhibitor, Ibrutinib on oxidative/inflammatory signaling in dermal/splenic neutrophils [phosphorylated BTK (p-BTK), inducible nitric oxide synthase (iNOS), nitrotyrosine], CD11c + DCs (p-BTK, iNOS, nitrotyrosine, MCP-1, TNF-α) and enzymatic antioxidants [superoxide dismutase (SOD), glutathione peroxidase (GPx), glutathione reductase (GR)] in imiquimod (IMQ)-induced psoriatic inflammation was evaluated using therapeutic mode. Our results show that IMQ treatment led to induction of p-BTK expression along with concomitant increase in oxidative stress in neutrophils, and CD11c + DCs in skin/periphery. Therapeutic treatment with Ibrutinib caused attenuation of IMQ-induced oxidative stress in CD11c + DCs and neutrophils. Further there were dysregulations in antioxidants enzymes (SOD/GPx/GR) in the skin of IMQ-treated mice, which were corrected by Ibrutinib. In short, our study reveals that BTK signaling in neutrophils and CD11c + DCs upregulates oxidative stress which is concomitant with psoriatic inflammation in mice. Ibrutinib attenuates psoriasis inflammation through downregulation of oxidative stress in these innate immune cells.
- Published
- 2020
23. Studies of Non-Protective Autophagy Provide Evidence that Recovery from Therapy-Induced Senescence is Independent of Early Autophagy
- Author
-
Nipa H. Patel, Moureq R. Alotaibi, Tareq Saleh, David A. Gewirtz, Liliya Tyutyunyk-Massey, and Emmanuel K. Cudjoe
- Subjects
Senescence ,autophagy ,senescence ,medicine.medical_treatment ,Cell fate determination ,Biology ,chemotherapy ,Article ,Catalysis ,Inorganic Chemistry ,lcsh:Chemistry ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,medicine ,Humans ,cancer ,Doxorubicin ,Physical and Theoretical Chemistry ,Molecular Biology ,lcsh:QH301-705.5 ,Cellular Senescence ,Spectroscopy ,Etoposide ,radiotherapy ,030304 developmental biology ,proliferative recovery ,0303 health sciences ,Chemotherapy ,Organic Chemistry ,Autophagy ,Cancer ,Chemoradiotherapy ,General Medicine ,HCT116 Cells ,medicine.disease ,Computer Science Applications ,Radiation therapy ,lcsh:Biology (General) ,lcsh:QD1-999 ,030220 oncology & carcinogenesis ,Cancer research ,medicine.drug - Abstract
Autophagy and senescence, predominant responses that may dictate cell fate after chemotherapy or radiation, often occur in tandem. Cells in states of senescence and/or autophagy are frequently growth arrested. We have previously reported that tumor cells induced into senescence by therapy can re-emerge from the growth-arrested state, a phenomenon termed proliferative recovery. The current work shows that, while tumor cells collaterally induced into senescence and autophagy by etoposide, doxorubicin, or radiation undergo proliferative recovery, neither pharmacological nor genetic inhibition of early autophagy alter the extent of senescence or the ability of cells to recover from senescence. These findings confirm and extend our previous observations, essentially dissociating senescence from autophagy, and further indicate that re-emergence from senescence does not appear to be facilitated by or dependent on autophagy. Our results also provide additional evidence for the promotion of the non-protective form of autophagy by both chemotherapeutic drugs and radiation, which may complicate current efforts to inhibit autophagy for therapeutic benefit.
- Published
- 2020
24. Inhibition of interleukin-2-inducible T-cell kinase causes reduction in imiquimod-induced psoriasiform inflammation through reduction of Th17 cells and enhancement of Treg cells in mice
- Author
-
Ahmed Nadeem, Homood M. As Sobeai, Moureq R. Alotaibi, Khalid E. Ibrahim, Sheikh F. Ahmad, Faleh Alqahtani, and Naif O. Al-Harbi
- Subjects
0301 basic medicine ,Male ,Inflammation ,Biochemistry ,T-Lymphocytes, Regulatory ,Pathogenesis ,03 medical and health sciences ,Immune system ,Psoriasis ,Medicine ,Animals ,STAT3 ,Intraepithelial Lymphocytes ,Skin ,Innate immune system ,Imiquimod ,integumentary system ,030102 biochemistry & molecular biology ,biology ,Kinase ,business.industry ,Tumor Necrosis Factor-alpha ,Interleukin-17 ,FOXP3 ,General Medicine ,Protein-Tyrosine Kinases ,Th1 Cells ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Cancer research ,biology.protein ,Th17 Cells ,medicine.symptom ,business ,Signal Transduction - Abstract
Psoriasis is a debilitating chronic skin disease with a worldwide prevalence. Its main features include well-marked silvery scales on the skin of hands and feet and back which arise due to hyperproliferation of keratinocytes and infiltration of immune cells in the skin. Multiple interactions exist between adaptive immune cells such as T cells and innate immune cells such as neutrophils and macrophages which are key players in the pathogenesis of psoriasis. Interleukin-2-inducible T-cell kinase (ITK) plays a key role in Th17 cell development through control of several transcription factors. ITK has been shown to control NFATc1, NFkB and STAT3 in CD4+ T cells. Effect of ITK inhibitor in imiquimod (IMQ)-induced psoriasiform inflammation remains to be explored. In the current examination, role of ITK signaling and its inhibition blockade were evaluated on NFATc1, NFkB and STAT3, IL-17A, TNF-α, IFN-γ, Foxp3, IL-10 in CD4+ T cells in IMQ model. Our data display that ITK signaling is involved in IMQ-induced psoriatic inflammation as paralleled by enhancement of p-ITK, NFATc1, p-NFkB and p-STAT3 in CD4+ T cells. It was associated with enhancement of Th17/Th1 cells and neutrophilic inflammation in the skin. Preventive treatment with ITK inhibitor led to a reduction in Th17/Th1 cells and enhancement of Treg cells. Overall, this study suggests that ITK signaling is an important modulator of transcription factor signaling in CD4+ T cells which is associated with Th17/Th1 cells and psoriasiform inflammation in mice. ITK signaling blockade could be a therapeutic target for the treatment of psoriatic inflammation.
- Published
- 2020
25. Pharmacological appraisal of ligustrazine based cyclohexanone analogs as inhibitors of inflammatory markers
- Author
-
Syed Nasir Abbas Bukhari, Abduaziz Ibrahim Alzarea, Moureq R. Alotaibi, Nabil K. Alruwaili, Khalid Saad Alharbi, Nawaf M. Alotaibi, Nasser Hadal Alotaibi, and Badriyah Shadid Alotaibi
- Subjects
Pharmaceutical Science ,Cyclohexanone ,Inflammation ,Pharmacology ,In Vitro Techniques ,Inhibitory postsynaptic potential ,Secretory Phospholipase A2 ,chemistry.chemical_compound ,Lipoxygenase ,Structure-Activity Relationship ,medicine ,Potency ,Animals ,Humans ,Cyclooxygenase Inhibitors ,Lipoxygenase Inhibitors ,chemistry.chemical_classification ,biology ,Cyclooxygenase 2 Inhibitors ,Chemistry ,Cyclohexanones ,Anti-Inflammatory Agents, Non-Steroidal ,Enzyme ,Pyrazines ,biology.protein ,Cyclooxygenase ,medicine.symptom - Abstract
The targeting of pro-inflammatory enzymes becomes a therapeutic intervention when acute inflammation is proliferating in pathological conditions. This research is intended to carry out an evaluation of inhibiting and inducing enzymes with inflammatory associations with 28 cyclohexanone analogs based on the ligustrazine. Tests were undertaken with inhibitor screening assay kits using a range of synthetic compounds to investigate how they could inhibit the activity of cyclooxygenase (COX) enzymes, secretory phospholipase A2 (sPLA2), and lipoxygenase (LOX) enzyme. Significant and similar inhibitory activities against sPLA2 with were noted with synthetic compounds which included 1f and 1g (IC50 = 2.2 μM). The optimal inhibitory activity regarding LOX enzyme was shown with compounds 1d (IC50 = 8.1 μM) and 1e (IC50 = 7.5 μM). Additionally, the compounds 1b, 1d, 1e, 2n, and 2o were shown to be significant inhibitors of COX-1 activity with IC50 values 0.09 to 0.7 μM. The outcomes of assays for COX inhibition demonstrated that the same compounds had a further strong inhibitive influence on the COX-2 enzyme, and certain compounds such as 1d, 1e, and 2n demonstrated enhanced potency compared with positive controls.
- Published
- 2019
26. Investigation of belinostat-induced genomic instability by molecular cytogenetic analysis and pathway-focused gene expression profiling
- Author
-
S. A. Bakheet, Ahmed Nadeem, Mohammed M. Attia, Mohammed A. Al-Hamamah, Mushtaq A. Ansari, Sheikh F. Ahmad, Gamaleldin I. Harisa, Sabry M. Attia, and Moureq R. Alotaibi
- Subjects
Male ,0301 basic medicine ,Genome instability ,DNA damage ,DNA repair ,Biology ,Hydroxamic Acids ,Toxicology ,medicine.disease_cause ,Genomic Instability ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,medicine ,Animals ,Pharmacology ,Sulfonamides ,Dose-Response Relationship, Drug ,Gene Expression Profiling ,Histone Deacetylase Inhibitors ,Gene expression profiling ,030104 developmental biology ,Histone ,chemistry ,Cytogenetic Analysis ,Cancer research ,biology.protein ,Chromosome breakage ,Carcinogenesis ,Belinostat ,DNA Damage ,Signal Transduction - 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.
- Published
- 2018
27. Abstract 2024: Studying the effects of interfering with doxorubicin-induced senescence in human colon cancer HCT116 cells
- Author
-
Tareq Saleh, Faten Abdullah Alaqil, Munirah Alohaydib, David A. Gewirtz, Moureq R. Alotaibi, Khalid Alhazzani, Ali Alhoshani, and Homood As Sobeia
- Subjects
Senescence ,Cancer Research ,Programmed cell death ,Colorectal cancer ,business.industry ,Cancer ,medicine.disease ,chemistry.chemical_compound ,Oncology ,chemistry ,Apoptosis ,Cancer cell ,Cancer research ,medicine ,Doxorubicin ,Propidium iodide ,business ,medicine.drug - Abstract
Colorectal cancer (CRC) is type of cancer that growing in the colon or the rectum. Although CRC can be treated and managed through several means of therapy, the disease mortality is still high in Saudi Arabia. Upon treatment, cancer cells undergo different form of cell death or growth arrest such as apoptosis and senescence. Senescence is a state of proliferation arrest that is considered as a cause of drug resistance. The current study was designed to interfere with Doxorubicin-induced senescence in cancer cells, and shift the cytotoxicity of doxorubicin from proliferation arrest to apoptosis. β-galactosidase (β-gal) staining has demonstrated that Doxorubicin induces senescence in wild-type HCT116 but not in p21 knock-out cells. (DAPI)/TUNEL stating and Annexin V assay indicate that treatment with Doxorubicin promotes apoptosis in p21 knock-out HCT116 significantly more than in wild-type HCT116. β-galactosidase staining and 5-Dodecanoylaminofluorescein Di-β-D-Galactopyranosid (C12FDG) demonstrated that combination of Doxorubicin with Sorafenib, Rapamycin, or Venetoclax has significantly reduced senescence and induced more apoptosis as shown by Propidium iodide (PI)/Annexin v assay and Caspase 3/7 Assay. PCR data analysis demonstrated that not all combination of Doxorubicin with other agents significantly decreased expression of senescence protein, however, the results of this study provide findings that interference with senescence pathways may shift cytotoxicity from senescence to apoptosis. The authors extend their appreciation to the Deputyship for Research and Innovation, Ministry of Education in Saudi Arabia for partly funding this work through the project number (DRI-KSU-1273). Citation Format: Moureq Rashed Alotaibi, Homood As Sobeia, Munirah Alohaydib, Faten Alaqil, Khalid Alhazzani, Ali Alhoshani, Tareq Saleh, David Gewirtz. Studying the effects of interfering with doxorubicin-induced senescence in human colon cancer HCT116 cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 2024.
- Published
- 2021
28. Abstract 3031: Low expression of immune checkpoint genes is significantly associated with early relapse in pediatric acute lymphoblastic leukemia
- Author
-
Basil Alotaibi, Homood M. As Sobeai, Moureq R. Alotaibi, Khalid Alhazzani, Ali Mohammed Alqasem, Ali Alhoshani, Razan Saad Alshahrani, Basil Nasser Alamri, and Munirah A Alkathiri
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Microarray ,business.industry ,Microarray analysis techniques ,medicine.medical_treatment ,Cancer ,Immunotherapy ,medicine.disease ,Immune checkpoint ,Clinical trial ,TIGIT ,Internal medicine ,medicine ,business ,Survival analysis - Abstract
Background: Acute lymphoblastic leukemia (ALL) is the most common type of childhood malignancy. Despite all the improvements in ALL treatment, cancer relapse remains a major dilemma. Immune checkpoint inhibitors are promising agents that are being evaluated in ALL clinical trials. The overall aim of our study is to evaluate the expression status of immune checkpoint genes in pediatric ALL patients as prognostic factors for the time of relapse and identify possible targets for immunotherapy. Methods: The expression of 21 regulatory immune checkpoint genes was extracted from two microarray datasets of precursor-B-ALL patients (Hogan et al. dataset (GSE28460) included 49 patients and Staal et al. dataset (GSE18497) contained 27 patients). The microarray data were run using the robust multichip average normalization method in RStudio (1.3.959). A 36 month-cutoff was determined to stratify patients into two groups, early relapse (< 36 months) and late relapse (≥ 36 months). The two datasets were normalized to a reference group (late relapse), then combined. The significant differentially expressed genes between the two groups were detected using Student's t-test. An expression score was generated for each patient based on the expression of the significant differentially expressed genes. Survival analysis and Pearson's correlation were performed to assess the impact of significant genes on the time of relapse. Results: 19 out of the 21 genes were downregulated in early relapsing patients compared to the late relapsing group, seven of which were statistically significant; BTLA4, KIR3DS, PDCD1, TIGIT, HAVCR, CTLA4, TNFRSF9. The expression of those seven genes was significantly associated with the time of relapse ( r = 0.381, P = 0.0007). Patients with a low expression profile of the significant differentially expressed genes were 2.3 times more likely to relapse early (CI 1.434-3.713, P = < 0.0001). Conclusion: The expression of immune checkpoint genes is significantly associated with the prognosis of ALL patients. BTLA4, KIR3DS, PDCD1, TIGIT, HAVCR, CTLA4, and TNFRSF9 might be prognostic factors for the time of relapse. Immunotherapy might be beneficial to the late relapsing patients where immune checkpoint genes were upregulated. The study was funded by the deputyship for Research and Innovation, Ministry of Education, Saudi Arabia (DRI-KSU-1273). Citation Format: Basil Jamal Alotaibi, Razan Saad Alshahrani, Munirah Abdulaziz Alkathiri, Ali Mohammed Alqasem, Basil Nasser Alamri, Ali Rashed Alhoshani, Moureq Rashed Alotaibi, Khalid Ahmed Alhazzani, Homood Moqbel As Sobeai. Low expression of immune checkpoint genes is significantly associated with early relapse in pediatric acute lymphoblastic leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 3031.
- Published
- 2021
29. Imbalance between the anti- and pro-inflammatory milieu in blood leukocytes of autistic children
- Author
-
Saleh A. Bakheet, Moureq R. Alotaibi, Mushtaq A. Ansari, Sheikh F. Ahmad, Khairy M.A. Zoheir, Adel R. A. Abd-Allah, Laila Y. Al-Ayadhi, Sabry M. Attia, Abdulaziz M.S. Alsaad, Mohammad Z. Alzahrani, and Ahmed Nadeem
- Subjects
Male ,0301 basic medicine ,CD14 ,Blotting, Western ,Immunology ,Real-Time Polymerase Chain Reaction ,Peripheral blood mononuclear cell ,Pathogenesis ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,Western blot ,IMMUNE IMBALANCE ,Humans ,Medicine ,CTLA-4 Antigen ,Autistic Disorder ,Child ,Molecular Biology ,Inflammation ,medicine.diagnostic_test ,business.industry ,Interleukins ,Flow Cytometry ,medicine.disease ,Cross-Sectional Studies ,030104 developmental biology ,chemistry ,Child, Preschool ,Ionomycin ,Leukocytes, Mononuclear ,Cytokines ,Autism ,Female ,business ,Biomarkers ,030217 neurology & neurosurgery - 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.
- Published
- 2017
30. Tumor cell escape from therapy-induced senescence
- Author
-
Amir A. Toor, Jason Reed, Tareq Saleh, David A. Gewirtz, Scott C. Henderson, Moureq R. Alotaibi, Lynne W. Elmore, Graeme F. Murray, Hisashi Harada, Joseph W. Landry, Zeinab Elsayed, Vasily A. Yakovlev, Liliya Tyutyunyk-Massey, and Ajinkya S. Kawale
- Subjects
0301 basic medicine ,Senescence ,Male ,Cell ,Antineoplastic Agents ,Mice, SCID ,Biology ,Biochemistry ,Flow cytometry ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Doxorubicin ,Etoposide ,Cellular Senescence ,Cell Proliferation ,Pharmacology ,Mice, Inbred BALB C ,medicine.diagnostic_test ,Cancer ,medicine.disease ,HCT116 Cells ,Phenotype ,Xenograft Model Antitumor Assays ,Tumor Burden ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,medicine.drug - Abstract
H460 non-small cell lung, HCT116 colon and 4T1 breast tumor cell lines induced into senescence by exposure to either etoposide or doxorubicin were able to recover proliferative capacity both in mass culture and when enriched for the senescence-like phenotype by flow cytometry (based on β-galactosidase staining and cell size, and a senescence-associated reporter, BTG1-RFP). Recovery was further established using both real-time microscopy and High-Speed Live-Cell Interferometry (HSLCI) and was shown to be accompanied by the attenuation of the Senescence-Associated Secretory Phenotype (SASP). Cells enriched for the senescence-like phenotype were also capable of forming tumors when implanted in both immunodeficient and immunocompetent mice. As chemotherapy-induced senescence has been identified in patient tumors, our results suggest that certain senescence-like phenotypes may not reflect a terminal state of growth arrest, as cells that recover with self-renewal capacity may ultimately contribute to disease recurrence.
- Published
- 2018
31. Genetic and epigenetic alterations induced by the small-molecule panobinostat: A mechanistic study at the chromosome and gene levels
- Author
-
Sabry M. Attia, Saleh A. Bakheet, Sheikh F. Ahmad, Moureq R. Alotaibi, Ahmed Nadeem, Homood M. As Sobeai, Mushtaq A. Ansari, Mohammed A. Al-Hamamah, and Mohamed S. Attia
- Subjects
Male ,DNA damage ,DNA repair ,Biology ,medicine.disease_cause ,Biochemistry ,Epigenesis, Genetic ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Panobinostat ,medicine ,Animals ,Epigenetics ,Molecular Biology ,Gene ,030304 developmental biology ,0303 health sciences ,DNA Breaks ,Cancer ,Cell Biology ,DNA Methylation ,medicine.disease ,Chromosomes, Mammalian ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,Carcinogenesis ,Transcriptome ,Oxidation-Reduction ,DNA hypomethylation ,Mutagens - Abstract
Increasing evidence supports the role of genetic and epigenetic alterations in a wide variety of human diseases, including cancer. Assessment of these alterations is hence essential for estimating the hazardous effects of human exposure to medications. Panobinostat received US Food and Drug Administration's approval in 2015 for treatment of certain tumors and its usefulness as part of a strategy to treat other diseases, such as human immunodeficiency virus infection, is currently investigated. Nevertheless, no data on in vivo genotoxical and epigenotoxical effects of panobinostat are available. The aim of the current study was to assess the genotoxical and epigenotoxical properties of panobinostat in murine bone marrow cells. Molecular mechanisms underlying these alterations were also evaluated. We show that mice treated with panobinostat doses recommended for human developed numerical chromosomal abnormalities, structural chromosomal damage, oxidative DNA damage, and DNA hypomethylation. These effects were dose-dependent. Further, panobinostat altered the expression of 23 genes implicated in DNA damage, as determined by RT² Profiler polymerase chain reaction (PCR) array, and confirmed by quantitative real-time PCR and western blotting. Collectively, these findings indicate that panobinostat exposure induces aneugenicity, clastogenicity, oxidative DNA damage, DNA hypomethylation, and down-regulation of repair gene expression, which may be responsible for panobinostat-induced genotoxical and epigenotoxical effects. Considering the potential toxicity of panobinostat, the medicinal use of panobinostat must be weighed against the risk of tumorigenesis and the demonstrated toxicity profile of panobinostat may support further development of chemotherapeutic treatments with reduced toxicity. Diminishing the metabolic liabilities associated with panobinostat exposure, and simultaneous use of panobinostat with DNA repair enhancers, are examples of strategies for drug design to reduce panobinostat carcinogenicity.
- Published
- 2018
32. Protease activated receptor-2 mediated upregulation of IL-17 receptor signaling on airway epithelial cells is responsible for neutrophilic infiltration during acute exposure of house dust mite allergens in mice
- Author
-
Sabry M. Attia, Ahmed Nadeem, Sheikh F. Ahmad, Naif O. Al-Harbi, Mohammad M. Al-Harbi, Moureq R. Alotaibi, Khalid E. Ibrahim, Nahid Siddiqui, and Shakir D. AlSharari
- Subjects
0301 basic medicine ,Male ,Chemokine ,Toxicology ,Piperazines ,Proinflammatory cytokine ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Downregulation and upregulation ,Medicine ,Animals ,Receptor, PAR-2 ,Antigens, Dermatophagoides ,Receptor ,Protease-activated receptor 2 ,Asthma ,House dust mite ,Inflammation ,Mice, Inbred BALB C ,Receptors, Interleukin-17 ,biology ,business.industry ,Epithelial Cells ,General Medicine ,respiratory system ,biology.organism_classification ,medicine.disease ,respiratory tract diseases ,Up-Regulation ,030104 developmental biology ,Neutrophil Infiltration ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,business ,Signal Transduction - Abstract
Asthma, a chronic inflammatory disease affecting the airways is primarily caused due to immune system dysfunction. Different inhaled allergens such as house dust mites (HDM), fungi, cockroach allergens are the main contributors to allergic asthma. Protease activated receptor-2 (PAR-2) signaling plays an important role in allergic asthma through modulation of immune mediators in airway epithelial cells (AECs). Interleukin-17A (IL-17A) signals via subunits of IL-17 receptor (IL-17R), i.e. interleukin-17 receptor A (IL-17RA) and interleukin-17 receptor C (IL-17RC), and plays a necessary role in neutrophilic infiltration in response to infectious/allergenic stimuli, however it is not known if PAR-2 activation affects IL-17A/IL-17R signaling during acute exposure to house dust mite (HDM) allergens. Therefore, our study exposed mice to HDM allergens for five days and evaluated its effect on IL-17A/IL-17R signaling, chemokine/cytokines and neutrophilic inflammation in mice. Our study shows that HDM allergens upregulate IL-17A levels in the lung and IL-17RA/IL-17RC expression in AECs. PAR-2 activation by trypsin also upregulates neutrophilic influx and IL-17A/IL-17R signaling in the lung. Upregulated IL-17A/IL-17R signaling was associated with increased BAL neutrophils, pulmonary MPO activity and proinflammatory chemokines and cytokines (IL-23, IL-6, and MCP-1 in AECs/lung) in HDM exposed mice. Further, HDM-induced IL-17A, IL-17R and chemokines/cytokines were attenuated by PAR-2 antagonist, ENMD-1068. Furthermore, HDM-primed mice treated with IL-17A had greater neutrophilic inflammation and higher levels of inflammatory cytokines/chemokines than PBS-exposed mice treated with IL-17A. This proposes that acute exposure to HDM allergens activate AECs at a very early stage where PAR-2/IL-17R signaling serves a crucial role in neutrophilic inflammation.
- Published
- 2018
33. DAPTA, a C-C chemokine receptor 5 (CCR5) antagonist attenuates immune aberrations by downregulating Th9/Th17 immune responses in BTBR T
- Author
-
Sheikh F, Ahmad, Mushtaq A, Ansari, Ahmed, Nadeem, Saleh A, Bakheet, Moureq R, Alotaibi, Abdullah F, Alasmari, Musaad A, Alshammari, Haneen A, Al-Mazroua, and Sabry M, Attia
- Subjects
Alanine ,Receptors, CCR5 ,Autism Spectrum Disorder ,Interleukins ,Brain ,Down-Regulation ,Forkhead Transcription Factors ,Mice, Inbred Strains ,Nuclear Receptor Subfamily 1, Group F, Member 3 ,Flow Cytometry ,Mice, Inbred C57BL ,Disease Models, Animal ,Mice ,Behavior Rating Scale ,CCR5 Receptor Antagonists ,Animals ,Cytokines ,Th17 Cells ,RNA, Messenger ,Spleen - Abstract
Autism spectrum disorder (ASD) is a prevalent neurodevelopmental disorder characterized by deficits in social interaction, communication, and repetitive behaviors. BTBR T
- Published
- 2018
34. Corrigendum to 'Apremilast prevent doxorubicin-induced apoptosis and inflammation in heart through inhibition of oxidative stress mediated activation of NF-κB signaling pathways' [Pharmacol. Rep. 70 (2018) 993–1000]
- Author
-
Abdullah F. Alasmari, Faisal Imam, Moureq R. Alotaibi, Mohd Nazam Ansari, Musaad A. Alshammari, Saleh A. Bahashwan, Naif O. Al-Harbi, Mushtaq A. Ansari, Wael A. Alanazi, Mohammad Rashid Khan, Abdulaziz M.S. Alsaad, Mohammad M. Al-Harbi, and Mashal M. Almutairi
- Subjects
Pharmacology ,business.industry ,Inflammation ,General Medicine ,medicine.disease_cause ,Nf κb signaling ,Apoptosis ,medicine ,Cancer research ,Doxorubicin ,Apremilast ,medicine.symptom ,business ,Oxidative stress ,medicine.drug - Published
- 2019
35. Differential Radiation Sensitivity in p53 Wild-Type and p53-Deficient Tumor Cells Associated with Senescence but not Apoptosis or (Nonprotective) Autophagy
- Author
-
Moureq R. Alotaibi, Yingliang Wu, Adam M. Hawkridge, Emmanuel K. Cudjoe, David A. Gewirtz, Nipa H. Patel, Jingwen Xu, Santiago Lima, and Tareq Saleh
- Subjects
0301 basic medicine ,Senescence ,Lung Neoplasms ,ATG5 ,Biophysics ,Apoptosis ,Biology ,Radiation Tolerance ,Article ,03 medical and health sciences ,Radiation sensitivity ,Tandem Mass Spectrometry ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Autophagy ,Humans ,Radiology, Nuclear Medicine and imaging ,Radiosensitivity ,Chromatography, High Pressure Liquid ,Radiation ,Wild type ,Genes, p53 ,030104 developmental biology ,Cell culture ,Cancer research ,Macrolides - Abstract
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.
- Published
- 2018
36. Rutin inhibits carfilzomib-induced oxidative stress and inflammation via the NOS-mediated NF-κB signaling pathway
- Author
-
Imran Kazmi, Faisal Imam, Naif O. Al-Harbi, Othman A. Al-Shabanah, Muhammad Afzal, Mohammed M. Al-Harbi, Moureq R. Alotaibi, Homood M. As Sobeai, and Ammar Cherkess Al Rikabi
- Subjects
0301 basic medicine ,Male ,Antioxidant ,medicine.medical_treatment ,Rutin ,Immunology ,Tetrazoles ,Apoptosis ,Pharmacology ,medicine.disease_cause ,Kidney ,Antioxidants ,Nephrotoxicity ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,White blood cell ,medicine ,Animals ,Pharmacology (medical) ,Rats, Wistar ,Flavonoids ,Inflammation ,business.industry ,Caspase 3 ,Imidazoles ,NF-kappa B ,Carfilzomib ,Rats ,Oxidative Stress ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Proteasome inhibitor ,Nitric Oxide Synthase ,business ,Olmesartan ,Oligopeptides ,030217 neurology & neurosurgery ,Oxidative stress ,medicine.drug ,Signal Transduction - Abstract
Carfilzomib (CFZ), a proteasome inhibitor approved by the FDA to treat multiple myeloma, may cause nephrotoxicity. Rutin is a bioflavonoid with antioxidant properties. We aimed to examine whether rutin protects the kidney from CFZ-induced nephrotoxicity. This study aimed to demonstrate the effect of rutin on CFZ-induced renal injury via the inhibition of oxidative stress and inflammation. Wistar albino rats were divided into six groups (n = 6): Group 1 (normal control; NC) was administered normal saline for 3 weeks; Group 2 (CFZ/toxic group) received CFZ [4 mg/kg, intraperitoneal (i.p.) injection] twice weekly for 3 weeks; Group 3 (standard treatment group) was administered CFZ (4 mg/kg, i.p.) and olmesartan (2 mg/kg, p.o.) for 3 weeks; Group 4 was administered CFZ (4 mg/kg, i.p.) and rutin (10 mg/kg, p.o.) for 3 weeks; Group 5 was administered CFZ (4 mg/kg, i.p.) and rutin (20 mg/kg, p.o.) for 3 weeks; and Group 6 was administered CFZ (4 mg/kg, i.p.) and rutin (40 mg/kg, p.o.) for 3 weeks. We carried out haematological and biochemical analyses, determined oxidative stress, caspase-3 activity, and protein levels, and performed a histopathological evaluation to confirm CFZ-induced nephrotoxicity and its prevention by rutin administration. Exposure to only CFZ significantly (p
- Published
- 2018
37. Characterization of Apoptosis in a Breast Cancer Cell Line after IL-10 Silencing
- Author
-
Moureq R, Alotaibi, Zeinab K, Hassan, Salim S, Al-Rejaie, Musaad A, Alshammari, Mashal M, Almutairi, Ali R, Alhoshani, Wael A, Alanazi, Mohamed M, Hafez, and Othman A, Al-Shabanah
- Subjects
Apoptosis ,Breast Neoplasms ,Interleukin-10 ,caspase-9 ,Gene Expression Regulation, Neoplastic ,Interleukin 10 ,Tumor Cells, Cultured ,Humans ,small interference RNA ,Female ,Gene Silencing ,RNA, Small Interfering ,real time PCR-caspase-3 ,Cell Proliferation ,Signal Transduction ,Research Article - Abstract
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.
- Published
- 2018
38. Short chain fatty acid, acetate ameliorates sepsis-induced acute kidney injury by inhibition of NADPH oxidase signaling in T cells
- Author
-
Mohammad M. Al-Harbi, Ahmed Nadeem, Moureq R. Alotaibi, Abdullah F. Alasmari, Naif O. Al-Harbi, Wael A. Alanazi, Khalid E. Ibrahim, Ahmad M. El-Sherbeeny, and Sheikh F. Ahmad
- Subjects
0301 basic medicine ,Male ,T cell ,T-Lymphocytes ,Immunology ,Pharmacology ,Acetates ,urologic and male genital diseases ,medicine.disease_cause ,Kidney ,Histone Deacetylases ,Blood Urea Nitrogen ,Sepsis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,medicine ,Immunology and Allergy ,Animals ,Humans ,Cells, Cultured ,Peroxidase ,Inflammation ,Mice, Inbred BALB C ,NADPH oxidase ,biology ,urogenital system ,Chemistry ,Acute kidney injury ,NADPH Oxidases ,Acute Kidney Injury ,medicine.disease ,female genital diseases and pregnancy complications ,Gastrointestinal Microbiome ,Oxidative Stress ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Creatinine ,biology.protein ,Histone deacetylase activity ,Reactive Oxygen Species ,Oxidative stress ,Signal Transduction - Abstract
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.
- Published
- 2017
39. Apremilast prevent doxorubicin-induced apoptosis and inflammation in heart through inhibition of oxidative stress mediated activation of NF-κB signaling pathways
- Author
-
Mashal M. Almutairi, Faisal Imam, Mohammad M. Al-Harbi, Mohammad Rashid Khan, Musaad A. Alshammari, Abdullah F. Alasmari, Abdulaziz M.S. Alsaad, Mohd Nazam Ansari, Naif O. Al-Harbi, Saleh A. Bahashwan, Moureq R. Alotaibi, Mushtaq A. Ansari, and Wael A. Alanazi
- Subjects
0301 basic medicine ,Male ,Inflammation ,Apoptosis ,Pharmacology ,medicine.disease_cause ,03 medical and health sciences ,Malondialdehyde ,medicine ,Animals ,Doxorubicin ,RNA, Messenger ,Cardiotoxicity ,Dose-Response Relationship, Drug ,business.industry ,Caspase 3 ,Myocardium ,NF-kappa B ,Phosphodiesterase ,General Medicine ,Catalase ,Glutathione ,Rats ,Thalidomide ,Oxidative Stress ,030104 developmental biology ,Glutathione Reductase ,Apremilast ,Signal transduction ,medicine.symptom ,business ,Oxidative stress ,medicine.drug ,Signal Transduction - Abstract
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.
- Published
- 2017
40. Corrigendum to 'Psoriatic inflammation causes hepatic inflammation with concomitant dysregulation in hepatic metabolism via IL-17A/IL-17 receptor signaling in a murine model' [Immunobiology 222 (2) (February 2017) 128-136]
- Author
-
Ahmed Nadeem, Moureq R. Alotaibi, Khalid Mashay Al-Anazi, Naif O. Al-Harbi, M. M. Al-Harbi, Sheikh F. Ahmad, Mushtaq A. Ansari, Khairy M.A. Zoheir, and Ahmad M. El-Sherbeeny
- Subjects
0301 basic medicine ,Immunology ,Inflammation ,Hematology ,Biology ,Hepatic inflammation ,03 medical and health sciences ,030104 developmental biology ,Il 17 receptor ,Murine model ,Concomitant ,medicine ,Immunology and Allergy ,medicine.symptom ,Drug metabolism - Published
- 2017
41. Neuro-protective effect of rutin against Cisplatin-induced neurotoxic rat model
- Author
-
Moureq R. Alotaibi, Wael A. Alanazi, Salim Salah Al-Rejaie, Musaad A. Alshammari, Ali Alhoshani, Mohamed M. Hafez, Othman A. Al-Shabanah, and Mashal M. Almutairi
- Subjects
0301 basic medicine ,Male ,Antioxidant ,Side effect ,medicine.medical_treatment ,Rutin ,Pharmacology ,medicine.disease_cause ,Thiobarbituric Acid Reactive Substances ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Medicine ,Animals ,PPAR delta ,Rats, Wistar ,chemistry.chemical_classification ,Cisplatin ,Brain Chemistry ,Glutathione Peroxidase ,business.industry ,Glutathione peroxidase ,Body Weight ,Neurotoxicity ,Brain ,lcsh:Other systems of medicine ,General Medicine ,lcsh:RZ201-999 ,medicine.disease ,Glutathione ,Rats ,Real time PCR ,Oxidative Stress ,030104 developmental biology ,Neuroprotective Agents ,Complementary and alternative medicine ,chemistry ,Toxicity ,Gene expression ,business ,030217 neurology & neurosurgery ,Oxidative stress ,medicine.drug ,Research Article - Abstract
Background Cisplatin is widely used chemotherapeutic agent for cancer treatment with limited uses due to its neurotoxic side effect. The aim of this study was to determine the potential preventive effects of rutin on the brain of cisplatin- neurotoxic rat model. Methods Forty rats were divided into four groups. Group-1 (control group) was intra-peritoneal (IP) injected with 2.5 ml/kg saline. Group-2 (rutin group) was orally administrated 30 mg/kg rutin dissolved in water for 14 days. Group-3 (cisplatin group) was IP received 5 mg/kg cisplatin single dose. Group-4 (rutin and cisplatin group) was orally administrated 30 mg/kg rutin dissolved in water for 14 days with a single dose of 5 mg/kg cisplatin IP on day ten. Brain tissues from frontal cortex was used to extract RNA, the gene expression levels of paraoxonase-1 (PON-1), PON-2, PON-3, peroxisome proliferator-activated receptor delta (PPAR-δ), and glutathione peroxidase (GPx) was investigated by Real-time PCR. Results Cisplatin significantly decreased the expression levels of PON-1, PON-3, PPAR-δ and GPX whereas significantly increased PON-2 expression levels. Co-administration of Rutin prevented the cisplatin-induced toxicity by restoring the alteration in the studied genes to normal values as in the control group. Conclusion This study showed that Rutin has neuroprotective effect and reduces cisplatin- neurotoxicity with possible mechanism via the antioxidant pathway.
- Published
- 2017
42. Protective effect of rutin supplementation against cisplatin-induced Nephrotoxicity in rats
- Author
-
Musaad A. Alshammari, Ali Alhoshani, Mohamed M. Hafez, Moureq R. Alotaibi, Othman A. Al-Shabanah, Abdel Malek Al-sheikh, Mashal M. Almutairi, Salim S. Al Rejaie, and Sufia Husain
- Subjects
P38 MAPK ,Male ,0301 basic medicine ,medicine.medical_specialty ,Pathology ,Rutin ,p38 mitogen-activated protein kinases ,Antineoplastic Agents ,Pharmacology ,lcsh:RC870-923 ,medicine.disease_cause ,Nephrotoxicity ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Rats, Wistar ,Blood urea nitrogen ,Cisplatin ,Kidney ,business.industry ,Acute Kidney Injury ,lcsh:Diseases of the genitourinary system. Urology ,Malondialdehyde ,Rats ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Nephrology ,030220 oncology & carcinogenesis ,Gene expression ,business ,Oxidative stress ,Research Article ,medicine.drug - Abstract
Background Cisplatin (CP) is commonly used in the treatment of different types of cancer but nephrotoxicity has been a major limiting factor. Therefore, the present study aimed to study the possible protective effect of rutin against nephrotoxicity induced by cisplatin in rats. Methods Forty male Wistar albino rats were randomly divided into 4 groups. Rats of group 1 control group intraperitoneal (i.p.) received 2.5 ml/kg, group 2 CP group received single dose 5 mg/kg cisplatin i.p. group 3 rutin group orally received 30 mg/kg rutin group 4 (CP plus rutin) received CP and rutin as in group 2 and 3. Kidneys were harvested for histopathology and for the study the gene expression of c-Jun N-terminal kinases (JNK), Mitogen-activated protein kinase 4 (MKK4), MKK7, P38 mitogen-activated protein kinases (P38), tumor necrosis factors alpha (TNF-α), TNF Receptor-Associated Factor 2 (TRAF2), and interleukin-1 alpha (IL-1-α). Results The cisplatin single dose administration to rats induced nephrotoxicity associated with a significant increase in blood urea nitrogen (BUN) and serum creatinine and significantly increase Malondialdehyde (MDA) in kidney tissues by 230 ± 5.5 nmol/g compared to control group. The animal treated with cisplatin showed a significant increase in the expression levels of the IL-1α (260%), TRFA2 (491%), P38 (410%), MKK4 (263%), MKK7 (412%), JNK (680%) and TNF-α (300%) genes compared to control group. Additionally, histopathological examination showed that cisplatin-induced interstitial congestion, focal mononuclear cell inflammatory, cell infiltrate, acute tubular injury with reactive atypia and apoptotic cells. Rutin administration attenuated cisplatin-induced alteration in gene expression and structural and functional changes in the kidney. Additionally, histopathological examination of kidney tissues confirmed gene expression data. Conclusion The present study suggested that the anti-oxidant and anti-inflammatory effect of rutin may prevent CP-induced nephrotoxicity via decreasing the oxidative stress, inhibiting the interconnected ROS/JNK/TNF/P38 MAPK signaling pathways, and repairing the histopathological changes against cisplatin administration.
- Published
- 2017
43. Cytotoxic Autophagy in Cancer Therapy
- Author
-
David A. Gewirtz, Moureq R. Alotaibi, Khushboo Sharma, and Ngoc Le
- Subjects
Drug ,Programmed cell death ,autophagy ,Lung Neoplasms ,medicine.medical_treatment ,media_common.quotation_subject ,Cell ,Antineoplastic Agents ,Breast Neoplasms ,Review ,Biology ,chemotherapy ,Catalysis ,Inorganic Chemistry ,lcsh:Chemistry ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Breast ,Physical and Theoretical Chemistry ,Lung cancer ,Lung ,Molecular Biology ,lcsh:QH301-705.5 ,Spectroscopy ,media_common ,Chemotherapy ,Brain Neoplasms ,Organic Chemistry ,Autophagy ,apoptosis ,Brain ,General Medicine ,medicine.disease ,Computer Science Applications ,Cell biology ,radiation ,medicine.anatomical_structure ,lcsh:Biology (General) ,lcsh:QD1-999 ,Apoptosis ,Female ,Glioblastoma - Abstract
Autophagy is a process of cellular self-digestion, whereby the cell degrades subcellular materials in order to generate energy and metabolic precursors in order to prolong survival, classically under conditions of nutrient deprivation. Autophagy can also involve the degradation of damaged or aged organelles, and misfolded or damaged proteins to eliminate these components that might otherwise be deleterious to cellular survival. Consequently, autophagy has generally been considered a prosurvival response. Many, if not most chemotherapeutic drugs and radiation also promote autophagy, which is generally considered a cytoprotective response, in that its inhibition frequently promotes apoptotic cells death. Furthermore, it has been shown that conventional chemotherapeutic drugs and radiation alone rarely induce a form of autophagy that leads to cell death. However, there are multiple examples in the literature where newer chemotherapeutic agents, drug combinations or drugs in combination with radiation promote autophagic cell death. This review will describe autophagic cell death induced in breast tumor cells, lung cancer cells as well as glioblastoma, demonstrating that it cannot be concluded that stress induced autophagy is, of necessity, cytoprotective in function.
- Published
- 2014
44. Abstract 901: Elimination of senescent tumor cells by ABT263 interferes with proliferative recovery and provides a two-hit therapeutic approach
- Author
-
Jason Reed, Zeinab Elsayed, Lynne W. Elmore, Vasily A. Yakovlev, Joseph W. Landry, Tareq Saleh, Amir A. Toor, Graeme F. Murray, Hisashi Harada, Scott C. Henderson, David A. Gewirtz, Moureq R. Alotaibi, Liliya Tyutyunyk-Massey, and Ajinkya S. Kawale
- Subjects
Senescence ,Cancer Research ,Chemotherapy ,medicine.diagnostic_test ,Cell division ,medicine.medical_treatment ,Cancer ,Biology ,medicine.disease ,Flow cytometry ,CXCL1 ,Oncology ,medicine ,Cancer research ,Senolytic ,Etoposide ,medicine.drug - Abstract
Senescence represents a fundamental response to cancer therapy. Accumulating senescent cells contribute to the deleterious outcomes of cancer therapy including cancer relapse, effects that may be largely mediated by the Senescence-Associated Secretory Phenotype (SASP). In this work, we show that tumor cells induced into senescence by etoposide retain proliferative capacity based on their capacity to generate proliferating colonies in culture as well as giving rise to viable tumors in vivo. Using a flow cytometry-based enrichment approach based on enlarged size and expression of Senescence-Associated β-galactosidase (SA-β-gal), we were able to utilize real time imaging to establish the re-emergence of non-small cell lung cancer cells from senescence-based arrest and the generation or proliferating daughter cells (i.e. self-renewal). Moreover, we implemented High-Speed Live-Cell Interferometry (HSLCI) to provide a single-cell lineage tracking of dividing senescent cells. The recovery from senescence was accompanied by resolution of several senescence-associated hallmarks, specifically SA-β-gal activity, p21Waf1/Cip1 and several components of the SASP (IL-1β, IL-6 and CXCL1). Our data suggests that Therapy-Induced Senescence (TIS) may ultimately be a transient process in that at least a subpopulation of tumor cells can recover proliferative capacity. We further demonstrate that the senolytic agent, ABT263, which has been shown to eliminate senescent cells from aging-related animal models can also eliminate senescent tumor cells that persistent after exposure to chemotherapy by shifting the response towards apoptotic cell death. Furthermore, sequential administration of ABT263 interferes with the ability of tumor cells induced into senescence by chemotherapy to recover growth potential. These studies suggest that senescent tumor cells can potentially contribute to cancer relapse by acquiring proliferative properties. The use of senolytic agents after induction of senescence by conventional or targeted therapies allows for the clearance of residual (possibly dormant) senescent tumor cells, which could serve to suppress disease recurrence and cancer mortality. Citation Format: Tareq Saleh, Liliya Tyutyunyk-Massey, Graeme F. Murray, Moureq R. Alotaibi, Ajinkya S. Kawale, Zeinab Elsayed, Scott C. Henderson, Vasily Yakovlev, Lynne W. Elmore, Amir Toor, Hisashi Harada, Jason Reed, Joseph W. Landry, David A. Gewirtz. Elimination of senescent tumor cells by ABT263 interferes with proliferative recovery and provides a two-hit therapeutic approach [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 901.
- Published
- 2019
45. Psoriasis-like inflammation leads to renal dysfunction via upregulation of NADPH oxidases and inducible nitric oxide synthase
- Author
-
Mushtaq A. Ansari, Abdulaziz M.S. Alsaad, Sheikh F. Ahmad, Moureq R. Alotaibi, Ahmed Nadeem, Mohammed M. Al-Harbi, and Naif O. Al-Harbi
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Immunology ,Renal function ,Nitric Oxide Synthase Type II ,Inflammation ,Systemic inflammation ,medicine.disease_cause ,Kidney ,Antioxidants ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Internal medicine ,medicine ,Immunology and Allergy ,Animals ,Humans ,Psoriasis ,Buthionine Sulfoximine ,Pharmacology ,Mice, Inbred BALB C ,Imiquimod ,Membrane Glycoproteins ,biology ,business.industry ,NOX4 ,NADPH Oxidases ,medicine.disease ,Acetylcysteine ,Nitric oxide synthase ,Disease Models, Animal ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,NADPH Oxidase 4 ,030220 oncology & carcinogenesis ,NADPH Oxidase 2 ,biology.protein ,Aminoquinolines ,Kidney Diseases ,medicine.symptom ,business ,Oxidative stress ,Kidney disease - 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.
- Published
- 2016
46. Psoriatic inflammation causes hepatic inflammation with concomitant dysregulation in hepatic metabolism via IL-17A/IL-17 receptor signaling in a murine model
- Author
-
Ahmed Nadeem, Mushtaq A. Ansari, Mohammed M. Al-Harbi, Moureq R. Alotaibi, Ahmed M. El-Sherbeeny, Khalid Mashay Al-Anazi, Khairy M.A. Zoheir, Naif O. Al-Harbi, and Sheikh F. Ahmad
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Immunology ,Inflammation ,Biology ,medicine.disease_cause ,Systemic inflammation ,Hepatitis ,Pathogenesis ,030207 dermatology & venereal diseases ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Internal medicine ,Psoriasis ,medicine ,Immunology and Allergy ,Animals ,Peroxidase ,Receptors, Interleukin-17 ,Cholesterol ,Interleukin-17 ,NF-kappa B ,Lipid metabolism ,Hematology ,medicine.disease ,Lipid Metabolism ,Disease Models, Animal ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,Cytokine ,chemistry ,Cytokines ,Lipid Peroxidation ,medicine.symptom ,Energy Metabolism ,Oxidative stress ,Biomarkers ,Signal Transduction - Abstract
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.
- Published
- 2016
47. Radiosensitization by PARP Inhibition in DNA Repair Proficient and Deficient Tumor Cells: Proliferative Recovery in Senescent Cells
- Author
-
David A. Gewirtz, Moureq R. Alotaibi, Tareq Saleh, Eric A. Hendrickson, Lawrence F. Povirk, and Khushboo Sharma
- Subjects
0301 basic medicine ,Senescence ,Programmed cell death ,Radiation-Sensitizing Agents ,DNA Ligases ,DNA Repair ,DNA damage ,DNA repair ,Biophysics ,Poly (ADP-Ribose) Polymerase-1 ,Apoptosis ,Biology ,Poly(ADP-ribose) Polymerase Inhibitors ,Article ,Olaparib ,03 medical and health sciences ,chemistry.chemical_compound ,DNA Ligase ATP ,Mice ,0302 clinical medicine ,Autophagy ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,DNA Breaks, Double-Stranded ,Cellular Senescence ,Cell Proliferation ,Radiation ,Cell growth ,Carcinoma ,HCT116 Cells ,Xenograft Model Antitumor Assays ,Cell biology ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Poly(ADP-ribose) Polymerases ,Cell aging ,DNA Damage - Abstract
Radiotherapy continues to be a primary modality in the treatment of cancer. DNA damage induced by radiation can promote apoptosis as well as both autophagy and senescence, where autophagy and senescence can theoretically function to prolong tumor survival. A primary aim of this work was to investigate the hypothesis that autophagy and/or senescence could be permissive for DNA repair, thereby facilitating tumor cell recovery from radiation-induced growth arrest and/or cell death. In addition, studies were designed to elucidate the involvement of autophagy and senescence in radiation sensitization by PARP inhibitors and the re-emergence of a proliferating tumor cell population. In the context of this work, the relationship between radiation-induced autophagy and senescence was also determined. Studies were performed using DNA repair proficient HCT116 colon carcinoma cells and a repair deficient Ligase IV (−/−) isogenic cell line. Irradiation promoted a parallel induction of autophagy and senescence that was strongly correlated with the extent of persistent H2AX phosphorylation in both cell lines; however inhibition of autophagy failed to suppress senescence, indicating that the two responses were dissociable. Irradiation resulted in a transient arrest in the HCT116 cells while arrest was prolonged in the Ligase IV (−/−) cells; however, both cell lines ultimately recovered proliferative function, which may reflect maintenance of DNA repair capacity. The PARP inhibitors (Olaparib) and (Niraparib) increased the extent of persistent DNA damage induced by radiation as well as the extent of both autophagy and senescence; neither cell line underwent significant apoptosis by radiation alone or in the presence of the PARP inhibitors. Inhibition of autophagy failed to attenuate radiation sensitization, indicating that autophagy was not involved in the action of the PARP inhibitors. As with radiation alone, despite sensitization by PARP inhibition, proliferative recovery was evident within a period of 10–20 days. While inhibition of DNA repair via PARP inhibition may initially sensitize tumor cells to radiation via the promotion of senescence, this strategy does not appear to interfere with proliferative recovery, which could ultimately contribute to disease recurrence.
- Published
- 2016
48. Abstract 1326: Nonprotective autophagy fails to confer resistance to radiation
- Author
-
Yingliang Wu, David A. Gewirtz, Moureq R. Alotaibi, Santiago Lima, Emmanuel K. Cudjoe, Tareq Saleh, Nipa H. Patel, and Jingwen Xu
- Subjects
Cancer Research ,Chemistry ,medicine.medical_treatment ,Autophagy ,ATG5 ,Cancer ,medicine.disease ,Radiation therapy ,Radiation sensitivity ,Oncology ,Apoptosis ,Cancer cell ,Cancer research ,medicine ,Gene silencing - Abstract
Radiation remains a predominant treatment option for advanced, inoperable lung cancer, however, resistance represents a major barrier against effective therapy. The exact mechanisms through which cancer cells exhibit resistance to radiotherapy still remains unresolved. Traditionally, radiation-induced autophagy is considered as a mechanism of resistance which attenuates the antitumor effects of radiotherapy i.e., autophagy plays a cytoprotective role on cell survival. To examine this concept further, we employed H460 non-small cell lung cancer (NSCLC) cells that were either p53 wild-type (p53 +/+) or p53 null (p53 -/-) to study the contribution of autophagy to radiation sensitivity. While autophagy was markedly higher in p53 +/+ cells (determined by p62 degradation, LC3B conversion, and acidic vesicle formation) the p53 +/+ cells were actually more radiosensitive than their counterpart H460 p53 -/- cells, which showed lower levels of radiation-induced autophagy. Despite modest radiosensitization by chloroquine, alternative pharmacological (3-methly adenine) and genetic (ATG5 silencing) inhibition of autophagy failed to radiosensitize either p53 +/+ or p53 -/- cells, indicating that radiation induced autophagy is not cytoprotective in function in these experimental models. Furthermore, secretome analysis (nLC-MS/MS) of p53 +/+ H460 cells showed no significant association with radioresistance-related proteins in comparison with p53 -/- H460 cells. The rate and extent of apoptosis was quite low and similar in the two cells lines, essentially ruling out apoptotic cell death as the basis for differential radiation sensitivity. Finally, Senescence was more pronounced in the p53 +/+ cells compared to the p53 -/- cells, which may contribute to the greater radiation sensitivity in the p53 +/+ cells. However, the most relevant finding in this work was that when autophagy is the nonprotective form, it does not confer inherent radiation resistance in tumor cells. Citation Format: Jingwen Xu, Emmanuel Kenneth Cudjoe, Tareq Saleh, Nipa Patel, Moureq Alotaibi, Yingliang Wu, Santiago Lima, David Gewirtz. Nonprotective autophagy fails to confer resistance to radiation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1326.
- Published
- 2018
49. Abstract 459: Therapy-induced senescence is reversible in vitro and in vivo
- Author
-
Scott C. Henderson, Emmanuel K. Cudjoe, Jingwen Xu, Ajinkya S. Kawale, Tareq Saleh, Zeinab Elsayed, Vasily A. Yakovlev, Lynne W. Elmore, Sarah L. Kyte, David A. Gewirtz, Moureq R. Alotaibi, and Joseph W. Landry
- Subjects
Senescence ,Cancer Research ,Colorectal cancer ,Cancer ,Biology ,medicine.disease ,Transplantation ,Oncology ,Downregulation and upregulation ,In vivo ,Cancer research ,medicine ,Lung cancer ,Etoposide ,medicine.drug - Abstract
While cellular senescence has long been recognized as an irreversible form of growth arrest, evidence in the literature has suggested that subpopulations of senescent tumor cells may retain proliferative capacity. To directly address this question, H460 non-small cell lung cancer cells induced into senescence by exposure to etoposide, and enriched based on β-galactosidase staining and size, were shown to recover reproductive capacity, which was accompanied by resolution of the DNA-damage-response (downregulation of p53 and p21Waf1/Cip1 induction), attenuation of the senescence-associated secretory phenotype (SASP) as well as downregulation of miRNA34 expression and an increase in c-myc. The senescence-enriched lung cancer cells were also capable of tumor growth upon transplantation into immunodeficient mice. Re-emergence from etoposide-induced senescence was also observed using HCT116 colon cancer cells, as evidenced by concomitant downregulation of a senescence-associated reporter (BTG1-RFP). Collectively, these findings indicate that therapy-induced senescence (TIS) may ultimately be a transient process in that at least a subpopulation of tumor cells can resume proliferation. We propose that some forms of tumor dormancy that lead to disease recurrence may reflect cells that enter into and ultimately escape from senescence. Citation Format: Tareq Saleh, Moureq R. Alotaibi, Sarah L. Kyte, Emmanuel K. Cudjoe, Ajinkya Kawale, Jingwen Xu, Zeinab Elsayed, Joseph W. Landry, Scott C. Henderson, Vasily Yakovlev, Lynne W. Elmore, David A. Gewirtz. Therapy-induced senescence is reversible in vitro and in vivo [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 459.
- Published
- 2018
50. Combined antiproliferative effects of the aminoalkylindole WIN55,212-2 and radiation in breast cancer cells
- Author
-
Aron H. Lichtman, Qing Tao, David A. Gewirtz, Moureq R. Alotaibi, Dana E. Selley, and Sean M. Emery
- Subjects
medicine.medical_treatment ,Morpholines ,Phytochemicals ,Antineoplastic Agents ,Breast Neoplasms ,Pharmacology ,Naphthalenes ,chemistry.chemical_compound ,Mice ,Sphingosine ,Cell Line, Tumor ,Synthetic cannabinoids ,medicine ,Autophagy ,Animals ,Humans ,Receptors, Cannabinoid ,Cellular Senescence ,Cell Proliferation ,Cannabinoid Receptor Agonists ,Cannabinoids ,Methanandamide ,Cancer ,Stereoisomerism ,medicine.disease ,Antineoplastic Agents, Phytogenic ,Benzoxazines ,Neoplasm Proteins ,Nabilone ,Radiation therapy ,Gene Expression Regulation, Neoplastic ,chemistry ,Molecular Medicine ,Female ,Cannabinoid ,Lysophospholipids ,Cannabidiol ,medicine.drug - Abstract
The potential antitumor activity of cannabinoid receptor agonists, such as the aminoalklylindole WIN55,212-2 (WIN2), has been studied extensively, but their potential interaction with conventional cancer therapies, such as radiation, remains unknown. In the present work, the influence of WIN2 on the antiproliferative activity of radiation in human (MCF-7 and MDA-MB231) and murine (4T1) breast cancer cells was investigated. The antiproliferative effects produced by combination of WIN2 and radiation were more effective than either agent alone. The stereoisomer of WIN2, WIN55,212-3 (WIN3), failed to inhibit growth or potentiate the growth-inhibitory effects of radiation, indicative of stereospecificity. Two other aminoalkylindoles, pravadoline and JWH-015 [(2-methyl-1-propyl-1H-indol-3-yl)-1-naphthalenyl-methanone], also enhanced the antiproliferative effects of radiation, but other synthetic cannabinoids (i.e., nabilone, CP55,940 [(+)-rel-5-(1,1-dimethylheptyl)-2-[(1R,2R,5R)-5-hydroxy-2-(3-hydroxypropyl)cyclohexyl]-phenol], and methanandamide) or phytocannabinoids [i.e., Δ9-tetrahydrocannabinol (THC) and cannabidiol] did not. The combination treatment of WIN2 + radiation promoted both autophagy and senescence but not apoptosis or necrosis. WIN2 also failed to alter radiation-induced DNA damage or the apparent rate of DNA repair. Although the antiproliferative actions of WIN2 were mediated through noncannabinoid receptor-mediated pathways, the observation that WIN2 interfered with growth stimulation by sphingosine-1-phosphate (S1P) implicates the potential involvement of S1P/ceramide signaling pathways. In addition to demonstrating that aminoalkylindole compounds could potentially augment the effectiveness of radiation treatment in breast cancer, the present study suggests that THC and nabilone are unlikely to interfere with the effectiveness of radiation therapy, which is of particular relevance to patients using cannabinoid-based drugs to ameliorate the toxicity of cancer therapies.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.