144 results on '"Kazim Husain"'
Search Results
2. Data from Selective Nuclear Export Inhibitor KPT-330 Enhances the Antitumor Activity of Gemcitabine in Human Pancreatic Cancer
- Author
-
Amit Mahipal, Daniel M. Sullivan, Tami Rashal, Yosef Landesman, Marsha Crochiere, Trinayan Kashyap, Sherma Zibadi, Kazim Husain, Domenico Coppola, Mokenge P. Malafa, and Sabiha Kazim
- Abstract
Pancreatic cancer is an aggressive and deadly malignancy responsible for the death of over 37,000 Americans each year. Gemcitabine-based therapy is the standard treatment for pancreatic cancer but has limited efficacy due to chemoresistance. In this study, we evaluated the in vitro and in vivo effects of gemcitabine combined with the selective nuclear export (CRM1) inhibitor KPT-330 on pancreatic cancer growth. Human pancreatic cancer MiaPaCa-2 and metastatic pancreatic cancer L3.6pl cell lines were treated with different concentrations of KPT-330 and gemcitabine alone or in combination, and anchorage–dependent/independent growth was recorded. In addition, L3.6pl cells with luciferase were injected orthotopically into the pancreas of athymic nude mice, which were treated with (i) vehicle (PBS 1 mL/kg i.p., 2/week and povidone/pluronic F68 1 mL/kg p.o., 3/week), (ii) KPT-330 (20 mg/kg p.o., 3/week), (iii) gemcitabine (100 mg/kg i.p., 2/week), or (iv) KPT-330 (10 mg/kg) + gemcitabine (50 mg/kg) for 4 weeks. KPT-330 and gemcitabine alone dose-dependently inhibited anchorage-dependent growth in vitro and tumor volume in vivo compared with vehicle treatment. However, the combination inhibited growth synergistically. In combination, KPT-330 and gemcitabine acted synergistically to enhance pancreatic cancer cell death greater than each single-agent therapy. Mechanistically, KPT-330 and gemcitabine promoted apoptosis, induced p27, depleted survivin, and inhibited accumulation of DNA repair proteins. Together, our data suggest that KPT-330 potentiates the antitumor activity of gemcitabine in human pancreatic cancer through inhibition of tumor growth, depletion of the antiapoptotic proteins, and induction of apoptosis. Mol Cancer Ther; 14(7); 1570–81. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
3. Data from Chemoprevention of Azoxymethane-induced Colon Carcinogenesis by Delta-Tocotrienol
- Author
-
Mokenge P. Malafa, Chung S. Yang, Domenico Coppola, Ashley Davis-Yadley, Steve Shivers, Anying Zhang, and Kazim Husain
- Abstract
This study evaluated the preclinical activity of δ-tocotrienol (DT3), a bioactive form of vitamin E, in the inhibition of colorectal cancer growth and development in vitro and in vivo. DT3 is the most bioactive isomer of vitamin E in inhibiting growth of colorectal cancer cells. However, it had little effect on the proliferation of normal colon mucosal cells NCM460. In HCT-116 and SW-620 colorectal cancer cells, DT3 (50 μmol/L) significantly inhibited malignant transformation (P < 0.02, P < 0.001), cell migration (P < 0.02, P < 0.05), and invasion (P < 0.05, P < 0.01) compared with vehicle. DT3 inhibited markers for epithelial (E-cadherin) to mesenchymal (vimentin) transition, metastasis (matrix metalloproteinase 9), angiogenesis VEGF, inflammation (NF-κB), and Wnt signaling (β-catenin) compared with vehicle in colorectal cancer cells. DT3 induced apoptosis selectively in colorectal cancer cells (SW-620 cells, HCT-116 cells, and HT-29) without affecting the normal colon cells. In the azoxymethane-induced colorectal carcinogenesis model in rats, DT3 (200 mg/kg orally twice a day) for 20 weeks significantly inhibited colorectal polyps by 70% and colorectal cancer by almost 99% compared with the vehicle treatment group (P < 0.02, P < 0.001), and the cancer inhibition effect was more potent than sulindac (50%). Taken together, these data demonstrate that DT3 is a potential chemopreventive agent in colorectal cancer, warranting further investigation into its clinical use in the prevention and treatment of colorectal cancer.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Table S2 from Plasma MicroRNAs as Novel Biomarkers for Patients with Intraductal Papillary Mucinous Neoplasms of the Pancreas
- Author
-
Mokenge Malafa, Domenico Coppola, Anthony M. Magliocco, Barbara Ann Centeno, Kazim Husain, Christina Georgeades, Yonghong Zhang, Sean J. Yoder, William J. Fulp, Dung-Tsa Chen, and Jennifer Permuth-Wey
- Abstract
Correlation between Paired Tissue and Plasma MiRNA Expression in 12 Individuals with IPMNs
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary legend from Selective Nuclear Export Inhibitor KPT-330 Enhances the Antitumor Activity of Gemcitabine in Human Pancreatic Cancer
- Author
-
Amit Mahipal, Daniel M. Sullivan, Tami Rashal, Yosef Landesman, Marsha Crochiere, Trinayan Kashyap, Sherma Zibadi, Kazim Husain, Domenico Coppola, Mokenge P. Malafa, and Sabiha Kazim
- Abstract
Supplemental legend for Fig. S1
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure Legends from Chemoprevention of Azoxymethane-induced Colon Carcinogenesis by Delta-Tocotrienol
- Author
-
Mokenge P. Malafa, Chung S. Yang, Domenico Coppola, Ashley Davis-Yadley, Steve Shivers, Anying Zhang, and Kazim Husain
- Abstract
This document contains the figure legends for Supplementary Figures 1-5.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure S2 from Plasma MicroRNAs as Novel Biomarkers for Patients with Intraductal Papillary Mucinous Neoplasms of the Pancreas
- Author
-
Mokenge Malafa, Domenico Coppola, Anthony M. Magliocco, Barbara Ann Centeno, Kazim Husain, Christina Georgeades, Yonghong Zhang, Sean J. Yoder, William J. Fulp, Dung-Tsa Chen, and Jennifer Permuth-Wey
- Abstract
miR-145-5p expression differentiates between cases and non-diseased controls. A) Box plot reveals miR-145-5p expression is higher in cases versus controls. B) ROC analysis reveals that miR-145-5p expression can differentiate between groups with an AUC=79.3 (95% CI: 68.3-90.3). Estimates of sensitivity, specificity, positive predictive value, and negative predictive value are 81%, 62.5%, 79%, and 65%, respectively.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure S3 from Plasma MicroRNAs as Novel Biomarkers for Patients with Intraductal Papillary Mucinous Neoplasms of the Pancreas
- Author
-
Mokenge Malafa, Domenico Coppola, Anthony M. Magliocco, Barbara Ann Centeno, Kazim Husain, Christina Georgeades, Yonghong Zhang, Sean J. Yoder, William J. Fulp, Dung-Tsa Chen, and Jennifer Permuth-Wey
- Abstract
A 5-miRNA signature discriminates Malignant (N=21) and Benign (N=21) IPMN Cases. A) Percentage of variation explained in the 5 principal components using the 5 miRNA signature. B) Association of the 5-miRNA signature with IPMN malignancy status. Box plots were used to display the distribution of the IPMN-risk malignancy score within each group. Two-sample t-tests were used to determine associations between the continuous PC1 score and IPMN malignancy status. C) ROC curve analysis of the 5-miRNA signature yielded an AUC of 73.2 (95% CI: 57.6-88.9) in differentiating between groups.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Table from Chemoprevention of Azoxymethane-induced Colon Carcinogenesis by Delta-Tocotrienol
- Author
-
Mokenge P. Malafa, Chung S. Yang, Domenico Coppola, Ashley Davis-Yadley, Steve Shivers, Anying Zhang, and Kazim Husain
- Abstract
This file contains Supplementary Table 1.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure S1 from Selective Nuclear Export Inhibitor KPT-330 Enhances the Antitumor Activity of Gemcitabine in Human Pancreatic Cancer
- Author
-
Amit Mahipal, Daniel M. Sullivan, Tami Rashal, Yosef Landesman, Marsha Crochiere, Trinayan Kashyap, Sherma Zibadi, Kazim Husain, Domenico Coppola, Mokenge P. Malafa, and Sabiha Kazim
- Abstract
Supplementary Figure S1. Effect of KPT-330 on human normal pancreatic epithelial (HPNE) cell proliferation (MTT assay).
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure Legends from Plasma MicroRNAs as Novel Biomarkers for Patients with Intraductal Papillary Mucinous Neoplasms of the Pancreas
- Author
-
Mokenge Malafa, Domenico Coppola, Anthony M. Magliocco, Barbara Ann Centeno, Kazim Husain, Christina Georgeades, Yonghong Zhang, Sean J. Yoder, William J. Fulp, Dung-Tsa Chen, and Jennifer Permuth-Wey
- Abstract
Please see this file for descriptions
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure S1 from Plasma MicroRNAs as Novel Biomarkers for Patients with Intraductal Papillary Mucinous Neoplasms of the Pancreas
- Author
-
Mokenge Malafa, Domenico Coppola, Anthony M. Magliocco, Barbara Ann Centeno, Kazim Husain, Christina Georgeades, Yonghong Zhang, Sean J. Yoder, William J. Fulp, Dung-Tsa Chen, and Jennifer Permuth-Wey
- Abstract
Flowchart for Statistical Analysis. Circulating miRNAs were evaluated in IPMN cases versus healthy controls and malignant versus benign IPMNs using linear models for microarray data (LIMMA), principal component analysis (PCA), and receiver operating characteristic (ROC) curve analyses, followed by pathway enrichment analyses. We also correlated abundance of plasma miRNAs and tissue miRNAs for cases with both sample types.
- Published
- 2023
- Full Text
- View/download PDF
13. Data from Plasma MicroRNAs as Novel Biomarkers for Patients with Intraductal Papillary Mucinous Neoplasms of the Pancreas
- Author
-
Mokenge Malafa, Domenico Coppola, Anthony M. Magliocco, Barbara Ann Centeno, Kazim Husain, Christina Georgeades, Yonghong Zhang, Sean J. Yoder, William J. Fulp, Dung-Tsa Chen, and Jennifer Permuth-Wey
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most fatal cancers worldwide, partly because methods are lacking to detect disease at an early, operable stage. Noninvasive PDAC precursors called intraductal papillary mucinous neoplasms (IPMN) exist, and strategies are needed to aid in their proper diagnosis and management. Data support the importance of miRNAs in the progression of IPMNs to malignancy, and we hypothesized that miRNAs may be shed from IPMN tissues and detected in blood. Our primary goals were to measure the abundance of miRNAs in archived preoperative plasma from individuals with pathologically confirmed IPMNs and healthy controls and discover plasma miRNAs that distinguish between IPMN patients and controls and between “malignant” and “benign” IPMNs. Using novel nCounter technology to evaluate 800 miRNAs, we showed that a 30-miRNA signature distinguished 42 IPMN cases from 24 controls [area underneath the curve (AUC) = 74.4; 95% confidence interval (CI), 62.3–86.5, P = 0.002]. The signature contained novel miRNAs and miRNAs previously implicated in pancreatic carcinogenesis that had 2- to 4-fold higher expression in cases than controls. We also generated a 5-miRNA signature that discriminated between 21 malignant (high-grade dysplasia and invasive carcinoma) and 21 benign (low- and moderate-grade dysplasia) IPMNs (AUC = 73.2; 95% CI, 57.6–88.9, P = 0.005), and showed that paired plasma and tissue samples from patients with IPMNs can have distinct miRNA expression profiles. This study suggests feasibility of using new cost-effective technology to develop a miRNA-based blood test to aid in the preoperative identification of malignant IPMNs that warrant resection while sparing individuals with benign IPMNs the morbidity associated with overtreatment. Cancer Prev Res; 8(9); 826–34. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
14. Supp Figures from Global Phosphoproteomics Reveal CDK Suppression as a Vulnerability to KRas Addiction in Pancreatic Cancer
- Author
-
Saïd M. Sebti, José Trevino, John Koomen, Eric A. Welsh, Mokenge Malafa, Jason B. Fleming, Patrick Underwood, Kazim Husain, Terence M. Williams, Bin Fang, Francisca Beato, Hua Yang, Rajanikanth Vangipurapu, Shengyan Xiang, Liwei Chen, and Aslamuzzaman Kazi
- Abstract
Supplementary Figures
- Published
- 2023
- Full Text
- View/download PDF
15. Data from Global Phosphoproteomics Reveal CDK Suppression as a Vulnerability to KRas Addiction in Pancreatic Cancer
- Author
-
Saïd M. Sebti, José Trevino, John Koomen, Eric A. Welsh, Mokenge Malafa, Jason B. Fleming, Patrick Underwood, Kazim Husain, Terence M. Williams, Bin Fang, Francisca Beato, Hua Yang, Rajanikanth Vangipurapu, Shengyan Xiang, Liwei Chen, and Aslamuzzaman Kazi
- Abstract
Purpose:Among human cancers that harbor mutant (mt) KRas, some, but not all, are dependent on mt KRas. However, little is known about what drives KRas dependency.Experimental Design:Global phosphoproteomics, screening of a chemical library of FDA drugs, and genome-wide CRISPR/Cas9 viability database analysis were used to identify vulnerabilities of KRas dependency.Results:Global phosphoproteomics revealed that KRas dependency is driven by a cyclin-dependent kinase (CDK) network. CRISPR/Cas9 viability database analysis revealed that, in mt KRas-driven pancreatic cancer cells, knocking out the cell-cycle regulators CDK1 or CDK2 or the transcriptional regulators CDK7 or CDK9 was as effective as knocking out KRas. Furthermore, screening of a library of FDA drugs identified AT7519, a CDK1, 2, 7, and 9 inhibitor, as a potent inducer of apoptosis in mt KRas-dependent, but not in mt KRas-independent, human cancer cells. In vivo AT7519 inhibited the phosphorylation of CDK1, 2, 7, and 9 substrates and suppressed growth of xenografts from 5 patients with pancreatic cancer. AT7519 also abrogated mt KRas and mt p53 primary and metastatic pancreatic cancer in three-dimensional (3D) organoids from 2 patients, 3D cocultures from 8 patients, and mouse 3D organoids from pancreatic intraepithelial neoplasia, primary, and metastatic tumors.Conclusions:A link between CDK hyperactivation and mt KRas dependency was uncovered and pharmacologically exploited to abrogate mt KRas-driven pancreatic cancer in highly relevant models, warranting clinical investigations of AT7519 in patients with pancreatic cancer.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Data from Global Phosphoproteomics Reveal CDK Suppression as a Vulnerability to KRas Addiction in Pancreatic Cancer
- Author
-
Saïd M. Sebti, José Trevino, John Koomen, Eric A. Welsh, Mokenge Malafa, Jason B. Fleming, Patrick Underwood, Kazim Husain, Terence M. Williams, Bin Fang, Francisca Beato, Hua Yang, Rajanikanth Vangipurapu, Shengyan Xiang, Liwei Chen, and Aslamuzzaman Kazi
- Abstract
Supplementary Figures Legend
- Published
- 2023
- Full Text
- View/download PDF
17. Role of environmental toxicants in the development of hypertensive and cardiovascular diseases
- Author
-
Ehsan, Habeeb, Saad, Aldosari, Shakil A, Saghir, Mariam, Cheema, Tahani, Momenah, Kazim, Husain, Yadollah, Omidi, Syed A A, Rizvi, Muhammad, Akram, and Rais A, Ansari
- Subjects
Health, Toxicology and Mutagenesis ,Toxicology - Abstract
The incidence of hypertension with diabetes mellitus (DM) as a co-morbid condition is on the rise worldwide. In 2000, an estimated 972 million adults had hypertension, which is predicted to grow to 1.56 billion by 2025. Hypertension often leads to diabetes mellitus that strongly puts the patients at an increased risk of cardiovascular, kidney, and/or atherosclerotic diseases. Hypertension has been identified as a major risk factor for the development of diabetes; patients with hypertension are at two-to-three-fold higher risk of developing diabetes than patients with normal blood pressure (BP). Causes for the increase in hypertension and diabetes are not well understood, environmental factors (e.g., exposure to environmental toxicants like heavy metals, organic solvents, pesticides, alcohol, and urban lifestyle) have been postulated as one of the reasons contributing to hypertension and cardiovascular diseases (CVD). The mechanism of action(s) of these toxicants in developing hypertension and CVDs is not well defined. Research studies have linked hypertension with the chronic consumption of alcohol and exposure to metals like lead, mercury, and arsenic have also been linked to hypertension and CVD. Workers chronically exposed to styrene have a higher incidence of CVD. Recent studies have demonstrated that exposure to particulate matter (PM) in diesel exhaust and urban air contributes to increased CVD and mortality. In this review, we have imparted the role of environmental toxicants such as heavy metals, organic pollutants, PM, alcohol, and some drugs in hypertension and CVD along with possible mechanisms and limitations in extrapolating animal data to humans.
- Published
- 2022
- Full Text
- View/download PDF
18. Protein kinase 2 (CK2): a potential regulator of immune cell development and function in cancer
- Author
-
Tanika T. Williamson, Nadine Nelson, Tomar Ghansah, and Kazim Husain
- Subjects
Immunology ,Protein Serine-Threonine Kinases ,cancer chemotherapy ,immune cells ,Neoplasms ,Tumor Microenvironment ,Humans ,Immunology and Allergy ,Casein Kinase II ,Protein kinase A ,Protein kinase B ,Chemistry ,Kinase ,fungi ,NF-kappa B ,Wnt signaling pathway ,RC581-607 ,Cell biology ,ck2 inhibitors and immunotherapy ,protein kinase ck2 ,Casein kinase 2 ,Signal transduction ,Immunologic diseases. Allergy ,Janus kinase ,Tyrosine kinase ,Signal Transduction - Abstract
Protein kinase CK2, formally known as casein kinase II, is ubiquitously expressed and highly conserved serine/threonine or tyrosine kinase enzyme that regulates diverse signaling pathways responsible for cellular processes (i.e., cell proliferation and apoptosis) via interactions with over 500 known substrates. The enzyme's physiological interactions and cellular functions have been widely studied, most notably in the blood and solid malignancies. CK2 has intrinsic role in carcinogenesis as overexpression of CK2 subunits (α, α`, and β) and deregulation of its activity have been linked to various forms of cancers. CK2 also has extrinsic role in cancer stroma or in the tumor microenvironment (TME) including the immune cells. However, very few research studies have focused on extrinsic role of CK2 in regulating immune responses as a therapeutic alternative for cancer. The following review discusses CK2's regulation of key signaling events [Nuclear factor kappa B (NF-κB), Janus kinase/signal transducer and activators of transcription (JAK/STAT), Hypoxia inducible factor-1alpha (HIF-1α), Cyclooygenase-2 (COX-2), Extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK), Notch, Protein kinase B/AKT, Ikaros and Wnt] that can influence the development and function of immune cells in cancer. Potential clinical trials using potent CK2 inhibitors will facilitate and improve the treatment of human malignancies.
- Published
- 2021
19. Global Phosphoproteomics Reveal CDK Suppression as a Vulnerability to KRas Addiction in Pancreatic Cancer
- Author
-
Terence M. Williams, Shengyan Xiang, Francisca Beato, Hua Yang, Jose G. Trevino, Jason B. Fleming, Bin Fang, Said M. Sebti, Kazim Husain, Aslamuzzaman Kazi, Mokenge P. Malafa, Liwei Chen, rajanikanth vangipurapu, John M. Koomen, Eric A. Welsh, and Patrick W. Underwood
- Subjects
0301 basic medicine ,Cancer Research ,endocrine system diseases ,Proteome ,Pancreatic Intraepithelial Neoplasia ,medicine.disease_cause ,Article ,Proto-Oncogene Proteins p21(ras) ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cyclin-dependent kinase ,Pancreatic cancer ,medicine ,Animals ,Humans ,Phosphorylation ,neoplasms ,Cyclin-dependent kinase 1 ,biology ,Kinase ,Phosphoproteomics ,medicine.disease ,digestive system diseases ,Cyclin-Dependent Kinases ,Pancreatic Neoplasms ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,KRAS ,Cyclin-dependent kinase 7 - Abstract
Purpose: Among human cancers that harbor mutant (mt) KRas, some, but not all, are dependent on mt KRas. However, little is known about what drives KRas dependency. Experimental Design: Global phosphoproteomics, screening of a chemical library of FDA drugs, and genome-wide CRISPR/Cas9 viability database analysis were used to identify vulnerabilities of KRas dependency. Results: Global phosphoproteomics revealed that KRas dependency is driven by a cyclin-dependent kinase (CDK) network. CRISPR/Cas9 viability database analysis revealed that, in mt KRas-driven pancreatic cancer cells, knocking out the cell-cycle regulators CDK1 or CDK2 or the transcriptional regulators CDK7 or CDK9 was as effective as knocking out KRas. Furthermore, screening of a library of FDA drugs identified AT7519, a CDK1, 2, 7, and 9 inhibitor, as a potent inducer of apoptosis in mt KRas-dependent, but not in mt KRas-independent, human cancer cells. In vivo AT7519 inhibited the phosphorylation of CDK1, 2, 7, and 9 substrates and suppressed growth of xenografts from 5 patients with pancreatic cancer. AT7519 also abrogated mt KRas and mt p53 primary and metastatic pancreatic cancer in three-dimensional (3D) organoids from 2 patients, 3D cocultures from 8 patients, and mouse 3D organoids from pancreatic intraepithelial neoplasia, primary, and metastatic tumors. Conclusions: A link between CDK hyperactivation and mt KRas dependency was uncovered and pharmacologically exploited to abrogate mt KRas-driven pancreatic cancer in highly relevant models, warranting clinical investigations of AT7519 in patients with pancreatic cancer.
- Published
- 2021
- Full Text
- View/download PDF
20. Role of Flavonoids as Epigenetic Modulators in Cancer Prevention and Therapy
- Author
-
Nishat Fatima, Syed Shabihe Raza Baqri, Atrayee Bhattacharya, Nii Koney-Kwaku Koney, Kazim Husain, Ata Abbas, and Rais A. Ansari
- Subjects
Review ,QH426-470 ,Biology ,DNA sequencing ,chemistry.chemical_compound ,Gene expression ,Genetics ,medicine ,cancer ,Epigenetics ,Genetics (clinical) ,DNA methylation ,Cancer prevention ,epigenetics ,histone modifications ,food and beverages ,Cancer ,medicine.disease ,Histone ,chemistry ,flavonoids ,Cancer research ,biology.protein ,non-coding RNAs ,Molecular Medicine ,DNA - Abstract
Epigenetic regulation involves reversible changes in histones and DNA modifications that can be inherited without any changes in the DNA sequence. Dysregulation of normal epigenetic processes can lead to aberrant gene expression as observed in many diseases, notably cancer. Recent insights into the mechanisms of DNA methylation, histone modifications, and non-coding RNAs involved in altered gene expression profiles of tumor cells have caused a paradigm shift in the diagnostic and therapeutic approaches towards cancer. There has been a surge in search for compounds that could modulate the altered epigenetic landscape of tumor cells, and to exploit their therapeutic potential against cancers. Flavonoids are naturally occurring phenol compounds which are abundantly found among phytochemicals and have potentials to modulate epigenetic processes. Knowledge of the precise flavonoid-mediated epigenetic alterations is needed for the development of epigenetics drugs and combinatorial therapeutic approaches against cancers. This review is aimed to comprehensively explore the epigenetic modulations of flavonoids and their anti-tumor activities.
- Published
- 2021
- Full Text
- View/download PDF
21. Targeting the IκB Kinase Enhancer and Its Feedback Circuit in Pancreatic Cancer☆
- Author
-
Jin Q. Cheng, Domenico Coppola, Surinder K. Batra, Richard B. Kim, Sridevi Challa, Kazim Husain, and Mokenge P. Malafa
- Subjects
0301 basic medicine ,Trametinib ,MAPK/ERK pathway ,Cancer Research ,Original article ,business.industry ,MEK inhibitor ,IκB kinase ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Cancer stem cell ,030220 oncology & carcinogenesis ,Pancreatic cancer ,Cancer research ,medicine ,IKBKE ,ERBB3 ,business - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease with an overall median 5-year survival rate of 8%. This poor prognosis is because of the development of resistance to chemotherapy and radiation therapy and lack of effective targeted therapies. IκB kinase enhancer (IKBKE) overexpression was previously implicated in chemoresistance. Because IKBKE is frequently elevated in PDAC and IKBKE inhibitors are currently in clinical trials, we evaluated IKBKE as a therapeutic target in this disease. Depletion of IKBKE was found to significantly reduce PDAC cell survival, growth, cancer stem cell renewal, and cell migration and invasion. Notably, IKBKE inhibitor CYT387 and IKBKE knockdown dramatically activated the MAPK pathway. Phospho-RTK array analyses showed that IKBKE inhibition leads to rapid upregulation of ErbB3 and IGF-1R expression, which results in MAPK-ERK pathway activation-thereby limiting the efficacy of IKBKE inhibitors. Furthermore, IKBKE inhibition leads to stabilization of FOXO3a, which is required for RTK upregulation on IKBKE inhibition. Finally, we demonstrated that the IKBKE inhibitors synergize with the MEK inhibitor trametinib to significantly induce cell death and inhibit tumor growth and liver metastasis in an orthotopic PDAC mouse model.
- Published
- 2020
22. Recombinant Protein Production: from Bench to Biopharming
- Author
-
Rais A. Ansari, Shakil A. Saghir, Rebecca Torisky, and Kazim Husain
- Published
- 2021
- Full Text
- View/download PDF
23. Apigenin Targets MicroRNA-155, Enhances SHIP-1 Expression, and Augments Anti-Tumor Responses in Pancreatic Cancer
- Author
-
Kazim Husain, Krystal Villalobos-Ayala, Valentina Laverde, Oscar A. Vazquez, Bradley Miller, Samra Kazim, George Blanck, Margaret L. Hibbs, Gerald Krystal, Isra Elhussin, Joakin Mori, Clayton Yates, and Tomar Ghansah
- Subjects
Cancer Research ,Oncology ,SHIP-1 ,miR-155 ,tumor microenvironment ,myelopoiesis ,MDSC ,TAM ,pancreatic cancer ,apigenin - Abstract
Pancreatic cancer (PC) is a deadly disease with a grim prognosis. Pancreatic tumor derived factors (TDF) contribute to the induction of an immunosuppressive tumor microenvironment (TME) that impedes the effectiveness of immunotherapy. PC-induced microRNA-155 (miRNA-155) represses expression of Src homology 2 (SH2) domain-containing Inositol 5′-phosphatase-1 (SHIP-1), a regulator of myeloid cell development and function, thus impacting anti-tumor immunity. We recently reported that the bioflavonoid apigenin (API) increased SHIP-1 expression which correlated with the expansion of tumoricidal macrophages (TAM) and improved anti-tumor immune responses in the TME of mice with PC. We now show that API transcriptionally regulates SHIP-1 expression via the suppression of miRNA-155, impacting anti-tumor immune responses in the bone marrow (BM) and TME of mice with PC. We discovered that API reduced miRNA-155 in the PC milieu, which induced SHIP-1 expression. This promoted the restoration of myelopoiesis and increased anti-tumor immune responses in the TME of heterotopic, orthotopic and transgenic SHIP-1 knockout preclinical mouse models of PC. Our results suggest that manipulating SHIP-1 through miR-155 may assist in augmenting anti-tumor immune responses and aid in the therapeutic intervention of PC.
- Published
- 2022
- Full Text
- View/download PDF
24. Apigenin Increases SHIP-1 Expression, Promotes Tumoricidal Macrophages and Anti-Tumor Immune Responses in Murine Pancreatic Cancer
- Author
-
Margaret L. Hibbs, DeVon DeLoach, Kun Jiang, Gerald Krystal, Kazim Husain, Ciara Alvarez, Krystal Villalobos-Ayala, Ivannie Ortiz Rivera, and Tomar Ghansah
- Subjects
Cancer Research ,Myeloid ,medicine.medical_treatment ,pancreatic cancer ,Inflammation ,lcsh:RC254-282 ,Article ,Immune system ,Pancreatic cancer ,medicine ,Macrophage ,apigenin ,Tumor microenvironment ,Cell growth ,business.industry ,Immunotherapy ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,tumor immunity ,medicine.anatomical_structure ,Oncology ,Cancer research ,medicine.symptom ,SHIP-1 ,business ,myeloid homeostasis - Abstract
Pancreatic cancer (PC) has an extremely poor prognosis due to the expansion of immunosuppressive myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM) in the inflammatory tumor microenvironment (TME), which halts the recruitment of effector immune cells and renders immunotherapy ineffective. Thus, the identification of new molecular targets that can modulate the immunosuppressive TME is warranted for PC intervention. Src Homology-2 (SH2) domain-containing Inositol 5&prime, Phosphatase-1 (SHIP-1) is a lipid signaling protein and a regulator of myeloid cell development and function. Herein, we used the bioflavonoid apigenin (API) to reduce inflammation in different PC models. Wild type mice harboring heterotopic or orthotopic PC were treated with API, which induced SHIP-1 expression, reduced inflammatory tumor-derived factors (TDF), increased the proportion of tumoricidal macrophages and enhanced anti-tumor immune responses, resulting in a reduction in tumor burden compared to vehicle-treated PC mice. In contrast, SHIP-1-deficient mice exhibited an increased tumor burden and displayed augmented proportions of pro-tumor macrophages. These results provide further support for the importance of SHIP-1 expression in promoting pro-tumor macrophage development in the pancreatic TME. Our findings suggest that agents augmenting SHIP-1 expression may provide novel therapeutic options for the treatment of PC.
- Published
- 2020
- Full Text
- View/download PDF
25. δ-Tocotrienol, a natural form of vitamin E, inhibits pancreatic cancer stem-like cells and prevents pancreatic cancer metastasis
- Author
-
Mokenge P. Malafa, Domenico Coppola, Said M. Sebti, Jose G. Trevino, Kazim Husain, and Barbara A. Centeno
- Subjects
0301 basic medicine ,Genetically modified mouse ,Pathology ,medicine.medical_specialty ,Epithelial-Mesenchymal Transition ,endocrine system diseases ,Angiogenesis ,pancreatic cancer ,Antineoplastic Agents ,Apoptosis ,Metastasis ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,SOX2 ,Cell Movement ,Cell Line, Tumor ,Pancreatic cancer ,medicine ,metastasis ,Animals ,Humans ,Vitamin E ,Cell Self Renewal ,Neoplasm Metastasis ,Transcription factor ,Cell Proliferation ,Neovascularization, Pathologic ,business.industry ,EMT ,Cancer ,invasion ,medicine.disease ,Xenograft Model Antitumor Assays ,digestive system diseases ,3. Good health ,Pancreatic Neoplasms ,Disease Models, Animal ,VEDT ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Cancer research ,Tocotrienol ,business ,Research Paper - Abstract
// Kazim Husain 1 , Barbara A. Centeno 2 , Domenico Coppola 2 , Jose Trevino 4 , Said M. Sebti 3 and Mokenge P. Malafa 1 1 Departments of Gastrointestinal Oncology, Tampa, FL, USA 2 Departments of Pathology, Tampa, FL, USA 3 Departments of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA 4 Department of Surgery, University of Florida, Gainesville, FL, USA Correspondence to: Mokenge P. Malafa, email: mokenge.malafa@moffitt.org Keywords: VEDT, pancreatic cancer, metastasis, invasion, EMT Received: November 16, 2016 Accepted: January 27, 2017 Published: February 28, 2017 ABSTRACT The growth, metastasis, and chemotherapy resistance of pancreatic ductal adenocarcinoma (PDAC) is characterized by the activation and growth of tumor-initiating cells in distant organs that have stem-like properties. Thus, inhibiting growth of these cells may prevent PDAC growth and metastases. We have demonstrated that δ-tocotrienol, a natural form of vitamin E (VEDT), is bioactive against cancer, delays progression, and prevents metastases in transgenic mouse models of PDAC. In this report, we provide the first evidence that VEDT selectively inhibits PDAC stem-like cells. VEDT inhibited the viability, survival, self-renewal, and expression of Oct4 and Sox2 transcription factors in 3 models of PDAC stem-like cells. In addition, VEDT inhibited the migration, invasion, and several biomarkers of epithelial-to-mesenchymal transition and angiogenesis in PDAC cells and tumors. These processes are critical for tumor metastases. Furthermore, in the L3.6pl orthotopic model of PDAC metastases, VEDT significantly inhibited growth and metastases of these cells. Finally, in an orthotopic xenograft model of human PDAC stem-like cells, we showed that VEDT significantly retarded the growth and metastases of gemcitabine-resistant PDAC human stem-like cells. Because VEDT has been shown to be safe and to reach bioactive levels in humans, this work supports investigating VEDT for chemoprevention of PDAC metastases.
- Published
- 2017
- Full Text
- View/download PDF
26. Vitamin E δ-tocotrienol sensitizes human pancreatic cancer cells to TRAIL-induced apoptosis through proteasome-mediated down-regulation of c-FLIPs
- Author
-
Surinder K. Batra, Said M. Sebti, Sean Z. Hutchinson, Mokenge P. Malafa, Rony A. Francois, Chen Wang, Michael Kongnyuy, Anying Zhang, Domenico Coppola, and Kazim Husain
- Subjects
Cancer Research ,c-FLIP ,medicine.medical_treatment ,Apoptosis ,TRAIL ,lcsh:RC254-282 ,03 medical and health sciences ,Vitamin E Compound ,chemistry.chemical_compound ,0302 clinical medicine ,Pancreatic tumor ,Pancreatic cancer ,Genetics ,medicine ,lcsh:QH573-671 ,lcsh:Cytology ,Chemistry ,Vitamin E ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,3. Good health ,δ-Tocotrienol ,Oncology ,Proteasome ,030220 oncology & carcinogenesis ,Cancer research ,Tumor necrosis factor alpha ,Tocotrienol ,Primary Research - Abstract
Background Vitamin E δ-tocotrienol (VEDT), a vitamin E compound isolated from sources such as palm fruit and annatto beans, has been reported to have cancer chemopreventive and therapeutic effects. Methods We report a novel function of VEDT in augmenting tumor necrosis factor-related apoptosis-inducing ligand- (TRAIL-) induced apoptosis in pancreatic cancer cells. The effects of VEDT were shown by its ability to trigger caspase-8-dependent apoptosis in pancreatic cancer cells. Results When combined with TRAIL, VEDT significantly augmented TRAIL-induced apoptosis of pancreatic cancer cells. VEDT decreased cellular FLICE inhibitory protein (c-FLIP) levels without consistently modulating the expression of decoy death receptors 1, 2, 3 or death receptors 4 and 5. Enforced expression of c-FLIP substantially attenuated VEDT/TRAIL-induced apoptosis. Thus, c-FLIP reduction plays an important part in mediating VEDT/TRAIL-induced apoptosis. Moreover, VEDT increased c-FLIP ubiquitination and degradation but did not affect its transcription, suggesting that VEDT decreases c-FLIP levels through promoting its degradation. Of note, degradation of c-FLIP and enhanced TRAIL-induced apoptosis in pancreatic cancer cells were observed only with the anticancer bioactive vitamin E compounds δ-, γ-, and β-tocotrienol but not with the anticancer inactive vitamin E compounds α-tocotrienol and α-, β-, γ-, and δ-tocopherol. Conclusions c-FLIP degradation is a key event for death receptor-induced apoptosis by anticancer bioactive vitamin E compounds in pancreatic cancer cells. Moreover, VEDT augmented TRAIL inhibition of pancreatic tumor growth and induction of apoptosis in vivo. Combination therapy with TRAIL agonists and bioactive vitamin E compounds may offer a novel strategy for pancreatic cancer intervention. Electronic supplementary material The online version of this article (10.1186/s12935-019-0876-0) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
27. Chemoprevention of Azoxymethane-induced Colon Carcinogenesis by Delta-Tocotrienol
- Author
-
Kazim Husain, Chung S. Yang, Anying Zhang, Domenico Coppola, Ashley H. Davis-Yadley, Steven C. Shivers, and Mokenge P. Malafa
- Subjects
0301 basic medicine ,Cancer Research ,Colorectal cancer ,Angiogenesis ,Carcinogenesis ,Azoxymethane ,Apoptosis ,Malignant transformation ,Metastasis ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Movement ,medicine ,Tumor Cells, Cultured ,Animals ,Vitamin E ,Wnt Signaling Pathway ,Cell Proliferation ,business.industry ,Cancer ,medicine.disease ,Rats, Inbred F344 ,Rats ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Cancer research ,Carcinogens ,Female ,Tocotrienol ,business - Abstract
This study evaluated the preclinical activity of δ-tocotrienol (DT3), a bioactive form of vitamin E, in the inhibition of colorectal cancer growth and development in vitro and in vivo. DT3 is the most bioactive isomer of vitamin E in inhibiting growth of colorectal cancer cells. However, it had little effect on the proliferation of normal colon mucosal cells NCM460. In HCT-116 and SW-620 colorectal cancer cells, DT3 (50 μmol/L) significantly inhibited malignant transformation (P < 0.02, P < 0.001), cell migration (P < 0.02, P < 0.05), and invasion (P < 0.05, P < 0.01) compared with vehicle. DT3 inhibited markers for epithelial (E-cadherin) to mesenchymal (vimentin) transition, metastasis (matrix metalloproteinase 9), angiogenesis VEGF, inflammation (NF-κB), and Wnt signaling (β-catenin) compared with vehicle in colorectal cancer cells. DT3 induced apoptosis selectively in colorectal cancer cells (SW-620 cells, HCT-116 cells, and HT-29) without affecting the normal colon cells. In the azoxymethane-induced colorectal carcinogenesis model in rats, DT3 (200 mg/kg orally twice a day) for 20 weeks significantly inhibited colorectal polyps by 70% and colorectal cancer by almost 99% compared with the vehicle treatment group (P < 0.02, P < 0.001), and the cancer inhibition effect was more potent than sulindac (50%). Taken together, these data demonstrate that DT3 is a potential chemopreventive agent in colorectal cancer, warranting further investigation into its clinical use in the prevention and treatment of colorectal cancer.
- Published
- 2018
28. A Phase I Safety, Pharmacokinetic, and Pharmacodynamic Presurgical Trial of Vitamin E δ-tocotrienol in Patients with Pancreatic Ductal Neoplasia
- Author
-
Kazim Husain, Said M. Sebti, Dung-Tsa Chen, Anthony Neuger, Mokenge P. Malafa, Richard Lush, Barbara A. Centeno, Gregory M. Springett, and Tai Hutchinson
- Subjects
Oncology ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Presurgical trial ,lcsh:Medicine ,Antineoplastic Agents ,Apoptosis ,Chemoprevention ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacokinetics ,Internal medicine ,Pancreatic cancer ,Preoperative Care ,medicine ,Humans ,Vitamin E ,Adverse effect ,030304 developmental biology ,Aged ,Aged, 80 and over ,0303 health sciences ,lcsh:R5-920 ,business.industry ,Tocotrienols ,lcsh:R ,General Medicine ,Middle Aged ,medicine.disease ,3. Good health ,Clinical trial ,Pancreatic Neoplasms ,Treatment Outcome ,Tolerability ,chemistry ,030220 oncology & carcinogenesis ,Neoplastic cell ,Female ,Tocotrienol ,business ,lcsh:Medicine (General) ,Biomarkers ,Research Article ,Carcinoma, Pancreatic Ductal - Abstract
Background Vitamin E δ-tocotrienol (VEDT), a natural vitamin E from plants, has shown anti-neoplastic and chemoprevention activity in preclinical models of pancreatic cancer. Here, we investigated VEDT in patients with pancreatic ductal neoplasia in a window-of-opportunity preoperative clinical trial to assess its safety, tolerability, pharmacokinetics, and apoptotic activity. Methods Patients received oral VEDT at escalating doses (from 200 to 3200 mg) daily for 13 days before surgery and one dose on the day of surgery. Dose escalation followed a three-plus-three trial design. Our primary endpoints were safety, VEDT pharmacokinetics, and monitoring of VEDT-induced neoplastic cell apoptosis (ClinicalTrials.gov number NCT00985777). Findings In 25 treated patients, no dose-limiting toxicity was encountered; thus no maximum-tolerated dose was reached. One patient had a drug-related adverse event (diarrhea) at a 3200-mg daily dose level. The effective half-life of VEDT was ~ 4 h. VEDT concentrations in plasma and exposure profiles were quite variable but reached levels that are bioactive in preclinical models. Biological activity, defined as significant induction of apoptosis in neoplastic cells as measured by increased cleaved caspase-3 levels, was seen in the majority of patients at the 400-mg to 1600-mg daily dose levels. Interpretation VEDT from 200 to 1600 mg daily taken orally for 2 weeks before pancreatic surgery was well tolerated, reached bioactive levels in blood, and significantly induced apoptosis in the neoplastic cells of patients with pancreatic ductal neoplasia. These promising results warrant further clinical investigation of VEDT for chemoprevention and/or therapy of pancreatic cancer., Highlights • Vitamin E δ-tocotrienol is the bioactive form of one of the natural vitamin E with activity against cancer cells • Vitamin E δ-tocotrienol is safe in patients up to 3200 mg • Vitamin E δ-tocotrienol selectively kills pancreatic tumor cells when compared with normal cells at 400, 600, and 800 mg/day • The biomarker effect of vitamin E δ-tocotrienol suggest significant anticancer activity in patients, justifying further study Vitamin E has been an intriguing vitamin to humans for its potential to promote human health. However, large-scale research with vitamin E to prevent cancer has had mixed results. Because recent laboratory studies have shown that the form of vitamin E used in previous interventions to reduce cancer risk have not been the active tocotrienol form of vitamin E, there is a question as to whether the lack of vitamin activity is due to the use of inactive forms of vitamin E in clinical trials. Based on our laboratory data, which showed that the vitamin E δ-tocotrienol (VEDT) form of vitamin E was active against pancreatic cancer, we tested the ability of VEDT to kill pancreatic tumor cells in patients using a window-of-opportunity design, with measurement of apoptosis as an intermediate endpoint. We found that VEDT was well tolerated at up to 3200 mg when taken for 2 weeks before surgery. We also found that, at doses of 400 to 800 mg, VEDT selectively killed pancreatic tumor cells.
- Published
- 2015
29. Plasma MicroRNAs as Novel Biomarkers for Patients with Intraductal Papillary Mucinous Neoplasms of the Pancreas
- Author
-
Dung-Tsa Chen, Mokenge P. Malafa, Kazim Husain, Sean J. Yoder, Domenico Coppola, Jennifer Permuth-Wey, William J. Fulp, Barbara A. Centeno, Anthony M. Magliocco, Yonghong Zhang, and Christina Georgeades
- Subjects
Male ,Quality Control ,Cancer Research ,Pathology ,medicine.medical_specialty ,Databases, Factual ,endocrine system diseases ,Malignancy ,Article ,Biomarkers, Tumor ,medicine ,Carcinoma ,Humans ,Blood test ,Prospective Studies ,Stage (cooking) ,Prospective cohort study ,Aged ,Retrospective Studies ,medicine.diagnostic_test ,business.industry ,Retrospective cohort study ,Middle Aged ,medicine.disease ,Carcinoma, Papillary ,Pancreatic Neoplasms ,MicroRNAs ,medicine.anatomical_structure ,Oncology ,Dysplasia ,Area Under Curve ,Preoperative Period ,Disease Progression ,Female ,Pancreas ,business ,Carcinoma, Pancreatic Ductal - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most fatal cancers worldwide, partly because methods are lacking to detect disease at an early, operable stage. Noninvasive PDAC precursors called intraductal papillary mucinous neoplasms (IPMN) exist, and strategies are needed to aid in their proper diagnosis and management. Data support the importance of miRNAs in the progression of IPMNs to malignancy, and we hypothesized that miRNAs may be shed from IPMN tissues and detected in blood. Our primary goals were to measure the abundance of miRNAs in archived preoperative plasma from individuals with pathologically confirmed IPMNs and healthy controls and discover plasma miRNAs that distinguish between IPMN patients and controls and between “malignant” and “benign” IPMNs. Using novel nCounter technology to evaluate 800 miRNAs, we showed that a 30-miRNA signature distinguished 42 IPMN cases from 24 controls [area underneath the curve (AUC) = 74.4; 95% confidence interval (CI), 62.3–86.5, P = 0.002]. The signature contained novel miRNAs and miRNAs previously implicated in pancreatic carcinogenesis that had 2- to 4-fold higher expression in cases than controls. We also generated a 5-miRNA signature that discriminated between 21 malignant (high-grade dysplasia and invasive carcinoma) and 21 benign (low- and moderate-grade dysplasia) IPMNs (AUC = 73.2; 95% CI, 57.6–88.9, P = 0.005), and showed that paired plasma and tissue samples from patients with IPMNs can have distinct miRNA expression profiles. This study suggests feasibility of using new cost-effective technology to develop a miRNA-based blood test to aid in the preoperative identification of malignant IPMNs that warrant resection while sparing individuals with benign IPMNs the morbidity associated with overtreatment. Cancer Prev Res; 8(9); 826–34. ©2015 AACR.
- Published
- 2015
- Full Text
- View/download PDF
30. Contributors
- Author
-
Bharat Bhushan Aggarwal, Navneet Agnihotri, Nur S. Amirruddin, Ritu Aneja, Sanjeev Banerjee, Chetna Bhandari, Alok Chandra Bharti, Anjali Bhat, Devivasha Bordoloi, Arup Chakraborty, Rahul Checker, Arpit Dheeraj, Patrick J. Gilhooley, Subash Chandra Gupta, Kuzhuvelil B. Harikumar, Kazim Husain, Mohit Jadli, Sankar Jagadeeshan, Abhimanyu Kumar Jha, Vijay P. Kale, Alan P. Kumar, Sandeep Kumar, Rani Kumari, Ajaikumar B. Kunnumakkara, Sukh Mahendra Singh, Mokenge P. Malafa, Sabira Mohammed, Ali Nabavizadeh, Shivakumari Asha Nair, Deepti Pande, Manoj K. Pandey, Mansi A. Parasramka, Shivani B. Paruthy, Raghavendra S. Patwardhan, Sangita Phadtare, M. Manu Prasad, Sahdeo Prasad, Vipin Rai, Priyanka Rajan, Kavita Rawat, Santosh K. Sandur, Roopali Saxena, Gautam Sethi, Deepak Sharma, Prerna Sharma, Anju Shrivastava, Anand Swaroop Shukla, Rana P. Singh, Surya P. Singh, Tejveer Singh, Bokyung Sung, Saima Syeda, Dhanir Tailor, Adrian K.K. Teo, Amit K. Tyagi, and Kanchan Vishnoi
- Published
- 2018
- Full Text
- View/download PDF
31. Role of Tocotrienols in Chemosensitization of Cancer
- Author
-
Mokenge P. Malafa and Kazim Husain
- Subjects
Bortezomib ,business.industry ,Vitamin E ,medicine.medical_treatment ,Cancer ,Pharmacology ,medicine.disease ,Capecitabine ,Gefitinib ,Docetaxel ,Chemosensitization ,medicine ,Erlotinib ,business ,medicine.drug - Abstract
Chemotherapy is one of the standard methods of treatment in many cancers. However, most of the tumors develop resistance to chemotherapeutic agents leading to recurrence, metastasis, and ultimately death of the patients. Therefore, a strategy to overcome chemoresistance and sensitize tumor cells to antitumor agents is urgently required. One promising strategy is to use dietary nutraceutical that sensitizes tumors to the chemotherapeutics. Tocotrienols are unsaturated vitamin E nutraceuticals found in fruits, vegetables, cereal grains, and essential oils and have potent antitumor activities without any clinical toxicity. In this monograph, we discussed the role of tocotrienols in chemosensitization of tumors to chemotherapeutic agents. The tumors sensitized by tocotrienols are pancreatic, breast, lung, colorectal, gastric, prostate, melanoma, bladder, hepatic, malignant mesothelioma, and multiple myeloma. The chemotherapeutic agents used are gemcitabine, capecitabine, tamoxifen, paclitaxel/docetaxel, doxorubicin/epirubicin, cisplatin, erlotinib/gefitinib, thalidomide/bortezomib, Met inhibitor, TRAIL, autophagy inducer, celecoxib, and statins. The chemosensitization of tumor cells by tocotrienols is mediated through modulating multiple signaling molecules such as cell proliferative proteins, cell survival proteins, nuclear factor-kappa B (NF-κB), signal transducer and activator of transcription-3 (STAT-3), HMG-CoA reductase, growth factor receptor, and apoptosis signaling pathways. These studies suggest that tocotrienols are promising agents used to sensitize tumors to standard chemotherapeutics. Future studies should be focused on both the efficacy and the safety of tocotrienols used in combination with chemotherapeutic agents in cancer patients.
- Published
- 2018
- Full Text
- View/download PDF
32. Response of Antioxidant System to Physical and Chemical Stress
- Author
-
Satu M. Somani, Kazim Husain, and Eric C. Schlorff
- Subjects
Stress (mechanics) ,Antioxidant ,Chemistry ,medicine.medical_treatment ,medicine ,Food science - Published
- 2017
- Full Text
- View/download PDF
33. Adiponectin expression and the cardioprotective role of the vitamin D receptor activator paricalcitol and the angiotensin converting enzyme inhibitor enalapril in ApoE-deficient mice
- Author
-
Kazim Husain, León Ferder, and Edu B Suárez-Martínez
- Subjects
Paricalcitol ,medicine.medical_specialty ,Blotting, Western ,Adipokine ,Angiotensin-Converting Enzyme Inhibitors ,Coronary Artery Disease ,Polymerase Chain Reaction ,Calcitriol receptor ,Article ,Mice ,Apolipoproteins E ,Enalapril ,Internal medicine ,medicine ,Animals ,Pharmacology (medical) ,biology ,Adiponectin ,business.industry ,Activator (genetics) ,Mice, Inbred C57BL ,Nitric oxide synthase ,Disease Models, Animal ,Oxidative Stress ,Endocrinology ,Gene Expression Regulation ,Ergocalciferols ,biology.protein ,RNA ,Receptors, Calcitriol ,Drug Therapy, Combination ,Female ,P22phox ,Cardiology and Cardiovascular Medicine ,business ,medicine.drug - Abstract
Background: Coronary heart disease (CHD) is the number one cause of death in the US. The adipokine adiponectin has been studied intensively for presenting and inversed association with almost every stage of CHD. For instance, the evaluation of molecules capable of enhancing endogenous adiponectin expression is well justified. In this study, we investigated the effect of the vitamin D receptor activator (VDRA) paricalcitol and the angiotensin-converting enzyme inhibitor (ACEI) enalapril on adiponectin expression, lipid profiles, adenosine monophosphate activated protein kinase (AMPK) expression, monocyte chemo-attractant protein-1 (MCP-1), tumor necrosis factor-alpha (TNFα),cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), antioxidant capacity, CuZn-superoxide dismutase (CuZn-SOD), Mn-SOD, NADPH p22phox subunits, inducible nitric oxidesynthase (iNOS), endothelial marker eNOS, and 81 atherosclerosis-related genes in ApoE-deficient mice. Method: Seven-week-old ApoE-deficient mice were treated for 16 weeks as follows: Group 1, ApoE vehicle control (intraperitoneal [i.p.] 100 µl propylene glycol); Group 2, ApoE-paricalcitol (200 ng i.p., 3/week); Group 3, ApoE-Enalapril (30 mg/kg daily); Group 4, ApoE-paricalcitol + enalapril (described dosing); and Group 5, wild-type control (C57BLV). Results: All treated groups presented significant changes in circulating and cardiac adiponectin, cardiac cholesterol levels, AMPK, MCP-1, TNF-α, COX-2, iNOS, eNOS, CuZn-SOD, Mn-SOD and p22phox. There were 15 genes that differed in their expression, 5 of which are involved in cardioprotection and antithrombotic mechanisms: Bcl2a1a, Col3a1, Spp1 (upregulated), Itga2, and Vwf (downregulated). Conclusion: Together, our data presented a novel role for VDRA and ACEI in reducing factors associated with CHD that may lead to the discovery of new therapeutic venues.
- Published
- 2014
- Full Text
- View/download PDF
34. MK-2206, an Akt inhibitor, enhances carboplatinum/paclitaxel efficacy in gastric cancer cell lines
- Author
-
Christopher L. Cubitt, Khaldoun Almhanna, Sabiha Kazim, Shumin M. Zhang, Said M. Sebti, Daniel C. Sullivan, Kazim Husain, and Mokenge P. Malafa
- Subjects
Cancer Research ,Paclitaxel ,Cell Survival ,medicine.medical_treatment ,Antineoplastic Agents ,Pharmacology ,Carboplatin ,chemistry.chemical_compound ,Stomach Neoplasms ,Cell Line, Tumor ,Humans ,Medicine ,Cytotoxicity ,Cell Proliferation ,Chemotherapy ,business.industry ,Cell growth ,Cancer ,Drug Synergism ,Combination chemotherapy ,medicine.disease ,Oncology ,chemistry ,MK-2206 ,Molecular Medicine ,Drug Screening Assays, Antitumor ,business ,Heterocyclic Compounds, 3-Ring ,Proto-Oncogene Proteins c-akt ,Research Paper - Abstract
Background Several molecularly-targeted agents are being evaluated in gastric cancer cell lines. In this study we evaluated the synergistic potential of MK-2206, an oral potent allosteric Akt inhibitor, in combination with chemotherapeutic agents in human gastric cancer cell lines. Materials and Methods We evaluated effects of MK-2206 on cell growth and cell signaling using a panel of gastric cancer cell lines AGS, SNU-1 and SNU 16. The analysis of drug combinations was conducted by using CellTiter-Blue™ Cell Viability Assay which yielded the combination index (CI). MK-2206 and representative chemotherapy agent were further evaluated regarding their effects on Akt inhibition and downstream targets using western blots probed with the appropriate antibodies. We assessed the combination of MK-2206 and chemotherapy in three different treatment sequences. Results We demonstrated in vitro synergistic efficacy of MK-2206 when combined with carboplatinum and paclitaxel in the three cell lines examined. Efficacy was dose dependent. We assessed the combination of MK-2206 and carboplatinum/paclitaxel in three different treatment sequences; 24 h of exposure to combination chemotherapy followed by a 48 h exposure to MK-2206 resulted in the highest synergistic antiproliferative effect in all cell lines. On the other hand, the reverse sequence (MK-2206 followed by chemotherapy) and the concurrent treatment schedule were slightly synergistic or additive as well. The effects of MK-2206 on p-Akt and other downstream targets was reported. Conclusions Our findings suggest that Akt inhibition augments the efficacy of existing gastric cancer therapeutics (carboplatinum and paclitaxel); thus, MK-2206 is a promising agent to treat gastric cancer patients who receive these cytotoxic agents. The magnitude of synergy depended on the treatment sequence; a schedule of MK-2206 dosed before or concurrently with chemotherapy was not as effective as being dosed after chemotherapy. Further experiments addressing MK-2206’s mechanism of action in combination with chemotherapy are needed.
- Published
- 2013
- Full Text
- View/download PDF
35. Vitamin E δ-Tocotrienol Prolongs Survival in the LSL-KrasG12D/+;LSL-Trp53R172H/+;Pdx-1-Cre (KPC) Transgenic Mouse Model of Pancreatic Cancer
- Author
-
Said M. Sebti, Kazim Husain, Dung Tsa Chen, Barbara A. Centeno, Mokenge P. Malafa, and Sunil R. Hingorani
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Male ,Genetically modified mouse ,Cancer Research ,medicine.medical_specialty ,Epithelial-Mesenchymal Transition ,Genotype ,medicine.medical_treatment ,Apoptosis ,Enzyme-Linked Immunosorbent Assay ,Mice, Transgenic ,Biology ,Deoxycytidine ,Article ,Mice ,Random Allocation ,chemistry.chemical_compound ,Internal medicine ,Pancreatic cancer ,Biomarkers, Tumor ,medicine ,Animals ,Vitamin E ,Cell Proliferation ,bcl-2-Associated X Protein ,Cell growth ,Body Weight ,medicine.disease ,Immunohistochemistry ,Gemcitabine ,Pancreatic Neoplasms ,Platelet Endothelial Cell Adhesion Molecule-1 ,Disease Models, Animal ,Genes, ras ,Treatment Outcome ,Endocrinology ,Oncology ,chemistry ,Drug Therapy, Combination ,Female ,Tocotrienol ,Poly(ADP-ribose) Polymerases ,Cyclin-Dependent Kinase Inhibitor p27 ,Signal Transduction ,medicine.drug - Abstract
Previous work has shown that vitamin E δ-tocotrienol (VEDT) prolongs survival and delays progression of pancreatic cancer in the LSL-KrasG12D/+;Pdx-1-Cre mouse model of pancreatic cancer. However, the effect of VEDT alone or in combination with gemcitabine in the more aggressive LSL-KrasG12D/+;LSL-Trp53R172H/+;Pdx-1-Cre (KPC) mouse model is unknown. Here, we studied the effects of VEDT and the combination of VEDT and gemcitabine in the KPC mice. KPC mice were randomized into four groups: (i) vehicle [olive oil, 1.0 mL/kg per os twice a day and PBS 1.0 mL/kg intrapertoneally (i.p.) twice a week], (ii) gemcitabine (100 mg/kg i.p. twice a week), (iii) VEDT (200 mg/kg per os twice a day), and (iv) gemcitabine + VEDT. Mice received treatment until they displayed symptoms of impending death from pancreatic cancer, at which point animals were euthanized. At 16 weeks, survival was 10% in the vehicle group, 30% in the gemcitabine group, 70% in the VEDT group (P < 0.01), and 90% in the VEDT combined with gemcitabine group (P < 0.05). VEDT alone and combined with gemcitabine resulted in reversal of epithelial-to-mesenchymal transition in tumors. Biomarkers of apoptosis (plasma CK18), PARP1 cleavage, and Bax expression were more greatly induced in tumors subjected to combined treatment versus individual treatment. Combined treatment induced cell-cycle inhibitors (p27Kip1 and p21Cip1) and inhibited VEGF, vascularity (CD31), and oncogenic signaling (pAKT, pMEK, and pERK) greater than individual drugs. No significant differences in body weight gain between drug treatment and control mice were observed. These results strongly support further investigation of VEDT alone and in combination with gemcitabine for pancreatic cancer prevention and treatment. Cancer Prev Res; 6(10); 1074–83. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
36. Role of Transcription Factors in Steatohepatitis and Hypertension after Ethanol: The Epicenter of Metabolism
- Author
-
Rais A Ansari, Syed A. A. Rizvi, and Kazim Husain
- Subjects
0301 basic medicine ,Alcoholic liver disease ,medicine.medical_specialty ,hypertension ,Alcohol Drinking ,lcsh:QR1-502 ,steatohepatitis ,Review ,Biochemistry ,lcsh:Microbiology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Humans ,Ethanol metabolism ,STAT3 ,Molecular Biology ,Transcription factor ,transcription factor ,Alcohol dehydrogenase ,biology ,Acetaldehyde ,medicine.disease ,Sterol regulatory element-binding protein ,Fatty Liver ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,Hypoxia-inducible factors ,chemistry ,biology.protein ,030211 gastroenterology & hepatology ,ethanol ,gene regulation ,Transcription Factors ,alcoholic liver disease - Abstract
Chronic alcohol consumption induces multi-organ damage, including alcoholic liver disease (ALD), pancreatitis and hypertension. Ethanol and ethanol metabolic products play a significant role in the manifestation of its toxicity. Ethanol metabolizes to acetaldehyde and produces reduced nicotinamide adenine dinucleotide (NADH) by cytosolic alcohol dehydrogenase. Ethanol metabolism mediated by cytochrome-P450 2E1 causes oxidative stress due to increased production of reactive oxygen species (ROS). Acetaldehyde, increased redox cellular state and ROS activate transcription factors, which in turn activate genes for lipid biosynthesis and offer protection of hepatocytes from alcohol toxicity. Sterol regulatory element binding proteins (SREBPs) and peroxisome proliferator activated-receptors (PPARs) are two key lipogenic transcription factors implicated in the development of fatty liver in alcoholic and non-alcoholic steatohepatitis. SREBP-1 is activated in the livers of chronic ethanol abusers. An increase in ROS activates nuclear factor erythroid-2-related factor-2 (Nrf2) and hypoxia inducible factor (HIF) to provide protection to hepatocytes from ethanol toxicity. Under ethanol exposure, due to increased gut permeability, there is release of gram-negative bacteria-derived lipopolysaccharide (LPS) from intestine causing activation of immune response. In addition, the metabolic product, acetaldehyde, modifies the proteins in hepatocyte, which become antigens inviting auto-immune response. LPS activates macrophages, especially the liver resident macrophages, Kupffer cells. These Kupffer cells and circulating macrophages secrete various cytokines. The level of tumor necrosis factor-α (TNFα), interleukin-1beta (IL-1β), IL-6, IL-8 and IL-12 have been found elevated among chronic alcoholics. In addition to elevation of these cytokines, the peripheral iron (Fe(2+)) is also mobilized. An increased level of hepatic iron has been observed among alcoholics. Increased ROS, IL-1β, acetaldehyde, and increased hepatic iron, all activate nuclear factor-kappa B (NF-κB) transcription factor. Resolution of increased reactive oxygen species requires increased expression of genes responsible for dismutation of increased ROS which is partially achieved by IL-6 mediated activation of signal transducers and activators of transcription 3 (STAT3). In addition to these transcription factors, activator protein-1 may also be activated in hepatocytes due to its association with resolution of increased ROS. These transcription factors are central to alcohol-mediated hepatotoxicity.
- Published
- 2016
37. Role of Angiotensin Peptides Precursor in Ethanol Mediated Hepato toxicity: Perspective on Angiotensinogen
- Author
-
Kazim Husain, Rais A Ansari, Syed A. A. Rizvi, and Natalia Osna
- Subjects
Hepatitis ,medicine.medical_specialty ,Ethanol ,Cirrhosis ,business.industry ,medicine.disease ,chemistry.chemical_compound ,Liver disease ,Endocrinology ,chemistry ,Fibrosis ,Internal medicine ,Toxicity ,Renin–angiotensin system ,Medicine ,Steatosis ,business - Abstract
Alcohol usage poses serious social and economical challenges. Its overuse is responsible for loss of billions of dollars due to fatal accidents and causes multiple diseases. Chronic alcoholism leads to liver disorders, including steatosis, hepatitis, fibrosis and cirrhosis. Both binge and chronic alcohol drinking result in blood pressure elevation and hyperten- sion. As a significant mechanism, changes in the levels of angiotensinogen- released peptides play an important role in advanced alcohol liver disease. This review summarizes some of the studies that have demonstrated their role in liver fi- brosis, cirrhosis and hypertension. Further, the individual variants and polymorphic sites may play a role in alcohol- mediated regulation of angiotensinogen. Possible role of human angiotensinogen on alcohol hepatotoxicity is also dis- cussed.
- Published
- 2012
- Full Text
- View/download PDF
38. Effect of Sulfhydryl Modification on Rat Kidney Basolateral Plasma Membrane Transport Function
- Author
-
Syed A. A. Rizvi, William O. Berndt, Anastasios Lymperopoulos, Kazim Husain, and Rais A Ansari
- Subjects
Health, Toxicology and Mutagenesis ,Kidney ,Toxicology ,Hazardous Substances ,Dithiothreitol ,Nephrotoxicity ,Cell membrane ,chemistry.chemical_compound ,medicine ,Animals ,Sulfhydryl Compounds ,Cell Membrane ,Biological Transport ,General Medicine ,Basolateral plasma membrane ,Glutathione ,Pollution ,In vitro ,Rats ,Membrane ,medicine.anatomical_structure ,chemistry ,Biochemistry ,p-Aminohippuric Acid - Abstract
Transport processes are the hallmark of functioning kidney. Various nephrotoxicants disrupt the transport processes to manifest nephrotoxicity. Of several nephrotoxicants, mercuric chloride (HgCl(2)) depletes the reduced glutathione (GSH) in kidney and has been observed to affect the in vitro p-aminohippurate (PAH) transport by basolateral (BL) membrane vesicles. The role of renal nonprotein sulfhydryls such as, reduced GSH has been demonstrated to affect the PAH transport by BL membrane vesicles. The role of protein sulfhydryls in transport process of PAH by BL membrane is not known. Due to mercury mediated effects on sulfhydryls, the effects of protein-sulfhydryls (-SH) modifying reagents in the current study were investigated on PAH transport by BL membrane. It was observed that modification of -SH by p-chloromercuribenzoate sulphate (pCMBS), and mercuric chloride (HgCl(2)) decreased while recovering the protein -SH with dithiothreitol treatment provided protection against the effects of pCMBS, and HgCl(2) on PAH transport by BL membrane vesicles.
- Published
- 2012
- Full Text
- View/download PDF
39. Chronic ethanol ingestion induces aortic inflammation/oxidative endothelial injury and hypertension in rats
- Author
-
León Ferder, Kazim Husain, Rais A Ansari, and Jainarine Lalla
- Subjects
Male ,medicine.medical_specialty ,Health, Toxicology and Mutagenesis ,Aorta, Thoracic ,Blood Pressure ,Inflammation ,Vasodilation ,Nitric Oxide ,Toxicology ,medicine.disease_cause ,Article ,Nitric oxide ,chemistry.chemical_compound ,Internal medicine ,medicine.artery ,medicine ,Animals ,Thoracic aorta ,chemistry.chemical_classification ,Reactive oxygen species ,Dose-Response Relationship, Drug ,Ethanol ,Superoxide Dismutase ,Tumor Necrosis Factor-alpha ,General Medicine ,Glutathione ,Rats, Inbred F344 ,Rats ,Oxidative Stress ,Endocrinology ,Blood pressure ,chemistry ,Hypertension ,Tumor necrosis factor alpha ,Endothelium, Vascular ,medicine.symptom ,Reactive Oxygen Species ,Oxidative stress - Abstract
The study aim was to investigate the relationship of chronic ethanol-induced inflammation leading to vascular endothelial injury and elevation of blood pressure (BP) in a rat model. Male Fisher rats were divided into two groups of six animals each and treated as follows: (1) Control (5% sucrose, orally) daily for 12 weeks and (2) 20% ethanol (4 g kg-1, orally) daily for 12 weeks. The mean arterial blood pressure was recorded every week. The animals were anesthetized with pentobarbital after 12 weeks; thoracic aorta were isolated and analyzed for aortic reactivity response, inflammatory mediators, oxidant/antioxidant enzyme protein expression and endothelial nitric oxide-generating system. The results show that the mean BP was significantly elevated 12 weeks after ethanol ingestion. The increased BP was related to increased aortic inflammation (tumor necrosis factor [TNF]-α; nitric oxide synthase [iNOS], COX-2 and MCP-1 protein expression) and elevated angiotensin II levels in alcohol-treated group compared to control. Aortic Nicotinamide adenine dinucleotide phosphate reduced (NADPH) oxidase activity, membrane and cytosolic subunits p22 phox and p47 phox expression and Mn-SOD activity and protein expression significantly increased, whereas nitric oxide (NO), endothelial NO synthase (eNOS), vascular endothelial growth factor (VEGF)-A and CuZn-SOD activity and protein expression significantly decreased in alcohol-treated group compared to control. The acetylcholine-mediated vasorelaxation response was depressed in the aorta of ethanol-treated rats compared to control. In conclusion, chronic ethanol-induced elevation in BP is related to increased aortic inflammation, elevated angiotensin II levels, induction of NADPH oxidase causing endothelial injury, depletion of CuZn-SOD, down-regulation of endothelial NO generating system and impaired vascular relaxation in rats.
- Published
- 2010
- Full Text
- View/download PDF
40. Mo1986 - Chemoprevention of Azoxymethane-Induced Colon Carcinogenesis by Delta-Tocotrienol
- Author
-
Mokenge P. Malafa, Ashley H. Davis-Yadley, Anying Zhang, Steve Shivers, Kazim Husain, and Domenico Coppola
- Subjects
Delta ,chemistry.chemical_compound ,Hepatology ,Chemistry ,Azoxymethane ,Gastroenterology ,Cancer research ,Tocotrienol ,Colon carcinogenesis - Published
- 2018
- Full Text
- View/download PDF
41. Vitamin E δ-Tocotrienol Levels in Tumor and Pancreatic Tissue of Mice after Oral Administration
- Author
-
Sean Z. Hutchinson, Domenico Coppola, Richard Lush, Anthony Neuger, Said M. Sebti, Rony A. Francois, Mokenge P. Malafa, and Kazim Husain
- Subjects
medicine.medical_specialty ,medicine.medical_treatment ,Transplantation, Heterologous ,Administration, Oral ,Mice, Nude ,Biology ,Neuroprotection ,Mice ,chemistry.chemical_compound ,Pharmacokinetics ,Oral administration ,Cell Line, Tumor ,Internal medicine ,medicine ,Animals ,Vitamin E ,Tissue Distribution ,Pancreas ,Pharmacology ,Original Paper ,General Medicine ,biochemical phenomena, metabolism, and nutrition ,Pancreatic Neoplasms ,Transplantation ,Endocrinology ,medicine.anatomical_structure ,Liver ,chemistry ,Toxicity ,Female ,Tocotrienol ,Neoplasm Transplantation ,Half-Life - Abstract
Tocotrienols are natural vitamin E compounds that are known to have a neuroprotective effect at nanomolar concentration and anti-carcinogenic effect at micromolar concentration. In this report, we investigated the pharmacokinetics, tumor and pancreatic tissue levels, and toxicity of δ-tocotrienol in mice because of its anti-tumor activity against pancreatic cancer. Following a single oral administration of δ-tocotrienol at 100 mg/kg, the peak plasma concentration (Cmax) was 57 ± 5 μmol/l, the time required to reach peak plasma concentration (Tmax) was 2 h and plasma half-life (t1/2) was 3.5 h. The δ-tocotrienol was cleared from plasma and liver within 24 h, but delayed from the pancreas. When mice were fed δ-tocotrienol for 6 weeks, the concentration in tumor tissue was 41 ± 3.5 nmol/g. This concentration was observed with the oral dose (100 mg/kg) of δ-tocotrienol which inhibited tumor growth by 80% in our previous studies. Interestingly, δ-tocotrienol was 10-fold more concentrated in the pancreas than in the tumor. We observed no toxicity due to δ-tocotrienol as mice gained normal weight with no histopathological changes in tissues. Our data suggest that bioactive levels of δ-tocotrienol can be achieved in the pancreas following oral administration and supports its clinical investigation in pancreatic cancer.
- Published
- 2009
- Full Text
- View/download PDF
42. Combination Therapy with Paricalcitol and Enalapril Ameliorates Cardiac Oxidative Injury in Uremic Rats
- Author
-
Kazim Husain, Eduardo Slatopolsky, Jane Finch, Masahide Mizobuchi, and León Ferder
- Subjects
Paricalcitol ,medicine.medical_specialty ,Heart Diseases ,Combination therapy ,Angiotensin-Converting Enzyme Inhibitors ,Antioxidants ,Rats, Sprague-Dawley ,Pharmacotherapy ,Enalapril ,Malondialdehyde ,Internal medicine ,medicine ,Animals ,Oxidative injury ,cardiovascular diseases ,Uremia ,integumentary system ,biology ,Superoxide Dismutase ,business.industry ,NADPH Oxidases ,medicine.disease ,Rats ,Oxidative Stress ,Endocrinology ,Nephrology ,Enzyme inhibitor ,Ergocalciferols ,biology.protein ,Vitamin D Analog ,Drug Therapy, Combination ,Female ,business ,hormones, hormone substitutes, and hormone antagonists ,circulatory and respiratory physiology ,medicine.drug - Abstract
Aims: This study investigated the protective effect of the angiotensin-converting enzyme inhibitor, enalapril, and the vitamin D analog, paricalcitol, alone or in combination, on cardiac oxidative stress in uremic rats. Methods: Rats were made uremic by 5/6 nephrectomy and treated for 4 months as follows: (1) uremic + vehicle (n = 11); (2) uremic + enalapril (30 mg/l in drinking water, n = 13); (3) uremic + paricalcitol (200 ng 3× week, n = 6); (4) uremic + enalapril + paricalcitol (n = 14), and (5) controls (n = 6). Results: Cardiac NADPH oxidase activity increased by 300% in uremic rats compared to normal controls. Treatment with enalapril, paricalcitol or the combination of the two protected uremic rats from cardiac oxidative stress by inhibiting enzyme activity. Cardiac malondialdehyde (MDA) levels were significantly increased in uremic rats compared to normal controls. Only the combination therapy inhibited the increase in MDA levels in uremic rats. Cardiac glutathione was significantly reduced in uremic rats compared to normal controls. Enalapril, paricalcitol or the two in combination all protected against this reduction in glutathione. Cardiac copper/zinc superoxide dismutase (CuZn-SOD) activity decreased whereas manganese (Mn-SOD) activity increased in uremic rats compared to controls. Both mono and combination therapies ameliorated the alterations in cardiac SOD activity seen in uremic rats. Conclusion: Enalapril, paricalcitol and their combined therapy afford protection against cardiac oxidative stress in uremia.
- Published
- 2008
- Full Text
- View/download PDF
43. Dose-Dependent Protection by Lipoic Acid against Cisplatin-Induced Nephrotoxicity in Rats: Antioxidant Defense System
- Author
-
Gary L. Trammell, Kazim Husain, Mokenge P. Malafa, Leonard P. Rybak, Satu M. Somani, and Craig Whitworth
- Subjects
Pharmacology ,Cisplatin ,Antioxidant ,Chemistry ,Health, Toxicology and Mutagenesis ,medicine.medical_treatment ,Dose dependence ,Toxicology ,Nephrotoxicity ,Lipoic acid ,chemistry.chemical_compound ,Cisplatin induced nephrotoxicity ,medicine ,medicine.drug - Published
- 2008
- Full Text
- View/download PDF
44. Interaction of SIV/SHIV infection and morphine on plasma oxidant/antioxidant balance in macaque
- Author
-
Antonio Pérez-Casanova, Kazim Husain, Vanessa Rivera-Amill, Richard J. Noel, and Anil Kumar
- Subjects
medicine.medical_specialty ,Clinical Biochemistry ,Simian Acquired Immunodeficiency Syndrome ,Biology ,medicine.disease_cause ,Macaque ,Antioxidants ,Superoxide dismutase ,chemistry.chemical_compound ,Internal medicine ,biology.animal ,medicine ,Animals ,Molecular Biology ,chemistry.chemical_classification ,Morphine ,Glutathione peroxidase ,Cell Biology ,General Medicine ,Glutathione ,Simian immunodeficiency virus ,Oxidants ,Malondialdehyde ,Endocrinology ,chemistry ,Immunology ,biology.protein ,Macaca ,Simian Immunodeficiency Virus ,Viral load ,Oxidative stress - Abstract
A homeostatic balance exists between the cellular generation of oxidant species and endogenous antioxidants under normal physiological conditions. Human Immunodeficiency Virus (HIV) infection is known to affect this balance causing oxidative stress. However, the interaction of HIV infection with a substance abuse on cellular oxidant/antioxidant system is sparse. This study was designed in order to investigate the interactive effect of morphine abuse and Simian Immunodeficiency Virus/ Simian Human Immunodeficiency Virus (SIV/SHIV) infection on plasma oxidant/antioxidant balance in rhesus macaques. Six rhesus macaques adapted to morphine dependence (20 weeks) along with three controls were infected with mixture of SHIV(KU-1B), SHIV(89.6P), and SIV(17E-Fr). Plasma samples from morphine-dependent and control macaques were analyzed for an array of oxidative stress indices after 16 weeks of infection. Morphine-dependence significantly increased plasma malondialdehyde (MDA) and 8-isoprostane levels (8-fold and 2-fold), but these animals showed higher MDA and 8-isoprostane levels after viral infection (18-fold and 4-fold) which was directly correlated with increase in viral load and decline in CD4+ cells. Plasma glutathione (GSH) level depleted (55%) with morphine dependence that was further depleted (25%) by the infection. Activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) were increased by 30% and 110%, respectively with morphine dependence, but that was decreased by the infection. Catalase (CAT) activity declined (25%) with morphine dependence that was further declined by infection. Our results clearly suggest that morphine interaction with SIV/SHIV infection causes higher oxidative tissue injury that might have implication in the pathogenesis of AIDS in morphine-dependent macaques.
- Published
- 2007
- Full Text
- View/download PDF
45. Morphine-Mediated Deterioration of Oxidative Stress Leads to Rapid Disease Progression in SIV/SHIV-Infected Macaques
- Author
-
Antonio Pérez-Casanova, Richard J. Noel, Vanessa Rivera-Amill, Kazim Husain, and Anil Kumar
- Subjects
Immunology ,Simian Acquired Immunodeficiency Syndrome ,medicine.disease_cause ,Superoxide dismutase ,chemistry.chemical_compound ,Malondialdehyde ,Virology ,medicine ,Animals ,biology ,Superoxide Dismutase ,HIV ,Viral Load ,Simian immunodeficiency virus ,Catalase ,biology.organism_classification ,CD4 Lymphocyte Count ,Oxidative Stress ,Infectious Diseases ,chemistry ,Lentivirus ,Disease Progression ,biology.protein ,Morphine ,Macaca ,Simian Immunodeficiency Virus ,Morphine Dependence ,Viral load ,Oxidative stress ,medicine.drug - Abstract
Oxidative stress is well documented in HIV infection, but the effect of concomitant substance abuse is largely unknown. We studied oxidative stress in our macaque model of morphine abuse and AIDS. In plasma, we found an approximately 50% decrease in catalase activity with morphine dependence that was exacerbated by infection in rapid progressors. Superoxide dismutase was decreased by a similar degree, but only in the presence of both morphine and viral infection. The loss of these antioxidant systems was coincident with significantly increased plasma malondialdehyde upon viral infection that displayed a synergistic increase in conjunction with morphine and rapid disease.
- Published
- 2007
- Full Text
- View/download PDF
46. Down regulation of aortic nitric oxide and antioxidant systems in chronic alcohol-induced hypertension in rats
- Author
-
Manuel Vazquez-Ortiz, Kazim Husain, and Jainarine Lalla
- Subjects
Male ,Vascular Endothelial Growth Factor A ,0301 basic medicine ,medicine.medical_specialty ,Nitric Oxide Synthase Type III ,Endothelium ,Health, Toxicology and Mutagenesis ,Down-Regulation ,Gene Expression ,Blood Pressure ,Nitric Oxide ,Toxicology ,Antioxidants ,Nitric oxide ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,medicine.artery ,medicine ,Animals ,Thoracic aorta ,RNA, Messenger ,Endothelial dysfunction ,Aorta ,chemistry.chemical_classification ,Glutathione Peroxidase ,Ethanol ,030102 biochemistry & molecular biology ,Superoxide Dismutase ,business.industry ,Glutathione peroxidase ,General Medicine ,Glutathione ,Catalase ,medicine.disease ,Rats, Inbred F344 ,Rats ,Disease Models, Animal ,Endocrinology ,Blood pressure ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Hypertension ,Endothelium, Vascular ,business - Abstract
The aim of this study was to investigate the relationship between chronic ethanol-induced increase in blood pressure (BP) and alterations in the aortic nitric oxide (NO) and the antioxidant systems in rats. Male Fisher rats (200—250 g) were divided into two groups of six animals each and treated as follows: 1) control (5% sucrose, orally) daily for 12 weeks and 2) 20% ethanol (4 g/kg, orally) daily for 12 weeks. The BP (systolic, diastolic and mean) was recorded every week through tail-cuff method. The animals were sacrificed 12 weeks after treatments and thoracic aorta was collected and analysed. The results show that systolic, diastolic and mean BP was significantly elevated 12 weeks after ethanol ingestion in rats compared to control. The endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF) expressions were down-regulated (50—55% of control) leading to depletion of aortic NO levels (69% of control) in ethanol treated rats compared to control. The ratio of reduced to oxidized glutathione (GSH/GSSG) was significantly depleted (58% of control) in the aorta of ethanol-treated rats compared to control. The decrease in aortic GSH/GSSG ratio was good correlated with increase in BP ( r = 0.69). The antioxidant enzymes: copper/zinc-superoxide dismutase (CuZn-SOD) and manganese (Mn)-SOD, catalase (CAT) and glutathione peroxidase (GSH-Px) activities were significantly depressed (36—53% of control) in the aorta of ethanol-treated rats compared to control. The study concluded that chronic ethanol ingestion induces hypertension which relates to the vascular endothelial dysfunction on account of the down-regulation of aortic endothelial antioxidants and NO generating system in rats. Human & Experimental Toxicology (2007) 26, 427—434
- Published
- 2007
- Full Text
- View/download PDF
47. Knockdown of CSE1L Gene in Colorectal Cancer Reduces Tumorigenesis in Vitro
- Author
-
Dung Tsa Chen, Kevin G Neill, Kazim Husain, Steven A. Eschrich, Evita Henderson-Jackson, Mokenge P. Malafa, Jose M. Pimiento, Domenico Coppola, and David Shibata
- Subjects
0301 basic medicine ,Adenoma ,Adult ,Male ,Cytoplasm ,Carcinogenesis ,Apoptosis ,Biology ,medicine.disease_cause ,Pathology and Forensic Medicine ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Cellular Apoptosis Susceptibility Protein ,Cell Line, Tumor ,medicine ,Humans ,neoplasms ,Cellular localization ,Aged ,Cell Proliferation ,Regulation of gene expression ,Aged, 80 and over ,Cell Nucleus ,Gene knockdown ,Tissue microarray ,Epithelial Cells ,Regular Article ,Middle Aged ,Molecular biology ,digestive system diseases ,3. Good health ,Gene Expression Regulation, Neoplastic ,Protein Transport ,030104 developmental biology ,Tissue Array Analysis ,030220 oncology & carcinogenesis ,Immunohistochemistry ,Female ,Colorectal Neoplasms ,Cellular apoptosis susceptibility protein - Abstract
Human cellular apoptosis susceptibility (chromosomal segregation 1-like, CSE1L) gene plays a role in nuclear-to-cytoplasm transport and chromosome segregation during mitosis, cellular proliferation, and apoptosis. CSE1L is involved in colon carcinogenesis. CSE1L gene expression was assessed with three data sets using Affymetrix U133 + gene chips on normal human colonic mucosa (NR), adenomas (ADs), and colorectal carcinoma (CRC). CSE1L protein expression in CRC, AD, and NR from the same patients was measured by immunohistochemistry using a tissue microarray. We evaluated CSE1L expression in CRC cells (HCT116, SW480, and HT29) and its biological functions. CSE1L mRNA was significantly increased in all AD and CRC compared with NR (P
- Published
- 2015
48. Triciribine Phosphate Monohydrate, an AKT Inhibitor, Enhances Gemcitabine Activity in Pancreatic Cancer Cells
- Author
-
Richard, Kim, Teruo, Yamauchi, Kazim, Husain, Said, Sebti, and Mokenge, Malafa
- Subjects
Dose-Response Relationship, Drug ,Staining and Labeling ,Acenaphthenes ,Cell Survival ,Apoptosis ,Drug Synergism ,Trypan Blue ,Ribonucleotides ,Deoxycytidine ,Gemcitabine ,Pancreatic Neoplasms ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,Cell Adhesion ,Humans ,Protein Kinase Inhibitors ,Proto-Oncogene Proteins c-akt ,Cell Proliferation - Abstract
Pancreatic cancer is a highly lethal cancer due to early metastasis and resistance to current chemotherapeutic agents. Abnormal protein kinase B (AKT) activation is an important mechanism of chemoresistance to gemcitabine, the most widely used agent in pancreatic cancer.In the study, we tested the hypothesis that combining an AKT inhibitor with gemcitabine would augment anti-tumor activity. We treated human pancreatic cancer MiaPaCa-2 cells with gemcitabine and the AKT inhibitor triciribine, alone and in combination, and evaluated treatment effects using trypan blue assay, 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium (MTT) assay, and cell death enzyme-linked immunosorbant assay. Colorimetric data of MTT assay were computationally analyzed for synergism of the combination therapy by CalcuSyn2 (Biosoft, Great Shelford, Cambridge, UK).Both gemcitabine and triciribine inhibited cell growth in a dose-dependent manner. Triciribine synergistically enhanced the cytotoxic activity of gemcitabine. The combination index (CI) provides the synergistic, additive, or antagonistic effects of the two-drug combination. CI at the 50% effective dose at 1:500 ratio of gemcitabine to triciribine was 0.74, indicating the synergistic effect of the drugs. The combination treatment with the non-apoptotic dose of each agent distinctly induced apoptosis, with gemcitabine in combination with triciribine, synergistically inhibiting pancreatic cancer cell growth and inducing apoptosis.These findings support the use of triciribine to overcome activated AKT-mediated resistance of pancreatic cancer to gemcitabine.
- Published
- 2015
49. Selective Nuclear Export Inhibitor KPT-330 Enhances the Antitumor Activity of Gemcitabine in Human Pancreatic Cancer
- Author
-
Sabiha Kazim, Sherma Zibadi, Kazim Husain, Amit Mahipal, Tami Rashal, Yosef Landesman, Domenico Coppola, Daniel M. Sullivan, Marsha Crochiere, Mokenge P. Malafa, and Trinayan Kashyap
- Subjects
Cancer Research ,medicine.medical_specialty ,Cell Survival ,Blotting, Western ,Active Transport, Cell Nucleus ,Mice, Nude ,Apoptosis ,Deoxycytidine ,Article ,chemistry.chemical_compound ,In vivo ,Internal medicine ,Pancreatic cancer ,Cell Line, Tumor ,Survivin ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Cell Adhesion ,Animals ,Humans ,Cell Proliferation ,Microscopy, Confocal ,Dose-Response Relationship, Drug ,Cell growth ,Drug Synergism ,Triazoles ,medicine.disease ,Xenograft Model Antitumor Assays ,Gemcitabine ,Tumor Burden ,Pancreatic Neoplasms ,medicine.anatomical_structure ,Endocrinology ,Hydrazines ,Oncology ,chemistry ,Cancer research ,Female ,Pancreas ,Cyclin-Dependent Kinase Inhibitor p27 ,medicine.drug - Abstract
Pancreatic cancer is an aggressive and deadly malignancy responsible for the death of over 37,000 Americans each year. Gemcitabine-based therapy is the standard treatment for pancreatic cancer but has limited efficacy due to chemoresistance. In this study, we evaluated the in vitro and in vivo effects of gemcitabine combined with the selective nuclear export (CRM1) inhibitor KPT-330 on pancreatic cancer growth. Human pancreatic cancer MiaPaCa-2 and metastatic pancreatic cancer L3.6pl cell lines were treated with different concentrations of KPT-330 and gemcitabine alone or in combination, and anchorage–dependent/independent growth was recorded. In addition, L3.6pl cells with luciferase were injected orthotopically into the pancreas of athymic nude mice, which were treated with (i) vehicle (PBS 1 mL/kg i.p., 2/week and povidone/pluronic F68 1 mL/kg p.o., 3/week), (ii) KPT-330 (20 mg/kg p.o., 3/week), (iii) gemcitabine (100 mg/kg i.p., 2/week), or (iv) KPT-330 (10 mg/kg) + gemcitabine (50 mg/kg) for 4 weeks. KPT-330 and gemcitabine alone dose-dependently inhibited anchorage-dependent growth in vitro and tumor volume in vivo compared with vehicle treatment. However, the combination inhibited growth synergistically. In combination, KPT-330 and gemcitabine acted synergistically to enhance pancreatic cancer cell death greater than each single-agent therapy. Mechanistically, KPT-330 and gemcitabine promoted apoptosis, induced p27, depleted survivin, and inhibited accumulation of DNA repair proteins. Together, our data suggest that KPT-330 potentiates the antitumor activity of gemcitabine in human pancreatic cancer through inhibition of tumor growth, depletion of the antiapoptotic proteins, and induction of apoptosis. Mol Cancer Ther; 14(7); 1570–81. ©2015 AACR.
- Published
- 2015
50. Interaction of exercise and adenosine receptor agonist and antagonist on rat heart antioxidant defense system
- Author
-
Satu M. Somani and Kazim Husain
- Subjects
Male ,medicine.medical_specialty ,Antioxidant ,medicine.medical_treatment ,Clinical Biochemistry ,Glutathione reductase ,Antioxidants ,Lipid peroxidation ,chemistry.chemical_compound ,Oxygen Consumption ,Theophylline ,Physical Conditioning, Animal ,Internal medicine ,Purinergic P1 Receptor Agonists ,medicine ,Animals ,Disulfides ,Molecular Biology ,chemistry.chemical_classification ,Glutathione Peroxidase ,Superoxide Dismutase ,Myocardium ,Glutathione peroxidase ,Cell Biology ,General Medicine ,Glutathione ,Catalase ,Adenosine ,Adenosine receptor ,Rats, Inbred F344 ,Rats ,Glutathione Reductase ,Endocrinology ,chemistry ,Phenylisopropyladenosine ,Lipid Peroxidation ,medicine.drug - Abstract
This study investigated the interactive effects of acute exercise and adenosine receptor agonist and antagonist on antioxidant enzyme activities, glutathione and lipid peroxidation in the heart of the rat. Male Fisher-344 rats were divided into six groups and treated as follows: (1) saline control; (2) acute exercise (100% VO2max); (3) R-Phenyl isopropyl adenosine (R-PIA) (3.46 micromol/kg, i.p.); (4) theophylline (1.70 micromol/kg, i.p.) plus acute exercise; (5) theophylline plus R-PIA; and (6) theophylline. Animals were sacrificed 1 h after treatments; hearts were isolated and analyzed. The results show that acute exercise as well as adenosine receptor agonist R-PIA significantly enhanced cardiac superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), and glutathione reductase (GR) activity by 36-135% and 16-51%, respectively. Adenosine receptor agonist R-PIA significantly decreased cardiac GSSG concentration and enhanced GSH/GSSG ratio by 22 and 30%, respectively. Whereas theophylline treatment blocked the activation of antioxidant enzyme activities enhanced by acute exercise and R-PIA. Theophylline treatment significantly increased lipid peroxidation by 43% in the heart of exercised rats. The study concluded that the adenosine receptors are involved in the upregulation of cardiac antioxidant defense system and attenuation of lipid peroxidation due to acute exercise in rats.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.