Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 240
Filter
1.
Nanomedicine ; 44: 102580, 2022 08.
Article in English | MEDLINE | ID: mdl-35768037

ABSTRACT

Ultraviolet Beam (UVB) radiation is the main cause of skin cancer worldwide. Besides biocompatibility, the instability and limited skin permeability are the most challenging features of many effective photochemopreventive agents. (-)-Epigallocatechin-3-gallate (EGCG) is a natural polyphenolic compound extracted from Camellia sinensis that has been demonstrated to have antioxidant, anti-inflammatory, and anti-cancer properties. We evaluated the efficacy of three innovative EGCG nanoformulations in chemoprevention of UVB-induced DNA damage in keratinocytes. Results indicated that the EGCG nanoformulations reduced UVB-induced oxidative stress elevation and DNA damage. The nanoformulations also reduced the UVB-induced formation of pyrimidine and pyrimidone photoproducts in 2D human immortalized HaCaT keratinocytes and SKH-1 hairless mice through antioxidant effects and possibly through absorption of UVB radiation. In addition, EGCG nanoformulations inhibited UVB-induced chemokine/cytokine activation and promoted EGCG skin permeability and stability. Taken together, the results suggest the use of EGCG nanoformulations as potential natural chemopreventive agents during exposure to UVB radiation.


Subject(s)
Catechin , Animals , Antioxidants/pharmacology , Catechin/analogs & derivatives , Catechin/pharmacology , DNA Damage , Humans , Keratinocytes , Mice , Mice, Hairless , Skin , Ultraviolet Rays
2.
Semin Cancer Biol ; 83: 413-421, 2022 08.
Article in English | MEDLINE | ID: mdl-33484869

ABSTRACT

Living host system possess mechanisms like innate immune system to combat against inflammation, stress singling, and cancer. These mechanisms are initiated by PAMP and DAMP mediated recognition by PRR. PRR is consist of variety of nucleic acid sensors like-RNA sensors. They play crucial role in identifying exogenous and endogenous RNA molecules, which subsequently mediate pro/inflammatory cytokine, IFN and ISGs response in traumatized or tumorigenic conditions. The sensors can sensitize wide range of nucleic acid particle in term of size and structure, while each category sensors belongs subclasses with differentially expressed in cell and distinguished functioning mechanisms. They are also able to make comparison between self and non-self-nucleic acid molecules through specific mechanisms. Besides exhibiting anti-inflammatory and anti-tumorigenic responses, RNA sensors cover the broad spectrum of response mechanisms. Transcriptionally RNA sensors undergo with tight epigenetic regulations. In this review study, we will be going to discuss about the details of RNA sensors, their functional mechanisms and epi-transactional regulations.


Subject(s)
Nucleic Acids , RNA , Epigenesis, Genetic , Humans , Immunity, Innate/genetics , Inflammation , RNA/genetics
3.
Cancer Rep (Hoboken) ; 5(2): e1459, 2022 02.
Article in English | MEDLINE | ID: mdl-34245130

ABSTRACT

BACKGROUND: Data are steadily accruing that demonstrate that intestinal tumors are frequently derived from multiple founding cells, resulting in tumors comprised of distinct ancestral clones that might cooperate or alternatively compete, thereby potentially impacting different phases of the disease process. AIM: We sought to determine whether tumors with a multi-ancestral architecture involving at least two distinct clones show increased tumor number, growth, progression, or resistance to drug intervention. METHODS: Mice carrying the Min allele of Apc were generated that were mosaic with only a subset of cells in the intestinal epithelium expressing an activated form of PI3K, a key regulatory kinase affecting several important cellular processes. These cells were identifiable as they fluoresced green, whereas all other cells fluoresced red. RESULTS: Cell lineage tracing revealed that many intestinal tumors from our mouse model were derived from at least two founding cells, those expressing the activated PI3K (green) and those which did not (red). Heterotypic tumors with a multi-ancestral architecture as evidenced by a mixture of green and red cells exhibited increased tumor growth and invasiveness. Clonal architecture also had an impact on tumor response to low-dose aspirin. Aspirin treatment resulted in a greater reduction of heterotypic tumors derived from multiple founding cells as compared to tumors derived from a single founding cell. CONCLUSION: These data indicate that genetically distinct tumor-founding cells can contribute to early intratumoral heterogeneity. The coevolution of the founding cells and their progeny enhances colon tumor progression and impacts the response to aspirin. These findings are important to a more complete understanding of tumorigenesis with consequences for several distinct models of tumor evolution. They also have practical implications to the clinic. Mouse models with heterogenous tumors are likely better for predicting drug efficacy as compared to models in which the tumors are highly homogeneous. Moreover, understanding how interactions among different populations in a single heterotypic tumor with a multi-ancestral architecture impact response to a single agent and combination therapies are necessary to fully develop personalized medicine.


Subject(s)
Cell Transformation, Neoplastic/genetics , Intestinal Neoplasms/genetics , Animals , Antineoplastic Agents/pharmacology , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Disease Progression , Drug Resistance, Neoplasm/genetics , Intestinal Neoplasms/drug therapy , Intestinal Neoplasms/pathology , Mice , Mice, Transgenic
4.
Sci Rep ; 11(1): 11935, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099820

ABSTRACT

Maytenus roylanus (MEM) is a plant with anti-proliferative effects against prostate cancer. We aimed to explore the mechanism of action of MEM in prostate cancer (PCa) by employing an in vitro global proteome approach to get useful information of various signaling pathways and effected genes to define the mechanism of MEM action in prostate cancer. We conducted a global proteome analysis of CWR22Rv1after treatment with methanolic extract of MEM. The result of the proteomic profiling of in vitro PCa cells demonstrated the reduction in tumor protein D52 (TPD52) expression after treatment with methanolic extract of MEM. Down-regulation of TPD52 expression at mRNA level was observed by MEM treatment in CWR22Rν1 and C4-2 cells in a dose-dependent fashion probably by cleavage of Caspase 3 and PARP, or by modulation of cyclin-dependent kinases in CWR22Rν1 and C4-2 cells. The progressive character of the TRAMP model demonstrates a chance to evaluate the potential of chemo-preventive agents for both initial and late stages of prostate cancer development, and induction in TPD52 protein expression with development as well as the progression of prostate cancer was observed in the TRAMP model. Analyses of the tissue microarray collection of 25 specimens confirmed the clinical significance of our findings identifying TPD52 as a potential marker for PCa progression. We determined that knockdown of TPD52 (CWR22Rν1 cells), a considerable downregulation was seen at the protein level. Downregulation of TPD52 inhibited the migration and invasive behavior of prostate cancer cells as observed. Moreover, we observed that the siRNA-TPD52 transfection of CWR22Rν1 cells resulted in tumor growth inhibition with a marked reduction in the secretion of prostate-specific antigen (PSA) in the serum. Intraperitoneal injection of MEM considerably slowed tumor growth in athymic mice, inhibited TPD52 expression, and caused a marked reduction in PSA levels of serum as demonstrated by immunoblot screening and immune-histochemical staining. This report illustrates a molecular overview of pathological processes in PCa, indicating possible new disease biomarkers and therapeutic targets.


Subject(s)
Maytenus/chemistry , Neoplasm Proteins/metabolism , Plant Extracts/pharmacology , Prostatic Neoplasms/metabolism , Proteomics/methods , Tissue Array Analysis/methods , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice, Nude , Neoplasm Proteins/genetics , PC-3 Cells , Plant Extracts/administration & dosage , Plant Leaves/chemistry , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , RNA Interference , Xenograft Model Antitumor Assays/methods
5.
Semin Cancer Biol ; 69: 129-139, 2021 02.
Article in English | MEDLINE | ID: mdl-31866477

ABSTRACT

Nano metal organic frameworks (NMOFs) belong to the group of nanoporous materials. Over the decades, the conducted researches explored the area for the potential applications of NMOFs in areas like biomedical, chemical engineering and materials science. Recently, NMOFs have been explored for their potential use in cancer diagnosis and therapeutics. The excellent physico-chemical features of NMOFs also make them a potential candiadate to facilitate drug design, delivery and storage against cancer cells. In this review, we have explored the characterstic features, synthesis methods, NMOFs based drug delivery, diagnosis and imaging in various cancer types. In addition to this, we have also pondered on the stability and toxicological concerns of NMOFs. Despite, a significant research has been done for the potential use of NMOFs in cancer diagonostic and therapeutics, more information regarding the stability, in-vivo clearance, toxicology, and pharmacokinetics is still needed to ehnace the use of NMOFs in cancer diagonostic and therapeutics.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Metal-Organic Frameworks/administration & dosage , Nanomedicine , Nanoparticles/administration & dosage , Neoplasms/drug therapy , Animals , Humans , Metal-Organic Frameworks/chemistry , Nanoparticles/chemistry , Neoplasms/pathology
6.
Surgeon ; 19(5): e146-e152, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33121877

ABSTRACT

AIM: Early diagnosis of colorectal cancer (CRC) improves outcome. Straight-To-Test (STT) pathway was introduced in Whittington Hospital in 2012. The aim was to reduce the time to first oncological treatment and minimise unnecessary outpatient clinic appointments. However, this pathway has added significant burden to the trust in terms of number of procedures to be done.We assessed the diagnostic yield and the effectiveness of this pathway in improving the time to diagnosis of colorectal cancer. We also performed a cost-effective analysis and discussed the current literature along with interventions to further improve the benefits of STT investigations. METHOD: This is a prospectively collected data of all patients who underwent STT examinations in a single centre from January 2012 till December 2018. The parameters collected were patient details, procedures performed, findings and discharge plan. We also performed a cost-effective analysis. RESULTS: A total 1648 (90.8%) of patients identified suitable for STT pathway underwent colonoscopy or flexible sigmoidoscopy. From this, 764 (50.2%) patients had diagnosed pathology and CRC was detected in 50(3%) of the patients. We also estimated annual savings of £ 21,599.54 (£151,196.76 in seven years). Patients on the STT pathway took 25 days to obtain results as compared to 40 days in the standard pathway. The decision to take the patient off the cancer pathway was shortened by 3 weeks. CONCLUSION: STT pathway has proven to be safe and cost-effective means of investigation. However, further improvement is needed in the implementation to make it a sustainable. mode of investigation in long run and increase the pickup rate of colorectal cancer through STT.


Subject(s)
Colorectal Neoplasms , Early Detection of Cancer , Colonoscopy , Colorectal Neoplasms/diagnosis , Cost-Benefit Analysis , Humans , Mass Screening , Sigmoidoscopy
7.
Prostate ; 81(1): 41-49, 2021 01.
Article in English | MEDLINE | ID: mdl-33095939

ABSTRACT

INTRODUCTION OR OBJECTIVE: Men with favorable-risk prostate cancer (PCa) on active surveillance may benefit from intervention strategies to slow or prevent disease progression and the need for definitive treatment. Pomegranate and its extracts have shown antiproliferative and proapoptotic effects in cell lines and animal models, but its effect on human prostate cancer as a target tissue remain unclear. Objectives of this trial include pomegranate's ability to alter serum and prostate tissue biomarkers and the ability of an active surveillance cohort to adhere to a chemoprevention trial for 1 year. METHODS: Men with organ-confined, favorable-risk PCa on AS were randomly assigned to receive pomegranate fruit extract (PFE) 1000 mg (n = 15) or placebo (n = 15) once daily for twelve months. Prostate biopsies were performed at study entry and upon completion of the 1-year intervention. Plasma and urinary biomarkers were analyzed utilizing immunoassays and HPLC. Tissue proteins were assessed by immunohistochemistry (IHC) and measured by automated quantitation. RESULTS: PFE was well-tolerated with no significant toxicities. One patient withdrew before study initiation and 29 completed the 1-year intervention. No differences in plasma insulin-like growth factor-1 (IGF-1) levels, prostate-specific antigen doubling time, or biopsy kinetics were observed. Metabolites including urolithin A and urolithin A-gluc were detected more frequently in the PFE arm in both urine and plasma (p < .001 and p = .006, respectively). IHC analyses revealed reductions from baseline in 8-OHdG (a DNA damage marker) (p = .01) and androgen receptor expression (p = .04) in prostate tumor associated with PFE treatment. CONCLUSION: PFE administration for 12-month was well-tolerated and the protocol followed in an active surveillance population. Analyses suggest that PFE contains bioactive compounds capable of altering biomarkers involving oxidative stress and androgen signaling in prostate tumor and normal-appearing adjacent tissue. No alterations in the IGF axis were noted. This finding of study adherence and target activity provides a rationale for the further investigation of PFE in the active surveillance population.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Plant Extracts/administration & dosage , Pomegranate/chemistry , Prostatic Neoplasms/drug therapy , Biomarkers, Tumor/blood , Biomarkers, Tumor/urine , Biopsy , Fruit/chemistry , Humans , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor I/metabolism , Kallikreins/blood , Male , Middle Aged , Phytotherapy , Placebos , Plant Extracts/isolation & purification , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , Prostatic Neoplasms/urine , Watchful Waiting
8.
Oxid Med Cell Longev ; 2020: 9535426, 2020.
Article in English | MEDLINE | ID: mdl-33178389

ABSTRACT

Trastuzumab (TZM) is a humanized monoclonal antibody that has been approved for the clinical management of HER2-positive metastatic breast and gastric cancers but its use is limited by its cumulative dose and off-target cardiotoxicity. Unfortunately, till date, there is no approved antidote to this off-target toxicity. Therefore, an acute study was designed at investigating the protective potential and mechanism(s) of CVE and IGE in TZM-induced cardiotoxicity utilizing cardiac enzyme and oxidative stress markers and histopathological endpoints. 400 mg/kg/day CVE and IGE dissolved in 5% DMSO in sterile water were investigated in Wistar rats injected with 2.25 mg/kg/day/i.p. route of TZM for 7 days, using serum cTnI and LDH, complete lipid profile, cardiac tissue oxidative stress markers assays, and histopathological examination of TZM-intoxicated heart tissue. Results showed that 400 mg/kg/day CVE and IGE profoundly attenuated increases in the serum cTnI and LDH levels but caused no significant alterations in the serum lipids and weight gain pattern in the treated rats. CVE and IGE profoundly attenuated alterations in the cardiac tissue oxidative stress markers' activities while improving TZM-associated cardiac histological lesions. These results suggest that CVE and IGE could be mediating its cardioprotection via antioxidant, free radical scavenging, and antithrombotic mechanisms, thus, highlighting the therapeutic potentials of CVE and IGE in the management of TZM-mediated cardiotoxicity.


Subject(s)
Cardiotoxicity , Cellulose/chemistry , Clerodendrum/chemistry , Plant Extracts/pharmacology , Seeds/chemistry , Trastuzumab/adverse effects , Africa , Animals , Cardiotoxicity/drug therapy , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Male , Plant Extracts/chemistry , Rats , Rats, Wistar , Trastuzumab/pharmacology
9.
Oxid Med Cell Longev ; 2020: 1602816, 2020.
Article in English | MEDLINE | ID: mdl-33149803

ABSTRACT

Cardiotoxicity as an off-target effect of doxorubicin therapy is a major limiting factor for its clinical use as a choice cytotoxic agent. Seeds of Irvingia gabonensis have been reported to possess both nutritional and medicinal values which include antidiabetic, weight losing, antihyperlipidemic, and antioxidative effects. Protective effects of Irvingia gabonensis ethanol seed extract (IGESE) was investigated in doxorubicin (DOX)-mediated cardiotoxicity induced with single intraperitoneal injection of 15 mg/kg of DOX following the oral pretreatments of Wistar rats with 100-400 mg/kg/day of IGESE for 10 days, using serum cardiac enzyme markers (cardiac troponin I (cTI) and lactate dehydrogenase (LDH)), cardiac tissue oxidative stress markers (catalase (CAT), malonyldialdehyde (MDA), superoxide dismutase (SOD), glutathione-S-transferase (GST), glutathione peroxidase (GSH-Px), and reduced glutathione (GSH)), and cardiac histopathology endpoints. In addition, both qualitative and quantitative analyses to determine IGESE's secondary metabolites profile and its in vitro antioxidant activities were also conducted. Results revealed that serum cTnI and LDH were significantly elevated by the DOX treatment. Similarly, activities of tissue SOD, CAT, GST, and GSH levels were profoundly reduced, while GPx activity and MDA levels were profoundly increased by DOX treatment. These biochemical changes were associated with microthrombi formation in the DOX-treated cardiac tissues on histological examination. However, oral pretreatments with 100-400 mg/kg/day of IGESE dissolved in 5% DMSO in distilled water significantly attenuated increases in the serum cTnI and LDH, prevented significant alterations in the serum lipid profile and the tissue activities and levels of oxidative stress markers while improving cardiovascular disease risk indices and DOX-induced histopathological lesions. The in vitro antioxidant studies showed IGESE to have good antioxidant profile and contained 56 major secondary metabolites prominent among which are γ-sitosterol, Phytol, neophytadiene, stigmasterol, vitamin E, hexadecanoic acid and its ethyl ester, Phytyl palmitate, campesterol, lupeol, and squalene. Overall, both the in vitro and in vivo findings indicate that IGESE may be a promising prophylactic cardioprotective agent against DOX-induced cardiotoxicity, at least in part mediated via IGESE's antioxidant and free radical scavenging and antithrombotic mechanisms.


Subject(s)
Cardiotoxicity/drug therapy , Cellulose/chemistry , Doxorubicin/adverse effects , Plant Extracts/therapeutic use , Seeds/chemistry , Animals , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Biomarkers/metabolism , Biphenyl Compounds/chemistry , Cardiotoxicity/blood , Free Radical Scavengers/chemistry , Gas Chromatography-Mass Spectrometry , Lipids/blood , Male , Metabolome , Myocardium/pathology , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Phytochemicals/analysis , Picrates/chemistry , Plant Extracts/pharmacology , Rats, Wistar , Risk Factors , Secondary Metabolism/drug effects
10.
J Toxicol ; 2020: 8859716, 2020.
Article in English | MEDLINE | ID: mdl-32714390

ABSTRACT

Doxorubicin is widely applied in hematological and solid tumor treatment but limited by its off-target cardiotoxicity. Thus, cardioprotective potential and mechanism(s) of CVE in DOX-induced cardiotoxicity were investigated using cardiac and oxidative stress markers and histopathological endpoints. 50-400 mg/kg/day CVE in 5% DMSO in distilled water were investigated in Wistar rats intraperitoneally injected with 2.5 mg/kg DOX on alternate days for 14 days, using serum troponin I and LDH, complete lipid profile, cardiac tissue oxidative stress marker assays, and histopathological examination of DOX-treated cardiac tissue. Preliminary qualitative and quantitative assays of CVE's secondary metabolites were also conducted. Phytochemical analyses revealed the presence of flavonoids (34.79 ± 0.37 mg/100 mg dry extract), alkaloids (36.73 ± 0.27 mg/100 mg dry extract), reducing sugars (07.78 ± 0.09 mg/100 mg dry extract), and cardiac glycosides (24.55 ± 0.12 mg/100 mg dry extract). 50-400 mg/kg/day CVE significantly attenuated increases in the serum LDH and troponin I levels. Similarly, the CVE dose unrelatedly decreased serum TG and VLDL-c levels without significant alterations in the serum TC, HDL-c, and LDL-c levels. Also, CVE profoundly attenuated alterations in the cardiac tissue oxidative stress markers' activities while improving DOX-associated cardiac histological lesions that were possibly mediated via free radical scavenging and/or antioxidant mechanisms. Overall, CVE may play a significant therapeutic role in the management of DOX-induced cardiotoxicity in humans.

11.
Nanomedicine (Lond) ; 15(11): 1147-1162, 2020 05.
Article in English | MEDLINE | ID: mdl-32292109

ABSTRACT

Many dietary polyphenols have been investigated for their therapeutic potential either as single agents or in combinations. Despite the significant anticancer potential of these polyphenols in in vitro cell culture and in vivo animal models, their clinical applications have been limited because of challenges such as ineffective systemic delivery, stability and low bioavailability. Nanoencapsulation of these polyphenols could prolong circulation, improve localization, enhance efficacy and reduce the chances of multidrug resistance. This review summarized the use of various polyphenols especially epigallocatechin gallate, quercetin, curcumin and resveratrol as nanoformulations for cancer prevention and treatment. Despite some success, more research is warranted to design a nanoencapsulated combination of polyphenols, effective in in vitro, in vivo and human systems.


Subject(s)
Catechin , Curcumin , Neoplasms , Animals , Catechin/pharmacology , Curcumin/pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/prevention & control , Polyphenols/pharmacology , Quercetin , Resveratrol
12.
Oxid Med Cell Longev ; 2019: 6726312, 2019.
Article in English | MEDLINE | ID: mdl-31827691

ABSTRACT

Plant-based therapies are being explored to prevent or treat several cancer types. The antioxidant properties of Polyalthia longifolia plant are well established. In our previous work, we demonstrated the presence of cytotoxic compounds in the methanol extract of Polyalthia longifolia (MEP) with potent activity against human leukemia cells. In the present study, we evaluated the efficacy of MEP against prostate cancer (PCa) and established the molecular basis of its effect in in vitro and in vivo models. We observed that MEP treatment resulted in a significant decrease in the growth and viability of PCa cells, associated with arrest in the G1/S phase of the cell cycle. Apoptosis was confirmed as the primary mode of MEP-induced cell death through activation of the intrinsic apoptotic machinery. Proteomic and biochemical studies identified BiP as an important target of MEP with the activation of the ER stress pathway, as a potential mechanism driving MEP-induced apoptosis. The extract exhibited strong efficacy in the PCa xenograft mouse model with significant inhibition of tumor growth and reduced tumor burden. Taken together, our findings indicate that MEP-induced apoptosis in PCa cells concomitant with the activation of the ER stress pathways results in the inhibition of tumor growth, in vitro and in vivo. Our studies provide initial evidence of the efficacy of MEP against PCa and advocate for in-depth studies in other preclinical models for its possible use in clinical settings.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Phytotherapy , Plant Extracts/pharmacology , Polyalthia/chemistry , Prostatic Neoplasms/drug therapy , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Humans , In Vitro Techniques , Male , Mice , Mice, Nude , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Sci Rep ; 9(1): 15761, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31673045

ABSTRACT

Stearoyl CoA desaturase 1 (SCD1) is a key enzyme in lipogenesis as it catalyzes the synthesis of monounsaturated fatty acids (MUFAs), mainly oleate (18:1n9) and palmitoleate (16:1n7) from saturated fatty acids (SFA), stearate (18:0) and palmitate (16:0), respectively. Studies on SCD1 deficiency in mouse models demonstrated beneficial metabolic phenotypes such as reduced adiposity and improved glucose tolerance. Even though, SCD1 represents a potential target to resolve obesity related metabolic diseases; SCD1 deficiency causes endoplasmic reticulum (ER) stress and activates unfolded protein response (UPR). The induction of ER stress in response to SCD1 deficiency is governed by the cofactor, PGC-1α. However, the mechanism by which SCD1 deficiency increases PGC-1α and subsequently induces ER stress still remains elusive. The present study demonstrates that despite reduced lipogenesis, liver specific SCD1 deficiency activates the mechanistic target of rapamycin complex 1 (mTORC1) along with induction of PGC-1α and ER stress. Further, mTORC1 inhibition attenuates SCD1 deficiency-mediated induction of both PGC-1α and ER stress. Similar observations were seen by restoring endogenously synthesized oleate, but not palmitoleate, suggesting a clear mTORC1-mediated regulation of ER stress during SCD1 deficiency. Overall, our results suggest a model whereby maintaining adequate levels of hepatic oleate is required to suppress mTORC1-mediated ER stress. In addition, the activation of mTORC1 by SCD1 deficiency reveals an important function of fatty acids in regulating different cellular processes through mTORC1 signaling.


Subject(s)
Dietary Carbohydrates/pharmacology , Endoplasmic Reticulum Stress , Liver/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Signal Transduction , Stearoyl-CoA Desaturase/deficiency , Animals , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Knockout , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Stearoyl-CoA Desaturase/metabolism
14.
J Biol Chem ; 294(51): 19475-19485, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31690632

ABSTRACT

Increased carbohydrate consumption increases hepatic de novo lipogenesis, which has been linked to the development of chronic metabolic diseases, including obesity, hepatic steatosis, and insulin resistance. Stearoyl CoA desaturase 1 (SCD1) is a critical lipogenic enzyme that catalyzes the synthesis of two monounsaturated fatty acids, oleate and palmitoleate, from the saturated fatty acids stearate and palmitate, respectively. SCD1-deficient mouse models are protected against diet-induced adiposity, hepatic steatosis, and hyperglycemia. However, the mechanism of this protection by SCD1 deficiency is unclear. Using liver-specific SCD1 knockout (LKO) mice fed a high-carbohydrate, low-fat diet, we show that hepatic SCD1 deficiency increases systemic glucose uptake. Hepatic SCD1 deficiency enhanced glucose transporter type 1 (GLUT1) expression in the liver and also up-regulated GLUT4 and adiponectin expression in adipose tissue. The enhanced glucose uptake correlated with increased liver expression and elevated plasma levels of fibroblast growth factor 21 (FGF21), a hepatokine known to increase systemic insulin sensitivity and regulate whole-body lipid metabolism. Feeding LKO mice a triolein-supplemented but not tristearin-supplemented high-carbohydrate, low-fat diet reduced FGF21 expression and plasma levels. Consistently, SCD1 inhibition in primary hepatocytes induced FGF21 expression, which was repressed by treatment with oleate but not palmitoleate. Moreover, deletion of the transcriptional coactivator PPARγ coactivator 1α (PGC-1α) reduced hepatic and plasma FGF21 and white adipocyte tissue-specific GLUT4 expression and raised plasma glucose levels in LKO mice. These results suggest that hepatic oleate regulates glucose uptake in adipose tissue either directly or partially by modulating the hepatic PGC-1α-FGF21 axis.


Subject(s)
Adipose Tissue/metabolism , Fibroblast Growth Factors/metabolism , Glucose/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Stearoyl-CoA Desaturase/genetics , Adiponectin/blood , Adiposity , Animals , Carbohydrate Metabolism , Diet , Fatty Acids, Monounsaturated/metabolism , Fatty Liver/metabolism , Insulin/metabolism , Lipid Metabolism , Lipogenesis , Liver/metabolism , Male , Mice , Mice, Knockout , Oleic Acid/pharmacology , Positron Emission Tomography Computed Tomography , Stearoyl-CoA Desaturase/metabolism
15.
Cells ; 8(9)2019 09 15.
Article in English | MEDLINE | ID: mdl-31540162

ABSTRACT

Psoriasis is a chronic immune-mediated skin disease that involves the interaction of immune and skin cells, and is characterized by cytokine-driven epidermal hyperplasia, deviant differentiation, inflammation, and angiogenesis. Because the available treatments for psoriasis have significant limitations, dietary products are potential natural sources of therapeutic molecules, which can repair the molecular defects associated with psoriasis and could possibly be developed for its management. Fisetin (3,7,3',4'-tetrahydroxyflavone), a phytochemical naturally found in pigmented fruits and vegetables, has demonstrated proapoptotic and antioxidant effects in several malignancies. This study utilized biochemical, cellular, pharmacological, and tissue engineering tools to characterize the effects of fisetin on normal human epidermal keratinocytes (NHEKs), peripheral blood mononuclear cells (PBMC), and CD4+ T lymphocytes in 2D and 3D psoriasis-like disease models. Fisetin treatment of NHEKs dose- and time-dependently induced differentiation and inhibited interleukin-22-induced proliferation, as well as activation of the PI3K/Akt/mTOR pathway. Fisetin treatment of TNF-α stimulated NHEKs also significantly inhibited the activation of p38 and JNK, but had enhanced effect on ERK1/2 (MAPK). In addition, fisetin treatment significantly decreased the secretion of Th1/Th-17 pro-inflammatory cytokines, particularly IFN-γ and IL-17A by 12-O-tetradecanolylphorbol 13-acetate (TPA)-stimulated NHEKs and anti-CD3/CD28-activated human PBMCs. Furthermore, we established the in vivo relevance of fisetin functions, using a 3D full-thickness human skin model of psoriasis (FTRHSP) that closely mimics in vivo human psoriatic skin lesions. Herein, fisetin significantly ameliorated psoriasis-like disease features, and decreased the production of IL-17 by CD4+ T lymphocytes co-cultured with FTRHSP. Collectively, our data identify the prodifferentiative, antiproliferative, and anti-inflammatory effects of fisetin, via modulation of the PI3K-Akt-mTOR and p38/JNK pathways and the production of cytokines in 2D and 3D human skin models of psoriasis. These results suggest that fisetin has a great potential to be developed as an effective and inexpensive agent for the treatment of psoriasis and other related inflammatory skin disorders.


Subject(s)
Flavonoids/pharmacology , Inflammation/drug therapy , MAP Kinase Signaling System/drug effects , Models, Biological , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Psoriasis/drug therapy , Skin/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Cell Survival/drug effects , Cells, Cultured , Flavonols , Humans , Inflammation/metabolism , Inflammation/pathology , Keratinocytes/drug effects , Keratinocytes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Psoriasis/metabolism , Psoriasis/pathology , Skin/pathology , TOR Serine-Threonine Kinases/metabolism
16.
Int J Cancer ; 145(11): 3022-3032, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31018249

ABSTRACT

The normal colon epithelium is transformed into its neoplastic counterpart through a series of genetic alterations in driver genes including activating mutations in PIK3CA. Treatment often involves surgery followed by 5-fluorouracil (5-FU) based therapy, which has limited efficiency and serious side effects. We sought to determine whether fisetin, a dietary flavonoid, alone or in combination with 5-FU affected tumorigenesis in the mammalian intestine. We first determined the effect of fisetin, 5-FU or their combination on PIK3CA-mutant and PIK3CA wild-type colon cancer cells by assessing cell viability, colony formation, apoptosis and effects on PI3K/AKT/mTOR signaling. Treatment of PIK3CA-mutant cells with fisetin and 5-FU reduced the expression of PI3K, phosphorylation of AKT, mTOR, its target proteins, constituents of mTOR signaling complex and this treatment increased the phosphorylation of AMPKα. We then determined whether fisetin and 5-FU together or singly affected tumorigenesis in ApcMin/+ mice that also express constitutively active PI3K in the distal small intestine and colon. Tumor incidence was markedly lower in fisetin-treated FC1 3K1 ApcMin/+ mice that also express constitutively active PI3K in distal small intestine and colon, as compared to control animals, indicating that fisetin is a strong preventive agent. In addition, the combination of fisetin and 5-FU also reduced the total number of intestinal tumors. Fisetin could be used as a preventive agent plus an adjuvant with 5-FU for the treatment of PIK3CA-mutant colorectal cancer.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Colorectal Neoplasms/drug therapy , Flavonoids/administration & dosage , Fluorouracil/administration & dosage , Mutation , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Drug Synergism , Flavonoids/pharmacology , Flavonols , Fluorouracil/pharmacology , HCT116 Cells , HT29 Cells , Humans , Mice , Phosphorylation/drug effects , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
17.
FASEB J ; 33(3): 3198-3211, 2019 03.
Article in English | MEDLINE | ID: mdl-30379590

ABSTRACT

Growth factor receptor-binding protein 10 (GRB10) is a well-known adaptor protein and a recently identified substrate of the mammalian target of rapamycin (mTOR). Depletion of GRB10 increases insulin sensitivity and overexpression suppresses PI3K/Akt signaling. Because the major reason for the limited efficacy of PI3K/Akt-targeted therapies in prostate cancer (PCa) is loss of mTOR-regulated feedback suppression, it is therefore important to assess the functional importance and regulation of GRB10 under these conditions. On the basis of these background observations, we explored the status and functional impact of GRB10 in PCa and found maximum expression in phosphatase and tensin homolog (PTEN)-deficient PCa. In human PCa samples, GRB10 inversely correlated with PTEN and positively correlated with pAKT levels. Knockdown of GRB10 in nontumorigenic PTEN null mouse embryonic fibroblasts and tumorigenic PCa cell lines reduced Akt phosphorylation and selectively activated a panel of receptor tyrosine kinases. Similarly, overexpression of GRB10 in PTEN wild-type PCa cell lines accelerated tumorigenesis and induced Akt phosphorylation. In PTEN wild-type PCa, GRB10 overexpression promoted mediated PTEN interaction and degradation. PI3K (but not mTOR) inhibitors reduced GRB10 expression, suggesting primarily PI3K-driven regulation of GRB10. In summary, our results suggest that GRB10 acts as a major downstream effector of PI3K and has tumor-promoting effects in prostate cancer.-Khan, M. I., Al Johani, A., Hamid, A., Ateeq, B., Manzar, N., Adhami, V. M., Lall, R. K., Rath, S., Sechi, M., Siddiqui, I. A., Choudhry, H., Zamzami, M. A., Havighurst, T. C., Huang, W., Ntambi, J. M., Mukhtar, H. Proproliferatve function of adaptor protein GRB10 in prostate carcinoma.


Subject(s)
GRB10 Adaptor Protein/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Carcinogens/antagonists & inhibitors , Carcinogens/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , GRB10 Adaptor Protein/antagonists & inhibitors , GRB10 Adaptor Protein/genetics , Gene Knockdown Techniques , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Models, Biological , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/genetics , RNA, Messenger , Signal Transduction
18.
Mol Cancer Ther ; 18(2): 356-363, 2019 02.
Article in English | MEDLINE | ID: mdl-30446585

ABSTRACT

Hyperactivated AKT kinase due to loss of its negative regulator PTEN influences many aspects of cancer biology, including chromatin. AKT primarily regulates acetyl-CoA production and phosphorylates many histone-modulating enzymes, resulting in their activation or inhibition. Therefore, understanding the therapeutic impact of AKT inhibition on chromatin-related events is essential. Here, we report that AKT inhibition in prostate-specific PTEN knockout mice significantly induces di- and trimethylation of H3K4 with concomitant reduction in H3K9 acetylation. Mechanistically, we observed that AKT inhibition reduces expression of the H3K4 methylation-specific histone demethylases KDM5 family, especially KDM5B expression at transcriptional levels. Furthermore, we observed that AKT negatively regulates miR-137 levels, which transcriptionally represses KDM5B expression. Overexpression of miR-137 significantly reduced KDM5B and increased H3K4 methylation levels but failed to change AKT phosphorylation. Overall, we observed that AKT transcriptionally regulates KDM5B mainly via repression of miR-137. Our data identify a mechanism by which AKT kinase modulates the prostate cancer epigenome through regulating H3K4 methylation. Additional studies on AKT inhibition-mediated induction of H3K4 methylation will help in designing strategies to enhance the therapeutic efficacy of PI3K/AKT inhibitors.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/genetics , MicroRNAs/genetics , Nuclear Proteins/genetics , PTEN Phosphohydrolase/genetics , Phosphorylcholine/analogs & derivatives , Prostatic Neoplasms/drug therapy , Repressor Proteins/genetics , Acetylation/drug effects , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , Humans , Male , Methylation/drug effects , Mice , Phosphorylcholine/administration & dosage , Phosphorylcholine/pharmacology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Xenograft Model Antitumor Assays
19.
Nutrients ; 11(1)2018 Dec 25.
Article in English | MEDLINE | ID: mdl-30585192

ABSTRACT

Tea is the most widely used beverage worldwide. Japanese and Chinese people have been drinking tea for centuries and in Asia, it is the most consumed beverage besides water. It is a rich source of pharmacologically active molecules which have been implicated to provide diverse health benefits. The three major forms of tea are green, black and oolong tea based on the degree of fermentation. The composition of tea differs with the species, season, leaves, climate, and horticultural practices. Polyphenols are the major active compounds present in teas. The catechins are the major polyphenolic compounds in green tea, which include epigallocatechin-3-gallate (EGCG), epigallocatechin, epicatechin-3-gallate and epicatechin, gallocatechins and gallocatechin gallate. EGCG is the predominant and most studied catechin in green tea. There are numerous evidences from cell culture and animal studies that tea polyphenols have beneficial effects against several pathological diseases including cancer, diabetes and cardiovascular diseases. The polyphenolic compounds present in black tea include theaflavins and thearubigins. In this review article, we will summarize recent studies documenting the role of tea polyphenols in the prevention of cancer, diabetes, cardiovascular and neurological diseases.


Subject(s)
Polyphenols/chemistry , Polyphenols/pharmacology , Tea/chemistry , Cardiovascular Diseases/prevention & control , Central Nervous System Diseases/prevention & control , Diabetes Mellitus/prevention & control , Humans , Neoplasms/prevention & control
20.
Sci Rep ; 8(1): 15726, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30356079

ABSTRACT

The anti-proliferative activity of dietary flavonoid fisetin has been validated in various cancer models. Establishing its precise mechanism of action has proved somewhat challenging given the multiplicity of its targets. We demonstrated that YB-1 promotes epithelial-to-mesenchymal transition and its inhibition suppressed tumor cell proliferation and invasion. The p90 ribosomal S6 kinase (RSK), an important ERK effector, activates YB-1 to drive melanoma growth. We found that fisetin treatment of monolayer/3-D melanoma cultures resulted in YB-1 dephosphorylation and reduced transcript levels. In parallel, fisetin suppressed mesenchymal markers and matrix-metalloproteinases in melanoma cells. Data from cell-free/cell-based systems indicated that fisetin inhibited RSK activity through binding to the kinase. Affinity studies for RSK isoforms evaluated stronger interaction for RSK2 than RSK1. Competition assays performed to monitor binding responses revealed that YB-1 and RSK2 do not compete, rather binding of fisetin to RSK2 promotes its binding to YB-1. Fisetin suppressed YB-1/RSK signaling independent of its effect on ERK, and reduced MDR1 levels. Comparable efficacy of fisetin and vemurafenib for inhibiting melanoma growth was noted albeit through divergent modulation of ERK. Our studies provide insight into additional modes of regulation through which fisetin interferes with melanoma growth underscoring its potential therapeutic efficacy in disease progression.


Subject(s)
Flavonoids/pharmacology , Melanoma/drug therapy , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Y-Box-Binding Protein 1/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Cell Culture Techniques/methods , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Flavonols , Humans , MAP Kinase Signaling System , Melanoma/pathology , Phosphorylation , Signal Transduction , Transcription, Genetic/drug effects , Y-Box-Binding Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...