Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Mol Cell ; 63(3): 526-38, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27453044

ABSTRACT

Intratumor genetic heterogeneity underlies the ability of tumors to evolve and adapt to different environmental conditions. Using CRISPR/Cas9 technology and specific DNA barcodes, we devised a strategy to recapitulate and trace the emergence of subpopulations of cancer cells containing a mutation of interest. We used this approach to model different mechanisms of lung cancer cell resistance to EGFR inhibitors and to assess effects of combined drug therapies. By overcoming intrinsic limitations of current approaches, CRISPR-barcoding also enables investigation of most types of genetic modifications, including repair of oncogenic driver mutations. Finally, we used highly complex barcodes inserted at a specific genome location as a means of simultaneously tracing the fates of many thousands of genetically labeled cancer cells. CRISPR-barcoding is a straightforward and highly flexible method that should greatly facilitate the functional investigation of specific mutations, in a context that closely mimics the complexity of cancer.


Subject(s)
Biomarkers, Tumor/genetics , CRISPR-Cas Systems , Carcinoma, Non-Small-Cell Lung/genetics , DNA, Neoplasm/genetics , Gene Editing/methods , Genetic Heterogeneity , Lung Neoplasms/genetics , Oncogenes , Point Mutation , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Lineage , Clone Cells/drug effects , Clone Cells/metabolism , Clone Cells/pathology , DNA Mutational Analysis , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Genetic Predisposition to Disease , HCT116 Cells , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MCF-7 Cells , Male , Mice, SCID , Multiplex Polymerase Chain Reaction , Phenotype , Protein Kinase Inhibitors/pharmacology , Time Factors , Tumor Microenvironment , Xenograft Model Antitumor Assays
2.
Cell Tissue Res ; 391(1): 127-144, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36227376

ABSTRACT

Obesity (Ob) depicts a state of energy imbalance(s) being characterized by the accumulation of excessive fat and which predisposes to several metabolic diseases. Mesenchymal stem cells (MSCs) represent a promising option for addressing obesity and its associated metabolic co-morbidities. The present study aims at assessing the beneficial effects of human placental MSCs (P-MSCs) in mitigating Ob-associated insulin resistance (IR) and mitochondrial dysfunction both in vivo and in vitro. Under obesogenic milieu, adipocytes showed a significant reduction in glucose uptake, and impaired insulin signaling with decreased expression of UCP1 and PGC1α, suggestive of dysregulated non-shivering thermogenesis vis-a-vis mitochondrial biogenesis respectively. Furthermore, obesogenic adipocytes demonstrated impaired mitochondrial respiration and energy homeostasis evidenced by reduced oxygen consumption rate (OCR) and blunted ATP/NAD+/NADP+ production respectively. Interestingly, co-culturing adipocytes with P-MSCs activated PI3K-Akt signaling, improved glucose uptake, diminished ROS production, enhanced mitochondrial OCR, improved ATP/NAD+/NADP+ production, and promoted beiging of adipocytes evidenced by upregulated expression of PRDM16, UCP1, and PGC1α expression. In vivo, P-MSCs administration increased the peripheral blood glucose uptake and clearance, and improved insulin sensitivity and lipid profile with a coordinated increase in the ratio of ATP/ADP and NAD+ and NADP+ in the white adipose tissue (WAT), exemplified in WNIN/GR-Ob obese mutant rats. In line with in vitro findings, there was a significant reduction in adipocyte hypertrophy, increased mitochondrial staining, and thermogenesis. Our findings advocate for a therapeutic application of P-MSCs for improving glucose and energy homeostasis, i.e., probably restoring non-shivering thermogenesis towards obesity management.


Subject(s)
Adipocytes , Energy Metabolism , Glucose , Insulin Resistance , Mesenchymal Stem Cells , Obesity , Placenta , Animals , Female , Humans , Rats , Adenosine Triphosphate/metabolism , Adipocytes/metabolism , Glucose/metabolism , Homeostasis , Insulin Resistance/physiology , Mesenchymal Stem Cells/metabolism , NAD/metabolism , NADP/metabolism , Obesity/metabolism , Obesity/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Placenta/metabolism , Glucose Metabolism Disorders/pathology , Energy Metabolism/physiology
3.
Inflammopharmacology ; 31(4): 2103-2120, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37266812

ABSTRACT

OBJECTIVE AND DESIGN: Inflammatory bowel disease (IBD) is an idiopathic inflammatory condition of the digestive system marked by oxidative stress, leukocyte infiltration, and elevation of inflammatory mediators. In this study, we demonstrate the protective effect of ethyl gallate (EG), a phytochemical, and propyl gallate (PG), an anti-oxidant, given through normal drinking water (DW) and copper water (CW) in various combinations, which had a positive effect on the amelioration of DSS-induced ulcerative colitis in C57BL/6 J mice. MATERIALS AND METHODS: We successfully determined the levels of proinflammatory cytokines and anti-oxidant enzymes by ELISA, tracked oxidative/nitrosative stress (RO/NS) by in vivo imaging (IVIS) using L-012 chemiluminescent probe, disease activity index (DAI), and histopathological and morphometric analysis of colon in DSS-induced colitis in a model. RESULTS: The results revealed that oral administration of ethyl gallate and propyl gallate at a dose of 50 mg/kg considerably reduced the severity of colitis and improved both macroscopic and microscopic clinical symptoms. The level of proinflammatory cytokines (TNF-α, IL-6, IL-1ß, and IFN-γ) in colonic tissue was considerably reduced in the DSS + EG-treated and DSS + PG-treated groups, compared to the DSS alone-treated group. IVIS imaging of animals from the DSS + EG and DSS + PG-treated groups showed a highly significant decrease in RO/NS species relative to the DSS control group, with the exception of the DSS + PG/CW and DSS + EG + PG/CW-treated groups. We also observed lower levels of myeloperoxidase (MPO), nitric oxide (NO), and lipid peroxidation (LPO), and restored levels of GST and superoxide dismutase (SOD) in DSS + EG-DW/CW, DSS + PG/DW, and DSS + EG + PG/DW groups compared to DSS alone-treated group. In addition, we showed that the EG, PG, and EG + PG treatment significantly reduced the DAI score, and counteracted the body weight loss and colon shortening in mice compared to DSS alone-treated group. In this 21-day study, mice were treated daily with test substances and were challenged to DSS from day-8 to 14. CONCLUSION: Our study highlights the protective effect of ethyl gallate and propyl gallate in various combinations which, in pre-clinical animals, serve as an anti-inflammatory drug against the severe form of colitis, indicating its potential for the treatment of IBD in humans. In addition, propyl gallate was investigated for the first time in this study for its anti-colitogenic effect with normal drinking water and reduced effect with copper water.


Subject(s)
Colitis, Ulcerative , Colitis , Drinking Water , Inflammatory Bowel Diseases , Humans , Animals , Mice , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Propyl Gallate/adverse effects , Dextran Sulfate/pharmacology , Antioxidants/pharmacology , Antioxidants/therapeutic use , Copper/adverse effects , Drinking Water/adverse effects , Mice, Inbred C57BL , Colitis/drug therapy , Colon , Cytokines , Inflammatory Bowel Diseases/pathology , Disease Models, Animal
4.
Semin Cancer Biol ; 69: 178-189, 2021 02.
Article in English | MEDLINE | ID: mdl-31419527

ABSTRACT

Cancer is an outrageous disease with uncontrolled differentiation, growth, and migration to the other parts of the body. It is the second-most common cause of death both in the U.S. and worldwide. Current conventional therapies, though much improved and with better prognosis, have several limitations. Chemotherapeutic agents, for instance, are cytotoxic to both tumor and healthy cells, and the non-specific distribution of drugs at tumor sites limits the dose administered. Nanotechnology, which evolved from the coalescence and union of varied scientific disciplines, is a novel science that has been the focus of much research. This technology is generating more effective cancer therapies to overcome biomedical and biophysical barriers against standard interventions in the body; its unique magnetic, electrical, and structural properties make it a promising tool. This article reviews endogenous- and exogenous-based stimulus-responsive drug delivery systems designed to overcome the limitations of conventional therapies. The article also summarizes the study of nanomaterials, including polymeric, gold, silver, magnetic, and quantum dot nanoparticles. Though an array of drug delivery systems has so far been proposed, there remain many challenges and concerns that should be addressed in order to fill the gaps in the field. Prominence is given to drug delivery systems that employ external- and internal-based stimuli and that are emerging as promising tools for cancer therapeutics in clinical settings.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Nanomedicine , Nanoparticles/administration & dosage , Nanostructures/chemistry , Neoplasms/drug therapy , Animals , Humans , Nanoparticles/chemistry , Neoplasms/pathology
5.
Genes Dev ; 27(17): 1868-85, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-24013501

ABSTRACT

The p53 tumor suppressor is a transcription factor that mediates varied cellular responses. The C terminus of p53 is subjected to multiple and diverse post-translational modifications. An attractive hypothesis is that differing sets of combinatorial modifications therein determine distinct cellular outcomes. To address this in vivo, a Trp53(ΔCTD/ΔCTD) mouse was generated in which the endogenous p53 is targeted and replaced with a truncated mutant lacking the C-terminal 24 amino acids. These Trp53(ΔCTD/ΔCTD) mice die within 2 wk post-partum with hematopoietic failure and impaired cerebellar development. Intriguingly, the C terminus acts via three distinct mechanisms to control p53-dependent gene expression depending on the tissue. First, in the bone marrow and thymus, the C terminus dampens p53 activity. Increased senescence in the Trp53(ΔCTD/ΔCTD) bone marrow is accompanied by up-regulation of Cdkn1 (p21). In the thymus, the C-terminal domain negatively regulates p53-dependent gene expression by inhibiting promoter occupancy. Here, the hyperactive p53(ΔCTD) induces apoptosis via enhanced expression of the proapoptotic Bbc3 (Puma) and Pmaip1 (Noxa). In the liver, a second mechanism prevails, since p53(ΔCTD) has wild-type DNA binding but impaired gene expression. Thus, the C terminus of p53 is needed in liver cells at a step subsequent to DNA binding. Finally, in the spleen, the C terminus controls p53 protein levels, with the overexpressed p53(ΔCTD) showing hyperactivity for gene expression. Thus, the C terminus of p53 regulates gene expression via multiple mechanisms depending on the tissue and target, and this leads to specific phenotypic effects in vivo.


Subject(s)
Gene Expression Regulation , Genes, p53/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cellular Senescence/genetics , Cerebellum/growth & development , Cerebellum/metabolism , Gene Knock-In Techniques , Growth and Development/genetics , Liver/metabolism , Mice , Mice, Inbred C57BL , Mutation/genetics , Protein Binding , Protein Processing, Post-Translational , Sequence Deletion/genetics , Thymocytes/cytology , Thymocytes/metabolism , Time Factors
6.
J Biol Chem ; 294(44): 16109-16122, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31511328

ABSTRACT

Growth hormone (GH) plays a significant role in normal renal function and overactive GH signaling has been implicated in proteinuria in diabetes and acromegaly. Previous results have shown that the glomerular podocytes, which play an essential role in renal filtration, express the GH receptor, suggesting the direct action of GH on these cells. However, the exact mechanism and the downstream pathways by which excess GH leads to diabetic nephropathy is not established. In the present article, using immortalized human podocytes in vitro and a mouse model in vivo, we show that excess GH activates Notch1 signaling in a γ-secretase-dependent manner. Pharmacological inhibition of Notch1 by γ-secretase inhibitor DAPT (N-[N-(3,5-Difluorophenacetyl)-l-alanyl]-S-phenyl glycine t-butylester) abrogates GH-induced epithelial to mesenchymal transition (EMT) and is associated with a reduction in podocyte loss. More importantly, our results show that DAPT treatment blocks cytokine release and prevents glomerular fibrosis, all of which are induced by excess GH. Furthermore, DAPT prevented glomerular basement membrane thickening and proteinuria induced by excess GH. Finally, using kidney biopsy sections from people with diabetic nephropathy, we show that Notch signaling is indeed up-regulated in such settings. All these results confirm that excess GH induces Notch1 signaling in podocytes, which contributes to proteinuria through EMT as well as renal fibrosis. Our studies highlight the potential application of γ-secretase inhibitors as a therapeutic target in people with diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/metabolism , Growth Hormone/pharmacology , Podocytes/metabolism , Proteinuria/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Animals , Cells, Cultured , Cytokines/metabolism , Diabetic Nephropathies/drug therapy , Dipeptides/pharmacology , Dipeptides/therapeutic use , Epithelial-Mesenchymal Transition , Humans , Male , Mice , Podocytes/drug effects , Podocytes/pathology , Proteinuria/drug therapy , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics
7.
J Immunol ; 201(6): 1727-1734, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30068593

ABSTRACT

Glatiramer acetate (GA; Copaxone) is a copolymer therapeutic that is approved by the Food and Drug Administration for the relapsing-remitting form of multiple sclerosis. Despite an unclear mechanism of action, studies have shown that GA promotes protective Th2 immunity and stimulates release of cytokines that suppress autoimmunity. In this study, we demonstrate that GA interacts with murine paired Ig-like receptor B (PIR-B) on myeloid-derived suppressor cells and suppresses the STAT1/NF-κB pathways while promoting IL-10/TGF-ß cytokine release. In inflammatory bowel disease models, GA enhanced myeloid-derived suppressor cell-dependent CD4+ regulatory T cell generation while reducing proinflammatory cytokine secretion. Human monocyte-derived macrophages responded to GA by reducing TNF-α production and promoting CD163 expression typical of alternative maturation despite the presence of GM-CSF. Furthermore, GA competitively interacts with leukocyte Ig-like receptors B (LILRBs), the human orthologs of PIR-B. Because GA limited proinflammatory activation of myeloid cells, therapeutics that target LILRBs represent novel treatment modalities for autoimmune indications.


Subject(s)
Antigens, CD/immunology , Glatiramer Acetate/pharmacology , Myeloid-Derived Suppressor Cells/immunology , Receptors, Immunologic/immunology , Animals , Antigens, CD/genetics , Autoimmune Diseases/drug therapy , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cytokines/genetics , Cytokines/immunology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Myeloid-Derived Suppressor Cells/pathology , NF-kappa B/genetics , NF-kappa B/immunology , Receptors, Immunologic/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th2 Cells/immunology , Th2 Cells/pathology
8.
J Cell Physiol ; 234(5): 6503-6518, 2019 05.
Article in English | MEDLINE | ID: mdl-30238984

ABSTRACT

The glomerular filtration barrier (GFB) plays a critical role in ensuing protein free urine. The integrity of the GFB is compromised during hypoxia that prevails during extreme physiological conditions. However, the mechanism by which glomerular permselectivity is compromised during hypoxia remains enigmatic. Rats exposed to hypoxia showed a decreased glomerular filtration rate, podocyte foot-processes effacement, and proteinuria. Accumulation of hypoxia-inducible factor-1α (HIF1α) in podocytes resulted in elevated expression of zinc finger E-box binding homeobox 2 (ZEB2) and decreased expression of E- and P-cadherin. We also demonstrated that HIF1α binds to hypoxia response element localized in the ZEB2 promoter. Furthermore, HIF1α also induced the expression of ZEB2-natural antisense transcript, which is known to increase the efficiency of ZEB2 translation. Ectopic expression of ZEB2 induced loss of E- and P-cadherin and is associated with enhanced motility of podocytes during hypoxic conditions. ZEB2 knockdown abrogated hypoxia-induced decrease in podocyte permselectivity. This study suggests that hypoxia leads to activation of HIF1α-ZEB2 axis, resulting in podocyte injury and poor renal outcome.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Podocytes/metabolism , Proteinuria/metabolism , Proteinuria/physiopathology , Zinc Finger E-box Binding Homeobox 2/metabolism , Animals , Hypoxia/metabolism , Hypoxia/physiopathology , Podocytes/pathology , Rats , Rats, Wistar
9.
J Cell Biochem ; 120(5): 7667-7678, 2019 May.
Article in English | MEDLINE | ID: mdl-30387200

ABSTRACT

Glomerular podocytes are the major components of the renal filtration barrier, and altered podocyte permselectivity is a key event in the pathogenesis of proteinuric conditions. Clinical conditions such as ischemia and sleep apnea and extreme physiological conditions such as high-altitude sickness are presented with renal hypoxia and are associated with significant proteinuria. Hypoxia is considered as an etiological factor in the progression of acute renal injury. A sustained increase in hypoxia-inducible factor 1α (HIF1α) is a major adaptive stimulus to the hypoxic conditions. Although the temporal association between hypoxia and proteinuria is known, the mechanism by which hypoxia elicits proteinuria remains to be investigated. Furthermore, stabilization of HIF1α is being considered as a therapeutic option to treat anemia in patients with chronic kidney disease. Therefore, in this study, we induced stabilization of HIF1α in glomerular regions in vivo and in podocytes in vitro upon exposure to cobalt chloride. The elevated HIF1α expression is concurrence with diminished expression of nephrin and podocin, podocyte foot-processes effacement, and significant proteinuria. Podocytes exposed to cobalt chloride lost their arborized morphology and cell-cell connections and also displayed cytoskeletal derangements. Elevation in expression of HIF1α is in concomitance with loss of nephrin and podocin in patients with diabetic nephropathy and chronic kidney disease. In summary, the current study suggests that HIF1α stabilization impairs podocyte function vis-à-vis glomerular permselectivity.

11.
PLoS Genet ; 9(8): e1003603, 2013.
Article in English | MEDLINE | ID: mdl-23966864

ABSTRACT

The role of Wnt signaling in embryonic development and stem cell maintenance is well established and aberrations leading to the constitutive up-regulation of this pathway are frequent in several types of human cancers. Upon ligand-mediated activation, Wnt receptors promote the stabilization of ß-catenin, which translocates to the nucleus and binds to the T-cell factor/lymphoid enhancer factor (TCF/LEF) family of transcription factors to regulate the expression of Wnt target genes. When not bound to ß-catenin, the TCF/LEF proteins are believed to act as transcriptional repressors. Using a specific lentiviral reporter, we identified hematopoietic tumor cells displaying constitutive TCF/LEF transcriptional activation in the absence of ß-catenin stabilization. Suppression of TCF/LEF activity in these cells mediated by an inducible dominant-negative TCF4 (DN-TCF4) inhibited both cell growth and the expression of Wnt target genes. Further, expression of TCF1 and LEF1, but not TCF4, stimulated TCF/LEF reporter activity in certain human cell lines independently of ß-catenin. By a complementary approach in vivo, TCF1 mutants, which lacked the ability to bind to ß-catenin, induced Xenopus embryo axis duplication, a hallmark of Wnt activation, and the expression of the Wnt target gene Xnr3. Through generation of different TCF1-TCF4 fusion proteins, we identified three distinct TCF1 domains that participate in the ß-catenin-independent activity of this transcription factor. TCF1 and LEF1 physically interacted and functionally synergized with members of the activating transcription factor 2 (ATF2) family of transcription factors. Moreover, knockdown of ATF2 expression in lymphoma cells phenocopied the inhibitory effects of DN-TCF4 on the expression of target genes associated with the Wnt pathway and on cell growth. Together, our findings indicate that, through interaction with ATF2 factors, TCF1/LEF1 promote the growth of hematopoietic malignancies in the absence of ß-catenin stabilization, thus establishing a new mechanism for TCF1/LEF1 transcriptional activity distinct from that associated with canonical Wnt signaling.


Subject(s)
Activating Transcription Factor 2/genetics , Carcinogenesis/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Neoplasms/genetics , beta Catenin/genetics , Activating Transcription Factor 2/metabolism , Animals , Cell Line, Tumor , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Neoplasms/pathology , Promoter Regions, Genetic , Signal Transduction , Transcriptional Activation/genetics , Wnt Signaling Pathway/genetics , Xenopus laevis
12.
EMBO J ; 29(24): 4118-31, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21113129

ABSTRACT

Reactive oxygen species (ROS) participate in normal intracellular signalling and in many diseases including cancer and aging, although the associated mechanisms are not fully understood. Forkhead Box O (FoxO) 3 transcription factor regulates levels of ROS concentrations, and is essential for maintenance of hematopoietic stem cells. Here, we show that loss of Foxo3 causes a myeloproliferative syndrome with splenomegaly and increased hematopoietic progenitors (HPs) that are hypersensitive to cytokines. These mutant HPs contain increased ROS, overactive intracellular signalling through the AKT/mammalian target of rapamycin signalling pathway and relative deficiency of Lnk, a negative regulator of cytokine receptor signalling. In vivo treatment with ROS scavenger N-acetyl-cysteine corrects these biochemical abnormalities and relieves the myeloproliferation. Moreover, enforced expression of Lnk by retroviral transfer corrects the abnormal expansion of Foxo3(-/-) HPs in vivo. Our combined results show that loss of Foxo3 causes increased ROS accumulation in HPs. In turn, this inhibits Lnk expression that contributes to exaggerated cytokine responses that lead to myeloproliferation. Our findings could explain the mechanisms by which mutations that alter Foxo3 function induce malignancy. More generally, the work illustrates how deregulated ROS may contribute to malignant progression.


Subject(s)
Forkhead Transcription Factors/deficiency , Myeloproliferative Disorders/pathology , Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/metabolism , Acetylcysteine/therapeutic use , Adaptor Proteins, Signal Transducing , Animals , Forkhead Box Protein O3 , Free Radical Scavengers/therapeutic use , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Mice , Mice, Knockout , Myeloproliferative Disorders/drug therapy , Signal Transduction , Splenomegaly/pathology
13.
Am J Hematol ; 89(10): 954-63, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24966026

ABSTRACT

Ineffective erythropoiesis is observed in many erythroid disorders including ß-thalassemia and anemia of chronic disease in which increased production of erythroblasts that fail to mature exacerbate the underlying anemias. As loss of the transcription factor FOXO3 results in erythroblast abnormalities similar to the ones observed in ineffective erythropoiesis, we investigated the underlying mechanisms of the defective Foxo3(-/-) erythroblast cell cycle and maturation. Here we show that loss of Foxo3 results in overactivation of the JAK2/AKT/mTOR signaling pathway in primary bone marrow erythroblasts partly mediated by redox modulation. We further show that hyperactivation of mTOR signaling interferes with cell cycle progression in Foxo3 mutant erythroblasts. Importantly, inhibition of mTOR signaling, in vivo or in vitro enhances significantly Foxo3 mutant erythroid cell maturation. Similarly, in vivo inhibition of mTOR remarkably improves erythroid cell maturation and anemia in a model of ß-thalassemia. Finally we show that FOXO3 and mTOR are likely part of a larger metabolic network in erythroblasts as together they control the expression of an array of metabolic genes some of which are implicated in erythroid disorders. These combined findings indicate that a metabolism-mediated regulatory network centered by FOXO3 and mTOR control the balanced production and maturation of erythroid cells. They also highlight physiological interactions between these proteins in regulating erythroblast energy. Our results indicate that alteration in the function of this network might be implicated in the pathogenesis of ineffective erythropoiesis.


Subject(s)
Erythroblasts/metabolism , Erythropoiesis , Forkhead Transcription Factors/metabolism , Homeostasis , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Disease Models, Animal , Erythroblasts/pathology , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Mice , Mice, Knockout , TOR Serine-Threonine Kinases/genetics , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , beta-Thalassemia/pathology
14.
Article in English | MEDLINE | ID: mdl-38598120

ABSTRACT

Aflatoxin (AF) poisoning of staple foods, such as rice, is caused by fungal contamination by Aspergillus species. These AFs are genotoxic, carcinogenic and suppress the immune system. Hence, the present study was conducted to elucidate the prevalence of AF contamination in rice samples collected from local markets of Hyderabad, Telangana, India. The rice samples collected were analysed for AF by using HPLC-fluorescence detection (HPLC-FLD). Based on AF contamination levels and dietary intake of rice, the health risk was assessed by the margin of exposure (MOE) and liver cancer risk in adults, adolescence and children. The percentage detected contamination with AFB1 and AFB2 of rice samples was 54% and 34%, with the concentration ranging between 0-20.35 µg/kg and 0-1.54 µg/kg, respectively. Three rice samples exceeded the Food Safety and Standards Authority of India (FSSAI) total AF acceptable limit of 15 µg/kg. The average MOE values were 53.73, 50.58 and 35.69 (all <10,000) for adults, adolescence and children, respectively. The average liver cancer risk associated with rice consumption in the population of Hyderabad was found to be 0.27, 0.28 and 0.40 hepatocellular carcinoma (HCC) cases/year/100,000 individuals in adults, adolescence and children, respectively. This study revealed an adverse health risk to population of Hyderabad due to consumption of AF contaminated rice.


Subject(s)
Aflatoxins , Food Contamination , Oryza , Oryza/chemistry , Aflatoxins/analysis , India , Food Contamination/analysis , Humans , Risk Assessment , Child , Adult , Adolescent , Dietary Exposure/analysis , Chromatography, High Pressure Liquid , Liver Neoplasms/chemically induced
15.
J Immunol ; 187(12): 6428-36, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22105999

ABSTRACT

Several direct target genes of the p53 tumor suppressor have been identified within pathways involved in viral sensing, cytokine production, and inflammation, suggesting a potential role of p53 in antiviral immunity. The increasing need to identify immune factors to devise host-targeted therapies against pandemic influenza A virus (IAV) led us to investigate the role of endogenous wild-type p53 on the immune response to IAV. We observed that the absence of p53 resulted in delayed cytokine and antiviral gene responses in lung and bone marrow, decreased dendritic cell activation, and reduced IAV-specific CD8(+) T cell immunity. Consequently, p53(-/-) mice showed a more severe IAV-induced disease compared with their wild-type counterparts. These findings establish that p53 influences the antiviral response to IAV, affecting both innate and adaptive immunity. Thus, in addition to its established functions as a tumor suppressor gene, p53 serves as an IAV host antiviral factor that might be modulated to improve anti-IAV therapy and vaccines.


Subject(s)
Adaptive Immunity , Gene Expression Regulation, Viral/immunology , Immunity, Innate , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Orthomyxoviridae Infections/immunology , Tumor Suppressor Protein p53/physiology , Viral Regulatory and Accessory Proteins/physiology , Adaptive Immunity/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Immunity, Innate/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Pneumonia, Viral/immunology , Pneumonia, Viral/metabolism , Pneumonia, Viral/pathology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Viral Regulatory and Accessory Proteins/deficiency , Viral Regulatory and Accessory Proteins/genetics
16.
Hum Cell ; 35(2): 557-571, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35091972

ABSTRACT

Obesity poses a significant risk factor for the onset of metabolic syndrome with allied complications, wherein mesenchymal stem cell therapy is seen as a promising treatment for obesity-induced metabolic syndrome. In the present study, we aim to explore the beneficial effects of the human placental mesenchymal stromal cells (P-MSCs) on obesity-associated insulin resistance (IR) including inflammation. To understand this, we have analyzed the peripheral blood glucose, serum insulin levels by ELISA, and the glucose uptake capacity of skeletal muscle by a 2-NBDG assay using flow cytometry in WNIN/GR-Ob rats treated with and without P-MSCs. Also, we have studied insulin signaling and cytokine profile in the skeletal muscle by western blotting, dot blotting, and Multiplex-ELISA techniques. The skeletal muscle of WNIN/GR-Ob rats demonstrates dysregulation of cytokines, altered glucose uptake vis-a-vis insulin signaling. However, P-MSCs' treatment was effective in WNIN/GR-Ob rats as compared to its control, to restore HOMA-IR, re-establishes dysregulated cytokines and PI3K-Akt pathway in addition to enhanced Glut4 expression and glucose uptake studied in skeletal muscle. Overall, our data advocate the beneficial effects of P-MSCs to ameliorate inflammatory milieu, improve insulin sensitivity, and normalize glucose homeostasis underlining the Ob-T2D conditions, and we attribute for immunomodulatory, paracrine, autocrine, and multipotent functions of P-MSCs.


Subject(s)
Diabetes Mellitus, Type 2 , Mesenchymal Stem Cells , Animals , Cytokines , Female , Humans , Insulin/metabolism , Mesenchymal Stem Cells/metabolism , Muscle, Skeletal/metabolism , Obesity/therapy , Phosphatidylinositol 3-Kinases , Placenta , Pregnancy , Rats
17.
Crit Rev Oncog ; 26(1): 35-49, 2021.
Article in English | MEDLINE | ID: mdl-33641283

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a pathological condition, wherein fat deposition exceeds the allowable limits of the healthy person. If the condition persists for a long time, the patient will eventually develop NASH (nonalcoholic steatohepatitis), which will probably lead to HCC (hepatocellular carcinoma). The incidence of NAFLD is rising at an alarming rate, and still there are no drugs approved by the U.S. Food and Drug Administration for this devastating health condition. To combat and treat NAFLD successfully, it is essential to understand how routine lipid metabolism in the liver is altered under these conditions. In this review, we discuss specifically during the NAFLD progression how the signaling pathways leading to excess fat accumulation in the liver are changed. We also address variations in the mechanisms underlying hepatic lipid uptake and changes in fatty acid oxidation mechanisms. We will also highlight the role of transcription factors and other lipolytic enzymes that stringently regulate the hepatic de novo lipolysis (DNL) and emphasize how they are altered during NAFLD progression. Finally, we will also touch upon how the lipid disposal from the liver goes wrong during the NAFLD progression. A comprehensive understanding of the changes in lipid metabolism is essential for developing successful therapies for NAFLD.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Disease Progression , Humans , Lipid Metabolism , Lipids
18.
Sci Rep ; 11(1): 16983, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34417511

ABSTRACT

Obesity (Ob) poses a significant risk factor for the onset of metabolic syndrome with associated complications, wherein the Mesenchymal Stem Cell (MSC) therapy shows pre-clinical success. Here, we explore the therapeutic applications of human Placental MSCs (P-MSCs) to address Ob-associated Insulin Resistance (IR) and its complications. In the present study, we show that intramuscular injection of P-MSCs homed more towards the visceral site, restored HOMA-IR and glucose homeostasis in the WNIN/GR-Ob (Ob-T2D) rats. P-MSC therapy was effective in re-establishing the dysregulated cytokines. We report that the P-MSCs activates PI3K-Akt signaling and regulates the Glut4-dependant glucose uptake and its utilization in WNIN/GR-Ob (Ob-T2D) rats compared to its control. Our data reinstates P-MSC treatment's potent application to alleviate IR and restores peripheral blood glucose clearance evidenced in stromal vascular fraction (SVF) derived from white adipose tissue (WAT) of the WNIN/GR-Ob rats. Gaining insights, we show the activation of the PI3K-Akt pathway by P-MSCs both in vivo and in vitro (palmitate primed 3T3-L1 cells) to restore the insulin sensitivity dysregulated adipocytes. Our findings suggest a potent application of P-MSCs in  pre-clinical/Ob-T2D management.


Subject(s)
Diabetes Mellitus, Experimental/prevention & control , Diabetes Mellitus, Experimental/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Models, Biological , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Cells, Cultured , Cytokines/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/prevention & control , Diabetes Mellitus, Type 2/therapy , Female , Glucose Transporter Type 4/metabolism , Homeostasis , Humans , Insulin/metabolism , Macrophages/metabolism , Obesity/complications , Phosphatidylinositol 3-Kinases/metabolism , Placenta/cytology , Pregnancy , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction
19.
Crit Rev Oncog ; 25(1): 57-70, 2020.
Article in English | MEDLINE | ID: mdl-32865911

ABSTRACT

The gut microbiome (GM) is a multifaceted environment wherein nearly 1014 microorganisms play various roles in host immune regulation, intestinal cell proliferation, bone mineralization, xenobiotics metabolism, and protection against pathogens. GM is also strongly coupled with the development and progression of nutrition-related diseases such as nonalcoholic fatty liver disease (NAFLD), wherein the gut-liver axis plays a major role as the gut and liver are functionally and anatomically associated through the portal vein. Dysbiosis causes leaky gut, resulting in the activation of inflammatory processes in the liver. Disruption of the gut barrier enhances microbial infiltration into the sub-mucosae, which through the bloodstream causes harmful microbial metabolites, such as butyrate, long-chain fatty acids, endotoxins, and indole-3-acetic acid, to seep into the liver. In NAFLD patients, these metabolites can lead to the development of nonalcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). In this review, we will discuss the important molecular pathways through which various metabolites and other signaling substances released by the GM regulate liver biology, under both physiological and pathological conditions. Finally, we highlight numerous therapeutic attempts, such as probiotics, prebiotics, and fecal microbial transplantation (FMT), to reprogram the gut-liver axis for decreasing liver diseases.


Subject(s)
Gastrointestinal Microbiome/physiology , Non-alcoholic Fatty Liver Disease/etiology , Animals , Choline/metabolism , Disease Progression , Dysbiosis , Fecal Microbiota Transplantation , Humans , Oxidative Stress , Probiotics/therapeutic use
20.
Cancer Res ; 66(9): 4715-24, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16651424

ABSTRACT

Notch signaling is believed to promote cell survival in general. However, the mechanism is not clearly understood. Here, we show that cells expressing intracellular domain of human Notch1 (NIC-1) are chemoresistant in a wild-type p53-dependent manner. NIC-1 inhibited p53 by inhibiting its activating phosphorylations at Ser(15), Ser(20), and Ser(392) as well as nuclear localization. In addition, we found that inhibition of p53 by NIC-1 mainly occurs through mammalian target of rapamycin (mTOR) using phosphatidylinositol 3-kinase (PI3K)-Akt/protein kinase B (PKB) pathway as the mTOR inhibitor, rapamycin treatment abrogated NIC-1 inhibition of p53 and reversed the chemoresistance. Consistent with this, rapamycin failed to reverse NIC-1-induced chemoresistance in cells expressing rapamycin-resistant mTOR. Further, ectopic expression of eukaryotic initiation factor 4E (eIF4E), a translational regulator that acts downstream of mTOR, inhibited p53-induced apoptosis and conferred protection against p53-mediated cytotoxicity to similar extent as that of NIC-1 overexpression but was not reversed by rapamycin, which indicates that eIF4E is the major target of mTOR in Notch1-mediated survival signaling. Finally, we show that MCF7 (breast cancer) and MOLT4 (T-cell acute lymphoblastic leukemia) cells having aberrant Notch1 signaling are chemoresistant, which can be reversed by both PI3K and mTOR inhibitors. These results establish that Notch1 signaling confers chemoresistance by inhibiting p53 pathway through mTOR-dependent PI3K-Akt/PKB pathway and imply that p53 status perhaps is an important determinant in combination therapeutic strategies, which use mTOR inhibitors and chemotherapy.


Subject(s)
Protein Kinases/metabolism , Receptor, Notch1/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Nucleus/metabolism , Cell Survival/physiology , Drug Resistance, Neoplasm , Eukaryotic Initiation Factor-4E/biosynthesis , Humans , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Receptor, Notch1/biosynthesis , Receptor, Notch1/genetics , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL