Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Carcinog ; 61(7): 702-716, 2022 07.
Article in English | MEDLINE | ID: mdl-35452563

ABSTRACT

Emerging evidence suggests an important role for SIRT1, a nicotinamide adenine dinucleotide (NAD)-dependent deacetylase in cancer development, progression and therapeutic resistance; making it a viable therapeutic target. Here, we examined the impact of resveratrol-mediated pharmacological activation of SIRT1 on the progression of HGPIN lesions (using the Pten-/- mouse model) and on prostate tumor development (using an orthotopic model of prostate cancer cells stably silenced for SIRT1). We show that precise SIRT1 modulation could benefit both cancer prevention and treatment. Positive effect of SIRT1 activation can prevent Pten deletion-driven development of HGPIN lesions in mice if resveratrol is administered early (pre-cancer stage) with little to no benefit after the establishment of HGPIN lesions or tumor cell implantation. Mechanistically, our results show that under androgen deprivation conditions, SIRT1 inhibition induces senescence as evidenced by decreased gene signature associated with negative regulators of senescence and increased senescence-associated ß-galactosidase activity. Furthermore, pharmacological inhibition of SIRT1 potentiated growth inhibitory effects of clinical androgen receptor blockade agents and radiation. Taken together, our findings provide an explanation for the discrepancy regarding the role of SIRT1 in prostate tumorigenesis. Our results reveal that the bifurcated roles for SIRT1 may occur in stage and context-dependent fashion by functioning in an antitumor role in prevention of early-stage prostate lesion development while promoting tumor development and disease progression post-lesion development. Clinically, these data highlight the importance of precise SIRT1 modulation to provide benefits for cancer prevention and treatment including sensitization to conventional therapeutic approaches.


Subject(s)
Prostatic Intraepithelial Neoplasia , Prostatic Neoplasms , Androgen Antagonists/pharmacology , Animals , Cellular Senescence , Humans , Male , Mice , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/prevention & control , Resveratrol/pharmacology , Sirtuin 1/genetics
2.
Mol Carcinog ; 58(6): 854-861, 2019 06.
Article in English | MEDLINE | ID: mdl-30859654

ABSTRACT

Published evidence shows a correlation between several molecular markers and prostate cancer (PCa) progression including in African Americans (AAs) who are disproportionately affected. Our early detection efforts led to the identification of elevated levels of antiapoptotic protein, c-FLIP and its upstream regulatory factors such as androgen receptor (AR), recepteur d'origine nantais (RON), a receptor tyrosine kinase in human prostate tumors. The primary objective of this study was to explore whether these markers play a role in racial disparities using immunohistochemistry in prostatectomy samples from a cohort of AA, Hispanic Whites (HWs), and non-Hispanic Whites (NHWs). Bivariable and multivariable logistic regression analyses were used to identify a statistical association between molecular markers, possible correlation with risk factors including race, obesity, prostate-specific antigen (PSA) and disease aggressiveness. Further, changes in the levels and expression of these molecular markers were also evaluated using human PCa cell lines. We found significantly elevated levels of RON ( P = 0.0082), AR ( P = 0.0001), c-FLIP ( P = 0.0071) in AAs compared with HWs or NHWs. Furthermore, a higher proportion of HW and NHWs had a high Gleason score (>6) but not PSA as compared to AAs ( P = 0.032). In summary, our findings suggest that PSA was important in predicting aggressive disease for the cohort overall; however, high levels of RON may play a role in predisposing AA men to develop aggressive disease. Future research is needed using large datasets to confirm these findings and to explore whether all or any of these markers could aid in race-specific stratification of patients for treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Prostatic Neoplasms/ethnology , Prostatic Neoplasms/surgery , Receptor Protein-Tyrosine Kinases/metabolism , Up-Regulation , Adult , Black or African American , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Line, Tumor , Cohort Studies , Gene Expression Regulation, Neoplastic , Hispanic or Latino , Humans , Logistic Models , Male , Middle Aged , Neoplasm Invasiveness , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Androgen/metabolism , White People
3.
Lab Invest ; 95(10): 1092-104, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26214584

ABSTRACT

The balance of nitric oxide (NO) versus superoxide generation has a major role in the initiation and progression of endothelial dysfunction. Under conditions of high glucose, endothelial nitric oxide synthase (eNOS) functions as a chief source of superoxide rather than NO. In order to improve NO bioavailability within the vessel wall in type-1 diabetes, we investigated treatment strategies that improve eNOS phosphorylation and NO-dependent vasorelaxation. We evaluated methods to increase the eNOS activity by (1) feeding Ins2(Akita) spontaneously diabetic (type-1) mice with l-arginine in the presence of sepiapterin, a precursor of tetrahydrobiopterin; (2) preventing eNOS/NO deregulation by the inclusion of inhibitor kappa B kinase beta (IKKß) inhibitor, salsalate, in the diet regimen in combination with l-arginine and sepiapterin; and (3) independently increasing eNOS expression to improve eNOS activity and associated NO production through generating Ins2(Akita) diabetic mice that overexpress human eNOS predominantly in vascular endothelial cells. Our results clearly demonstrated that diet supplementation with l-arginine, sepiapterin along with salsalate improved phosphorylation of eNOS and enhanced vasorelaxation of thoracic/abdominal aorta in type-1 diabetic mice. More interestingly, despite the overexpression of eNOS, the in-house generated transgenic eNOS-GFP (TgeNOS-GFP)-Ins2(Akita) cross mice showed an unanticipated effect of reduced eNOS phosphorylation and enhanced superoxide production. Our results demonstrate that enhancement of endogenous eNOS activity by nutritional modulation is more beneficial than increasing the endogenous expression of eNOS by gene therapy modalities.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Dietary Supplements , Endothelium, Vascular/metabolism , Hypoglycemic Agents/therapeutic use , Nitric Oxide Synthase Type III/metabolism , Protein Kinase Inhibitors/therapeutic use , Protein Processing, Post-Translational , Animals , Aorta/cytology , Aorta/metabolism , Aorta/physiopathology , Arginine/metabolism , Arginine/therapeutic use , Cattle , Cells, Cultured , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiopathology , Female , Heterozygote , Humans , Hypoglycemic Agents/metabolism , Insulin/genetics , Insulin/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide Synthase Type III/chemistry , Nitric Oxide Synthase Type III/genetics , Phosphorylation , Protein Kinase Inhibitors/metabolism , Pterins/metabolism , Pterins/therapeutic use , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Salicylates/metabolism , Salicylates/therapeutic use , Weaning
4.
Proc Natl Acad Sci U S A ; 108(21): 8779-84, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21555558

ABSTRACT

Diabetes and the development of its complications have been associated with mitochondrial DNA (mtDNA) dysfunction, but causal relationships remain undetermined. With the objective of testing whether increased mtDNA mutations exacerbate the diabetic phenotype, we have compared mice heterozygous for the Akita diabetogenic mutation (Akita) with mice homozygous for the D257A mutation in mitochondrial DNA polymerase gamma (Polg) or with mice having both mutations (Polg-Akita). The Polg-D257A protein is defective in proofreading and increases mtDNA mutations. At 3 mo of age, the Polg-Akita and Akita male mice were equally hyperglycemic. Unexpectedly, as the Polg-Akita males aged to 9 mo, their diabetic symptoms decreased. Thus, their hyperglycemia, hyperphagia and urine output declined significantly. The decrease in their food intake was accompanied by increased plasma leptin and decreased plasma ghrelin, while hypothalamic expression of the orexic gene, neuropeptide Y, was lower and expression of the anorexic gene, proopiomelanocortin, was higher. Testis function progressively worsened with age in the double mutants, and plasma testosterone levels in 9-mo-old Polg-Akita males were significantly reduced compared with Akita males. The hyperglycemia and hyperphagia returned in aged Polg-Akita males after testosterone administration. Hyperglycemia-associated distal tubular damage in the kidney also returned, and Polg-D257A-associated proximal tubular damage was enhanced. The mild diabetes of female Akita mice was not affected by the Polg-D257A mutation. We conclude that reduced diabetic symptoms of aging Polg-Akita males results from appetite suppression triggered by decreased testosterone associated with damage to the Leydig cells of the testis.


Subject(s)
Appetite/genetics , DNA, Mitochondrial/genetics , DNA-Directed DNA Polymerase/genetics , Diabetes Mellitus/genetics , Mice, Inbred Strains/genetics , Mutation , Aging , Animals , DNA Polymerase gamma , Diabetes Mellitus/pathology , Diabetes Mellitus/physiopathology , Female , Hyperglycemia , Leydig Cells/pathology , Male , Mice , Phenotype , RNA Editing/genetics , Testis/pathology , Testosterone/blood
5.
Mol Carcinog ; 52(4): 275-85, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22213062

ABSTRACT

The prevalence of hepatocellular carcinoma (HCC) was diminished from 60% to 18% at 15 months of age in C3HeB/FeJ male transgenic mice expressing hMGMT in our previous studies. To directly test if the methyltransferase activity is required for diminished tumor prevalence, two separate lines of transgenic mice bearing an enzymatically inactive form of hMGMT were used. In these lines, cysteine 145 was substituted with alanine (C145A). Expression of the hMGMT C145A transgene in liver was demonstrated by Northern blots and Western blots. Immunohistochemistry revealed predominantly nuclear localization of the hMGMT C145A protein. hMGMT C145A transgenic mice were crossed with lacI transgenic mice to assess mutant frequencies in the presence of the mutant protein. Mutant frequencies were similar among livers of lacI × hMGMT C145A bi-transgenic mice and lacI × wild-type (WT) mice. DNA sequence analysis of recovered lacI mutants revealed similar mutation spectra for hMGMT C145A and WT mice. The prevalence of HCC was also similar for the two tested lines of hMGMT C145A mice, 45% and 48% prevalence with median tumor sizes of 11 and 8 mm, and WT mice, 40% prevalence and median tumor size of 10 mm. These results provide evidence that residue C145 in hMGMT is required to reduce the prevalence of HCC in C3HeB/FeJ mice transgenic for hMGMT.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver/pathology , O(6)-Methylguanine-DNA Methyltransferase/genetics , Amino Acid Substitution , Animals , Carcinoma, Hepatocellular/enzymology , Enzyme Activation , Humans , Liver/enzymology , Liver/metabolism , Liver Neoplasms/enzymology , Male , Mice , Mice, Transgenic , O(6)-Methylguanine-DNA Methyltransferase/analysis , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Transgenes
6.
PLoS One ; 17(12): e0279694, 2022.
Article in English | MEDLINE | ID: mdl-36584001

ABSTRACT

A perfluorocarbon (PFC) investigated for treatment of traumatic brain injury (TBI) delivers oxygen to support brain function, but causes transient thrombocytopenia. TBI can cause acute inflammation with resulting thrombocytopenia; an interaction between the PFC effects and TBI inflammation might exacerbate thrombocytopenia. Therefore, PFC effects on platelet (PLT) function and hemostasis in a lipopolysaccharide (LPS) model of inflammation in the baboon were studied. Animals were randomized to receive saline ±LPS, and ± one of two doses of PFC. PLT count, transmission electron microscopy, and microparticle populations were quantified at baseline (BL) and at 2, 24, 48, 72, and 96 hours; hemostatic parameters for aggregometry and for blood clotting were measured at baseline (BL) and days 3 and 4. Injection of vehicle and LPS caused thrombocytopenia within hours; PFCs caused delayed thrombocytopenia beginning 48 hours post-infusion. LPS+PFC produced a more prolonged PLT decline and decreased clot strength. LPS+PFC increased ADP-stimulated aggregation, but PFC alone did not. Microparticle abundance was greatest in the LPS+PFC groups. LPS+PFC caused diffuse microvascular hemorrhage and death in 2 of 5 baboons in the low dose LPS-PFC group and 2 of 2 in the high dose LPS-PFC group. Necropsy and histology suggested death was caused by shock associated with hemorrhage in multiple organs. Abnormal morphology of platelets and red blood cells were notable for PFC inclusions. In summary, PFC infusion caused clinically significant thrombocytopenia and exacerbated LPS-induced platelet activation. The interaction between these effects resulted in decreased hemostatic capacity, diffuse bleeding, shock and death.


Subject(s)
Fluorocarbons , Inflammation , Animals , Disease Models, Animal , Erythrocytes/drug effects , Erythrocytes/pathology , Fluorocarbons/poisoning , Hemorrhage/chemically induced , Hemostatics , Inflammation/drug therapy , Lipopolysaccharides , Thrombocytopenia/chemically induced
7.
J Cutan Pathol ; 38(5): 420-31, 2011 May.
Article in English | MEDLINE | ID: mdl-21251037

ABSTRACT

Trichodysplasia spinulosa (TS) is a folliculocentric and clinically papular dermatological disorder occurring in the setting of immunosuppression typically in association with solid organ transplantation or hematolymphoid malignancies. We report the occurrence of TS in a 7-year-old girl with Down syndrome and pre-B-acute lymphoblastic leukemia who was completing chemotherapy at onset. The patient's affected follicles were dilated by an expansion of a dystrophic follicular inner root sheath cell population displaying enlarged trichohyaline cytoplasmic granules and progressing centrally to keratotic and parakeratotic debris, and superficially demonstrating some diminutive hair shaft-like material within the keratotic spicules. Electron microscopic studies of a follicular lesion showed extracellular viral particles suggestive of a polyomavirus within the central follicular keratotic debris. DNA polymerase chain reaction (PCR) and gene sequencing studies, performed on the tissue of the microscopic slide and paraffin block, for the recently identified TS-associated polyomavirus (TSPyV) resulted as positive for TSPyV. PCR for the Merkel cell polyomavirus was negative. To date, this case is unique in representing the first case of TS confirmed by electron microscopy in which a related viral pathogen has been molecularly identified. An additional 19 reported cases classifiable as TS are tabulated and reviewed.


Subject(s)
Hair Diseases/pathology , Hair Diseases/virology , Hair Follicle/pathology , Hair Follicle/virology , Polyomavirus Infections/pathology , Polyomavirus Infections/virology , Polyomavirus , Child , DNA, Viral/metabolism , Down Syndrome/complications , Down Syndrome/metabolism , Down Syndrome/pathology , Down Syndrome/therapy , Female , Hair Diseases/metabolism , Hair Follicle/metabolism , Humans , Polyomavirus Infections/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology
8.
J Gerontol A Biol Sci Med Sci ; 75(4): 647-653, 2020 03 09.
Article in English | MEDLINE | ID: mdl-30423026

ABSTRACT

Nuclear factor (NF)κB is a transcription factor that controls immune and inflammatory signaling pathways. In skeletal muscle, NFκB has been implicated in the regulation of metabolic processes and tissue mass, yet its affects on mitochondrial function in this tissue are unclear. To investigate the role of NFκB on mitochondrial function and its relationship with muscle mass across the life span, we study a mouse model with muscle-specific NFκB suppression (muscle-specific IκBα super-repressor [MISR] mice). In wild-type mice, there was a natural decline in muscle mass with aging that was accompanied by decreased mitochondrial function and mRNA expression of electron transport chain subunits. NFκB inactivation downregulated expression of PPARGC1A, and upregulated TFEB and PPARGC1B. NFκB inactivation also decreased gastrocnemius (but not soleus) muscle mass in early life (1-6 months old). Lower oxygen consumption rates occurred in gastrocnemius and soleus muscles from young MISR mice, whereas soleus (but not gastrocnemius) muscles from old MISR mice displayed increased oxygen consumption compared to age-matched controls. We conclude that the NFκB pathway plays an important role in muscle development and growth. The extent to which NFκB suppression alters mitochondrial function is age dependent and muscle specific. Finally, mitochondrial function and muscle mass are tightly associated in both genotypes and across the life span.


Subject(s)
Mitochondria, Muscle/physiology , Muscle Development/physiology , NF-kappa B/physiology , Aging/genetics , Aging/pathology , Aging/physiology , Animals , Citrate (si)-Synthase/metabolism , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Muscle/genetics , Muscle Development/genetics , Muscle, Skeletal/physiology , Muscle, Skeletal/ultrastructure , NF-kappa B/antagonists & inhibitors , Oxygen Consumption , Signal Transduction
9.
Commun Biol ; 3: 12, 2020.
Article in English | MEDLINE | ID: mdl-31909204

ABSTRACT

NAD[P]H:quinone oxidoreductase 1 (NQO1) regulates cell fate decisions in response to stress. Oxidative stress supports cancer maintenance and progression. Previously we showed that knockdown of NQO1 (NQO1low) prostate cancer cells upregulate pro-inflammatory cytokines and survival under hormone-deprived conditions. Here, we tested the ability of NQO1low cells to form tumors. We found NQO1low cells form aggressive tumors compared with NQO1high cells. Biopsy specimens and circulating tumor cells showed biochemical recurrent prostate cancer was associated with low NQO1. NQO1 silencing was sufficient to induce SMAD-mediated TGFß signaling and mesenchymal markers. TGFß treatment decreased NQO1 levels and induced molecular changes similar to NQO1 knockdown cells. Functionally, NQO1 depletion increased migration and sensitivity to oxidative stress. Collectively, this work reveals a possible new gatekeeper role for NQO1 in counteracting cellular plasticity in prostate cancer cells. Further, combining NQO1 with TGFß signaling molecules may serve as a better signature to predict biochemical recurrence.


Subject(s)
Cell Plasticity/genetics , NAD(P)H Dehydrogenase (Quinone)/genetics , Oxidative Stress , Prostatic Neoplasms/physiopathology , Transforming Growth Factor beta/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Nude , NAD(P)H Dehydrogenase (Quinone)/metabolism , Prostatic Neoplasms/genetics , Transforming Growth Factor beta/metabolism , Up-Regulation/physiology
10.
Prostate ; 69(7): 755-60, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19170049

ABSTRACT

BACKGROUND: Transgenic adenocarcinoma of mouse prostate (TRAMP) mice, derived by prostate specific expression of SV40 large T antigen using the rat probasin promoter, all develop prostate tumors akin to human prostate cancers. More recently, epithelial-stromal (ES) tumors resembling phyllodes tumors have been described in the seminal vesicles of TRAMP mice. We report malignancy arising in these ES tumors of the seminal vesicles in TRAMP mice. METHODS: H&E stained sections from 28-week-old TRAMP mice autopsies were examined. Immunostains (cytokeratin, vimentin, desmin, and MIB-1) and electron microscopy were performed on selected blocks of the genitourinary system and metastatic tumor nodules. RESULTS: The seminal vesicles frequently develop tumors containing broad papillae, with bland epithelium and a cellular spindled stroma just beneath the epithelium. The stromal cells have high nuclear to cytoplasmic ratio, frequent apoptotic cells and mitoses. In some cases, the stromal cells become large mass lesions that overgrow the prostate. The epithelium can also proliferate and become malignant. The tumors have high proliferation indices by MIB-1. Some metastatic tumors have characteristics similar to the seminal vesicle ES tumor. CONCLUSIONS: Metastatic tumors in TRAMP mice show three patterns: (1) A definite adenocarcinoma pattern metastatic from the prostate; (2) poorly differentiated tumor without epithelial differentiation; (3) carcinosarcomatous pattern. The carcinosarcomatous pattern and some of the poorly differentiated tumors likely arise from seminal vesicle ES tumors.


Subject(s)
Adenocarcinoma/pathology , Prostatic Neoplasms/pathology , Seminal Vesicles/pathology , Adenocarcinoma/genetics , Adenocarcinoma/ultrastructure , Animals , Disease Models, Animal , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Prostatic Neoplasms/genetics , Prostatic Neoplasms/ultrastructure , Seminal Vesicles/ultrastructure
11.
J Diabetes Complications ; 33(1): 23-32, 2019 01.
Article in English | MEDLINE | ID: mdl-30424931

ABSTRACT

Previous studies demonstrated that global deficiency of eNOS in diabetic mice exacerbated renal lesions and that overexpression of eNOS may protect against tissue injury. Our study revealed for the first time overexpression of eNOS leads to disease progression rather than protection. Transgenic mice selectively expressing eNOS in endothelial cells (eNOSTg) were cross bred with Ins2Akita type-1 (AK) diabetic mice to generate eNOS overexpressing eNOSTg/AK mice. Wild type, eNOSTg, AK and eNOSTg/AK mice were assessed for kidney function and blood glucose levels. Remarkably, overexpressing eNOSTg mice showed evidence of unpredicted glomerular injury with segmental mesangiolysis and occasional microaneurysms. Notably, in eNOSTg/AK mice overexpression of eNOS led to increased glomerular/endothelial injury that was associated with increased superoxide levels and renal dysfunction. Results indicate for the first time that overexpressing eNOS in endothelial cells cannot ameliorate diabetic lesions, but paradoxically leads to progression of nephropathy likely due to eNOS uncoupling and superoxide upsurge. This novel finding has a significant impact on current therapeutic strategies to improve endothelial function and prevent progression of diabetic renal disease. Further, the eNOSTg/AK model developed in this study has significant translational potentials for elucidating the underlying mechanism implicated in the deflected function of eNOS in diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/metabolism , Endothelium, Vascular/metabolism , Kidney Glomerulus/metabolism , Nitric Oxide Synthase Type III/biosynthesis , Animals , Diabetic Nephropathies/diagnostic imaging , Diabetic Nephropathies/pathology , Disease Models, Animal , Disease Progression , Endothelium, Vascular/diagnostic imaging , Endothelium, Vascular/pathology , Insulin/genetics , Kidney Glomerulus/diagnostic imaging , Kidney Glomerulus/pathology , Mice , Mice, Inbred Strains , Mice, Transgenic , Microscopy, Electron
12.
Nat Commun ; 10(1): 4720, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31624249

ABSTRACT

Understanding the molecular components of insulin signaling is relevant to effectively manage insulin resistance. We investigated the phenotype of the TMEM127 tumor suppressor gene deficiency in vivo. Whole-body Tmem127 knockout mice have decreased adiposity and maintain insulin sensitivity, low hepatic fat deposition and peripheral glucose clearance after a high-fat diet. Liver-specific and adipose-specific Tmem127 deletion partially overlap global Tmem127 loss: liver Tmem127 promotes hepatic gluconeogenesis and inhibits peripheral glucose uptake, while adipose Tmem127 downregulates adipogenesis and hepatic glucose production. mTORC2 is activated in TMEM127-deficient hepatocytes suggesting that it interacts with TMEM127 to control insulin sensitivity. Murine hepatic Tmem127 expression is increased in insulin-resistant states and is reversed by diet or the insulin sensitizer pioglitazone. Importantly, human liver TMEM127 expression correlates with steatohepatitis and insulin resistance. Our results suggest that besides tumor suppression activities, TMEM127 is a nutrient-sensing component of glucose/lipid homeostasis and may be a target in insulin resistance.


Subject(s)
Adipose Tissue/metabolism , Genes, Tumor Suppressor , Insulin Resistance/genetics , Liver/metabolism , Membrane Proteins/genetics , Adipogenesis/genetics , Animals , Diet, High-Fat , Gene Expression Profiling/methods , Gluconeogenesis/genetics , Humans , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Specificity/genetics
13.
Lab Invest ; 88(5): 515-28, 2008 May.
Article in English | MEDLINE | ID: mdl-18391994

ABSTRACT

Functional consequences of impaired endothelial nitric oxide synthase (eNOS) activity causing organ-specific abnormalities on a diabetic setting are not completely understood. In this study, we extensively characterized a diabetic mouse model (lepr(db/db)) in which eNOS expression is genetically disrupted (eNOS-/-). The eNOS-/-/ lepr(db/db) double-knockout (DKO) mice developed obesity, hyperglycemia, hyperinsulinemia and hypertension. Analysis of tissues from DKO mice showed large islets in the pancreas and fat droplets in hepatocytes. Interestingly, the aorta was normal and atherogenic lesions were not observed. Abnormalities in the aorta including poor re-endothelialization and increased medial wall thickness were evident only in response to deliberate injury. In contrast, significant glomerular capillary damage in the kidney was identified, with DKO mice demonstrating a robust diabetic nephropathy similar to human disease. The vascular and renal impairments in DKO mice were pronounced despite lower fasting plasma glucose levels compared to lepr(db/db) mice, indicating that eNOS is a critical determinant of hyperglycemia-induced organ-specific complications and their severity in diabetes. Results provide the first evidence that absence of eNOS in diabetes has a greater deleterious effect on the renal microvasculature than on the larger aortic vessel. The DKO model may suggest novel therapeutic strategies to prevent both vascular and renal complications of diabetes.


Subject(s)
Diabetes Complications , Diabetes Mellitus/enzymology , Diabetes Mellitus/genetics , Diabetic Angiopathies/etiology , Nitric Oxide Synthase Type III/deficiency , Albuminuria/physiopathology , Animals , Aorta, Abdominal/injuries , Aorta, Abdominal/pathology , Body Weight , Creatinine/urine , Diabetes Mellitus/pathology , Diabetes Mellitus/physiopathology , Diabetic Angiopathies/pathology , Diabetic Angiopathies/physiopathology , Endothelium, Vascular/pathology , Genotype , Hypertension/etiology , Insulin Resistance , Kidney/blood supply , Kidney/pathology , Kidney Glomerulus/pathology , Kidney Glomerulus/ultrastructure , Macrophages/pathology , Mice , Mice, Knockout , Mice, Mutant Strains , Microcirculation
14.
J Clin Endocrinol Metab ; 103(8): 2802-2806, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29878124

ABSTRACT

Context: Mutations in genes encoding for the succinate dehydrogenase (SDH) complex are linked to hereditary paraganglioma syndromes. Paraganglioma syndrome 3 is associated with mutations in SDHC and typically manifests as benign, nonfunctional head and neck paragangliomas. Design: We describe a case of a 51-year-old woman who initially presented with diarrhea and hypertension and was found to have a retroperitoneal mass, which was resected with a pathology consistent with paraganglioma. Five years later, her symptoms recurred, and she was found to have new retroperitoneal lymphadenopathy and lytic lesions in the first lumbar vertebral body and the right iliac crest, which were visualized on CT scan and octreoscan but not on iodine-123-meta-iodobenzylguanidine (123I-MIBG) and bone scans. She had significantly elevated 24-hour urine norepinephrine and dopamine. The patient received external beam radiation and a series of different antineoplastic agents. Her disease progressed, and she eventually expired within 2 years. Genetic testing revealed a heterozygous SDHC c.43C>T, p.Arg15X mutation, which was also detected in her daughter and her grandson, both of whom have no biochemical or imaging evidence of paraganglioma syndrome yet. Conclusion: We report a unique case of functional, metastatic abdominal paraganglioma associated with SDHC germline mutation. Our case exemplifies that SDHC germline mutation has variable penetrance, which may manifest with an aggressive biology that could be missed by a 123I-MIBG scan.


Subject(s)
Adrenal Gland Neoplasms/genetics , Germ-Line Mutation , Membrane Proteins/genetics , Paraganglioma/genetics , Adrenal Gland Neoplasms/diagnosis , Adrenal Gland Neoplasms/pathology , Fatal Outcome , Female , Genetic Testing , Humans , Middle Aged , Neoplasm Metastasis , Paraganglioma/diagnosis , Paraganglioma/pathology , Pheochromocytoma/diagnosis , Pheochromocytoma/genetics , Pheochromocytoma/pathology
15.
Cancer Lett ; 425: 101-115, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29608984

ABSTRACT

Molecular targeted compounds are emerging as a strategy to improve classical chemotherapy. Herein, we describe that using low dose of the multikinase inhibitor sorafenib improves cyclophosphamide antitumor activity by inhibiting angiogenesis, metastasis and promoting tumor healing in MDA-MB231 xenografts and the 4T1-12B syngeneic breast cancer metastasis model. Mechanistic studies in MDA-MB231 cells revealed that alkylation upregulates inflammatory genes/proteins such as COX-2, IL8, CXCL2 and MMP1 in a MEK1/2-ERK1/2-dependent manner. These proteins enrich the secretome of cancer cells, stimulating cell invasion and angiogenesis via autocrine and paracrine mechanisms. Sorafenib inhibits MEK1/2-ERK1/2 pathway thereby decreasing inflammatory genes and mitigating cell invasion and angiogenesis at basal and alkylation-induced conditions whereas NRF2 and ER stress pathways involved in alkylation survival are not affected. In non-invasive/non-angiogenic breast cancer cells (SKBR3 and MCF7), alkylation did not elicit inflammatory responses with the only sorafenib effect being ERK1/2-independent ROS-dependent cytotoxicity when using higher drug concentrations. In summary, our data show that alkylating agents may elicit inflammatory responses that seems to contribute to malignant progression in specific breast cancer cells. Identifying and targeting drivers of this phenotype may offer opportunities to optimize combined drug regimens between classical chemotherapeutics and targeted agents.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Breast Neoplasms/drug therapy , Cyclophosphamide/administration & dosage , Neovascularization, Pathologic/drug therapy , Sorafenib/administration & dosage , Animals , Antineoplastic Agents, Alkylating/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclophosphamide/pharmacology , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Mice , Signal Transduction/drug effects , Sorafenib/pharmacology , Xenograft Model Antitumor Assays
16.
Cancer Lett ; 433: 232-241, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30003927

ABSTRACT

Radiation therapy (XRT) is a standard treatment for prostate cancer (PCa). Although dose escalation increases local control, toxicity hampers further escalation. Broader improvement will be possible by the addition of adjuvant therapies, which can synergize with radiation and thus improve efficacy. We have identified a natural compound (Nexrutine, Nx) that inhibits the survival and growth of PCa cells in combination with radiation. Combination studies demonstrated strong interaction between Nx and radiation both in vitro in multiple PCa cell lines and in the Transgenic adenocarcinoma of mouse prostate (TRAMP) model. Nx potentiated growth inhibitory effects of IR by down regulating ribosomal protein S6K (RPS6KB1), CyclinD1, Chk1 and HIF-1 α and prolonging G2/M checkpoint block. RPS6KB1 is upregulated in prostate cancers and its expression is correlated with tumor grade. Knockdown of RPS6KB1 in PCa cells increased their sensitivity toward radiation-induced survival inhibition. Overall, we provide scientific evidence (i) in support of Nx as an adjuvant in PCa patients receiving XRT (ii) suggesting that RPS6KB1 is an important player in Nx-mediated combinatorial benefits and emphasizes that RPS6KB1 is a novel target for PCa treatment. These data underscore the need to test the agent in additional preclinical models to validate these observations.


Subject(s)
Antineoplastic Agents/pharmacology , Plant Extracts/pharmacology , Prostatic Neoplasms/radiotherapy , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Checkpoint Kinase 1/biosynthesis , Cyclin D1/biosynthesis , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Male , Mice , PC-3 Cells , Ribosomal Protein S6 Kinases, 70-kDa/biosynthesis
17.
J Clin Invest ; 113(6): 856-66, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15067318

ABSTRACT

The prevailing paradigm is that in human rheumatoid arthritis (RA), the accumulation of monocytes and T cells in the joint, mediated in part by such CC chemokine receptors (CCRs) as CCR2 and CCR5, respectively, plays a central role in disease pathogenesis. To further validate this paradigm, we conducted proof-of-principle studies and tested the hypothesis that gene inactivation of Ccr2 or Ccr5 will ameliorate experimental RA. Contrary to our expectations, we found that in two well-established murine models of experimental RA, CCR2 expression in the hematopoietic cell compartment served as a negative regulator of autoantibody production as well as arthritic disease onset, severity, and resolution. In contrast, the RA phenotype in Ccr5-null mice was similar to that of WT mice. Remarkably, the collagen-induced arthritis phenotype of Ccr2-/- mice mimicked closely that of severe human RA, including production of rheumatoid factor, enhanced T cell production, and monocyte/macrophage accumulation in the joints. Our findings demonstrate an essential protective role of CCR2 expression in RA, indicate the existence of alternative receptors responsible for monocyte/macrophage accumulation to inflamed joints, and emphasize the need to clarify carefully the complex effects of the chemokine system in RA before they can be considered as therapeutic targets.


Subject(s)
Arthritis, Experimental/genetics , Arthritis, Rheumatoid/genetics , Receptors, Chemokine/genetics , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Collagen Type II/immunology , Humans , Mice , Models, Animal , Receptors, CCR2 , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism
18.
J Mol Med (Berl) ; 84(6): 503-12, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16520943

ABSTRACT

The host factors that influence autoimmune arthritides such as rheumatoid arthritis have not been fully elucidated. We previously found that genetic inactivation of CC chemokine receptor 2 (CCR2) in the arthritis-prone DBA/1j mouse strain significantly increases the susceptibility of this strain to autoimmune arthritis induced by immunization with collagen type II (CII) and complete Freund's adjuvant (CFA). Here, we show that following intradermal infection with Mycobacterium avium, a similar arthritis phenotype was detected in Ccr2-null mice in the DBA/1j, but not in the BALB/c background. The failure to develop arthritis in Ccr2-null BALB/c mice occurred in the face of high bacterial burdens and low interferon gamma (IFNgamma) production. By contrast, Ccr2-null DBA/1j mice had low bacterial burdens, produced normal amounts of IFNgamma, and had high titers of autoantibodies against CII. Thus, the Ccr2-null state in an arthritic-prone genetic background leads to increased arthritis susceptibility following infectious (M. avium) and noninfectious (CII/CFA) challenges. Because CCR2 serves as a negative regulator of murine arthritis, caution might need to be exercised while testing CCR2 blockers in human arthritis or other diseases. These findings also indicate that Ccr2-null DBA/1j mice might serve as a valuable model system to uncover the immunological determinants of arthritis and to test novel antiarthritic agents.


Subject(s)
Arthritis, Experimental/metabolism , Mycobacterium avium , Receptors, Chemokine/physiology , Tuberculosis, Cutaneous/complications , Animals , Arthritis, Experimental/etiology , Arthritis, Experimental/pathology , Collagen Type II/immunology , Interferon-gamma/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Knockout , Receptors, CCR2 , Receptors, Chemokine/genetics
20.
Hum Gene Ther ; 17(9): 949-59, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16972763

ABSTRACT

Macrophages have the potential to deliver therapeutic genes to many target tissues. Macrophage-specific synthetic promoters (SPs) generated by random ligation of myeloid/macrophage cis elements had activity up to 100-fold that of a native macrophage promoter in macrophage cell lines, but were minimally active in nonmyeloid cells. Mouse bone marrow cells (BMCs) transduced ex vivo with lentivectors expressing green fluorescent protein (GFP) driven either by an SP (SP-GFP) or a cytomegalovirus (CMV) promoter (CMV-GFP) were used for syngeneic transplantation of lethally irradiated mice. Blood leukocytes showed stable GFP expression for up to 15 months after transplantation. SP-GFP expression was selective for CD11b+ macrophages, whereas CMV-GFP expression was observed in erythrocytes, as well as in both CD11b+ and CD11b- leukocytes. Furthermore, SP-GFP expression was much stronger than CMV-GFP expression in CD11b+ macrophages. apoE-/- BMCs transduced with the lentiviral vector encoding human apoE were used to transplant apoE-/- mice. Macrophage expression of apoE from 10 to 26 weeks of age significantly reduced atherosclerotic lesions in recipient apoE-/- mice. Thus, the novel SPs, especially when combined with lentivectors, are useful for macrophage-specific delivery of therapeutic genes.


Subject(s)
Genetic Therapy , Macrophages, Peritoneal/cytology , Promoter Regions, Genetic , Animals , Apolipoproteins E/biosynthesis , Apolipoproteins E/genetics , Atherosclerosis/therapy , Base Sequence , Blotting, Western , Bone Marrow/metabolism , Cell Line , DNA Primers , Female , Flow Cytometry , Genetic Vectors , Humans , Lentivirus/genetics , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL