Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
PLoS One ; 15(2): e0228005, 2020.
Article in English | MEDLINE | ID: mdl-32027681

ABSTRACT

Targeted gene therapy using recombinant adeno-associated virus (rAAV) vectors is a potential therapeutic strategy for treating cancer, and tissue-specific promoters may help with tissue targeting. Medullary thyroid carcinoma (MTC) is a disease of the calcitonin secreting thyroid C cells, and calcitonin is highly expressed in MTC tumors compared to other cells. To target MTC cells, we evaluated an rAAV serotype 2 vector (rAAV2-pM+104-GFP) containing a modified calcitonin/calcitonin gene related peptide promoter (pM+104) and a green fluorescent protein (GFP) reporter gene. In vitro transduction experiments comparing the MTC TT cell line with non-MTC cell lines demonstrated that rAAV2-pM+104-GFP infection yielded significantly (p < 0.05) higher GFP expression in TT cells than in non-MTC cell lines (HEK293 and HeLa), and significantly higher expression than in TT cells infected with the positive control rAAV2-pCBA-GFP vector. The rAAV2-pCBA-GFP control vector included a well-characterized, ubiquitously expresses control promoter, the chicken beta actin promoter with a cytomegalovirus enhancer (pCBA). In vivo experiments using a TT cell xenograft tumor mouse model showed that tumors directly injected with 2 x 1010 vg of rAAV2-pM+104-GFP vector resulted in GFP expression detected in 21.7% of cells, 48 hours after the injection. Furthermore, GFP expression was significantly higher for rAAV-pM+104-GFP treatments with a longer vector treatment duration and higher vector dose, with up to 52.6% (q < 0.05) GFP cells detected 72 hours after injecting 1x 1011 vg/tumor. These data show that we have developed an rAAV vector with improved selectivity for MTC.


Subject(s)
Calcitonin/genetics , Carcinoma, Neuroendocrine/therapy , Dependovirus/genetics , Genetic Vectors/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Thyroid Neoplasms/therapy , Animals , Calcitonin Gene-Related Peptide/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Luciferases/metabolism , Male , Mice, SCID , Promoter Regions, Genetic , Transgenes , Xenograft Model Antitumor Assays
2.
Proc Natl Acad Sci U S A ; 114(5): 1117-1122, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096371

ABSTRACT

In recent years, it has been established that programmed cell death protein ligand 1 (PD-L1)-mediated inhibition of activated PD-1+ T lymphocytes plays a major role in tumor escape from immune system during cancer progression. Lately, the anti-PD-L1 and -PD-1 immune therapies have become an important tool for treatment of advanced human cancers, including bladder cancer. However, the underlying mechanisms of PD-L1 expression in cancer are not fully understood. We found that coculture of murine bone marrow cells with bladder tumor cells promoted strong expression of PD-L1 in bone marrow-derived myeloid cells. Tumor-induced expression of PD-L1 was limited to F4/80+ macrophages and Ly-6C+ myeloid-derived suppressor cells. These PD-L1-expressing cells were immunosuppressive and were capable of eliminating CD8 T cells in vitro. Tumor-infiltrating PD-L1+ cells isolated from tumor-bearing mice also exerted morphology of tumor-associated macrophages and expressed high levels of prostaglandin E2 (PGE2)-forming enzymes microsomal PGE2 synthase 1 (mPGES1) and COX2. Inhibition of PGE2 formation, using pharmacologic mPGES1 and COX2 inhibitors or genetic overexpression of PGE2-degrading enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH), resulted in reduced PD-L1 expression. Together, our study demonstrates that the COX2/mPGES1/PGE2 pathway involved in the regulation of PD-L1 expression in tumor-infiltrating myeloid cells and, therefore, reprogramming of PGE2 metabolism in tumor microenvironment provides an opportunity to reduce immune suppression in tumor host.


Subject(s)
B7-H1 Antigen/biosynthesis , Bone Marrow Cells/metabolism , Cyclooxygenase 2/physiology , Dinoprostone/physiology , Macrophages/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Prostaglandin-E Synthases/physiology , Animals , B7-H1 Antigen/genetics , Cell Communication , Cell Line, Tumor , Coculture Techniques , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Hydroxyprostaglandin Dehydrogenases/biosynthesis , Hydroxyprostaglandin Dehydrogenases/genetics , Mice , Mice, Inbred C3H , Mice, Inbred NOD , Mice, SCID , Prostaglandin Antagonists/pharmacology , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology
3.
Front Neurol ; 4: 186, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24312074

ABSTRACT

Among the U.S. military personnel, blast injury is among the leading causes of brain injury. During the past decade, it has become apparent that even blast injury as a form of mild traumatic brain injury (mTBI) may lead to multiple different adverse outcomes, such as neuropsychiatric symptoms and long-term cognitive disability. Blast injury is characterized by blast overpressure, blast duration, and blast impulse. While the blast injuries of a victim close to the explosion will be severe, majority of victims are usually at a distance leading to milder form described as mild blast TBI (mbTBI). A major feature of mbTBI is its complex manifestation occurring in concert at different organ levels involving systemic, cerebral, neuronal, and neuropsychiatric responses; some of which are shared with other forms of brain trauma such as acute brain injury and other neuropsychiatric disorders such as post-traumatic stress disorder. The pathophysiology of blast injury exposure involves complex cascades of chronic psychological stress, autonomic dysfunction, and neuro/systemic inflammation. These factors render blast injury as an arduous challenge in terms of diagnosis and treatment as well as identification of sensitive and specific biomarkers distinguishing mTBI from other non-TBI pathologies and from neuropsychiatric disorders with similar symptoms. This is due to the "distinct" but shared and partially identified biochemical pathways and neuro-histopathological changes that might be linked to behavioral deficits observed. Taken together, this article aims to provide an overview of the current status of the cellular and pathological mechanisms involved in blast overpressure injury and argues for the urgent need to identify potential biomarkers that can hint at the different mechanisms involved.

4.
J Liver ; 2(1)2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23724364

ABSTRACT

Liver and kidney damage associated with polytrauma, endotoxic shock/sepsis, and organ transplantation, are among the leading causes of the multiple organ failure. Development of novel sensitive biomarkers that detect early stages of liver and kidney injury is vital for the effective diagnostics and treatment of these life-threatening conditions. Previously, we identified several hepatic proteins, including Argininosuccinate Synthase (ASS) and sulfotransferases which were degraded in the liver and rapidly released into circulation during Ischemia/Reperfusion (I/R) injury. Here we compared sensitivity and specificity of the newly developed sandwich ELISA assays for ASS and the sulfotransferase isoform SULT2A1 with the standard clinical liver and kidney tests Alanine Aminotransferase (ALT) and Aspartate Transaminase (AST) in various pre-clinical models of acute injury. Our data suggest that ASS and SULT2A1 have superior characteristics for liver and kidney health assessment in endotoxemia, Ischemia/Reperfusion (I/R), chemical and drug-induced liver injury and may be of high potential value for clinical applications.

5.
J Neurotrauma ; 30(22): 1881-7, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23805797

ABSTRACT

Severe blast exposures are frequently complicated with fatal intracranial hemorrhages. However, many more sustain low level blasts without tissue damage detectable by brain imaging. To investigate effects of nonlethal blast on thrombin-related biomarkers, rats were subjected to two different types of head-directed blast: 1) moderate "composite" blast with strong head acceleration or 2) moderate primary blast, without head acceleration. Thrombin generation (TG) ex vivo after blast was studied by calibrated automated thrombography (CAT). In the same blood samples, we assessed maximal concentration of TG (TGmax), start time, peak time, mean time, and concentrations of protein markers for vascular/hemostatic dysfunctions: integrin α/ß, soluble endothelial selectin (sE-selectin), soluble intercellular cell adhesion molecule-1 (sICAM-1), and matrix metalloproteinases (MMP)-2, MMP-8, and MMP-13. Blast remarkably affected all TG indices. In animals exposed to "composite" blast, TGmax peaked at 6 h (∼4.5-fold vs. control), sustained at day 1 (∼3.8-fold increase), and declined to a 2-fold increase over control at day 7 post-blast. After primary blast, TGmax also rose to ∼4.2-fold of control at 6 h, dropped to ∼1.7-fold of control at day 1, and then exhibited a slight secondary increase at 2-fold of control at day 7. Other TG indices did not differ significantly between two types of blast exposure. The changes were also observed in other microvascular/inflammatory/hemostatic biomarkers. Integrin α/ß and sICAM-1 levels were elevated after both "composite" and primary blast at 6 h, 1 day, and 7 days. sE-selectin exhibited near normal levels after "composite" blast, but increased significantly at 7 days after primary blast; MMP-2, MMP-8, and MMP-13 slightly rose after "composite" blast and significantly increased (∼2-4-fold) after primary blast. In summary, CAT may have a clinical diagnostic utility in combination with selected set of microvascular/inflammatory biomarkers in patients subjected to low/moderate level blast exposures.


Subject(s)
Blast Injuries/blood , Brain Injuries/blood , Thrombin/metabolism , Animals , Biomarkers/blood , Biomarkers/metabolism , Blast Injuries/etiology , Brain Injuries/etiology , Calibration , Disease Models, Animal , Fluorometry/methods , Rats , Thrombin/analysis , Time Factors
6.
Neurosci Lett ; 544: 62-7, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23570732

ABSTRACT

Explosive overpressure brain injury (OBI) impacts the lives of both military and civilian population. We hypothesize that a single exposure to OBI results in increased hypothalamic expression of oxidative stress and activation of the sympatho-adrenal medullary axis. Since a key component of blast-induced organ injury is the primary overpressure wave, we assessed selective biochemical markers of autonomic function and oxidative stress in male Sprague Dawley rats subjected to head-directed overpressure insult. Rats were subjected to single head-directed OBI with a 358kPa peak overpressure at the target. Control rats were exposed to just noise signal being placed at ~2m distance from the shock tube nozzle. Sympathetic nervous system activation of the adrenal medullae (AM) was evaluated at 6h following blast injury by assessing the expression of catecholamine biosynthesizing enzymes, tyrosine hydroxylase (TH), dopamine-ß hydroxylase (DßH), neuropeptide Y (NPY) along with plasma norepinephrine (NE). TH, DßH and NPY expression increased 20%, 25%, and 91% respectively, following OBI (P<0.05). Plasma NE was also significantly elevated by 23% (P<0.05) following OBI. OBI significantly elevated TH (49%, P<0.05) in the nucleus tractus solitarius (NTS) of the brain stem while AT1 receptor expression and NADPH oxidase activity, a marker of oxidative stress, was elevated in the hypothalamus following OBI. Collectively, the increased levels of TH, DßH and NPY expression in the rat AM, elevated TH in NTS along with increased plasma NE suggest that single OBI exposure results in increased sympathoexcitation. The mechanism may involve the elevated AT1 receptor expression and NADPH oxidase levels in the hypothalamus. Taken together, such effects may be important factors contributing to pathology of brain injury and autonomic dysfunction associated with the clinical profile of patients following OBI.


Subject(s)
Adrenal Medulla/injuries , Adrenal Medulla/metabolism , Blast Injuries/metabolism , Catecholamines/biosynthesis , Hypothalamus/injuries , Hypothalamus/metabolism , Reactive Oxygen Species/metabolism , Animals , Male , Oxidative Stress , Rats , Rats, Sprague-Dawley , Up-Regulation
7.
Cell Oncol (Dordr) ; 36(3): 247-57, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23619943

ABSTRACT

BACKGROUND: The heterogeneity of liver cancer, in particular hepatocellular carcinoma (HCC), portrays the requirement of multiple targets for both its treatment and prevention. Multifaceted agents, minimally or non-toxic for normal hepatocytes, are required to address the molecular diversity of HCC, including the resistance of putative liver cancer stem cells to chemotherapy. METHODS: We designed and synthesized two fatty acid ethers of isopropylamino propanol, C16:0-AIP-1 and C18:1-AIP-2 (jointly named AIPs), and evaluated their anti-proliferative effects on the human HCC cell line Huh7 and the murine hepatoma cell line BNL 1MEA.7R.1, both in vitro and in an in vivo allograft mouse model. RESULTS: We found that AIP-1 and AIP-2 inhibited proliferation and caused cell death in both Huh7 and BNL 1MEA.7R.1 cells. Importantly, AIP-1 and AIP-2 were found to block the activation of putative liver cancer stem cells as manifested by suppression of clonal 'carcinosphere' development in growth factor-free and anchorage-free medium. The AIPs exhibited a relatively low toxicity against normal human or rat hepatocytes in primary cultures. In addition, we found that the AIPs utilized multifaceted pathways that mediate both autophagy and apoptosis in HCC, including the inhibition of AKTs and CAMK-1. In immune-competent mice, the AIPs significantly reduced BNL 1MEA.7R.1 cell-driven tumor allograft development, with a higher efficiency than sorafenib. A combination of AIP-1 + AIP-2 was most effective in reducing the tumor allograft incidence. CONCLUSIONS: AIPs represent a novel class of simple fatty acid derivatives that are effective against liver tumors via diverse pathways. They show a low toxicity towards normal hepatocytes. The addition of AIPs may represent a new avenue towards the management of chronic liver injury and, ultimately, the prevention and treatment of HCC.


Subject(s)
Amides/pharmacology , Ether/pharmacology , Fatty Acids/pharmacology , Liver Neoplasms/pathology , Neoplasm Transplantation , Amides/chemical synthesis , Amides/chemistry , Amides/therapeutic use , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell-Free System , Disease Models, Animal , Down-Regulation/drug effects , Ether/chemical synthesis , Ether/chemistry , Ether/therapeutic use , Fatty Acids/therapeutic use , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Mice , Protein Kinase Inhibitors/pharmacology , Proteolysis/drug effects , Spectrin/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology
8.
Biomarkers ; 18(3): 242-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23510167

ABSTRACT

Argininosuccinate synthase (ASS) plays an important role in regulating metabolic functions in mammals. We previously reported that hepatic ASS is released into circulation at very high concentrations in response to endotoxin and acute liver injury. We propose that ASS may serve as a novel biomarker for various inflammatory conditions. Our data showed that ASS accumulated in serum and urine of septic, obese or tumor mice in a condition-dependent fashion. Moreover, ASS significantly increased in urine within the first week after tumor cell implantation in mice which subsequently develop tumors. These results suggest that ASS is a novel biomarker increased upon diverse inflammatory conditions.


Subject(s)
Argininosuccinate Lyase/blood , Argininosuccinate Lyase/urine , Carcinoma, Hepatocellular/urine , Liver Neoplasms, Experimental/urine , Obesity/urine , Sepsis/urine , Animals , Bile/chemistry , Biomarkers/blood , Biomarkers/urine , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/pathology , Inflammation , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/blood , Liver Neoplasms, Experimental/diagnosis , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/diagnosis , Obesity/pathology , Sepsis/blood , Sepsis/diagnosis , Sepsis/pathology
9.
Front Neurol ; 3: 15, 2012.
Article in English | MEDLINE | ID: mdl-22403567

ABSTRACT

A number of experimental models of blast brain injury have been implemented in rodents and larger animals. However, the variety of blast sources and the complexity of blast wave biophysics have made data on injury mechanisms and biomarkers difficult to analyze and compare. Recently, we showed the importance of rat position toward blast generated by an external shock tube. In this study, we further characterized blast producing moderate traumatic brain injury and defined "composite" blast and primary blast exposure set-ups. Schlieren optics visualized interaction between the head and a shock wave generated by external shock tube, revealing strong head acceleration upon positioning the rat on-axis with the shock tube (composite blast), but negligible skull movement upon peak overpressure exposure off-axis (primary blast). Brain injury signatures of a primary blast hitting the frontal head were assessed and compared to damage produced by composite blast. Low to negligible levels of neurodegeneration were found following primary blast compared to composite blast by silver staining. However, persistent gliosis in hippocampus and accumulation of GFAP/CNPase in circulation was detected after both primary and composite blast. Also, markers of vascular/endothelial inflammation integrin alpha/beta, soluble intercellular adhesion molecule-1, and L-selectin along with neurotrophic factor nerve growth factor-beta were increased in serum within 6 h post-blasts and persisted for 7 days thereafter. In contrast, systemic IL-1, IL-10, fractalkine, neuroendocrine peptide Orexin A, and VEGF receptor Neuropilin-2 (NRP-2) were raised predominantly after primary blast exposure. In conclusion, biomarkers of major pathological pathways were elevated at all blast set-ups. The most significant and persistent changes in neuro-glial markers were found after composite blast, while primary blast instigated prominent systemic cytokine/chemokine, Orexin A, and Neuropilin-2 release, particularly when primary blast impacted rats with unprotected body.

10.
Int Immunopharmacol ; 11(9): 1180-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21481813

ABSTRACT

Lipopolysaccharide (LPS), a structural component of Gram-negative bacteria, is implicated in the pathogenesis of endotoxemia/sepsis and multi-organ injury, including liver damage. We have shown that argininosuccinate synthase (ASS), a hepatic enzyme of the urea cycle, accumulates in circulation within 1h after treatment with both LPS alone and hepatotoxic combination of LPS and D-Galactosamine. These findings indicate ASS as a sensitive biomarker of liver responses to bacterial endotoxin. Furthermore, we suggest that the ASS release represents a potential counteracting liver reaction to LPS, and demonstrates anti-LPS activity of recombinant ASS (rASS) in vitro and in rodent models of endotoxemia in vivo. rASS physically bound to LPS, as indicated by a gel shift assay, and suppressed Escherichia coli growth in cultures consistent with direct antimicrobial properties of ASS. rASS reduced LPS cytotoxicity, TNF-α production, and increased cell viability in cultured mouse macrophages, even when added one hour following LPS challenge. Intraperitoneal injection of rASS (5 mg/kg) after treatment with a high dose of LPS remarkably increased survival of rodents, with a concomitant decrease of sepsis markers TNF-α, C-reactive protein (CRP), and lactate dehydrogenase (LDH) levels in blood. These results suggest that the endogenous ASS constitutes a novel liver-derived component of the innate immune response to bacterial LPS, and that recombinant ASS could mitigate the lethal effects of bacterial endotoxins during sepsis.


Subject(s)
Argininosuccinate Synthase/immunology , Bacterial Infections/immunology , Immunity, Innate/immunology , Lipopolysaccharides/antagonists & inhibitors , Liver/immunology , Animals , Argininosuccinate Synthase/blood , Argininosuccinate Synthase/metabolism , Bacterial Infections/metabolism , C-Reactive Protein/antagonists & inhibitors , Cell Survival/immunology , Cells, Cultured , Endotoxemia/blood , Endotoxemia/immunology , Endotoxemia/metabolism , Endotoxins/immunology , Endotoxins/metabolism , Escherichia coli/immunology , Escherichia coli/metabolism , Galactosamine/immunology , Humans , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/blood , Lipopolysaccharides/immunology , Lipopolysaccharides/toxicity , Liver/enzymology , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
11.
J Trauma ; 69(4): 795-804, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20215974

ABSTRACT

OBJECTIVES: Existing experimental approaches for studies of blast impact in small animals are insufficient and lacking consistency. Here, we present a comprehensive model, with repeatable blast signatures of controlled duration, peak pressure, and transmitted impulse, accurately reproducing blast impact in laboratory animals. MATERIALS: Rat survival, brain pathomorphology, and levels of putative biomarkers of brain injury glial fibrillary acid protein (GFAP), neuron-specific enolase, and ubiquitin C-terminal hydrolase (UCH)-L1 were examined in brain, cerebrospinal fluid (CSF), and blood after 10 msec of 358 kPa peak overpressure blast exposure. RESULTS: The high-speed imaging demonstrated a strong head acceleration/jolting accompanied by typical intracranial hematomas and brain swelling. Microscopic injury was revealed by prominent silver staining in deep brain areas, including the nucleus subthalamicus zone, suggesting both diffused and focal neurodegeneration. GFAP and 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase), markers of astroglia and oligodendroglia, accumulated substantially in the hippocampus 24 hours after blast and persisted for 30 days postblast. However, GFAP content in the blood significantly increased 24 hours after injury, followed by a decline and subsequent accumulation in CSF in a time-dependent fashion. A similar profile is shown for UCH-L1 increase in blood, whereas increased CSF levels of UCH-L1 persisted throughout 14 days after blast and varied significantly in individual rats. Neuron-specific enolase levels in blood were significantly elevated within 24 hours and 48 hours postblast. CONCLUSIONS: The proposed model of controlled nonpenetrating blast in rats demonstrates the critical pathologic and biochemical signatures of blast brain injury that may be triggered by cerebrovascular responses, including blood-brain barrier disruption, glia responses, and neuroglial alterations.


Subject(s)
Blast Injuries/pathology , Brain Concussion/pathology , Brain Injuries/pathology , Diffuse Axonal Injury/pathology , Disease Models, Animal , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Air Pressure , Animals , Biomarkers/metabolism , Blood-Brain Barrier/physiology , Blotting, Western , Cerebral Cortex/injuries , Cerebral Cortex/pathology , Enzyme-Linked Immunosorbent Assay , Glial Fibrillary Acidic Protein/metabolism , Gliosis/pathology , Hippocampus/injuries , Hippocampus/pathology , Phosphopyruvate Hydratase/metabolism , Rats , S100 Proteins/metabolism , Ubiquitin Thiolesterase/metabolism
12.
Pediatr Blood Cancer ; 47(6): 757-64, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16206189

ABSTRACT

BACKGROUND: The t(17;19)(q21;p13), which occurs in a small subset of acute lymphoblastic leukemias (ALLs) and is associated with a dismal prognosis, creates a chimeric E2A-HLF transcription factor with transforming properties. PROCEDURE: We used representational difference analysis to identify candidate E2A-HLF target genes. Transient transfection assays and an inducible expression model system were then used to evaluate the ability of E2A-HLF to modulate target gene expression. RESULTS: We identified ABCB1 (MDR1, P-glycoprotein) as a gene differentially expressed in ALL cell lines with and without E2A-HLF expression and demonstrated that t(17;19)+ ALL cell lines expressed high levels of ABCB1 protein and had a drug efflux-positive phenotype. Although ABCB1 transcription is regulated by C/EBPbeta via interaction with a DNA response element that shares significant homology with the optimal E2A-HLF binding site, E2A-HLF did not directly activate transcription of reporter genes under control of ABCB1 promoter elements in transient transfection assays. However, ABCB1 expression was induced in a DNA-binding independent manner by E2A-HLF, E2A-PBX1, and truncated E2A polypeptides consisting of those portions of E2A present in leukemic fusion proteins. CONCLUSIONS: E2A-HLF-mediated over-expression of ABCB1 may play a critical role in defining the clinical phenotype of ALLs with a t(17;19), suggesting pharmacologic modulation of ABCB1 activity as a rational therapeutic strategy for this chemotherapy resistant subtype of ALL.


Subject(s)
Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 19/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Oncogene Proteins, Fusion/genetics , Organic Anion Transporters/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription Factors/genetics , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Binding Sites , Biological Transport, Active/drug effects , Biological Transport, Active/genetics , Cell Line, Tumor , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Humans , Oncogene Proteins, Fusion/biosynthesis , Organic Anion Transporters/drug effects , Phenotype , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Prognosis , RNA, Messenger/drug effects , RNA, Messenger/genetics , Rhodamines/pharmacokinetics , Transcription Factors/biosynthesis , Transcription, Genetic , Translocation, Genetic
13.
Proc Natl Acad Sci U S A ; 100(24): 14217-22, 2003 Nov 25.
Article in English | MEDLINE | ID: mdl-14617771

ABSTRACT

Adiponectin (Acrp30) is a physiologically active polypeptide hormone secreted by adipose tissue that shows insulin-sensitizing, antiinflammatory, and antiatherogenic properties. In humans, Acrp30 levels are inversely related to the degree of adiposity. In the current study, we tested the long-term weight-reducing and insulin-enhancing effects of Acrp30 cDNA delivered peripherally by a viral vector. To this end, we have generated a series of recombinant adeno-associated virus vectors of serotypes 1 and 5 encoding mouse Acrp30 cDNAs. The long-term expression of recombinant adeno-associated virus-Acrp30 vectors was tested after intramuscular or intraportal injection in female Sprague-Dawley rats with diet-induced obesity. We show that a single peripheral injection of 10(12) physical particles of Acrp30-encoding vectors resulted in sustained (up to 280 days) significant reduction in body weight, concomitant with the reduction in daily food intake. Acrp30 treatment resulted in higher peripheral insulin sensitivity measured by the i.p. glucose tolerance test in fasted animals. Ectopic expression of the Acrp30 transgene resulted in modulation of hepatic gluconeogenesis and lipogenesis, as demonstrated by the reduction of the expression of two key genes: PEPCK (phosphoenolpyruvate carboxykinase) and SREBP-1c (sterol regulatory element-binding protein 1c) in the liver. These data show successful peripheral therapy in a clinically relevant model for human obesity and insulin resistance.


Subject(s)
Intercellular Signaling Peptides and Proteins , Obesity/etiology , Proteins/genetics , Transcription Factors , Adiponectin , Animals , Animals, Genetically Modified , Base Sequence , Body Weight , CCAAT-Enhancer-Binding Proteins/genetics , DNA, Complementary/genetics , DNA-Binding Proteins/genetics , Dependovirus/genetics , Diet/adverse effects , Eating , Female , Gene Expression , Genetic Therapy , Genetic Vectors , Gluconeogenesis , Insulin Resistance , Lipids/biosynthesis , Liver/metabolism , Obesity/physiopathology , Obesity/therapy , Protein Serine-Threonine Kinases/genetics , Proteins/physiology , Rats , Rats, Sprague-Dawley , Sterol Regulatory Element Binding Protein 1
14.
Am J Physiol Regul Integr Comp Physiol ; 285(5): R1011-20, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14557234

ABSTRACT

The objective of this study was to determine if central overexpression of leptin could overcome the leptin resistance caused by 100 days of high-fat feeding. Three-month old-F344XBN male rats were fed either control low fat chow (Chow), which provides 15% of energy as fat, or a high-fat/high-sucrose diet (HF), which provides 59% of energy as fat. Over several weeks, the HF-fed animals spontaneously split into two groups of animals: those that became obese on the HF diet (DIO) and those that did not gain extra weight on the HF diet [diet resistant (DR)]. After 100 days of HF feeding, animals were given a single intracerebroventricular injection containing 5.75E10 particles of rAAV encoding leptin (rAAV-leptin) or control virus (rAAV-con). Chow animals responded robustly to rAAV-leptin, including significant anorexia, weight loss, and lipopenia. In contrast, DIO were completely unresponsive to rAAV-leptin. DR rats responded to rAAV-leptin, but in a more variable fashion than Chow. Unlike what was observed in Chow, the anorectic response to rAAV-leptin rapidly attenuated and was no longer significant by day 14 postvector delivery. Both DIO and DR animals were found to have reduced long-form leptin receptor expression and enhanced basal P-STAT-3 in the hypothalamus with respect to Chow. rAAV-leptin caused an increase in STAT3 phosphorylation and proopiomelanocortin expression in the hypothalamus and an increase in uncoupling protein-1 in brown adipose tissue in both Chow and DR animals, but failed to do so in DIO. This suggests that central overexpression of leptin is not a viable strategy to reverse diet-induced obesity.


Subject(s)
Genetic Therapy , Leptin/genetics , Obesity/metabolism , Obesity/therapy , Adipose Tissue, Brown/metabolism , Animals , Body Weight , Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Dietary Fats/pharmacology , Eating , Fatty Acids, Nonesterified/blood , Gene Expression , Ion Channels , Leptin/blood , Male , Membrane Proteins/metabolism , Mitochondrial Proteins , Neuropeptide Y/genetics , Obesity/etiology , Oxygen Consumption , Pro-Opiomelanocortin/genetics , Rats , Rats, Inbred BN , Rats, Inbred F344 , Receptors, Cell Surface/genetics , Receptors, Leptin , STAT3 Transcription Factor , Trans-Activators/metabolism , Transgenes/physiology , Treatment Failure , Uncoupling Protein 1
15.
Endocrinology ; 143(8): 3026-35, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12130569

ABSTRACT

The purpose of this study was to determine whether leptin induces leptin resistance by examining the temporal attenuation of the anorexic and energy expenditure responses to leptin. We administered recombinant adeno-associated virus encoding rat leptin cDNA or control viral vector into mildly obese rats for 138 d and compared these results with those from pair-fed rats. We measured food consumption, body weight, oxygen consumption, leptin signal transduction, and brown adipose tissue uncoupling protein 1. The anorexic response attenuated by d 25, whereas the increase in energy expenditure persisted for 83 d before attenuating. Despite attenuation of physiological responses, phosphorylated signal transducer and activator of transcription-3 remained elevated for the duration of the study. The temporal differential attenuation of the anorexic and thermogenic responses allowed us to determine the relative contributions of each response to weight maintenance. The anorexic response predominantly mediated the initial loss of body weight, but only the energy expenditure response was necessary to maintain the reduced weight. This study provides evidence that leptin induces leptin resistance. The leptin resistance was associated with persistent elevation in hypothalamic phosphorylated signal transducer and activator of transcription-3 and was characterized by a rapid attenuation of the anorexic response and slower onset for the attenuation of the energy expenditure response. We propose that both elevated leptin and obesity may be necessary for the development of leptin resistance.


Subject(s)
Body Weight/drug effects , Eating/drug effects , Energy Metabolism/drug effects , Leptin/pharmacology , Adipose Tissue, Brown/metabolism , Animals , Carrier Proteins/genetics , Dependovirus/genetics , Hypothalamus/metabolism , Ion Channels , Leptin/genetics , Male , Membrane Proteins/genetics , Mitochondrial Proteins , RNA, Messenger/analysis , Rats , Rats, Inbred BN , Signal Transduction , Uncoupling Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...