Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Cell Rep Med ; : 101696, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39178856

ABSTRACT

Pancreatic adenocarcinoma (PDAC) is one the most intractable cancers, in part due to its highly inflammatory microenvironment and paucity of infiltrating dendritic cells (DCs). Here, we find that genetic ablation or antibody blockade of tumor necrosis factor receptor 1 (TNFR1) enhanced intratumor T cell activation and slowed PDAC growth. While anti-PD-1 checkpoint inhibition alone had little effect, it further enhanced intratumor T cell activation in combination with anti-TNFR1. The major cellular alteration in the tumor microenvironment in the absence of TNFR1 signaling was a large increase in DC number and immunostimulatory phenotype. This may reflect a direct effect on DCs, because TNF induced TNFR1-dependent apoptosis of bone-marrow-derived DCs. The therapeutic response to anti-TNFR1 alone was superior to the combination of DC-activating agonistic anti-CD40 and Flt3 ligand (Flt3L). These observations suggest that targeting TNFR1, perhaps in concert with other strategies that promote DC generation and mobilization, may have therapeutic benefits.

2.
Breast Cancer Res ; 24(1): 75, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36333737

ABSTRACT

BACKGROUND: Breast cancer is a heterogenous disease with several histological and molecular subtypes. Models that represent these subtypes are essential for translational research aimed at improving clinical strategy for targeted therapeutics. METHODS: Different combinations of genetic aberrations (Brca1 and Trp53 loss, and inhibition of proteins of the Rb family) were induced in the mammary gland by injection of adenovirus expressing Cre recombinase into the mammary ducts of adult genetically engineered mice. Mammary tumors with different genetic aberrations were classified into molecular subtypes based on expression of molecular markers and RNAseq analysis. In vitro potency assays and Western blots were used to examine their drug sensitivities. RESULTS: Induction of Brca1 and Trp53 loss in mammary ductal epithelium resulted in development of basal-like hormone receptor (HR)-negative mammary tumors. Inhibition of Rb and Trp53 loss or the combination of Rb, Trp53 and Brca1 aberrations resulted in development of luminal ductal carcinoma positive for ER, PR, and Her2 expression. HR positivity in tumors with Rb, Trp53 and Brca1 aberrations indicated that functionality of the Rb pathway rather than Brca1 status affected HR status in these models. Mammary tumor gene expression profiles recapitulated human basal-like or luminal B breast cancer signatures, but HR-positive luminal cancer models were endocrine resistant and exhibited upregulation of PI3K signaling and sensitivity to this pathway inhibition. Furthermore, both tumor subtypes were resistant to CDK4/6 inhibition. CONCLUSIONS: Examination of molecular expression profiles and drug sensitivities of tumors indicate that these breast cancer models can be utilized as a translational platform for evaluation of targeted combinations to improve chemotherapeutic response in patients that no longer respond to hormone therapy or that are resistant to CDK4/6 inhibition.


Subject(s)
Breast Neoplasms , Mammary Glands, Human , Mammary Neoplasms, Animal , Mice , Animals , Humans , Female , Mammary Glands, Human/metabolism , Phosphatidylinositol 3-Kinases , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Mammary Neoplasms, Animal/pathology , Epithelium/metabolism , Hormones , BRCA1 Protein/genetics
3.
Mol Cancer Ther ; 20(10): 1926-1940, 2021 10.
Article in English | MEDLINE | ID: mdl-34376576

ABSTRACT

The desmoplastic stroma of pancreatic cancers forms a physical barrier that impedes intratumoral drug delivery. Attempts to modulate the desmoplastic stroma to increase delivery of administered chemotherapy have not shown positive clinical results thus far, and preclinical reports in which chemotherapeutic drugs were coadministered with antistromal therapies did not universally demonstrate increased genotoxicity despite increased intratumoral drug levels. In this study, we tested whether TGFß antagonism can break the stromal barrier, enhance perfusion and tumoral drug delivery, and interrogated cellular and molecular mechanisms by which the tumor prevents synergism with coadministered gemcitabine. TGFß inhibition in genetically engineered murine models (GEMM) of pancreas cancer enhanced tumoral perfusion and increased intratumoral gemcitabine levels. However, tumors rapidly adapted to TGFß-dependent stromal modulation, and intratumoral perfusion returned to pre-treatment levels upon extended TGFß inhibition. Perfusion was governed by the phenotypic identity and distribution of cancer-associated fibroblasts (CAF) with the myelofibroblastic phenotype (myCAFs), and myCAFs which harbored unique genomic signatures rapidly escaped the restricting effects of TGFß inhibition. Despite the reformation of the stromal barrier and reversal of initially increased intratumoral exposure levels, TGFß inhibition in cooperation with gemcitabine effectively suppressed tumor growth via cooperative reprogramming of T regulatory cells and stimulation of CD8 T cell-mediated antitumor activity. The antitumor activity was further improved by the addition of anti-PD-L1 immune checkpoint blockade to offset adaptive PD-L1 upregulation induced by TGFß inhibition. These findings support the development of combined antistroma anticancer therapies capable of impacting the tumor beyond the disruption of the desmoplastic stroma as a physical barrier to improve drug delivery.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/immunology , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/immunology , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Stromal Cells/immunology , Tumor Microenvironment , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Combined Modality Therapy , Deoxycytidine/pharmacology , Humans , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Stromal Cells/drug effects , Stromal Cells/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Gemcitabine
4.
Mol Cancer Ther ; 20(10): 2082-2092, 2021 10.
Article in English | MEDLINE | ID: mdl-34315768

ABSTRACT

Antibody-based therapies designed for human use frequently fail to cross-react with the murine isoform of their target. Because of this problem, preclinical studies of antibody-based mesothelin (Msl)-targeted therapeutics in immunocompetent systems have been limited by the lack of suitable mouse models. Here, we describe two immunocompetent humanized mesothelin transgenic mouse lines that can act as tolerant hosts for C57Bl/6-syngeneic cell lines expressing the human isoform of mesothelin. Thyroid peroxidase (TPO) mice have thyroid-restricted human mesothelin expression. Mesothelin (Msl) mice express human mesothelin in the typical serosal membrane distribution and can additionally be utilized to assess on-target, off-tumor toxicity of human mesothelin-targeted therapeutics. Both transgenic strains shed human mesothelin into the serum like human mesothelioma and patients with ovarian cancer, and serum human mesothelin can be used as a blood-based surrogate of tumor burden. Using these models, we examined the on-target toxicity and antitumor activity of human mesothelin-targeted recombinant immunotoxins. We found that immunotoxin treatment causes acute and chronic histologic changes to serosal membranes in Msl mice, while human mesothelin-expressing thyroid follicular cells in TPO mice are resistant to immunotoxin despite excellent drug delivery. Furthermore, poor delivery of immunotoxin to syngeneic orthotopic human mesothelin-expressing pancreatic adenocarcinoma limits antitumor activity both alone and in combination with immune checkpoint inhibition. In summary, we have developed two high-fidelity, immunocompetent murine models for human cancer that allow for rigorous preclinical evaluation of human mesothelin-targeted therapeutics.


Subject(s)
Adenocarcinoma/therapy , Mesothelin/administration & dosage , Mesothelioma/therapy , Pancreatic Neoplasms/therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Apoptosis , Cell Proliferation , Female , Genetic Engineering , Humans , Male , Mesothelin/genetics , Mesothelin/metabolism , Mesothelioma/genetics , Mesothelioma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Sci Transl Med ; 12(530)2020 02 12.
Article in English | MEDLINE | ID: mdl-32051227

ABSTRACT

Solid tumors elicit a detectable immune response including the infiltration of tumor-associated macrophages (TAMs). Unfortunately, this immune response is co-opted into contributing toward tumor growth instead of preventing its progression. We seek to reestablish an antitumor immune response by selectively targeting surface receptors and endogenous signaling processes of the macrophage subtypes driving cancer progression. RP-182 is a synthetic 10-mer amphipathic analog of host defense peptides that selectively induces a conformational switch of the mannose receptor CD206 expressed on TAMs displaying an M2-like phenotype. RP-182-mediated activation of this receptor in human and murine M2-like macrophages elicits a program of endocytosis, phagosome-lysosome formation, and autophagy and reprograms M2-like TAMs to an antitumor M1-like phenotype. In syngeneic and autochthonous murine cancer models, RP-182 suppressed tumor growth, extended survival, and was an effective combination partner with chemo- or immune checkpoint therapy. Antitumor activity of RP-182 was also observed in CD206high patient-derived xenotransplantation models. Mechanistically, via selective reduction of immunosuppressive M2-like TAMs, RP-182 improved adaptive and innate antitumor immune responses, including increased cancer cell phagocytosis by reprogrammed TAMs.


Subject(s)
Mannose-Binding Lectins , Tumor-Associated Macrophages , Animals , Cell Line, Tumor , Humans , Immunity, Innate , Lectins, C-Type , Mannose Receptor , Mice , Receptors, Cell Surface
6.
J Pharm Biomed Anal ; 181: 113093, 2020 Mar 20.
Article in English | MEDLINE | ID: mdl-31931447

ABSTRACT

Synthetic host defense peptides (HDP) are a new class of promising therapeutic agents with potential application in a variety of diseases. RP-182 is a 10mer synthetic HDP design, which selectively reduces M2-like tumor associated macrophages via engagement with the cell surface lectin receptor MRC1/CD206 and is currently being developed as an innate immune defense regulator to improve anti-tumor immunity in immunologically cold tumors. Herein, we describe a sensitive and specific liquid chromatography (LC) coupled to quadrupole electron spray tandem mass spectrometry method to measure positively charged HDPs and HDP peptide fragments in complex biological matrices. Carboxylic acid magnetic beads were used as an affinity-capturing agent to extract the positively charged RP-182 from both mouse plasma and tissue homogenates. Beads were eluted with 0.1% (v/v) formic acid and chromatographic separation was achieved on a Waters 2.1 × 100 mm, 3.5 µm XSelect Peptide CSH C18 column with a Vanguard pre-column of the same phase. MS/MS was performed on a Thermo TSQ Quantiva triple quadrupole mass spectrometer operating in Selected Reaction Monitoring (SRM) mode fragmenting the plus three parent ion 458.9+3 and monitoring ions 624.0+2, 550.5+2, and 597.3+1 for RP-182 and 462.4+3 > 629.1+2, 555.5+2, and 607.3+1 for isotopic RP-182 standard. The assay had good linearity ranging from 1 ng to 1000 ng in mouse plasma with the lower limit of detection for RP-182 at 1 ng in mouse plasma with good intra- and inter-sample precision and accuracy. Recovery ranged from 66% to 77% with minimum matrix effects. The method was successfully applied to an abbreviated pharmacokinetic study in mice after single IP injection of RP-182. The method was successfully tested on a second HDP, the 17mer D4E1, and the cationic human peptide hormone ghrelin suggesting that it might be a general sensitive method applicable to quantifying HDP peptides that are difficult to extract.


Subject(s)
Antimicrobial Cationic Peptides/isolation & purification , Animals , Antimicrobial Cationic Peptides/blood , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacokinetics , Carboxylic Acids/chemistry , Chromatography, Affinity/methods , Chromatography, High Pressure Liquid/methods , Drug Evaluation, Preclinical/methods , Ghrelin/blood , Ghrelin/chemistry , Ghrelin/isolation & purification , Limit of Detection , Magnetic Phenomena , Mice , Reproducibility of Results , Tandem Mass Spectrometry/methods
7.
Cancer Res ; 80(8): 1630-1643, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31911550

ABSTRACT

Pancreatic cancer is a disease with limited therapeutic options. Resistance to chemotherapies poses a significant clinical challenge for patients with pancreatic cancer and contributes to a high rate of recurrence. Oncogenic KRAS, a critical driver of pancreatic cancer, promotes metabolic reprogramming and upregulates NRF2, a master regulator of the antioxidant network. Here, we show that NRF2 contributed to chemoresistance and was associated with a poor prognosis in patients with pancreatic cancer. NRF2 activation metabolically rewired and elevated pathways involved in glutamine metabolism. This curbed chemoresistance in KRAS-mutant pancreatic cancers. In addition, manipulating glutamine metabolism restrained the assembly of stress granules, an indicator of chemoresistance. Glutaminase inhibitors sensitized chemoresistant pancreatic cancer cells to gemcitabine, thereby improving the effectiveness of chemotherapy. This therapeutic approach holds promise as a novel therapy for patients with pancreatic cancer harboring KRAS mutation. SIGNIFICANCE: These findings illuminate the mechanistic features of KRAS-mediated chemoresistance and provide a rationale for exploiting metabolic reprogramming in pancreatic cancer cells to confer therapeutic opportunities that could be translated into clinical trials. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/8/1630/F1.large.jpg.


Subject(s)
Drug Resistance, Neoplasm/physiology , Glutamine/metabolism , NF-E2-Related Factor 2/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Glutaminase/antagonists & inhibitors , Heterografts , Humans , Mice , Mice, Nude , Mutation , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Prognosis , Random Allocation , Tissue Array Analysis , Up-Regulation , Gemcitabine
8.
Cancer Chemother Pharmacol ; 82(6): 1067-1080, 2018 12.
Article in English | MEDLINE | ID: mdl-30306263

ABSTRACT

PURPOSE: Metarrestin is a first-in-class small molecule clinical candidate capable of disrupting the perinucleolar compartment, a subnuclear structure unique to metastatic cancer cells. This study aims to define the pharmacokinetic (PK) profile of metarrestin and the pharmacokinetic/pharmacodynamic relationship of metarrestin-regulated markers. METHODS: PK studies included the administration of single or multiple dose of metarrestin at 3, 10, or 25 mg/kg via intravenous (IV) injection, gavage (PO) or with chow to wild-type C57BL/6 mice and KPC mice bearing autochthonous pancreatic tumors. Metarrestin concentrations were analyzed by UPLC-MS/MS. Pharmacodynamic assays included mRNA expression profiling by RNA-seq and qRT-PCR for KPC mice. RESULTS: Metarrestin had a moderate plasma clearance of 48 mL/min/kg and a large volume of distribution of 17 L/kg at 3 mg/kg IV in C57BL/6 mice. The oral bioavailability after single-dose (SD) treatment was > 80%. In KPC mice treated with SD 25 mg/kg PO, plasma AUC0-∞ of 14400 ng h/mL, Cmax of 810 ng/mL and half-life (t1/2) of 8.5 h were observed. At 24 h after SD of 25 mg/kg PO, the intratumor concentration of metarrestin was high with a mean value of 6.2 µg/g tissue (or 13 µM), well above the cell-based IC50 of 0.4 µM. At multiple dose (MD) 25 mg/kg/day PO in KPC mice, mean tissue/plasma AUC0-24h ratio for tumor, spleen and liver was 37, 30 and 31, respectively. There was a good linear relationship of dosage to AUC0-24h and C24h. AUC0-24h MD to AUC0-24h SD ratios ranged from two for liver to five for tumor indicating additional accumulation in tumors. Dose-dependent normalization of FOXA1 and FOXO6 mRNA expression was observed in KPC tumors. CONCLUSIONS: Metarrestin is an effective therapeutic candidate with a favorable PK profile achieving excellent intratumor tissue levels in a disease with known poor drug delivery.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Organelles/drug effects , Pancreatic Neoplasms/drug therapy , Pyrimidines/pharmacokinetics , Pyrroles/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Antineoplastic Agents/therapeutic use , Area Under Curve , Cell Line, Tumor , Dose-Response Relationship, Drug , Forkhead Transcription Factors/genetics , Half-Life , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Injections, Intravenous , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity , Organelles/metabolism , Organelles/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pyrimidines/administration & dosage , Pyrimidines/blood , Pyrimidines/therapeutic use , Pyrroles/administration & dosage , Pyrroles/blood , Pyrroles/therapeutic use , Tissue Distribution
9.
Cancer Res ; 72(22): 5921-33, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22969147

ABSTRACT

Patients with lung cancer with activating mutations in the EGF receptor (EGFR) kinase, who are treated long-term with tyrosine kinase inhibitors (TKI), often develop secondary mutations in EGFR associated with resistance. Mice engineered to develop lung adenocarcinomas driven by the human EGFR T790M resistance mutation are similarly resistant to the EGFR TKI erlotinib. By tumor volume endpoint analysis, these mouse tumors respond to BIBW 2992 (an irreversible EGFR/HER2 TKI) and rapamycin combination therapy. To correlate EGFR-driven changes in the lung with response to drug treatment, we conducted an integrative analysis of global transcriptome and metabolite profiling compared with quantitative imaging and histopathology at several time points during tumor progression and treatment. Responses to single-drug treatments were temporary, whereas combination therapy elicited a sustained response. During tumor development, metabolomic signatures indicated a shift to high anabolic activity and suppression of antitumor programs with 11 metabolites consistently present in both lung tissue and blood. Combination drug treatment reversed many of the molecular changes found in tumored lung. Data integration linking cancer signaling networks with metabolic activity identified key pathways such as glutamine and glutathione metabolism that signified response to single or dual treatments. Results from combination drug treatment suggest that metabolic transcriptional control through C-MYC and SREBP, as well as ELK1, NRF1, and NRF2, depends on both EGFR and mTORC1 signaling. Our findings establish the importance of kinetic therapeutic studies in preclinical assessment and provide in vivo evidence that TKI-mediated antiproliferative effects also manifest in specific metabolic regulation.


Subject(s)
Adenocarcinoma/drug therapy , Lung Neoplasms/drug therapy , Quinazolines/pharmacology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Afatinib , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Growth Processes/physiology , Disease Progression , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Gene Expression , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Protein Kinase Inhibitors/pharmacology , Quinazolines/administration & dosage , Sirolimus/administration & dosage , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Cancer Res ; 72(16): 4141-53, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22617326

ABSTRACT

The majority of human high-grade serous epithelial ovarian cancer (SEOC) is characterized by frequent mutations in p53 and alterations in the RB and FOXM1 pathways. A subset of human SEOC harbors a combination of germline and somatic mutations as well as epigenetic dysfunction for BRCA1/2. Using Cre-conditional alleles and intrabursal induction by Cre-expressing adenovirus in genetically engineered mice, we analyzed the roles of pathway perturbations in epithelial ovarian cancer initiation and progression. Inactivation of RB-mediated tumor suppression induced surface epithelial proliferation with progression to stage I carcinoma. Additional biallelic inactivation and/or missense p53 mutation in the presence or absence of Brca1/2 caused progression to stage IV disease. As in human SEOC, mice developed peritoneal carcinomatosis, ascites, and distant metastases. Unbiased gene expression and metabolomic profiling confirmed that Rb, p53, and Brca1/2-triple mutant tumors aligned with human SEOC, and not with other intraperitoneal cancers. Together, our findings provide a novel resource for evaluating disease etiology and biomarkers, therapeutic evaluation, and improved imaging strategies in epithelial ovarian cancer.


Subject(s)
BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Carcinoma, Ovarian Epithelial , Disease Models, Animal , Female , Gene Deletion , Immunohistochemistry , Mice , Mice, Transgenic , Mutation , Neoplasm Metastasis , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Tumor Suppressor Protein p53/genetics
11.
Cell Metab ; 11(3): 194-205, 2010 Mar 03.
Article in English | MEDLINE | ID: mdl-20197052

ABSTRACT

Adipose triglyceride lipase (ATGL) is the rate-limiting enzyme for triacylglycerol (TAG) hydrolysis in adipocytes. The precise mechanisms whereby ATGL is regulated remain uncertain. Here, we demonstrate that a protein encoded by G(0)/G(1) switch gene 2 (G0S2) is a selective regulator of ATGL. G0S2 is highly expressed in adipose tissue and differentiated adipocytes. When overexpressed in HeLa cells, G0S2 localizes to lipid droplets and prevents their degradation mediated by ATGL. Moreover, G0S2 specifically interacts with ATGL through the hydrophobic domain of G0S2 and the patatin-like domain of ATGL. More importantly, interaction with G0S2 inhibits ATGL TAG hydrolase activity. Knockdown of endogenous G0S2 accelerates basal and stimulated lipolysis in adipocytes, whereas overexpression of G0S2 diminishes the rate of lipolysis in both adipocytes and adipose tissue explants. Thus, G0S2 functions to attenuate ATGL action both in vitro and in vivo and by this mechanism regulates TAG hydrolysis.


Subject(s)
Adipocytes/enzymology , Adipose Tissue/enzymology , Carboxylic Ester Hydrolases/metabolism , Cell Cycle Proteins/genetics , Lipase/metabolism , Lipolysis/genetics , Adipocytes/ultrastructure , Animals , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Lipids , Mice , Protein Interaction Domains and Motifs , Vacuoles/metabolism
12.
Am J Physiol Cell Physiol ; 294(1): C295-305, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17977947

ABSTRACT

In the current study, we examined whether diabetes affected the ability of HDL to stimulate nitric oxide (NO) production. Using HDL isolated from both diabetic humans and diabetic mouse models, we found that female HDL no longer induced NO synthesis, despite containing equivalent amounts of estrogen as nondiabetic controls. Furthermore, HDL isolated from diabetic females and males prevented acetylcholine-induced stimulation of NO generation. Analyses of both the human and mouse diabetic HDL particles showed that the HDLs contained increased levels of myristic acid. To determine whether myristic acid associated with HDL particles was responsible for the decrease in NO generation, myristic acid was added to HDL isolated from nondiabetic humans and mice. Myristic acid-associated HDL inhibited the generation of NO in a dose-dependent manner. Importantly, diabetic HDL did not alter the levels of endothelial NO synthase or acetylcholine receptors associated with the cells. Surprisingly, diabetic HDL inhibited ionomycin-induced stimulation of NO production without affecting ionomycin-induced increases in intracellular calcium. Further analysis indicated that diabetic HDL prevented calmodulin from interacting with endothelial NO synthase (eNOS) but did not affect the activation of calmodulin kinase or calcium-independent mechanisms for stimulating eNOS. These studies are the first to show that a specific fatty acid associated with HDL inhibits the stimulation of NO generation. These findings have important implications regarding cardiovascular disease in diabetic patients.


Subject(s)
Acetylcholine/metabolism , Calmodulin/metabolism , Diabetes Mellitus/metabolism , Endothelial Cells/metabolism , Lipoproteins, HDL/metabolism , Myristic Acid/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Diabetes Mellitus/enzymology , Diabetes Mellitus/genetics , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Estrogens/metabolism , Female , Humans , Ionomycin/pharmacology , Ionophores/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myristic Acid/pharmacology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Receptors, Cholinergic/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Signal Transduction
13.
Am J Physiol Cell Physiol ; 291(6): C1271-8, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16822947

ABSTRACT

HIV protease inhibitors are important pharmacological agents used in the treatment of HIV-infected patients. One of the major disadvantages of HIV protease inhibitors is that they increase several cardiovascular risk factors, including the expression of CD36 in macrophages. The expression of CD36 in macrophages promotes the accumulation of cholesterol, the development of foam cells, and ultimately atherosclerosis. Recent studies have suggested that alpha-tocopherol can prevent HIV protease inhibitor-induced increases in macrophage CD36 levels. Because of the potential clinical utility of using alpha-tocopherol to limit some of the side effects of HIV protease inhibitors, we tested the ability of alpha-tocopherol to prevent ritonavir, a common HIV protease inhibitor, from inducing atherosclerosis in the LDL receptor (LDLR) null mouse model. Surprisingly, alpha-tocopherol did not prevent ritonavir-induced atherosclerosis. However, cotreatment with the nucleoside reverse transcriptase inhibitors (NRTIs), didanosine or D4T, did prevent ritonavir-induced atherosclerosis. Using macrophages isolated from LDLR null mice, we demonstrated that the NRTIs prevented the upregulation of CD36 and cholesterol accumulation in macrophages. Treatment of LDLR null mice with NRTIs promoted the ubiquitination and downregulation of protein kinase Calpha (PKC). Previous studies demonstrated that HIV protease inhibitor activation of PKC was necessary for the upregulation of CD36. Importantly, the in vivo inhibition of PKC with chelerythrine prevented ritonavir-induced upregulation of CD36, accumulation of cholesterol, and the formation of atherosclerotic lesions. These novel mechanistic studies suggest that NRTIs may provide protection from one of the negative side effects associated with HIV protease inhibitors, namely the increase in CD36 levels and subsequent cholesterol accumulation and atherogenesis.


Subject(s)
Atherosclerosis/chemically induced , Atherosclerosis/prevention & control , HIV Protease Inhibitors/adverse effects , Protein Kinase C-alpha/metabolism , Reverse Transcriptase Inhibitors/metabolism , Ritonavir/adverse effects , Ubiquitin/metabolism , Animals , Antioxidants/administration & dosage , Antioxidants/metabolism , Atherosclerosis/metabolism , CD36 Antigens/metabolism , Cholesterol/metabolism , Didanosine/metabolism , Enzyme Activation , HIV Infections/drug therapy , HIV Protease Inhibitors/metabolism , HIV Protease Inhibitors/therapeutic use , Humans , Macrophages/metabolism , Mice , Mice, Knockout , Protein Kinase C-alpha/antagonists & inhibitors , Receptors, LDL/genetics , Receptors, LDL/metabolism , Reverse Transcriptase Inhibitors/therapeutic use , Ritonavir/metabolism , Ritonavir/therapeutic use , Stavudine/metabolism , alpha-Tocopherol/administration & dosage , alpha-Tocopherol/metabolism
14.
J Clin Invest ; 111(10): 1579-87, 2003 May.
Article in English | MEDLINE | ID: mdl-12750408

ABSTRACT

Cardiovascular diseases remain the leading cause of death in the United States. Two factors associated with a decreased risk of developing cardiovascular disease are elevated HDL levels and sex - specifically, a decreased risk is found in premenopausal women. HDL and estrogen stimulate eNOS and the production of nitric oxide, which has numerous protective effects in the vascular system including vasodilation, antiadhesion, and anti-inflammatory effects. We tested the hypothesis that HDL binds to its receptor, scavenger receptor class B type I (SR-BI), and delivers estrogen to eNOS, thereby stimulating the enzyme. HDL isolated from women stimulated eNOS, whereas HDL isolated from men had minimal activity. Studies with ovariectomized and ovariectomized/estrogen replacement mouse models demonstrated that HDL-associated estradiol stimulation of eNOS is SR-BI dependent. Furthermore, female HDL, but not male HDL, promoted the relaxation of muscle strips isolated from C57BL/6 mice but not SR-BI null mice. Finally, HDL isolated from premenopausal women or postmenopausal women receiving estradiol replacement therapy stimulated eNOS, whereas HDL isolated from postmenopausal women did not stimulate eNOS. We conclude that HDL-associated estrodial is capable of the stimulating eNOS. These studies establish a new paradigm for examining the cardiovascular effects of HDL and estrogen.


Subject(s)
CD36 Antigens/metabolism , Endothelium, Vascular/drug effects , Estradiol/pharmacology , Lipoproteins, HDL/pharmacology , Membrane Proteins , Nitric Oxide Synthase/metabolism , Receptors, Immunologic , Receptors, Lipoprotein , Animals , CD36 Antigens/genetics , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Estradiol/metabolism , Female , Femoral Artery/drug effects , Femoral Artery/physiology , Hormone Replacement Therapy , Humans , In Vitro Techniques , Lipoproteins, HDL/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Ovariectomy , Receptors, Scavenger , Scavenger Receptors, Class B , Sex Factors , Vasodilation/drug effects
15.
J Clin Invest ; 111(3): 389-97, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12569165

ABSTRACT

Protease inhibitors decrease the viral load in HIV patients, however the patients develop hypertriglyceridemia, hypercholesterolemia, and atherosclerosis. It has been assumed that protease inhibitor-dependent increases in atherosclerosis are secondary to the dyslipidemia. Incubation of THP-1 cells or human PBMCs with protease inhibitors caused upregulation of CD36 and the accumulation of cholesteryl esters. The use of CD36-blocking antibodies, a CD36 morpholino, and monocytes isolated from CD36 null mice demonstrated that protease inhibitor-induced increases in cholesteryl esters were dependent on CD36 upregulation. These data led to the hypothesis that protease inhibitors induce foam cell formation and consequently atherosclerosis by upregulating CD36 and cholesteryl ester accumulation independent of dyslipidemia. Studies with LDL receptor null mice demonstrated that low doses of protease inhibitors induce an increase in the level of CD36 and cholesteryl ester in peritoneal macrophages and the development of atherosclerosis without altering plasma lipids. Furthermore, the lack of CD36 protected the animals from protease inhibitor-induced atherosclerosis. Finally, ritonavir increased PPAR-gamma and CD36 mRNA levels in a PKC- and PPAR-gamma-dependent manner. We conclude that protease inhibitors contribute to the formation of atherosclerosis by promoting the upregulation of CD36 and the subsequent accumulation of sterol in macrophages.


Subject(s)
Arteriosclerosis/chemically induced , CD36 Antigens/biosynthesis , Cholesterol Esters/metabolism , HIV Protease Inhibitors/pharmacology , Macrophages/metabolism , Animals , CD36 Antigens/metabolism , Hyperlipidemias/drug therapy , Immunoglobulin M/metabolism , Leukocytes, Mononuclear/metabolism , Male , Mice , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Time Factors , Transcription Factors/metabolism , Up-Regulation
16.
J Biol Chem ; 277(26): 23525-33, 2002 Jun 28.
Article in English | MEDLINE | ID: mdl-11976335

ABSTRACT

Numerous studies have implicated either the presence or absence of CD36 in the development of hypertension. In addition, hypercholesterolemia is associated with the loss of nitric oxide-induced vasodilation and the subsequent increase in blood pressure. In the current study, we tested the hypothesis that diet-induced hypercholesterolemia promotes the disruption of agonist-stimulated nitric oxide generation and vasodilation in a CD36-dependent manner. To test this, C57BL/6, apoE null, CD36 null, and apoE/CD36 null mice were maintained on chow or high fat diets. In contrast to apoE null mice fed a chow diet, apoE null mice fed a high fat diet did not respond to acetylcholine with a decrease in blood pressure. Caveolae isolated from in vivo vessels did not contain endothelial nitric-oxide synthase and were depleted of cholesterol. Age-matched apoE/CD36 null mice fed a chow or high fat diet responded to acetylcholine with a decrease in blood pressure. The mechanism underlying the vascular dysfunction was reversible because vessels isolated from apoE null high fat-fed mice regained responsiveness to acetylcholine when incubated with plasma obtained from chow-fed mice. Further analysis demonstrated that the plasma low density lipoprotein fraction was responsible for depleting caveolae of cholesterol, removing endothelial nitric-oxide synthase from caveolae, and preventing nitric oxide production. In addition, the pharmacological removal of caveola cholesterol with cyclodextrin mimicked the effects caused by the low density lipoprotein fraction. We conclude that the ablation of CD36 prevented the negative impact of hypercholesterolemia on agonist-stimulated nitric oxide-mediated vasodilation in apoE null mice. These studies provide a direct link between CD36 and the early events that underlie hypercholesterolemia-mediated hypertension and mechanistic linkages between CD36 function, nitric-oxide synthase activation, caveolae integrity, and blood pressure regulation.


Subject(s)
Blood Vessels/physiopathology , CD36 Antigens/physiology , Hypercholesterolemia/physiopathology , Nitric Oxide Synthase/physiology , Acetylcholine/pharmacology , Animals , Caveolae/physiology , Cyclodextrins/toxicity , Hypercholesterolemia/complications , Hypertension/etiology , Lipoproteins, LDL/toxicity , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Phosphorylation , Vasodilation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL