Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
J Cell Physiol ; 235(6): 5305-5317, 2020 06.
Article in English | MEDLINE | ID: mdl-31875985

ABSTRACT

Although degenerative disc disease (DDD) and related low back pain (LBP) are growing public health problems, the underlying disease mechanisms remain unclear. An increase in the vascular endothelial growth factor (VEGF) levels in DDD has been reported. This study aimed to examine the role of VEGF receptors (VEGFRs) in DDD, using a mouse model of DDD. Progressive DDD was induced by anterior stabbing of lumbar intervertebral discs in wild type (WT) and VEGFR-1 tyrosine-kinase deficient mice (vegfr-1TK-/- ). Pain assessments were performed weekly for 12 weeks. Histological and immunohistochemical assessments were made for discs, dorsal root ganglions, and spinal cord. Both vegfr-1TK-/- and WT mice presented with similar pathological changes in discs with an increased expression of inflammatory cytokines and matrix-degrading enzymes. Despite the similar pathological patterns, vegfr-1TK-/- mice showed insensitivity to pain compared with WT mice. This insensitivity to discogenic pain was related to lower levels of pain factors in the discs and peripheral sensory neurons and lower spinal glial activation in the vegfr-1TK- /- mice than in the WT mice. Exogenous stimulation of bovine disc cells with VEGF increased inflammatory and cartilage degrading enzyme. Silencing vegfr-1 by small-interfering-RNA decreased VEGF-induced expression of pain markers, while silencing vegfr-2 decreased VEGF-induced expression of inflammatory and metabolic markers without changing pain markers. This suggests the involvement of VEGFR-1 signaling specifically in pain transmission. Collectively, our results indicate that the VEGF signaling is involved in DDD. Particularly, VEGFR-1 is critical for discogenic LBP transmission independent of the degree of disc pathology.


Subject(s)
Intervertebral Disc/metabolism , Low Back Pain/genetics , Lumbar Vertebrae/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Animals , Disease Models, Animal , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Gene Expression Regulation/genetics , Humans , Intervertebral Disc/injuries , Intervertebral Disc/pathology , Low Back Pain/pathology , Lumbar Vertebrae/injuries , Lumbar Vertebrae/pathology , Mice , Pain Measurement , Signal Transduction/genetics
2.
Liver Int ; 39(12): 2214-2229, 2019 12.
Article in English | MEDLINE | ID: mdl-31436873

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common causes of cancer-related deaths globally due, in part, to the majority of patients being diagnosed with intermediate or advanced stage disease. Our increased understanding of the heterogeneous molecular pathogenesis of HCC has led to significant developments in novel targeted therapies. Despite these advances, there remains a high unmet need for new treatment options. HCC is a complex disease with multiple pathogenic mechanisms caused by a variety of risk factors, making it difficult to characterize with a single biomarker. In fact, numerous biomarkers have been studied in HCC, but alpha-fetoprotein (AFP) remains the most widely used and accepted serum marker since its discovery over 60 years ago. This review summarizes the most relevant studies associated with the regulation of AFP at the gene and protein levels; the pathophysiology of AFP as a pro-proliferative protein; and the correlation of AFP with molecular HCC subclasses, the vascular endothelial growth factor pathway and angiogenesis. Also described are the historical and current uses of AFP for screening and surveillance, diagnosis, its utility as a prognostic and predictive biomarker and its role as a tumour antigen in HCC. Taken together, these data demonstrate the relevance of AFP for patients with HCC and identify several remaining questions that will benefit from future research.


Subject(s)
Carcinoma, Hepatocellular/blood , Liver Neoplasms/blood , alpha-Fetoproteins/metabolism , Antigens, Neoplasm/blood , Biomarkers/blood , Humans
3.
Mol Carcinog ; 50(9): 655-67, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21268135

ABSTRACT

Methyl 2-cyano-3,11-dioxo-18ß-olean-1,12-dien-30-oate (CDODA-Me) and the corresponding 2-trifluoromethyl analog (CF(3)DODA-Me) are derived synthetically from the triterpenoid glycyrrhetinic acid, a major component of licorice. CDODA-Me and CF(3)DODA-Me inhibited growth of highly invasive ARO, DRO, K-18, and HTh-74 thyroid cancer cells and this was due, in part, to decreased expression of specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4 that are overexpressed in these cells. CDODA-Me and CF(3)DODA-Me also decreased expression of Sp-dependent genes, such as survivin and vascular endothelial growth factor (VEGF), and induced apoptosis. In addition, pituitary tumor-transforming gene-1 (PTTG-1) protein and mRNA levels were also decreased in thyroid cancer cells treated with CDODA-Me or CF(3)DODA-Me and this was accompanied by decreased expression of PTTG-1-dependent c-Myc and fibroblast growth factor-2 (FGF-2) genes. RNA interference studies against Sp1, Sp3, and Sp4 proteins showed that in thyroid cancer cells, PTTG-1 was an Sp-dependent gene. This study demonstrates for the first time that drugs, such as CDODA-Me and CF(3)DODA-Me, that decrease Sp protein expression also downregulate PTTG-1 in thyroid cancer cells and therefore have potential for clinical treatment of thyroid cancer and other endocrine neoplasias where PTTG-1 is a major pro-oncogenic factor.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Glycyrrhetinic Acid/analogs & derivatives , Neoplasm Proteins/genetics , Thyroid Neoplasms/metabolism , Base Sequence , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Primers , Fluorine/chemistry , Glycyrrhetinic Acid/chemistry , Glycyrrhetinic Acid/pharmacology , Humans , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Securin , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
4.
BMC Cancer ; 11: 371, 2011 Aug 24.
Article in English | MEDLINE | ID: mdl-21864401

ABSTRACT

BACKGROUND: Betulinic acid (BA) inhibits growth of several cancer cell lines and tumors and the effects of BA have been attributed to its mitochondriotoxicity and inhibition of multiple pro-oncogenic factors. Previous studies show that BA induces proteasome-dependent degradation of specificity protein (Sp) transcription factors Sp1, Sp3 and Sp4 in prostate cancer cells and this study focused on the mechanism of action of BA in colon cancer cells. METHODS: The effects of BA on colon cancer cell proliferation and apoptosis and tumor growth in vivo were determined using standardized assays. The effects of BA on Sp proteins and Sp-regulated gene products were analyzed by western blots, and real time PCR was used to determine microRNA-27a (miR-27a) and ZBTB10 mRNA expression. RESULTS: BA inhibited growth and induced apoptosis in RKO and SW480 colon cancer cells and inhibited tumor growth in athymic nude mice bearing RKO cells as xenograft. BA also decreased expression of Sp1, Sp3 and Sp4 transcription factors which are overexpressed in colon cancer cells and decreased levels of several Sp-regulated genes including survivin, vascular endothelial growth factor, p65 sub-unit of NFκB, epidermal growth factor receptor, cyclin D1, and pituitary tumor transforming gene-1. The mechanism of action of BA was dependent on cell context, since BA induced proteasome-dependent and proteasome-independent downregulation of Sp1, Sp3 and Sp4 in SW480 and RKO cells, respectively. In RKO cells, the mechanism of BA-induced repression of Sp1, Sp3 and Sp4 was due to induction of reactive oxygen species (ROS), ROS-mediated repression of microRNA-27a, and induction of the Sp repressor gene ZBTB10. CONCLUSIONS: These results suggest that the anticancer activity of BA in colon cancer cells is due, in part, to downregulation of Sp1, Sp3 and Sp4 transcription factors; however, the mechanism of this response is cell context-dependent.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Colonic Neoplasms/drug therapy , Sp Transcription Factors/metabolism , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Pentacyclic Triterpenes , Proteasome Endopeptidase Complex/metabolism , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sp Transcription Factors/genetics , Xenograft Model Antitumor Assays , Betulinic Acid
5.
Cancer Discov ; 11(2): 424-445, 2021 02.
Article in English | MEDLINE | ID: mdl-33106316

ABSTRACT

Recent clinical and preclinical advances have highlighted the existence of a previously hypothesized lymphogenous route of metastasis. However, due to a lack of suitable preclinical modeling tools, its contribution to long-term disease outcome and relevance for therapy remain controversial. Here, we established a genetically engineered mouse model (GEMM) fragment-based tumor model uniquely sustaining a functional network of intratumoral lymphatics that facilitates seeding of fatal peripheral metastases. Multiregimen survival studies and correlative patient data identified primary tumor-derived Angiopoietin-2 (Ang2) as a potent therapeutic target to restrict lymphogenous tumor cell dissemination. Mechanistically, tumor-associated lymphatic endothelial cells (EC), in contrast to blood vascular EC, were found to be critically addicted to the Angiopoietin-Tie pathway. Genetic manipulation experiments in combination with single-cell mapping revealed agonistically acting Ang2-Tie2 signaling as key regulator of lymphatic maintenance. Correspondingly, acute presurgical Ang2 neutralization was sufficient to prolong survival by regressing established intratumoral lymphatics, hence identifying a therapeutic regimen that warrants further clinical evaluation. SIGNIFICANCE: Exploiting multiple mouse tumor models including a unique GEMM-derived allograft system in combination with preclinical therapy designs closely matching the human situation, this study provides fundamental insight into the biology of tumor-associated lymphatic EC and defines an innovative presurgical therapeutic window of migrastatic Ang2 neutralization to restrict lymphogenous metastasis.This article is highlighted in the In This Issue feature, p. 211.


Subject(s)
Angiopoietin-2/metabolism , Lung Neoplasms/pathology , Lymphatic Metastasis/pathology , Receptor, TIE-2/metabolism , Animals , Disease Models, Animal , Endothelial Cells/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Signal Transduction
6.
Dev Cell ; 56(11): 1677-1693.e10, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34038707

ABSTRACT

Single-cell transcriptomics (scRNA-seq) has revolutionized the understanding of the spatial architecture of tissue structure and function. Advancing the "transcript-centric" view of scRNA-seq analyses is presently restricted by the limited resolution of proteomics and genome-wide techniques to analyze post-translational modifications. Here, by combining spatial cell sorting with transcriptomics and quantitative proteomics/phosphoproteomics, we established the spatially resolved proteome landscape of the liver endothelium, yielding deep mechanistic insight into zonated vascular signaling mechanisms. Phosphorylation of receptor tyrosine kinases was detected preferentially in the central vein area, resulting in an atypical enrichment of tyrosine phosphorylation. Prototypic biological validation identified Tie receptor signaling as a selective and specific regulator of vascular Wnt activity orchestrating angiocrine signaling, thereby controlling hepatocyte function during liver regeneration. Taken together, the study has yielded fundamental insight into the spatial organization of liver endothelial cell signaling. Spatial sorting may be employed as a universally adaptable strategy for multiomic analyses of scRNA-seq-defined cellular (sub)-populations.


Subject(s)
Liver Regeneration/genetics , Liver/growth & development , Phosphoproteins/genetics , Transcriptome/genetics , Endothelial Cells/metabolism , Endothelium/growth & development , Flow Cytometry , Gene Expression Regulation, Developmental/genetics , Hepatocytes/metabolism , Humans , Liver/metabolism , Liver/pathology , Phosphorylation/genetics , Proteomics/methods , RNA-Seq , Regeneration/genetics , Single-Cell Analysis , Wnt Signaling Pathway/genetics
7.
Mol Pharmacol ; 77(3): 396-404, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20023005

ABSTRACT

Nerve growth factor-induced B (NGFI-B) genes are orphan nuclear receptors, and NGFI-B alpha (Nur77, TR3) is overexpressed in bladder tumors and bladder cancer cells compared with nontumorous bladder tissue. 1,1-Bis(3'-indolyl)-1-(p-methoxyphenyl)-methane (DIM-C-pPhOCH(3)) and 1,1-bis(3'-indolyl)-1-(p-phenyl)methane have previously been identified as activators of Nur77, and both compounds inhibited growth and induced apoptosis of UC-5 and KU7 bladder cancer cells. The proapoptotic effects of methylene-substituted diindolylmethanes (C-DIMs) were unaffected by cotreatment with leptomycin B and were dependent on nuclear Nur77, and RNA interference with a small inhibitory RNA for Nur77 (iNur77) demonstrated that C-DIM-induced activation of apoptosis was Nur77-dependent. Microarray analysis of DIM-C-pPhOCH(3)-induced genes in UC-5 bladder cancer cells showed that this compound induced multiple Nur77-dependent proapoptotic or growth inhibitory genes including tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), cystathionase, p21, p8, and sestrin-2. DIM-C-pPhOCH(3) (25 mg/kg/d) also induced apoptosis and inhibited tumor growth in athymic nude mice bearing KU7 cells as xenografts, demonstrating that Nur77-active C-DIMs exhibit potential for bladder cancer chemotherapy by targeting Nur77, which is overexpressed in this tumor type.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/physiology , Growth Inhibitors/pharmacology , Indoles/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Urinary Bladder Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Growth Inhibitors/therapeutic use , Humans , Indoles/therapeutic use , Male , Mice , Mice, Nude , Random Allocation , Urinary Bladder Neoplasms/drug therapy
8.
MAbs ; 12(1): 1831880, 2020.
Article in English | MEDLINE | ID: mdl-33183151

ABSTRACT

CXCR1 and CXCR2 signaling play a critical role in neutrophil migration, angiogenesis, and tumorigenesis and are therefore an attractive signaling axis to target in a variety of indications. In human, a total of seven chemokines signal through these receptors and comprise the ELR+CXC chemokine family, so named because of the conserved ELRCXC N-terminal motif. To fully antagonize CXCR1 and CXCR2 signaling, an effective therapeutic should block either both receptors or all seven ligands, yet neither approach has been fully realized clinically. In this work, we describe the generation and characterization of LY3041658, a humanized monoclonal antibody that binds and neutralizes all seven human and cynomolgus monkey ELR+CXC chemokines and three of five mouse and rat ELR+CXC chemokines with high affinity. LY3041658 is able to block ELR+CXC chemokine-induced Ca2+ mobilization, CXCR2 internalization, and chemotaxis in vitro as well as neutrophil mobilization in vivo without affecting other neutrophil functions. In addition to the in vitro and in vivo activity, we characterized the epitope and structural basis for binding in detail through alanine scanning, crystallography, and mutagenesis. Together, these data provide a robust preclinical characterization of LY3041658 for which the efficacy and safety is being evaluated in human clinical trials for neutrophilic skin diseases.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Receptors, Interleukin-8A/antagonists & inhibitors , Receptors, Interleukin-8B/antagonists & inhibitors , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Antibody Affinity , Chemotaxis, Leukocyte/immunology , Humans , Macaca fascicularis , Mice , Neutrophils/immunology , Rats
9.
EMBO Mol Med ; 12(6): e11164, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32302470

ABSTRACT

The angiopoietin (Ang)-Tie pathway has been intensely pursued as candidate second-generation anti-angiogenic target. While much of the translational work has focused on the ligand Ang2, the clinical efficacy of Ang2-targeting drugs is limited and failed to improve patient survival. In turn, the orphan receptor Tie1 remains therapeutically unexplored, although its endothelial-specific genetic deletion has previously been shown to result in a strong reduction in metastatic growth. Here, we report a novel Tie1 function-blocking antibody (AB-Tie1-39), which suppressed postnatal retinal angiogenesis. During primary tumor growth, neoadjuvant administration of AB-Tie1-39 strongly impeded systemic metastasis. Furthermore, the administration of AB-Tie1-39 in a perioperative therapeutic window led to a significant survival advantage as compared to control-IgG-treated mice. Additional in vivo experimental metastasis and in vitro transmigration assays concurrently revealed that AB-Tie1-39 treatment suppressed tumor cell extravasation at secondary sites. Taken together, the data phenocopy previous genetic work in endothelial Tie1 KO mice and thereby validate AB-Tie1-39 as a Tie1 function-blocking antibody. The study establishes Tie1 as a therapeutic target for metastasis in a perioperative or neoadjuvant setting.


Subject(s)
Neoplasms , Receptor, TIE-1 , Angiopoietin-1 , Angiopoietin-2 , Animals , Gene Deletion , Humans , Mice , Neovascularization, Pathologic , Receptor, TIE-1/genetics , Receptor, TIE-2
10.
J Clin Invest ; 130(4): 1977-1990, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32149735

ABSTRACT

Angiopoietin-2 (Ang2), a ligand of the endothelial Tie2 tyrosine kinase, is involved in vascular inflammation and leakage in critically ill patients. However, the role of Ang2 in demyelinating central nervous system (CNS) autoimmune diseases is unknown. Here, we report that Ang2 is critically involved in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), a rodent model of multiple sclerosis. Ang2 expression was induced in CNS autoimmunity, and transgenic mice overexpressing Ang2 specifically in endothelial cells (ECs) developed a significantly more severe EAE. In contrast, treatment with Ang2-blocking Abs ameliorated neuroinflammation and decreased spinal cord demyelination and leukocyte infiltration into the CNS. Similarly, Ang2-binding and Tie2-activating Ab attenuated the development of CNS autoimmune disease. Ang2 blockade inhibited expression of EC adhesion molecules, improved blood-brain barrier integrity, and decreased expression of genes involved in antigen presentation and proinflammatory responses of microglia and macrophages, which was accompanied by inhibition of α5ß1 integrin activation in microglia. Taken together, our data suggest that Ang2 provides a target for increasing Tie2 activation in ECs and inhibiting proinflammatory polarization of CNS myeloid cells via α5ß1 integrin in neuroinflammation. Thus, Ang2 targeting may serve as a therapeutic option for the treatment of CNS autoimmune disease.


Subject(s)
Angiopoietin-2/immunology , Blood-Brain Barrier/immunology , Cell Movement/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Endothelial Cells/immunology , Leukocytes/immunology , Multiple Sclerosis/immunology , Angiopoietin-2/genetics , Animals , Blood-Brain Barrier/pathology , Cell Movement/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelial Cells/pathology , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Integrin alpha5beta1/genetics , Integrin alpha5beta1/immunology , Leukocytes/pathology , Mice , Mice, Transgenic , Microglia/immunology , Microglia/pathology , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology
11.
Carcinogenesis ; 30(7): 1193-201, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19406933

ABSTRACT

The non-steroidal anti-inflammatory drug tolfenamic acid (TA) inhibits proliferation of SEG-1 and BIC-1 esophageal cancer cells with half-maximal growth inhibitory concentration values of 36 and 48 muM, respectively. TA also increased Annexin V staining in both cell lines, indicative of proapoptotic activity. Treatment of SEG-1 and BIC-1 cells with TA for up to 72 h decreased expression of specificity protein (Sp) transcription factors Sp1, Sp3 and Sp4 and this was accompanied by decreased expression of the well-characterized Sp-regulated genes cyclin D1, vascular endothelial growth factor and survivin. TA also decreased hepatocyte growth factor receptor, (c-Met), a receptor tyrosine kinase that is overexpressed in esophageal cancer cells and tumors and is an important drug target. Knockdown of Sp1, Sp3 and Sp4 by RNA interference in SEG-1 and BIC-1 cells also decreased c-Met expression, demonstrating that c-Met is an Sp-regulated gene in esophageal cancer cells. Sp1 was overexpressed in esophageal cancer cells and tumors and increased Sp1 staining was observed in esophageal tumors from patients. TA (20 mg/kg/day) also decreased tumor growth and weight in athymic nude mice bearing SEG-1 cells as xenografts and this was accompanied by increased apoptosis and decreased Sp1 and c-Met staining in tumors from treated mice. Thus, TA-dependent downregulation of Sp transcription factors and c-Met defines a novel chemotherapeutic approach for treatment of esophageal cancer.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Esophageal Neoplasms/metabolism , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Sp Transcription Factors/antagonists & inhibitors , ortho-Aminobenzoates/pharmacology , Animals , Cell Line, Tumor , Down-Regulation , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Proto-Oncogene Proteins c-met/metabolism , Sp Transcription Factors/metabolism , Transplantation, Heterologous
12.
Int J Cancer ; 125(8): 1965-74, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19582879

ABSTRACT

Methyl 2-cyano-3,11-dioxo-18beta-olean-1,12-dien-30-oate (CDODA-Me) is a synthetic derivative of glycyrrhetinic acid, a triterpenoid phytochemical found in licorice extracts. CDODA-Me inhibited growth of RKO and SW480 colon cancer cells and this was accompanied by decreased expression of Sp1, Sp3 and Sp4 protein and mRNA and several Sp-dependent genes including survivin, vascular endothelial growth factor (VEGF), and VEGF receptor 1 (VEGFR1 or Flt-1). CDODA-Me also induced apoptosis, arrested RKO and SW480 cells at G(2)/M, and inhibited tumor growth in athymic nude mice bearing RKO cells as xenografts. CDODA-Me decreased expression of microRNA-27a (miR-27a), and this was accompanied by increased expression of 2 miR-27a-regulated mRNAs, namely ZBTB10 (an Sp repressor) and Myt-1 which catalyzes phosphorylation of cdc2 to inhibit progression of cells through G(2)/M. Both CDODA-Me and antisense miR-27a induced comparable responses in RKO and SW480 cells, suggesting that the potent anticarcinogenic activity of CDODA-Me is due to repression of oncogenic miR-27a.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Glycyrrhetinic Acid/analogs & derivatives , MicroRNAs/metabolism , Oncogenes , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Blotting, Northern , Blotting, Western , Cell Cycle , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Glycyrrhetinic Acid/chemical synthesis , Glycyrrhetinic Acid/chemistry , Glycyrrhetinic Acid/pharmacology , Humans , Mice , Mice, Nude , MicroRNAs/genetics , PPAR gamma/agonists , PPAR gamma/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Sp Transcription Factors/antagonists & inhibitors , Sp Transcription Factors/genetics , Sp Transcription Factors/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Mol Carcinog ; 48(8): 692-702, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19125423

ABSTRACT

Methyl 2-cyano-3,11-dioxo-18beta-olean-1,12-dien-30-oate (CDODA-Me) is a synthetic triterpenoid derived from glycyrrhetinic acid, a bioactive phytochemical in licorice, CDODA-Me inhibits growth of Panc1 and Panc28 pancreatic cancer cell lines and activates peroxisome proliferator-activated receptor gamma (PPARgamma)-dependent transactivation in these cells. CDODA-Me also induced p21 and p27 protein expression and downregulates cyclin D1; however, these responses were receptor-independent. CDODA-Me induced apoptosis in Panc1 and Panc28 cells, and this was accompanied by receptor-independent induction of the proapoptotic proteins early growth response-1 (Egr-1), nonsteroidal anti-inflammatory drug-activated gene-1 (NAG-1), and activating transcription factor-3 (ATF3). Induction of NAG-1 and Egr-1 by CDODA-Me was dependent on activation of phosphatidylinositol-3-kinase (PI3-K) and/or p42 and p38 mitogen-activated protein kinase (MAPK) pathways but there were differences between Panc28 and Panc1 cells. Induction of NAG-1 in Panc28 cells was p38-MAPK- and PI3-K-dependent but Egr-1-independent, whereas induction in Panc1 cells was associated with activation of p38-MAPK, PI3-K, and p42-MAPK and was only partially Egr-1-dependent. This is the first report of the induction of the proapoptotic protein NAG-1 in pancreatic cancer cells.


Subject(s)
Apoptosis/drug effects , Glycyrrhetinic Acid/pharmacology , Growth Differentiation Factor 15/metabolism , Pancreatic Neoplasms/pathology , Activating Transcription Factor 3/metabolism , Adipose Tissue/cytology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Blotting, Western , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Early Growth Response Protein 1/metabolism , Glycyrrhetinic Acid/analogs & derivatives , Humans , Luciferases/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , PPAR gamma/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , Signal Transduction , Transfection , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Cancer Res ; 67(6): 2816-23, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17363604

ABSTRACT

Betulinic acid is a pentacyclic triterpene natural product initially identified as a melanoma-specific cytotoxic agent that exhibits low toxicity in animal models. Subsequent studies show that betulinic acid induces apoptosis and antiangiogenic responses in tumors derived from multiple tissues; however, the underlying mechanism of action is unknown. Using LNCaP prostate cancer cells as a model, we now show that betulinic acid decreases expression of vascular endothelial growth (VEGF) and the antiapoptotic protein survivin. The mechanism of these betulinic acid-induced antiangiogenic and proapoptotic responses in both LNCaP cells and in tumors is due to activation of selective proteasome-dependent degradation of the transcription factors specificity protein 1 (Sp1), Sp3, and Sp4, which regulate VEGF and survivin expression. Thus, betulinic acid acts as a novel anticancer agent through targeted degradation of Sp proteins that are highly overexpressed in tumors.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Prostatic Neoplasms/drug therapy , Sp Transcription Factors/antagonists & inhibitors , Triterpenes/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Humans , Inhibitor of Apoptosis Proteins , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Nude , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/drug therapy , Pentacyclic Triterpenes , Promoter Regions, Genetic/drug effects , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Sp Transcription Factors/metabolism , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/metabolism , Sp4 Transcription Factor/antagonists & inhibitors , Sp4 Transcription Factor/genetics , Sp4 Transcription Factor/metabolism , Survivin , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays , Betulinic Acid
15.
Cancer Res ; 67(2): 674-83, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17234778

ABSTRACT

Nerve growth factor-induced Balpha (NGFI-Balpha, Nur77) is an orphan nuclear receptor with no known endogenous ligands; however, recent studies on a series of methylene-substituted diindolylmethanes (C-DIM) have identified 1,1-bis(3'-indolyl)-1-(phenyl)methane (DIM-C-Ph) and 1,1-bis(3'-indolyl)-1-(p-anisyl)methane (DIM-C-pPhOCH3) as Nur77 agonists. Nur77 is expressed in several colon cancer cell lines (RKO, SW480, HCT-116, HT-29, and HCT-15), and we also observed by immunostaining that Nur77 was overexpressed in colon tumors compared with normal colon tissue. DIM-C-Ph and DIM-C-pPhOCH3 decreased survival and induced apoptosis in RKO colon cancer cells, and this was accompanied by induction of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) protein. The induction of apoptosis and TRAIL by DIM-C-pPhOCH3 was significantly inhibited by a small inhibitory RNA for Nur77 (iNur77); however, it was evident from RNA interference studies that DIM-C-pPhOCH3 also induced Nur77-independent apoptosis. Analysis of DIM-C-pPhOCH3-induced gene expression using microarrays identified several proapoptotic genes, and analysis by reverse transcription-PCR in the presence or absence of iNur77 showed that induction of programmed cell death gene 1 was Nur77 dependent, whereas induction of cystathionase and activating transcription factor 3 was Nur77 independent. DIM-C-pPhOCH3 (25 mg/kg/d) also inhibited tumor growth in athymic nude mice bearing RKO cell xenografts. These results show that Nur77-active C-DIM compounds represent a new class of anti-colon cancer drugs that act through receptor-dependent and receptor-independent pathways.


Subject(s)
Apoptosis/drug effects , Apoptosis/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , DNA-Binding Proteins/agonists , Indoles/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Steroid/agonists , Transcription Factors/agonists , Animals , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HT29 Cells , Humans , Male , Methane/analogs & derivatives , Methane/pharmacology , Mice , Mice, Nude , Nuclear Receptor Subfamily 4, Group A, Member 1 , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Steroid/biosynthesis , Transcription Factors/biosynthesis , Xenograft Model Antitumor Assays
16.
Mol Cancer Ther ; 7(12): 3825-33, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19074857

ABSTRACT

Nurr1 is an orphan nuclear receptor and a member of the nerve growth factor I-B subfamily of transcription factors with no known endogenous ligand or stimulator. We show, for the first time, evidence that Nurr1 is expressed in a panel of 11 human bladder cancer cell lines. A new class of methylene-substituted diindolylmethanes (C-DIM) were screened and 1,1-bis(3'-indolyl)-1-(p-chlorophenyl)methane (DIM-C-pPhCl) activated the ligand-binding domain of Nurr1. Treatment of bladder cancer cells with Nurr1-active C-DIM resulted in decreased cell survival (MTT assay) and induction of cell death pathways, resulting in poly(ADP-ribose) polymerase cleavage and DNA fragmentation. The specificity of the Nurr1-active compound was shown using RNA interference in 253J B-V cells, whereby small interfering RNA against Nurr1 attenuated ligand-dependent activation of Nurr1 and poly(ADP-ribose) polymerase cleavage. Furthermore, activation of Nurr1 resulted in stimulation of tumor necrosis factor-related apoptosis-inducing ligand and small interfering RNA experiments attenuated tumor necrosis factor-related apoptosis-inducing ligand production. In an orthotopic model of human bladder tumors established in nude mice, administration of a Nurr1-active C-DIM suppressed bladder cancer growth. These results identify Nurr1 as a potential target for bladder cancer therapy and also identify a novel agent for activating Nurr1.


Subject(s)
Benzhydryl Compounds/pharmacology , DNA-Binding Proteins/physiology , Indoles/pharmacology , Transcription Factors/physiology , Urinary Bladder Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Survival , Humans , Indoles/chemistry , Ligands , Male , Mice , Mice, Nude , Models, Biological , Neoplasm Transplantation , Nuclear Receptor Subfamily 4, Group A, Member 2 , RNA Interference , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Urinary Bladder Neoplasms/pathology
17.
Mol Cancer Ther ; 7(7): 2109-20, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18645021

ABSTRACT

Bis(3'-indolyl)methane (DIM) is a metabolite of the phytochemical indole-3-carbinol, and both compounds exhibit a broad spectrum of anticancer activities. We have developed a series of synthetic symmetrical ring-substituted DIM analogues, including 5,5'-dibromoDIM, which are more potent than DIM as inhibitors of cancer cell and tumor growth. In colon cancer cells, 5,5'-dibromoDIM decreased cell proliferation and inhibited G(0)-G(1)- to S-phase progression, and this was accompanied by induction of the cyclin-dependent kinase inhibitor p21 in HT-29 and RKO colon cancer cells. Mechanistic studies showed that induction of p21 in both RKO (p53 wild-type) and HT-29 (p53 mutant) cells by 5,5'-dibromoDIM was Krüppel-like factor 4 (KLF4) dependent, and induction of p53 in RKO cells was also KLF4 dependent. Analysis of the p21 promoter in p53-dependent RKO cells showed that 5,5'-dibromoDIM activated p21 gene expression through the proximal GC-rich sites 1 and 2, and chromatin immunoprecipitation assays showed that KLF4 and p53 bound to this region of the promoter, whereas in HT-29 cells unidentified upstream cis-elements were required for induction of p21. 5,5'-DibromoDIM (30 mg/kg/d) also inhibited tumor growth and induced p21 in athymic nude mice bearing RKO cells as xenografts, showing that ring-substituted DIM such as 5,5'-dibromoDIM represent a novel class of mechanism-based drugs for clinical treatment of colon cancer.


Subject(s)
Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Indoles/pharmacology , Kruppel-Like Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kruppel-Like Factor 4 , Male , Mice , Mice, Nude , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
18.
Mol Cancer Ther ; 18(4): 856-867, 2019 04.
Article in English | MEDLINE | ID: mdl-30787172

ABSTRACT

Inhibition of VEGFR signaling is an effective treatment for renal cell carcinoma, but resistance continues to be a major problem. Recently, the sphingosine phosphate (S1P) signaling pathway has been implicated in tumor growth, angiogenesis, and resistance to antiangiogenic therapy. S1P is a bioactive lipid that serves an essential role in developmental and pathologic angiogenesis via activation of the S1P receptor 1 (S1P1). S1P1 signaling counteracts VEGF signaling and is required for vascular stabilization. We used in vivo and in vitro angiogenesis models including a postnatal retinal angiogenesis model and a renal cell carcinoma murine tumor model to test whether simultaneous inhibition of S1P1 and VEGF leads to improved angiogenic inhibition. Here, we show that inhibition of S1P signaling reduces the endothelial cell barrier and leads to excessive angiogenic sprouting. Simultaneous inhibition of S1P and VEGF signaling further disrupts the tumor vascular beds, decreases tumor volume, and increases tumor cell death compared with monotherapies. These studies suggest that inhibition of angiogenesis at two stages of the multistep process may maximize the effects of antiangiogenic therapy. Together, these data suggest that combination of S1P1 and VEGFR-targeted therapy may be a useful therapeutic strategy for the treatment of renal cell carcinoma and other tumor types.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Drug Therapy, Combination , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/pathology , Lysophospholipids/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Neovascularization, Pathologic/drug therapy , Sphingosine/analogs & derivatives , Sphingosine/antagonists & inhibitors , Sunitinib/pharmacology , Treatment Outcome , Tumor Burden/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Xenograft Model Antitumor Assays
19.
Mol Pharmacol ; 73(2): 553-65, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17989348

ABSTRACT

Methyl 2-cyano-3,11-dioxo-18beta-olean-1,12-diene-30-oate (beta-CDODA-Me) is a synthetic analog of the naturally occurring triterpenoid glycyrrhetinic acid, which contains a 2-cyano substituent in the A-ring. beta-CDODA-Me was a potent inhibitor of LNCaP prostate cancer cell growth (IC(50) approximately 1 muM) and activated peroxisome proliferator-activated receptor gamma (PPARgamma), whereas analogs without the cyano group were weakly cytotoxic and did not activate PPARgamma. beta-CDODA-Me induced p21 and p27, down-regulated cyclin D1 protein expression, and induced two other proapoptotic proteins, namely nonsteroidal anti-inflammatory drug-activated gene-1 and activating transcription factor-3. However, induction of these responses by beta-CDODA-Me was PPARgamma-independent and due to activation of phosphatidylinositol-3-kinase, mitogen-activated protein kinase, and jun N-terminal kinase pathways by this compound. In contrast, beta-CDODA-Me also decreased androgen receptor (AR) and prostate-specific antigen (PSA) mRNA and protein levels through kinase-independent pathways. beta-CDODA-Me repressed AR mRNA transcription, whereas decreased PSA mRNA levels were dependent on protein synthesis and were reversed by cycloheximide. Thus, potent inhibition of LNCaP cell survival by beta-CDODA-Me is due to PPARgamma-independent activation of multiple pathways that selectively activate growth-inhibitory and proapoptotic responses.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Glycyrrhetinic Acid/analogs & derivatives , Glycyrrhetinic Acid/pharmacology , PPAR gamma/agonists , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/physiology , Cell Line, Tumor , Glycyrrhetinic Acid/therapeutic use , Humans , Male , PPAR gamma/metabolism , Prostatic Neoplasms/metabolism
20.
Carcinogenesis ; 29(6): 1139-47, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18460448

ABSTRACT

1,1-Bis(3'-indolyl)-1-(p-substituted phenyl)methanes (C-DIMs) activate the orphan receptors peroxisome proliferator-activated receptor gamma (PPARgamma) and Nur77 and induce receptor-dependent and -independent apoptotic pathways in colon and other cancer cells. Structure-activity studies show that the p-bromo (DIM-C-pPhBr) and p-fluoro (DIM-C-pPhF) analogs, which exhibit minimal activation of Nur77 and PPARgamma, induce expression of CCAAT/enhancer-binding protein homologous protein (CHOP/GADD153) in colon cancer cells. Moreover, among a series of bromo and fluoro C-DIM analogs, their induction of CHOP was dependent on the position of the phenyl substituents (para >/= meta >/= ortho) and required a free indole group. DIM-C-pPhBr and DIM-C-pPhF not only induced CHOP but also activated death receptor 5 (CHOP dependent), cleavage of caspase 8 and poly (ADP ribose) polymerase (PARP) that is consistent with activation of the extrinsic pathway of apoptosis. These responses were associated with the activation of c-jun N-terminal kinase (JNK) pathway since inhibition of JNK inhibited induction of the extrinsic apoptotic pathway by these C-DIMs. However, in contrast to classical inducers of endoplasmic reticulum (ER) stress such as tunicamycin and thapsigargin, the C-DIM compounds did not induce glucose-related protein 78 that is a marker of ER stress. Proapoptotic and anticarcinogenic effects were also observed in athymic nude mice bearing RKO cell xenografts and treated with 30 mg/kg/day DIM-C-pPhBr and this was accompanied by increased JNK phosphorylation in the tumors. Thus, the anticarcinogenic activity of DIM-C-pPhBr in colon cancer cells and tumors is related to a novel ER stress-independent activation of JNK.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/enzymology , Enzyme Activation/drug effects , Indoles/pharmacology , JNK Mitogen-Activated Protein Kinases/drug effects , Animals , Antineoplastic Agents/chemistry , Blotting, Western , Cell Line, Tumor , Collagen Type XI/drug effects , Collagen Type XI/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Humans , Immunohistochemistry , Immunoprecipitation , Indoles/chemistry , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Nude , Structure-Activity Relationship , Transcription Factor CHOP/drug effects , Transcription Factor CHOP/metabolism , Transfection , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL