Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
2.
Sci Rep ; 12(1): 22255, 2022 12 23.
Article in English | MEDLINE | ID: mdl-36564457

ABSTRACT

Triple-negative breast cancer (TNBC) is associated with high mortality due to the high expression of pro-inflammatory cytokines and lack of targeted therapies. N-acylethanolamine acid amidase (NAAA) is an N-terminal cysteine hydrolase that promotes inflammatory responses through the deactivation of Palmitoylethanolamide (PEA), an endogenous bioactive lipid mediator. Here, we examined NAAA expression in TNBC cells (MDA-MB-231 and MDA-MB-BrM2 cells) and the effects of NAAA inhibition on TNBC tumor growth, using a selective NAAA inhibitor AM11095 (IC50 = 20 nM). TNBC cells expressed elevated levels of full-length and splice mRNAs naaa variants. TNBC cells also express the N-acyl ethanol amides and elevated levels of the two fatty acid cores arachidonic (AA) and docosahexaenoic (DHA). PEA or AM11095 inhibited the secretion of IL-6 and IL-8, reduced the activation of the NF-kB pathway, decreased the expression of VEGF and Placental growth factor (PLGF) in TNBCs, and inhibited tumor cell migration in vitro. Using cellular magnetic resonance imaging (MRI), body images of mice administered with human MDA-MB-BrM2 cells treated with AM11095 showed a significant decrease in tumor numbers with a lower volume of tumors and increased mice survival. Mice untreated or treated with vehicle control showed a high number of tumors with high volumes in multiple organs. Thus, NAAA inhibition may constitute a potential therapeutic approach in the management of TNBC-associated inflammation and tumor growth.


Subject(s)
Triple Negative Breast Neoplasms , Mice , Humans , Female , Animals , Triple Negative Breast Neoplasms/drug therapy , Amidohydrolases/genetics , Amidohydrolases/metabolism , Placenta Growth Factor/therapeutic use , Inflammation/drug therapy , Amides/therapeutic use
3.
Sci Rep ; 12(1): 5328, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35351947

ABSTRACT

While the prevalence of breast cancer metastasis in the brain is significantly higher in triple negative breast cancers (TNBCs), there is a lack of novel and/or improved therapies for these patients. Monoacylglycerol lipase (MAGL) is a hydrolase involved in lipid metabolism that catalyzes the degradation of 2-arachidonoylglycerol (2-AG) linked to generation of pro- and anti-inflammatory molecules. Here, we targeted MAGL in TNBCs, using a potent carbamate-based inhibitor AM9928 (hMAGL IC50 = 9 nM) with prolonged pharmacodynamic effects (46 h of target residence time). AM9928 blocked TNBC cell adhesion and transmigration across human brain microvascular endothelial cells (HBMECs) in 3D co-cultures. In addition, AM9928 inhibited the secretion of IL-6, IL-8, and VEGF-A from TNBC cells. TNBC-derived exosomes activated HBMECs resulting in secretion of elevated levels of IL-8 and VEGF, which were inhibited by AM9928. Using in vivo studies of syngeneic GFP-4T1-BrM5 mammary tumor cells, AM9928 inhibited tumor growth in the mammary fat pads and attenuated blood brain barrier (BBB) permeability changes, resulting in reduced TNBC colonization in brain. Together, these results support the potential clinical application of MAGL inhibitors as novel treatments for TNBC.


Subject(s)
Monoacylglycerol Lipases , Triple Negative Breast Neoplasms , Brain/metabolism , Endothelial Cells/metabolism , Humans , Inflammation , Monoacylglycerol Lipases/metabolism , Triple Negative Breast Neoplasms/pathology
4.
J Pathol ; 232(3): 369-81, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24421076

ABSTRACT

Although the incidence of breast cancer metastasis (BCM) in brain has increased significantly in triple-negative breast cancer (TNBC), the mechanisms remain elusive. Using in vivo mouse models for BCM in brain, we observed that TNBC cells crossed the blood-brain barrier (BBB), lodged in the brain microvasculature and remained adjacent to brain microvascular endothelial cells (BMECs). Breaching of the BBB in vivo by TNBCs resulted in increased BBB permeability and changes in ZO-1 and claudin-5 tight junction (TJ) protein structures. Angiopoietin-2 expression was elevated in BMECs and was correlated with BBB disruption. Secreted Ang-2 impaired TJ structures and increased BBB permeability. Treatment of mice with the neutralizing Ang-2 peptibody trebananib prevented changes in the BBB integrity and BMEC destabilization, resulting in inhibition of TNBC colonization in brain. Thus, Ang-2 is involved in initial steps of brain metastasis cascade, and inhibitors for Ang-2 may serve as potential therapeutics for brain metastasis.


Subject(s)
Angiopoietin-2/metabolism , Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Mammary Neoplasms, Experimental/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Blood-Brain Barrier/pathology , Capillary Permeability/physiology , Female , Heterografts , Humans , Immunohistochemistry , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Tight Junctions/metabolism , Tight Junctions/pathology , Triple Negative Breast Neoplasms/pathology
5.
Int J Cancer ; 134(5): 1034-44, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-23934616

ABSTRACT

Neuropeptide substance P (SP) has been implicated in inflammation, pain, depression and breast cancer cell (BCC) growth. Here, we examined the role of SP in trafficking of BCCs (human MDA-MB-231 and MDA-MB-231BrM2 cells) across the blood-brain barrier (BBB) and brain microvascular endothelial cells (BMECs) using in vitro and in vivo models. SP was secreted from BCCs and mediated adhesion and transmigration of BCCs across human BMECs (HBMECs) in vitro. SP induced activation of HBMECs, leading to secretion of Tumor Necrosis Factor alpha (TNF-α) and angiopoietin-2 (Ang-2) from HBMECs, resulting in changes in localization and distribution of tight junction (TJ) ZO-1 (tight junction protein zonula occludins-1) and claudin-5 structures as well as increased permeability of HBMECs. Using spontaneous breast cancer metastasis mouse model (syngeneic) of GFP-4T1-BrM5 mammary tumor cells administered into mammary fat pads of Balb/c mice, SP inhibitor spantide III inhibited in vivo changes in permeability of the BBB and BMEC-TJs ZO-1 and claudin-5 structures as well as decreased tumor cell colonization in brain. Thus, SP secreted from BCCs induces transmigration of BCCs across the BBB, leading to activation of BMECs and secretion of TNF-α and Ang-2, resulting in BBB impairment and colonization of tumor cells in brain. Therefore, therapies based on SP inhibition in combination with other therapies may prevent breaching of the BBB by BCCs and their colonization in brain.


Subject(s)
Blood-Brain Barrier , Breast Neoplasms/pathology , Endothelial Cells/physiology , Substance P/physiology , Tight Junctions/physiology , Animals , Cell Adhesion , Cell Movement , Cytoskeleton/chemistry , Female , Humans , Mice , Microvessels/cytology , Permeability , Substance P/analogs & derivatives , Substance P/antagonists & inhibitors , Substance P/pharmacology , Tumor Necrosis Factor-alpha/metabolism
6.
Br J Pharmacol ; 171(2): 468-79, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24148086

ABSTRACT

BACKGROUND AND PURPOSE: HIV-1 glycoprotein Gp120 induces apoptosis in rodent and human neurons in vitro and in vivo. HIV-1/Gp120 is involved in the pathogenesis of HIV-associated dementia (HAD) and inhibits proliferation of adult neural progenitor cells (NPCs) in glial fibrillary acidic protein (GFAP)/Gp120 transgenic (Tg) mice. As cannabinoids exert neuroprotective effects in several model systems, we examined the protective effects of the CB2 receptor agonist AM1241 on Gp120-mediated insults on neurogenesis. EXPERIMENTAL APPROACH: We assessed the effects of AM1241 on survival and apoptosis in cultures of human and murine NPCs with immunohistochemical and TUNEL techniques. Neurogenesis in the hippocampus of GFAP/Gp120 transgenic mice in vivo was also assessed by immunohistochemistry. KEY RESULTS: AM1241 inhibited in vitro Gp120-mediated neurotoxicity and apoptosis of primary human and murine NPCs and increased their survival. AM1241 also promoted differentiation of NPCs to neuronal cells. While GFAP/Gp120 Tg mice exhibited impaired neurogenesis, as indicated by reduction in BrdU⁺ cells and doublecortin⁺ (DCX⁺) cells, and a decrease in cells with proliferating cell nuclear antigen (PCNA), administration of AM1241 to GFAP/Gp120 Tg mice resulted in enhanced in vivo neurogenesis in the hippocampus as indicated by increase in neuroblasts, neuronal cells, BrdU⁺ cells and PCNA⁺ cells. Astrogliosis and gliogenesis were decreased in GFAP/Gp120 Tg mice treated with AM1241, compared with those treated with vehicle. CONCLUSIONS AND IMPLICATIONS: The CB2 receptor agonist rescued impaired neurogenesis caused by HIV-1/Gp120 insult. Thus, CB2 receptor agonists may act as neuroprotective agents, restoring impaired neurogenesis in patients with HAD.


Subject(s)
Glial Fibrillary Acidic Protein/genetics , HIV Envelope Protein gp120/genetics , Neurogenesis/drug effects , Receptor, Cannabinoid, CB2/agonists , Animals , Apoptosis/drug effects , Cannabinoids/pharmacology , Cell Differentiation/drug effects , Cell Survival/drug effects , Doublecortin Protein , Hippocampus/cytology , Hippocampus/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Stem Cells/drug effects , Neurogenesis/genetics , Neurogenesis/physiology , Primary Cell Culture , Real-Time Polymerase Chain Reaction , Receptor, Cannabinoid, CB2/biosynthesis
7.
Transfusion ; 51 Suppl 4: 65S-71S, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22074629

ABSTRACT

The cannabinoid receptors CB(1) and CB(2) are seven-transmembrane Gαi protein-coupled receptors and are expressed in certain mature hematopoietic cells. We recently showed that these receptors are expressed in murine and human hematopoietic stem cells (HSCs) and that CB(2) agonists induced chemotaxis, enhanced colony formation of marrow cells, as well as caused in vivo mobilization of murine HSCs with short- and long-term repopulating abilities. Based on these observations, we have further explored the role of CB(2) and its agonist AM1241 on hematopoietic recovery following sublethal irradiation in mice. Cannabinoid receptor 2 knockout mice (Cnr2(-/-) deficient mice) exhibited impaired recovery following sublethal irradiation as compared with irradiated wild-type (WT) mice, as determined by low colony-forming units and low peripheral blood counts. WT mice treated with CB(2) agonist AM1241 following sublethal irradiation demonstrated accelerated marrow recovery and increased total marrow cells (approximately twofold) and total lineage- c-kit(+) cells (approximately sevenfold) as well as enhanced HSC survival as compared with vehicle control-treated mice. When the CB(2) agonist AM1241 was administered to WT mice 12 days before their sublethal irradiation, analysis of hematopoiesis in these mice showed decreased apoptosis of HSCs, enhanced survival of HSCs, as well as increase in total marrow cells and c-kit+ cells in the marrow. Thus, CB(2) agonist AM1241 promoted recovery after sublethal irradiation by inhibiting apoptosis of HSCs and promoting survival, as well as enhancing the number of HSCs entering the cell cycle.


Subject(s)
Cannabinoid Receptor Modulators/physiology , Endocannabinoids , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Receptor, Cannabinoid, CB1/physiology , Receptor, Cannabinoid, CB2/physiology , Animals , Apoptosis , Cannabinoids/pharmacology , Cell Cycle , Cell Movement/physiology , Colony-Forming Units Assay , Hematopoiesis/radiation effects , Humans , Mice , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Stem Cell Niche , Whole-Body Irradiation
8.
Blood ; 117(3): 827-38, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21063029

ABSTRACT

Endocannabinoids are arachidonic acid derivatives and part of a novel bioactive lipid signaling system, along with their G-coupled cannabinoid receptors (CB1 and CB2) and the enzymes involved in their biosynthesis and degradation. However, their roles in hematopoiesis and hematopoietic stem and progenitor cell (HSPC) functions are not well characterized. Here, we show that bone marrow stromal cells express endocannabinoids (anandamide and 2-arachidonylglycerol), whereas CB2 receptors are expressed in human and murine HSPCs. On ligand stimulation with CB2 agonists, CB2 receptors induced chemotaxis, migration, and enhanced colony formation of bone marrow cells, which were mediated via ERK, PI3-kinase, and Gαi-Rac1 pathways. In vivo, the CB2 agonist AM1241 induced mobilization of murine HSPCs with short- and long-term repopulating abilities. In addition, granulocyte colony-stimulating factor -induced mobilization of HSPCs was significantly decreased by specific CB2 antagonists and was impaired in Cnr2(-/-) cannabinoid type 2 receptor knockout mice. Taken together, these results demonstrate that the endocannabinoid system is involved in hematopoiesis and that CB2/CB2 agonist axis mediates repopulation of hematopoiesis and mobilization of HSPCs. Thus, CB2 agonists may be therapeutically applied in clinical conditions, such as bone marrow transplantation.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/physiology , Receptor, Cannabinoid, CB2/metabolism , Animals , Bone Marrow Cells/metabolism , Cannabinoid Receptor Modulators/metabolism , Cannabinoids/pharmacology , Cell Movement/drug effects , Cyclohexanols/pharmacology , Female , Flow Cytometry , Hematopoiesis/drug effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/genetics , Stromal Cells/metabolism
9.
J Biol Chem ; 285(46): 35471-8, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20826813

ABSTRACT

Endocannabinoids are lipid signaling molecules that act via G-coupled receptors, CB(1) and CB(2). The endocannabinoid system is capable of activation of distinct signaling pathways on demand in response to pathogenic events or stimuli, hereby enhancing cell survival and promoting tissue repair. However, the role of endocannabinoids in hematopoietic stem and progenitor cells (HSPCs) and their interaction with hematopoietic stem cells (HSC) niches is not known. HSPCs are maintained in the quiescent state in bone marrow (BM) niches by intrinsic and extrinsic signaling. We report that HSPCs express the CB(1) receptors and that BM stromal cells secrete endocannabinoids, anandamide (AEA) (35 pg/10(7) cells), and 2-AG (75.2 ng/10(7) cells). In response to the endotoxin lipopolysaccharide (LPS), elevated levels of AEA (75.6 pg/10(7) cells) and 2-AG (98.8 ng/10(7) cells) were secreted from BM stromal cells, resulting in migration and trafficking of HSPCs from the BM niches to the peripheral blood. Furthermore, administration of exogenous cannabinoid CB(1) agonists in vivo induced chemotaxis, migration, and mobilization of human and murine HSPCs. Cannabinoid receptor knock-out mice Cnr1(-/-) showed a decrease in side population (SP) cells, whereas fatty acid amide hydrolase (FAAH)(-/-) mice, which have elevated levels of AEA, yielded increased colony formation as compared with WT mice. In addition, G-CSF-induced mobilization in vivo was modulated by endocannabinoids and was inhibited by specific cannabinoid antagonists as well as impaired in cannabinoid receptor knock-out mice Cnr1(-/-), as compared with WT mice. Thus, we propose a novel function of the endocannabinoid system, as a regulator of HSPC interactions with their BM niches, where endocannabinoids are expressed in HSC niches and under stress conditions, endocannabinoid expression levels are enhanced to induce HSPC migration for proper hematopoiesis.


Subject(s)
Bone Marrow Cells/metabolism , Cannabinoid Receptor Modulators/biosynthesis , Endocannabinoids , Hematopoietic Stem Cells/metabolism , Stromal Cells/metabolism , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , Arachidonic Acids/biosynthesis , Blotting, Western , Bone Marrow Cells/cytology , Cannabinoid Receptor Modulators/physiology , Cell Communication/physiology , Cell Movement/drug effects , Cells, Cultured , Cyclohexanols/pharmacology , Female , Flow Cytometry , Glycerides/biosynthesis , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polyunsaturated Alkamides , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Side-Population Cells/cytology , Side-Population Cells/metabolism , Stem Cell Niche/cytology , Stem Cell Niche/metabolism , Stromal Cells/cytology
10.
J Biol Chem ; 285(34): 26190-8, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20511222

ABSTRACT

Reactive molecules have diverse effects on cells and contribute to several pathological conditions. Cells have evolved complex protective systems to neutralize these molecules and restore redox homeostasis. Previously, we showed that association of nuclear factor (NF)-erythroid-derived 2 (E2)-related factor 2 (NRF2) with the nuclear matrix protein NRP/B was essential for the transcriptional activity of NRF2 target genes in tumor cells. The present study demonstrates the molecular mechanism by which NRP/B, via NRF2, modulates the transcriptional activity of antioxidant response element (ARE)-driven genes. NRP/B is localized in the nucleus of primary brain tissue and human neuroblastoma (SH-SY5Y) cells. Treatment with hydrogen peroxide (H(2)O(2)) enhances the nuclear colocalization of NRF2 and NRP/B and induces heme oxygenase 1 (HO1). Treatment of NRP/B or NRF2 knockdowns with H(2)O(2) induced apoptosis. Co-expression of NRF2 with members of the Kelch protein family, NRP/B, MAYVEN, or MAYVEN-related protein 2 (MRP2), revealed that the NRF2-NRP/B complex is important for the transcriptional activity of ARE-driven genes HO1 and NAD(P)H:quinine oxidoreductase 1 (NQO1). NRP/B interaction with Nrf2 was mapped to NRF2 ECH homology 4 (Neh4)/Neh5 regions of NRF2. NRP/B mutations that resulted in low binding affinity to NRF2 were unable to activate NRF2-modulated transcriptional activity of the ARE-driven genes, HO1 and NQO1. Thus, the interaction of NRP/B with the Neh4/Neh5 domains of NRF2 is indispensable for activation of NRF2-mediated ARE-driven antioxidant and detoxifying genes that confer cellular defense against oxidative stress-induced damage.


Subject(s)
Microfilament Proteins/physiology , NF-E2-Related Factor 2/metabolism , Neuropeptides/physiology , Nuclear Matrix-Associated Proteins/physiology , Nuclear Proteins/physiology , Oxidative Stress/genetics , Transcriptional Activation , Antioxidants , Brain Chemistry , Cell Line, Tumor , Gene Expression Regulation , Humans , Neuroblastoma/chemistry , Oxidation-Reduction , Response Elements
11.
PLoS One ; 5(3): e9707, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20300573

ABSTRACT

BACKGROUND: Mammary stem cells are maintained within specific microenvironments and recruited throughout lifetime to reconstitute de novo the mammary gland. Mammary stem cells have been isolated through the identification of specific cell surface markers and in vivo transplantation into cleared mammary fat pads. Accumulating evidence showed that during the reformation of mammary stem cell niches by dispersed epithelial cells in the context of the intact epithelium-free mammary stroma, non-mammary epithelial cells may be sequestered and reprogrammed to perform mammary epithelial cell functions and to adopt mammary epithelial characteristics during reconstruction of mammary epithelium in regenerating mammary tissue in vivo. METHODOLOGY/PRINCIPAL FINDINGS: To examine whether other types of progenitor cells are able to contribute to mammary branching morphogenesis, we examined the potential of murine embryonic stem (mES) cells, undergoing hematopoietic differentiation, to support mammary reconstitution in vivo. We observed that cells from day 14 embryoid bodies (EBs) under hematopoietic differentiation condition, but not supernatants derived from these cells, when transplanted into denuded mammary fat pads, were able to contribute to both the luminal and myoepithelial lineages in branching ductal structures resembling the ductal-alveolar architecture of the mammary tree. No teratomas were observed when these cells were transplanted in vivo. CONCLUSIONS/SIGNIFICANCE: Our data provide evidence for the dominance of the tissue-specific mammary stem cell niche and its role in directing mES cells, undergoing hematopoietic differentiation, to reprogram into mammary epithelial cells and to promote mammary epithelial morphogenesis. These studies should also provide insights into regeneration of damaged mammary gland and the role of the mammary microenvironment in reprogramming cell fate.


Subject(s)
Embryonic Stem Cells/cytology , Hematopoietic Stem Cells/cytology , Animals , Cell Differentiation , Cell Lineage , Collagen/chemistry , Drug Combinations , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , Hematopoiesis , Immunohistochemistry/methods , Laminin/chemistry , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred BALB C , Polymerase Chain Reaction , Proteoglycans/chemistry , Time Factors , Y Chromosome
12.
Mol Cancer Res ; 8(3): 407-20, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20215423

ABSTRACT

BRCA1 is an essential caretaker protein in the surveillance of DNA damage, is mutated in approximately 50% of all hereditary breast cancer cases, and its expression is frequently decreased in sporadic breast cancer. beta-Catenin is a multifunctional protein that forms adhesion complex with E-cadherins, alpha-catenin, and actin, and plays a central role in Wnt signaling through its nuclear translocation and activation of beta-catenin-responsive genes. Although significant progress has been made in understanding the Wnt/beta-catenin and BRCA1 signaling cascades, it is not known whether there is a link between beta-catenin and BRCA1. We observed that the expression of the active nuclear form of beta-catenin (also known as ABC, Ser37/Thr41-nonphosphorylated beta-catenin, dephosphorylated beta-catenin) was lower or absent in the nucleus in most BRCA1 familial breast cancer tissues (17 cases) compared with sporadic breast cancer (14 samples) and normal breast tissues. Wild-type-BRCA1, but not mutated BRCA1, interacted with beta-catenin and increased the levels of beta-catenin protein expression in vitro. Furthermore, H(2)O(2) induced the interaction of the nuclear form of beta-catenin with BRCA1. The active form of beta-catenin protein was downregulated upon exposure to H(2)O(2) in the nucleus of BRCA1-deficient HCC1937 breast cancer cells, whereas reconstitution of WT-BRCA1 in HCC1937 cells inhibited this downregulation. This study provides evidence of a novel interaction between BRCA1 and beta-catenin, and that loss of BRCA1 leads to impaired expression of the nuclear form of beta-catenin, which may contribute to the pathogenesis of breast cancer.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/genetics , Carcinoma/genetics , Gene Expression Regulation, Neoplastic/genetics , beta Catenin/metabolism , BRCA1 Protein/metabolism , Breast Neoplasms/metabolism , Carcinoma/metabolism , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Humans , Hydrogen Peroxide/pharmacology , Mutation/genetics , Oxidants/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/genetics , Signal Transduction/genetics , beta Catenin/genetics
13.
Patholog Res Int ; 2011: 920509, 2010 Dec 29.
Article in English | MEDLINE | ID: mdl-21253507

ABSTRACT

Brain metastasis, an important cause of cancer morbidity and mortality, occurs in at least 30% of patients with breast cancer. A key event of brain metastasis is the migration of cancer cells through the blood-brain barrier (BBB). Although preventing brain metastasis is immensely important for survival, very little is known about the early stage of transmigration and the molecular mechanisms of breast tumor cells penetrating the BBB. The brain endothelium plays an important role in brain metastasis, although the mechanisms are not clear. Brain Microvascular Endothelial Cells (BMECs) are the major cellular constituent of the BBB. BMECs are joined together by intercellular tight junctions (TJs) that are responsible for acquisition of highly selective permeability. Failure of the BBB is a critical event in the development and progression of several diseases that affect the CNS, including brain tumor metastasis development. Here, we have delineated the mechanisms of BBB impairment and breast cancer metastasis to the brain. Understanding the molecular mediators that cause changes in the BBB should lead to better strategies for effective treatment modalities targeted to inhibition of brain tumors.

14.
Clin Exp Metastasis ; 27(2): 97-105, 2010 Feb.
Article in English | MEDLINE | ID: mdl-18814042

ABSTRACT

Breast cancer is the most common malignancy and second leading cause of cancer death in women. Ninety percent of mortality in breast cancer is often associated with metastatic progression or relapse in patients. Critical stages in the development of aggressive breast cancer include the growth of primary tumors and their ability to spread to foreign organs and form metastases, as well as the establishment of an independent blood supply within the new tumors. Hence, it is imperative to characterize the key molecules that regulate the metastasis of human breast cancer cells. The expression of CXCR4/CXCL12 in breast tumors has been correlated with a poor prognosis, increased metastasis, resistance to conventional therapeutic agents and a poor outcome in the pathogenesis of breast cancer. However, effective anti-CXCR4 therapy remains a challenge. Here, we will review the putative involvement of the CXCR4/CXCL12 signaling axis in breast cancer metastasis to the brain. Characterization of signaling events important for breast cancer cell growth and their metastasis to the brain should provide insights into breast cancer therapies and improved, successful treatments for breast cancer.


Subject(s)
Brain Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chemokine CXCL12/metabolism , Receptors, CXCR4/metabolism , Signal Transduction/physiology , Brain Neoplasms/metabolism , Chemokine CXCL12/physiology , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Receptors, CXCR4/physiology
15.
PLoS One ; 4(9): e7091, 2009 Sep 21.
Article in English | MEDLINE | ID: mdl-19768112

ABSTRACT

BACKGROUND: BRCA1 is a key regulatory protein participating in cell cycle checkpoint and DNA damage repair networks. BRCA1 plays important roles in protecting numerous cellular processes in response to cell damaging signals. Transforming growth factor-beta (TGF-beta) is a potent regulator of growth, apoptosis and invasiveness of tumor cells. TFG-beta activates Smad signaling via its two cell surface receptors, the TbetaRII and ALK5/TbetaRI, leading to Smad-mediated transcriptional regulation. METHODOLOGY/PRINCIPAL FINDINGS: Here, we report an important role of BRCA1 in modulating TGF-beta signaling during oxidative stress responses. Wild-type (WT) BRCA1, but not mutated BRCA1 failed to activate TGF-beta mediated transactivation of the TGF-beta responsive reporter, p3TP-Lux. Further, WT-BRCA1, but not mutated BRCA1 increased the expression of Smad3 protein in a dose-dependent manner, while silencing of WT-BRCA1 by siRNA decreased Smad3 and Smad4 interaction induced by TGF-beta in MCF-7 breast cancer cells. BRCA1 interacted with Smad3 upon TGF-beta1 stimulation in MCF-7 cells and this interaction was mediated via the domain of 298-436aa of BRCA1 and Smad3 domain of 207-426aa. In addition, H(2)O(2) increased the colocalization and the interaction of Smad3 with WT-BRCA1. Interestingly, TGF-beta1 induced Smad3 and Smad4 interaction was increased in the presence of H(2)O(2) in cells expressing WT-BRCA1, while the TGF-beta1 induced interaction between Smad3 and Smad4 was decreased upon H(2)O(2) treatment in a dose-dependent manner in HCC1937 breast cancer cells, deficient for endogenous BRCA1. This interaction between Smad3 and Smad4 was increased in reconstituted HCC1937 cells expressing WT-BRCA1 (HCC1937/BRCA1). Further, loss of BRCA1 resulted in H(2)O(2) induced nuclear export of phosphor-Smad3 protein to the cytoplasm, resulting decreased of Smad3 and Smad4 interaction induced by TGF-beta and in significant decrease in Smad3 and Smad4 transcriptional activities. CONCLUSIONS/SIGNIFICANCE: These results strongly suggest that loss or reduction of BRCA1 alters TGF-beta growth inhibiting activity via Smad3 during oxidative stress responses.


Subject(s)
BRCA1 Protein/metabolism , Gene Expression Regulation , Oxidative Stress , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Apoptosis , COS Cells , Cell Proliferation , Chlorocebus aethiops , Germ-Line Mutation , Humans , Hydrogen Peroxide/metabolism , Neoplasm Invasiveness , Smad Proteins/metabolism , Transcription, Genetic
16.
J Immunol ; 181(9): 6406-16, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941231

ABSTRACT

HIV-1 infection has significant effect on the immune system as well as on the nervous system. Breakdown of the blood-brain barrier (BBB) is frequently observed in patients with HIV-associated dementia (HAD) despite lack of productive infection of human brain microvascular endothelial cells (HBMEC). Cellular products and viral proteins secreted by HIV-1 infected cells, such as the HIV-1 Gp120 envelope glycoprotein, play important roles in BBB impairment and HIV-associated dementia development. HBMEC are a major component of the BBB. Using cocultures of HBMEC and human astrocytes as a model system for human BBB as well as in vivo model, we show for the first time that cannabinoid agonists inhibited HIV-1 Gp120-induced calcium influx mediated by substance P and significantly decreased the permeability of HBMEC as well as prevented tight junction protein down-regulation of ZO-1, claudin-5, and JAM-1 in HBMEC. Furthermore, cannabinoid agonists inhibited the transmigration of human monocytes across the BBB and blocked the BBB permeability in vivo. These results demonstrate that cannabinoid agonists are able to restore the integrity of HBMEC and the BBB following insults by HIV-1 Gp120. These studies may lead to better strategies for treatment modalities targeted to the BBB following HIV-1 infection of the brain based on cannabinoid pharmacotherapies.


Subject(s)
Anti-HIV Agents/pharmacology , Arachidonic Acids/pharmacology , Brain/blood supply , Brain/virology , Cannabinoid Receptor Modulators/pharmacology , Endothelium, Vascular/virology , Glycerides/pharmacology , HIV Envelope Protein gp120/antagonists & inhibitors , HIV-1/drug effects , AIDS Dementia Complex/enzymology , AIDS Dementia Complex/pathology , AIDS Dementia Complex/prevention & control , AIDS Dementia Complex/virology , Amidohydrolases/antagonists & inhibitors , Anti-HIV Agents/therapeutic use , Arachidonic Acids/physiology , Benzamides/pharmacology , Benzamides/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Brain/drug effects , Brain/pathology , Cannabinoid Receptor Modulators/agonists , Cannabinoid Receptor Modulators/physiology , Carbamates/pharmacology , Carbamates/therapeutic use , Cell Line , Coculture Techniques , Endocannabinoids , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiology , Glycerides/physiology , HIV Envelope Protein gp120/physiology , HIV-1/physiology , Humans , Microcirculation/drug effects , Microcirculation/physiology , Receptor, Cannabinoid, CB1/physiology
17.
J Cell Mol Med ; 12(5A): 1517-26, 2008.
Article in English | MEDLINE | ID: mdl-18363848

ABSTRACT

Metastasis contributes to more than 90% of mortality in breast cancer. Critical stages in the development of aggressive breast cancer include growth of the primary tumours, and their abilities to spread to distant organs, colonize and establish an independent blood supply. The integrin family of cell adhesion receptors is essential to breast cancer progression. Furthermore, integrin-linked kinase can 'convert' localized breast cancer cells into invasive and metastatic cells. Upon stimulation by growth factors and chemokine ligands, integrin-linked kinase mediates the phosphorylation of Akt Ser473, and glycogen synthase kinase-3. The current notion is that overexpression of integrin-linked kinase resulted in an invasive, metastatic phenotype in several cancer model systems in vivo and in vitro, thus, implicating a role for integrin-linked kinase in oncogenic transformation, angiogenesis and metastasis. Here, we will review the role of integrin-linked kinase in breast cancer metastasis. Elucidation of signalling events important for breast tumour metastasis should provide insights into successful breast cancer therapies.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Protein Serine-Threonine Kinases/metabolism , Animals , Humans , Neoplasm Metastasis/pathology , Receptor, ErbB-2/metabolism , Vascular Endothelial Growth Factor A/metabolism
18.
Int J Oncol ; 32(3): 619-23, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18292939

ABSTRACT

Neuroblastoma is the second most common pediatric malignancy. The clinical course of this disease ranges from spontaneous regression and good survival to highly malignant therapy-resistant tumors. There is a continuous need for genetic and biologic markers for the diverse clinical phenotypes observed in neuroblastoma patients. One of the known markers in neuroblastoma is expression of the CXCR4 chemokine receptor. CXCR4 expression correlates with high-stage disease, and the autocrine stimulation of CXCR4 by its ligand (CXCL12) was shown to be necessary for the survival of some neuroblastoma cells in vitro. However, the mechanisms responsible for activation of the CXCL12-CXCR4 autocrine pathway in neuroblastoma remain uncertain. Our previous findings suggest that Csk homologous kinase (CHK) is a physiological inhibitor of CXCR4 expression. Since CHK is highly expressed in neurons, we evaluated changes in CHK expression in human neuroblastoma. CHK protein expression was below detectable levels based on Western blot analyses in 13 out of 16 human neuroblastoma cell lines and in 6 out of 16 primary neuroblastoma tissues. When CHK expression was restored in IMR32 neuroblastoma cells by retrovirus-mediated cDNA transfer, diminished CXCR4 mRNA and protein levels were observed, as assessed by RT-PCR and flow cytometry analyses, respectively. Furthermore, exogenous expression of CHK markedly suppressed the mRNA levels and secretion of the CXCL12 chemokine from IMR32 cells as well as inhibited the growth rate of these cells. Taken together, our data strongly suggest that CHK is capable of inhibiting the CXCL12-CXCR4 pathway in neuroblastoma.


Subject(s)
Chemokine CXCL12/metabolism , Neuroblastoma/metabolism , Proto-Oncogene Proteins pp60(c-src)/physiology , Receptors, CXCR4/metabolism , Cell Proliferation , Chemokine CXCL12/genetics , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Neuroblastoma/genetics , Proto-Oncogene Proteins pp60(c-src)/genetics , RNA, Messenger/metabolism , Receptors, CXCR4/genetics , Signal Transduction/genetics , Transfection , Tumor Cells, Cultured
19.
Cancer Res ; 67(18): 8596-604, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875699

ABSTRACT

The transcription factor NF-E2-related factor 2 (Nrf2) translocates into the nucleus and activates phase II genes encoding detoxification enzymes and antioxidant proteins, resulting in the protection of cells from oxidative insults. However, the involvement of Nrf2-mediated oxidative stress responses in breast cancer cells is largely unknown. Notably, during our study of the Nrf2 pathway in breast cancer cells, we observed that the nuclear matrix protein NRP/B was expressed and colocalized with Nrf2 in these cells, suggesting that NRP/B is involved in Nrf2-mediated oxidative stress responses. The expression level of NRP/B was variable in different breast cancer cells and breast cancer tissues, and was found to be localized in the nucleus. NRP/B expression was increased after exposure to the oxidative stress agent, hydrogen peroxide (H(2)O(2)), particularly in the highly aggressive MDA-MB-231 breast cancer cells. Association of NRP/B with Nrf2 in vitro and in vivo was observed in MDA-MB-231 breast cancer cells, and this association was up-regulated upon exposure to H(2)O(2), but not to sodium nitroprusside, SIN-1, and DETA-NO. NRP/B also enhanced Nrf2-mediated NAD(P)H:quinine oxidoreductase 1 promoter activity. Thus, this study reveals that NRP/B enhances oxidative stress responses in breast cancer cells via the Nrf2 pathway, identifying a novel role of nuclear matrix protein(s) in oxidative stress responses.


Subject(s)
Breast Neoplasms/metabolism , Microfilament Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Neuropeptides/metabolism , Nuclear Proteins/metabolism , Animals , Breast Neoplasms/genetics , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydrogen Peroxide/pharmacology , Microfilament Proteins/biosynthesis , Microfilament Proteins/genetics , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , Neuropeptides/biosynthesis , Neuropeptides/genetics , Nitric Oxide Donors/pharmacology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Oxidative Stress , Transfection , Up-Regulation
20.
PLoS One ; 2(7): e641, 2007 Jul 25.
Article in English | MEDLINE | ID: mdl-17653268

ABSTRACT

BACKGROUND: Characterization of intrinsic and extrinsic factors regulating the self-renewal/division and differentiation of stem cells is crucial in determining embryonic stem (ES) cell fate. ES cells differentiate into multiple hematopoietic lineages during embryoid body (EB) formation in vitro, which provides an experimental platform to define the molecular mechanisms controlling germ layer fate determination and tissue formation. METHODS AND FINDINGS: The cannabinoid receptor type 1 (CB1) and cannabinoid receptor type 2 (CB2) are members of the G-protein coupled receptor (GPCR) family, that are activated by endogenous ligands, the endocannabinoids. CB1 receptor expression is abundant in brain while CB2 receptors are mostly expressed in hematopoietic cells. However, the expression and the precise roles of CB1 and CB2 and their cognate ligands in ES cells are not known. We observed significant induction of CB1 and CB2 cannabinoid receptors during the hematopoietic differentiation of murine ES (mES)-derived embryoid bodies. Furthermore, mES cells as well as ES-derived embryoid bodies at days 7 and 14, expressed endocannabinoids, the ligands for both CB1 and CB2. The CB1 and CB2 antagonists (AM251 and AM630, respectively) induced mES cell death, strongly suggesting that endocannabinoids are involved in the survival of mES cells. Treatment of mES cells with the exogenous cannabinoid ligand Delta(9)-THC resulted in the increased hematopoietic differentiation of mES cells, while addition of AM251 or AM630 blocked embryoid body formation derived from the mES cells. In addition, cannabinoid agonists induced the chemotaxis of ES-derived embryoid bodies, which was specifically inhibited by the CB1 and CB2 antagonists. CONCLUSIONS: This work has not been addressed previously and yields new information on the function of cannabinoid receptors, CB1 and CB2, as components of a novel pathway regulating murine ES cell differentiation. This study provides insights into cannabinoid system involvement in ES cell survival and hematopoietic differentiation.


Subject(s)
Cannabinoid Receptor Modulators/pharmacology , Dronabinol/pharmacology , Embryonic Stem Cells/physiology , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/genetics , Animals , Cannabinoid Receptor Modulators/genetics , Cannabinoid Receptor Modulators/physiology , Cell Differentiation/drug effects , Chemotaxis/drug effects , Embryonic Development/genetics , Embryonic Development/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Gene Expression Regulation , Mice , RNA, Messenger/genetics , Receptor, Cannabinoid, CB1/physiology , Receptor, Cannabinoid, CB2/physiology , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...