201
|
Long-term low-dose exposure of human urothelial cells to sodium arsenite activates lipocalin-2 via promoter hypomethylation. Arch Toxicol 2014; 88:1549-59. [PMID: 24570342 DOI: 10.1007/s00204-014-1214-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 02/11/2014] [Indexed: 01/18/2023]
Abstract
We previously reported that the sustained exposure of human urothelial cells (HUCs) to low-dose sodium arsenite induces changes in the gene expression profile and neoplastic transformation. In this study, we used the HumanMethylation27 BeadChip to analyze genome-wide methylation profiles and 5-aza-2'-deoxycytidine to examine the involvement of promoter methylation in gene expression. Because the expression of lipocalin-2 (LCN2) was highly enhanced by promoter hypomethylation in inorganic arsenic (iAs)-HUCs cells as well as bladder cancer tissues, we further showed that mutations at the binding sequences for NF-κB and C/EBP-α significantly reduced LCN2 promoter activity. By chromatin immunoprecipitation assay, we demonstrated the significantly increased binding of RelA (p65) and NF-κB1 (p50) to the hypomethylated promoter of LCN2 in the iAs-HUCs. Furthermore, we also demonstrated that LCN2 overexpression was crucial for the neoplastic characteristics of the iAs-HUCs, such as enhanced anchorage-independent growth, resistance to serum deprivation and activation of NF-κB signaling. In addition, our results indicated that enhanced NF-κB activity in iAs-HUCs was via LCN2-mediated increase in intracellular iron and reactive oxygen species levels. Taken together, our results show that sustained low-dose arsenic exposure results in epigenetic changes and enhanced oncogenic potential via LCN2 overexpression.
Collapse
|
202
|
Guo P, You JO, Yang J, Jia D, Moses MA, Auguste DT. Inhibiting metastatic breast cancer cell migration via the synergy of targeted, pH-triggered siRNA delivery and chemokine axis blockade. Mol Pharm 2014; 11:755-65. [PMID: 24467226 PMCID: PMC3993942 DOI: 10.1021/mp4004699] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Because breast cancer patient survival inversely correlates with metastasis, we engineered vehicles to inhibit both the C-X-C chemokine receptor type 4 (CXCR4) and lipocalin-2 (Lcn2) mediated migratory pathways. pH-responsive liposomes were designed to protect and trigger the release of Lcn2 siRNA. Liposomes were modified with anti-CXCR4 antibodies to target metastatic breast cancer (MBC) cells and block migration along the CXCR4-CXCL12 axis. This synergistic approach--coupling the CXCR4 axis blockade with Lcn2 silencing--significantly reduced migration in triple-negative human breast cancer cells (88% for MDA-MB-436 and 92% for MDA-MB-231). The results suggested that drug delivery vehicles engineered to attack multiple migratory pathways may effectively slow progression of MBC.
Collapse
Affiliation(s)
- Peng Guo
- Department of Biomedical Engineering, The City College of New York , 160 Convent Avenue, New York, New York 10031, United States
| | | | | | | | | | | |
Collapse
|
203
|
Liao CJ, Li PT, Lee YC, Li SH, Chu ST. Lipocalin 2 induces the epithelial–mesenchymal transition in stressed endometrial epithelial cells: possible correlation with endometriosis development in a mouse model. Reproduction 2014; 147:179-87. [DOI: 10.1530/rep-13-0236] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lipocalin 2 (LCN2) is an induced stressor that promotes the epithelial–mesenchymal transition (EMT). We previously demonstrated that the development of endometriosis in mice correlates with the secretion of LCN2 in the uterus. Here, we sought to clarify the relationship between LCN2 and EMT in endometrial epithelial cells and to determine whether LCN2 plays a role in endometriosis. Antibodies that functionally inhibit LCN2 slowed the growth of ectopic endometrial tissue in a mouse model of endometriosis, suggesting that LCN2 promotes the formation of endometriotic lesions. Using nutrient deprivation as a stressor, LCN2 expression was induced in cultured primary endometrial epithelial cells. As LCN2 levels increased, the cells transitioned from a round to a spindle-like morphology and dispersed. Immunochemical analyses revealed decreased levels of cytokeratin and increased levels of fibronectin in these endometrial cells, adhesive changes that correlate with induction of cell migration and invasion.Lcn2knockdown also indicated that LCN2 promotes EMT and migration of endometrial epithelial cells. Our results suggest that stressful cellular microenvironments cause uterine tissues to secrete LCN2 and that this results in EMT of endometrial epithelial cells, which may correlate with the development of ectopic endometriosis. These findings shed light on the role of LCN2 in the pathology of endometrial disorders.
Collapse
|
204
|
Soliman NA, Zineldeen DH, El-Khadrawy OH. Effect of NUCKS-1 Overexpression on Cytokine Profiling in Obese Women with Breast Cancer. Asian Pac J Cancer Prev 2014; 15:837-45. [DOI: 10.7314/apjcp.2014.15.2.837] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
205
|
Abelson S, Shamai Y, Berger L, Skorecki K, Tzukerman M. Niche-dependent gene expression profile of intratumoral heterogeneous ovarian cancer stem cell populations. PLoS One 2013; 8:e83651. [PMID: 24358304 PMCID: PMC3866276 DOI: 10.1371/journal.pone.0083651] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 11/06/2013] [Indexed: 12/24/2022] Open
Abstract
Intratumoral heterogeneity challenges existing paradigms for anti-cancer therapy. We have previously demonstrated that the human embryonic stem cells (hESC)-derived cellular microenvironment in immunocompromised mice, enables functional distinction of heterogeneous tumor cells, including cells which do not grow into a tumor in a conventional direct tumor xenograft platform. We have identified and characterized six cancer cell subpopulations each clonally expanded from a single cell, derived from human ovarian clear cell carcinoma of a single tumor, to demonstrate striking intratumoral phenotypic heterogeneity that is dynamically dependent on the tumor growth microenvironment. These cancer cell subpopulations, characterized as cancer stem cell subpopulations, faithfully recapitulate the full spectrum of histological phenotypic heterogeneity known for human ovarian clear cell carcinoma. Each of the six subpopulations displays a different level of morphologic and tumorigenic differentiation wherein growth in the hESC-derived microenvironment favors growth of CD44+/aldehyde dehydrogenase positive pockets of self-renewing cells that sustain tumor growth through a process of tumorigenic differentiation into CD44-/aldehyde dehydrogenase negative derivatives. Strikingly, these derivative cells display microenvironment-dependent plasticity with the capacity to restore self-renewal markers and CD44 expression. In the current study, we delineate the distinct gene expression and epigenetic profiles of two such subpopulations, representing extremes of phenotypic heterogeneity in terms of niche-dependent self-renewal and tumorigenic differentiation. By combining Gene Set Enrichment, Gene Ontology and Pathway-focused array analyses with methylation status, we propose a suite of robust differences in tumor self-renewal and differentiation pathways that underlie the striking intratumoral phenotypic heterogeneity which characterize this and other solid tumor malignancies.
Collapse
Affiliation(s)
- Sagi Abelson
- Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Rambam Medical Center, Haifa, Israel
| | - Yeela Shamai
- Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Rambam Medical Center, Haifa, Israel
| | - Liron Berger
- Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Rambam Medical Center, Haifa, Israel
| | - Karl Skorecki
- Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Rambam Medical Center, Haifa, Israel
- Rambam Medical Center, Haifa, Israel
- * E-mail: (MT); (KS)
| | - Maty Tzukerman
- Rambam Medical Center, Haifa, Israel
- * E-mail: (MT); (KS)
| |
Collapse
|
206
|
Clinical implications of aldo-keto reductase family 1 member C3 and its relationship with lipocalin 2 in cancer of the uterine cervix. Gynecol Oncol 2013; 132:474-82. [PMID: 24316309 DOI: 10.1016/j.ygyno.2013.11.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 11/21/2013] [Accepted: 11/26/2013] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Over-expression of the aldo-keto reductase family 1 member C3 (AKR1C3) has been demonstrated in many human cancers. Lipocalin 2 (LCN2) is reported to inhibit cervical cancer metastasis but little is known regarding its relationship with AKR1C3 in the development and progression of uterine cervical cancer. This study aimed to investigate the involvement of AKR1C3 and its relationship with LCN2 in cervical cancer. METHODS The roles of AKR1C3 and LCN2 were investigated using the lentivirus shRNA system in SiHa and Caski cervical cancer cells. LCN2 and matrix metalloproteinase-2 (MMP-2) promoters were constructed to demonstrate transcriptional regulation by shAKR1C3 and shLCN2, respectively. The influences of metastatic phenotypes were analyzed by wound healing, Boyden chamber, and immunofluorescence assays. The activity of MMP-2 was determined by zymography assay. The impacts of AKR1C3 and LCN2 on patient prognosis were evaluated using tissue microarrays by Cox regression and Kaplan-Meier models. RESULTS Silencing of the AKR1C3 gene increased the expression of LCN2 and decreased the migratory and invasive abilities and changed the cytoskeleton of cervical cancer cells. When AKR1C3 was over-expressed, it decreased LCN2 promoter activity and LCN2 expression and increased cell migration. The mRNA level and enzyme activity of MMP-2 increased in silenced LCN2 cells. Positive AKR1C3 and negative LCN2 were correlated with higher recurrence and poorer survival of cervical cancer patients. CONCLUSIONS Silencing of AKR1C3 increases LCN2 expression and inhibits metastasis in cervical cancer. Both AKR1C3 and LCN2 serve as molecular targets for cancer therapy to improve the clinical outcome of cervical cancer patients.
Collapse
|
207
|
Cheng G, Sun X, Wang J, Xiao G, Wang X, Fan X, Zu L, Hao M, Qu Q, Mao Y, Xue Y, Wang J. HIC1 silencing in triple-negative breast cancer drives progression through misregulation of LCN2. Cancer Res 2013; 74:862-72. [PMID: 24295734 DOI: 10.1158/0008-5472.can-13-2420] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The tumor suppressor gene HIC1 is frequently deleted or epigenetically silenced in human cancer, where its restoration may improve cancer prognosis. Here, we report results illuminating how HIC1 silencing alters effect or signals in triple-negative breast cancer (TNBC), which are crucial for its pathogenesis. HIC1 expression was silenced only in TNBC compared with other molecular subtypes of breast cancer. Restoring HIC1 expression in TNBC cells reduced cell migration, invasion, and metastasis, whereas RNAi-mediated silencing of HIC1 in untransformed human breast cells increased their invasive capabilities. Mechanistic investigations identified the small-secreted protein lipocalin-2 (LCN2), as a critical downstream target of HIC1 in TNBC cells. Elevating LCN2 expression in cells expressing HIC1 partially rescued its suppression of cell invasion and metastasis. Notably, autocrine secretion of LCN2 induced by loss of HIC1 activated the AKT pathway through the neutrophil gelatinase-associated lipocalin receptor, which is associated with TNBC progression. Taken together, our findings revealed that the HIC1-LCN2 axis may serve as a subtype-specific prognostic biomarker, providing an appealing candidate target for TNBC therapy.
Collapse
Affiliation(s)
- Guangcun Cheng
- Authors' Affiliations: Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine; and Comprehensive Breast Health Center, Rui Jin Hospital, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
208
|
Lipocalin2 enhances the matrix metalloproteinase-9 activity and invasion of extravillous trophoblasts under hypoxia. Placenta 2013; 34:1036-43. [DOI: 10.1016/j.placenta.2013.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 07/24/2013] [Accepted: 08/06/2013] [Indexed: 11/21/2022]
|
209
|
Neutrophil Gelatinase-Associated Lipocalin and Its Influence on Tumor Progression in Systemic Malignancies. World J Surg 2013; 37:2727-8. [DOI: 10.1007/s00268-013-2131-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
210
|
El-Gazzar A, Cai X, Reeves RS, Dai Z, Caballero-Benitez A, McDonald DL, Vazquez J, Gooley TA, Sale GE, Spies T, Groh V. Effects on tumor development and metastatic dissemination by the NKG2D lymphocyte receptor expressed on cancer cells. Oncogene 2013; 33:4932-40. [PMID: 24141776 PMCID: PMC3994187 DOI: 10.1038/onc.2013.435] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 09/13/2013] [Indexed: 12/16/2022]
Abstract
The stimulatory NKG2D lymphocyte receptor together with its tumor-associated ligands enable the immune system to recognize and destroy cancer cells. However, with dynamic changes unfolding, cancers exploit NKG2D and its ligands for immune evasion and suppression. Recent findings have added yet another functional dimension wherein cancer cells themselves coopt NKG2D for their own benefit to complement the presence of its ligands for self stimulation of parameters of tumorigenesis. Those findings are here extended to in vivo tumorigenicity testing by employing orthotopic xenotransplant breast cancer models in mice. Using human cancer lines with ectopic NKG2D expression and RNAi-mediated protein depletion among other controls, we show that NKG2D self-stimulation has tumor promoting capacity. NKG2D signals had no notable effects on cancer cell proliferation and survival but acted at the level of angiogenesis, thus promoting tumor growth, tumor cell intravasation and dissemination. NKG2D-mediated effects on tumor initiation may represent another factor in the observed overall enhancement of tumor development. Altogether, these results may impact immunotherapy approaches, which currently do not account for such NKG2D effects in cancer patients and thus could be misdirected as underlying assumptions are incomplete.
Collapse
Affiliation(s)
- A El-Gazzar
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - X Cai
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - R S Reeves
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Z Dai
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - A Caballero-Benitez
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - D L McDonald
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - J Vazquez
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - T A Gooley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - G E Sale
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - T Spies
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - V Groh
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
211
|
Wang YP, Yu GR, Lee MJ, Lee SY, Chu IS, Leem SH, Kim DG. Lipocalin-2 negatively modulates the epithelial-to-mesenchymal transition in hepatocellular carcinoma through the epidermal growth factor (TGF-beta1)/Lcn2/Twist1 pathway. Hepatology 2013; 58:1349-61. [PMID: 23696034 DOI: 10.1002/hep.26467] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 04/12/2013] [Indexed: 12/18/2022]
Abstract
UNLABELLED Lipocalin-2 (Lcn2) is preferentially expressed in hepatocellular carcinoma (HCC). However, the functional role of Lcn2 in HCC progression is still poorly understood, particularly with respect to its involvement in invasion and metastasis. The purpose of this study was to investigate whether Lcn2 is associated with the epithelial-mesenchymal transition (EMT) in HCC and to elucidate the underlying signaling pathway(s). Lcn2 was preferentially expressed in well-differentiated HCC versus liver cirrhosis tissues, and its expression was positively correlated with the stage of HCC. The characteristics of EMT were reversed by adenoviral transduction of Lcn2 into SH-J1 cells, including the down-regulation of N-cadherin, vimentin, alpha-smooth muscle actin, and fibronectin, and the concomitant up-regulation of CK8, CK18, and desmoplakin I/II. Knockdown of Lcn2 by short hairpin RNA (shRNA) in HKK-2 cells expressing high levels of Lcn2 was associated with EMT. Epidermal growth factor (EGF) or transforming growth factor beta1 (TGF-β1) treatment resulted in down-regulation of Lcn2, accompanied by an increase in Twist1 expression and EMT in HCC cells. Stable Lcn2 expression in SH-J1 cells reduced Twist1 expression, inhibited cell proliferation and invasion in vitro, and suppressed tumor growth and metastasis in a mouse model. Furthermore, EGF or TGF-β1 treatment barely changed EMT marker expression in SH-J1 cells ectopically expressing Lcn2. Ectopic expression of Twist1 induced EMT marker expression even in cells expressing Lcn2, indicating that Lcn2 functions downstream of growth factors and upstream of Twist1. CONCLUSION Together, our findings indicate that Lcn2 can negatively modulate the EMT in HCC cells through an EGF (or TGF-β1)/Lcn2/Twist1 pathway. Thus, Lcn2 may be a candidate metastasis suppressor and a potential therapeutic target in HCC.
Collapse
Affiliation(s)
- Yun-Peng Wang
- Division of Gastroenterology and Hepatology, Research Institute of Clinical Medicine, Department of Internal Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Jeonbuk, South Korea
| | | | | | | | | | | | | |
Collapse
|
212
|
Chen W, Ayala-Orozco C, Biswal NC, Perez-Torres C, Bartels M, Bardhan R, Stinnet G, Liu XD, Ji B, Deorukhkar A, Brown LV, Guha S, Pautler RG, Krishnan S, Halas NJ, Joshi A. Targeting pancreatic cancer with magneto-fluorescent theranostic gold nanoshells. Nanomedicine (Lond) 2013; 9:1209-1222. [PMID: 24063415 PMCID: PMC4051872 DOI: 10.2217/nnm.13.84] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Indexed: 02/08/2023] Open
Abstract
AIM We report a magneto-fluorescent theranostic nanocomplex targeted to neutrophil gelatinase-associated lipocalin (NGAL) for imaging and therapy of pancreatic cancer. MATERIALS & METHODS Gold nanoshells resonant at 810 nm were encapsulated in silica epilayers doped with iron oxide and the near-infrared (NIR) dye indocyanine green, resulting in theranostic gold nanoshells (TGNS), which were subsequently conjugated with antibodies targeting NGAL in AsPC-1-derived xenografts in nude mice. RESULTS Anti-NGAL-conjugated TGNS specifically targeted pancreatic cancer cells in vitro and in vivo providing contrast for both NIR fluorescence and T2-weighted MRI with higher tumor contrast than can be obtained using long-circulating, but nontargeted, PEGylated nanoparticles. The nanocomplexes also enabled highly specific cancer cell death via NIR photothermal therapy in vitro. CONCLUSION TGNS with embedded NIR and magnetic resonance contrasts can be specifically targeted to pancreatic cancer cells with expression of early disease marker NGAL, and enable molecularly targeted imaging and photothermal therapy.
Collapse
Affiliation(s)
- Wenxue Chen
- Division of Molecular Imaging, Department of Radiology, Baylor College of Medicine, Mail: BCM 360, One Baylor Plaza, Houston, TX 77030 (USA)
- Department of Diagnostic Imaging, The Fourth Hospital of Hebei Medical University/Hebei Province Tumor Hospital, 12 Jiankang Road, Shijiazhuang, Hebei Province 050011 (China)
| | | | - Nrusingh C. Biswal
- Division of Molecular Imaging, Department of Radiology, Baylor College of Medicine, Mail: BCM 360, One Baylor Plaza, Houston, TX 77030 (USA)
| | - Carlos Perez-Torres
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Marc Bartels
- Division of Molecular Imaging, Department of Radiology, Baylor College of Medicine, Mail: BCM 360, One Baylor Plaza, Houston, TX 77030 (USA)
| | - Rizia Bardhan
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX 77005
| | - Gary Stinnet
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Xian-De Liu
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
| | - Baoan Ji
- Department of Biochemistry and Molecular Biology, Guggenheim 13-21B. Mayo Clinic, 200 First Street S.W., Rochester, MN 55905
| | - Amit Deorukhkar
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
| | - Lisa V. Brown
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX 77005
| | - Sushovan Guha
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
| | - Robia G. Pautler
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Sunil Krishnan
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
| | - Naomi J Halas
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX 77005
- Department of Electrical and Computer Engineering, Rice University, 6100 Main St, Houston, TX 77005
| | - Amit Joshi
- Division of Molecular Imaging, Department of Radiology, Baylor College of Medicine, Mail: BCM 360, One Baylor Plaza, Houston, TX 77030 (USA)
| |
Collapse
|
213
|
McPherson CA, Merrick BA, Harry GJ. In vivo molecular markers for pro-inflammatory cytokine M1 stage and resident microglia in trimethyltin-induced hippocampal injury. Neurotox Res 2013; 25:45-56. [PMID: 24002884 DOI: 10.1007/s12640-013-9422-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 08/13/2013] [Accepted: 08/20/2013] [Indexed: 12/18/2022]
Abstract
Microglia polarization to the classical M1 activation state is characterized by elevated pro-inflammatory cytokines; however, a full profile has not been generated in the early stages of a sterile inflammatory response recruiting only resident microglia. We characterized the initial M1 state in a hippocampal injury model dependent upon tumor necrosis factor (TNF) receptor signaling for dentate granule cell death. Twenty-one-day-old CD1 male mice were injected with trimethyltin (TMT 2.3 mg/kg, i.p.) and the hippocampus was examined at an early stage (24-h post-dosing) of neuronal death. Glia activation was assessed using a custom quantitative nuclease protection assay. We report elevated mRNA levels for glia response such as ionizing calcium-binding adapter molecule-1 and glial fibrillary acidic protein (Gfap); Fas, hypoxia inducible factor alpha, complement component 1qb, TNF-related genes (Tnf, Tnfaip3, Tnfrsfla); interleukin-1 alpha, Cd44, chemokine (C-C motif) ligand (Ccl)2, Cc14, integrin alpha M, lipocalin (Lcn2), and secreted phosphoprotein 1 (Spp1). These changes occurred in the absence of changes in matrix metalloproteinase 9 and 12, neural cell adhesion molecule, metabotropic glutamate receptor (Grm)3, and Ly6/neurotoxin 1 (Lynx1), as well as, a decrease in neurotrophin 3, glutamate receptor subunit epsilon (Grin)-2b, and neurotrophic tyrosine kinase receptor, type 3. The M2 anti-inflammatory marker, transforming growth factor beta-1 (Tgfb1) was elevated. mRNAs associated with early stage of injury-induced neurogenesis including fibroblast growth factor 21 and Mki67 were elevated. In the "non-injured" temporal cortex receiving projections from the hippocampus, Lynx1, Grm3, and Grin2b were decreased and Gfap increased. Formalin fixed-paraffin-embedded tissue did not generate a comparable profile.
Collapse
Affiliation(s)
- C A McPherson
- Neurotoxicology Group, Division of National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, P.O. Box 12233, MD E1-07, Research Triangle Park, NC, 27709, USA
| | | | | |
Collapse
|
214
|
Chien MH, Ying TH, Yang SF, Yu JK, Hsu CW, Hsieh SC, Hsieh YH. Lipocalin-2 induces apoptosis in human hepatocellular carcinoma cells through activation of mitochondria pathways. Cell Biochem Biophys 2013; 64:177-86. [PMID: 22707293 DOI: 10.1007/s12013-012-9370-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lipocalin 2 (LCN2) is a secreted, iron-binding glycoprotein that is abnormally expressed in some malignant human cancers. However, the roles of LCN2 in hepatocellular carcinoma (HCC) cells are unknown. In this study, we suggested the LCN2 and LCN2R were weak detected in the HCC cell lines, LCN2 and LCN2R were found to be down-regulated in tumor tissues in 16 HCC patients. MTT, DAPI, TUNEL, and flow cytometry analyses revealed that LCN2 overexpression dramatically inhibited cell viability, induced apoptosis features of cell-cycle arrest in sub-G1 phase, in DNA fragmentation, and in condensation of chromatin in Huh-7 and SK-Hep-1 cells. Western blots were used to detect the activation of caspase, pro-apoptosis, and anti-apoptosis protein expression in overexpress-LCN2 HCC cells. LCN2-induced apoptosis was characterized by cleavage of caspase-9, -8, -3, and PARP protein, and a reduction in the mitochondrial membrane potential (MMP). Furthermore, LCN2 also enhanced the down-regulated Bcl-2 and up-regulated the expression of Bax. In addition, our experiments with caspase inhibitors LEHD-FMK and IETD-FMK prevent LCN2-induced apoptosis. We also demonstrated that treatment of overexpress-LCN2 HCC cells with the LCN2 neutralized antibody also significantly attenuated LCN2-induced cell apoptosis. These findings indicate that LCN2 overexpression can effectively induce apoptosis of HCC cells and may be used as a potent therapy against human HCC.
Collapse
|
215
|
Frankfurt O, Platanias LC. Implications of lipocalin 2 expression in acute myeloid leukemia. Leuk Lymphoma 2013; 54:1573-4. [DOI: 10.3109/10428194.2013.786073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
216
|
Stewart CA, Wang Y, Bonilla-Claudio M, Martin JF, Gonzalez G, Taketo MM, Behringer RR. CTNNB1 in mesenchyme regulates epithelial cell differentiation during Müllerian duct and postnatal uterine development. Mol Endocrinol 2013; 27:1442-54. [PMID: 23904126 DOI: 10.1210/me.2012-1126] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Müllerian duct differentiation and development into the female reproductive tract is essential for fertility, but mechanisms regulating these processes are poorly understood. WNT signaling is critical for proper development of the female reproductive tract as evident by the phenotypes of Wnt4, Wnt5a, Wnt7a, and β-catenin (Ctnnb1) mutant mice. Here we extend these findings by determining the effects of constitutive CTNNB1 activation within the mesenchyme of the developing Müllerian duct and its differentiated derivatives. This was accomplished by crossing Amhr2-Cre knock-in mice with Ctnnb1 exon (ex) 3(f/f) mice. Amhr2-Cre(Δ/+); Ctnnb1 ex3(f/+) females did not form an oviduct, had smaller uteri, endometrial gland defects, and were infertile. At the cellular level, stabilization of CTNNB1 in the mesenchyme caused alterations within the epithelium, including less proliferation, delayed uterine gland formation, and induction of an epithelial-mesenchymal transition (EMT) event. This EMT event is observed before birth and is complete within 5 days after birth. Misexpression of estrogen receptor α in the epithelia correlated with the EMT before birth, but not after. These studies indicate that regulated CTNNB1 in mesenchyme is important for epithelial cell differentiation during female reproductive tract development.
Collapse
Affiliation(s)
- C Allison Stewart
- Department of Genetics, University of Texas MD Anderson Cancer Center, University of Texas Graduate School of Biomedical Sciences, Houston, Texas 77030, USA
| | | | | | | | | | | | | |
Collapse
|
217
|
Expression of neutrophil gelatinase-associated lipocalin in colorectal neoplastic progression: a marker of malignant potential? Br J Cancer 2013; 108:2537-41. [PMID: 23736029 PMCID: PMC3694245 DOI: 10.1038/bjc.2013.264] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background: Neutrophil gelatinase-associated lipocalin (NGAL) has a diverse functional repertoire, involved in the innate immune response as well as cell growth and differentiation. Expression has been linked to malignant disease development and progression. Methods: Neutrophil gelatinase-associated lipocalin expression was assessed immunohistochemically in 98 colorectal neoplastic lesions (52 cancer polyps (CaPs) and 46 sporadic adenoma/adjacent normal mucosa paired specimens) to investigate association with adenoma progression and early colorectal carcinogenesis. Results: Within CaPs, all adenomatous and carcinomatous epithelium expressed NGAL, with 92% (43 out of 47) and 58% (19 out of 33) epithelial positivity, respectively, as well as positive stromal cell expression. This was significantly increased compared with normal mucosal epithelium (P=0.0001). Neutrophil gelatinase-associated lipocalin positivity was also identified in sporadic low-grade adenomas, in both the epithelial and stromal compartments as compared with adjacent normal mucosa (P=0.0001 and 0.0002), and this increased along with adenoma size >1 cm (P=0.03). Conclusion: Neutrophil gelatinase-associated lipocalin is expressed by the majority of human neoplastic colorectal lesions. This phenotypic switch occurs at an early stage in neoplastic progression with clear differential expression between normal mucosa and adenomatous polyps, rather than further downstream in disease progression at the adenoma–carcinoma transformation. Thus, NGAL expression is not a useful biomarker for determining disease progression from adenomatous to malignant colorectal neoplasia.
Collapse
|
218
|
Kim DW, Walker RL, Meltzer PS, Cheng SY. Complex temporal changes in TGFβ oncogenic signaling drive thyroid carcinogenesis in a mouse model. Carcinogenesis 2013; 34:2389-400. [PMID: 23698635 DOI: 10.1093/carcin/bgt175] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Despite recent advances, understanding of molecular genetic alterations underlying thyroid carcinogenesis remains unclear. One key question is how dynamic temporal changes in global genomic expression affect carcinogenesis as the disease progresses. To address this question, we used a mouse model that spontaneously develops follicular thyroid cancer similar to human cancer (Thrb (PV/PV) mice). Using complementary DNA microarrays, we compared global gene expression profiles of thyroid tumors of Thrb (PV/PV) mice with the age- and gender-matched thyroids of wild-type mice at 3 weeks and at 2, 4, 6 and 14 months. These time points covered the pathological progression from early hyperplasia to capsular invasion, vascular invasion and eventual metastasis. Microarray data indicated that 462 genes were upregulated (Up-cluster genes) and 110 genes were downregulated (Down-cluster genes). Three major expression patterns (trending up, cyclical and spiking up and then down) and two (trending down and cyclical) were apparent in the Up-cluster and Down-cluster genes, respectively. Functional clustering of tumor-related genes followed by Ingenuity Pathways Analysis identified the transforming growth factor β (TGF β)-mediated network as key signaling pathways. Further functional analyses showed sustained activation of TGFβ receptor-pSMAD2/3 signaling, leading to decreased expression of E-cadherin and increased expression of fibronectin, vimentin, collagens and laminins. These TGFβ-induced changes facilitated epithelial-to-mesenchymal transition, which promotes cancer invasion and migration. Thus, complex temporal changes in gene expression patterns drive thyroid cancer progression, and persistent activation of TGFβ-TGFRβII-pSMAD2/3 signaling leads to EMT, thus promoting metastasis. This study provides new understanding of progression and metastatic spread of human thyroid cancer.
Collapse
Affiliation(s)
- Dong Wook Kim
- Gene Regulation Section, Laboratory of Molecular Biology and
| | | | | | | |
Collapse
|
219
|
Wang Y, Zeng T. Neutrophil gelatinase-associated lipocalin protein as a biomarker in the diagnosis of breast cancer: A meta-analysis. Biomed Rep 2013; 1:479-483. [PMID: 24648972 DOI: 10.3892/br.2013.89] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Accepted: 03/12/2013] [Indexed: 02/05/2023] Open
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL) has been thought to play an important role in breast cancer tumorigenesis and progression. Various studies have focused on the association between NGAL and breast cancer. The aim of this meta-analysis was to establish the overall accuracy of the NGAL test in the diagnosis of breast cancer. A comprehensive search of the literature was conducted using PubMed, OVID, ScienceDirect and the China National Knowledge Infrastructure (CNKI) databases, and our screening covered all published papers until November 2012. The relevant papers were selected according to stringent inclusion criteria. Essential data were abstracted from the included papers and further analysed by a systematic meta-analysis. The present meta-analysis included four study papers. The summary estimate was 64% (95% CI, 0.59-0.69) for sensitivity and 87% (95% CI, 0.81-0.92) for specificity. The positive likelihood (PLR), negative likelihood (NLR) and diagnostic odds (DOR) ratios were 5.63 (95% CI, 3.63-8.74), 0.32 (95% CI, 0.14-0.71) and 18.02 (95% CI, 9.84-32.98), respectively. The area under the summary ROC curve (AUC) for the diagnosis of breast cancer was 0.9008. Thus, NGAL is a potential biomarker for the diagnosis of breast cancer.
Collapse
Affiliation(s)
- Yu Wang
- Laboratory of Molecular Diagnosis of Cancer, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041
| | - Tingting Zeng
- Adverse Drug Reaction Monitoring Center of Chengdu, High-Tech District, Chengdu, Sichuan 610042, P.R. China
| |
Collapse
|
220
|
Chappell WH, Abrams SL, Montalto G, Cervello M, Martelli AM, Candido S, Libra M, Polesel J, Talamini R, Arlinghaus R, Steelman LS, McCubrey JA. Effects of ectopic expression of NGAL on doxorubicin sensitivity. Oncotarget 2013; 3:1236-45. [PMID: 23100449 PMCID: PMC3717946 DOI: 10.18632/oncotarget.691] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL, a.k.a Lnc2) is a member of the lipocalin family which has diverse roles including stabilizing matrix metalloproteinase-9 from auto-degradation and as siderocalins which are important in the transport of iron. NGAL also has important biological functions involved in immunity and inflammation as well as responses to kidney damage. NGAL expression has also been associated with certain neoplasia and is important in the metastasis of breast cancer. Many advanced cancer patients have elevated levels of NGAL in their urine and it has been proposed that NGAL may be a prognostic indicator for certain cancers (e.g. breast, brain, and others). NGAL expression is detected in response to various chemotherapeutic drugs including doxorubicin and docetaxel. We were interested in the roles of NGAL expression in cancer and whether it is associated with chemotherapeutic drug resistance. In the present study, we investigated whether increased NGAL expression led to resistance to the chemotherapeutic drug doxorubicin in normal breast epithelial cells (MCF-10A), breast cancer cells (MCF-7), and colorectal cancer cells (HT-29). We infected the various cell lines with a retrovirus encoding NGAL which we constructed. Increased NGAL expression was readily detected in the NGAL-infected cells but not the empty vector-infected cells. However, increased NGAL expression did not alter the sensitivity of the cells to the chemotherapeutic drug doxorubicin. Thus, although NGAL expression is often detected after chemotherapeutic drug treatment, it by itself, does not lead to doxorubicin resistance.
Collapse
Affiliation(s)
- William H Chappell
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
221
|
Lai HS, Wu YM, Lai SL, Lin WH. Lipocalin-2 gene expression during liver regeneration after partial hepatectomy in rats. Int J Surg 2013; 11:314-8. [PMID: 23481292 DOI: 10.1016/j.ijsu.2013.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 02/08/2013] [Accepted: 02/14/2013] [Indexed: 01/20/2023]
Abstract
BACKGROUND Lipocalin-2 (Lcn2) is related to cell proliferation. We studied Lcn2 gene expression during liver regeneration after partial hepatectomy (PH). METHODS Male Wistar rats were sacrificed before and 2, 4, 6, 12, 24, 72 h, 7 days after 70% or 40% PH. The remnant liver weight/body weight (RLW/BW) ratio, Lcn2 gene and mRNA expression in the remnant livers were measured. Hepatocytes and nonparenchymal cells were isolated from the remnant livers. Expression of Lcn2 related protein was detected by Western blot. RESULTS The RLW/BW ratio increased to nearly 90% of the original liver 72 h after PH. Lcn2 gene expression showed upward curves from 4 to 72 h after PH in both 70% and 40% PH rats and peaked at 12 h (8 times vs 0 h). Lcn2 mRNA expression showed parallel upward curves from 2 to 72 h. The peak was significantly higher in 70% PH rats (2(7) times vs 0 h) than in 40% PH rats (2(5) times vs 0 h) 12 h after PH (p < 0.05). Lcn2 related protein in the isolated liver cells was markedly enhanced 24 h after PH, more in hepatocytes than in nonparenchymal cells. CONCLUSION The expressions of Lcn2 gene and mRNA, and its related protein increased markedly after PH. Lcn2 might be important in the genetic regulation of liver regeneration after PH.
Collapse
Affiliation(s)
- Hong-Shiee Lai
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | | | | | | |
Collapse
|
222
|
Chappell WH, Abrams SL, Stadelman KM, LaHair MM, Franklin RA, Cocco L, Evangelisti C, Chiarini F, Martelli AM, Steelman LS, McCubrey JA. Increased NGAL (Lnc2) expression after chemotherapeutic drug treatment. Adv Biol Regul 2013; 53:146-155. [PMID: 23073564 DOI: 10.1016/j.jbior.2012.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 09/06/2012] [Indexed: 06/01/2023]
Affiliation(s)
- William H Chappell
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, 600 Moye Blvd., Greenville, NC 27834, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
223
|
Kaur H, Mao S, Li Q, Sameni M, Krawetz SA, Sloane BF, Mattingly RR. RNA-Seq of human breast ductal carcinoma in situ models reveals aldehyde dehydrogenase isoform 5A1 as a novel potential target. PLoS One 2012; 7:e50249. [PMID: 23236365 PMCID: PMC3516505 DOI: 10.1371/journal.pone.0050249] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 10/22/2012] [Indexed: 01/16/2023] Open
Abstract
Breast ductal carcinoma in situ (DCIS) is being found in great numbers of women due to the widespread use of mammography. To increase knowledge of DCIS, we determined the expression changes that are common among three DCIS models (MCF10.DCIS, SUM102 and SUM225) compared to the MCF10A model of non-tumorigenic mammary epithelial cells in three dimensional (3D) overlay culture with reconstituted basement membrane (rBM). Extracted mRNA was subjected to 76 cycles of deep sequencing (RNA-Seq) using Illumina Genome Analyzer GAIIx. Analysis of RNA-Seq results showed 295 consistently differentially expressed transcripts in the DCIS models. These differentially expressed genes encode proteins that are associated with a number of signaling pathways such as integrin, fibroblast growth factor and TGFβ signaling, show association with cell-cell signaling, cell-cell adhesion and cell proliferation, and have a notable bias toward localization in the extracellular and plasma membrane compartments. RNA-Seq data was validated by quantitative real-time PCR of selected differentially expressed genes. Aldehyde dehydrogenase 5A1 (ALDH5A1) which is an enzyme that is involved in mitochondrial glutamate metabolism, was over-expressed in all three DCIS models at both the mRNA and protein levels. Disulfiram and valproic acid are known to inhibit ALDH5A1 and are safe for chronic use in humans for other disorders. Both of these drugs significantly inhibited net proliferation of the DCIS 3D rBM overlay models, but had minimal effect on MCF10A 3D rBM overlay models. These results suggest that ALDH5A1 may play an important role in DCIS and potentially serve as a novel molecular therapeutic target.
Collapse
Affiliation(s)
- Hitchintan Kaur
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Shihong Mao
- Center for Molecular Medicine and Genetics, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Quanwen Li
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, United States of America
| | - Mansoureh Sameni
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Stephen A. Krawetz
- Center for Molecular Medicine and Genetics, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, United States of America
| | - Bonnie F. Sloane
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, United States of America
| | - Raymond R. Mattingly
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
224
|
Labbus K, Henning M, Borkham-Kamphorst E, Geisler C, Berger T, Mak TW, Knüchel R, Meyer HE, Weiskirchen R, Henkel C. Proteomic profiling in Lipocalin 2 deficient mice under normal and inflammatory conditions. J Proteomics 2012; 78:188-96. [PMID: 23219901 DOI: 10.1016/j.jprot.2012.11.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 10/31/2012] [Accepted: 11/12/2012] [Indexed: 01/17/2023]
Abstract
Lipocalin 2 (LCN2) belongs to the superfamily of lipocalins which represent a group of small secreted proteins classified as extracellular transport proteins expressed in many tissues. LCN2 is strongly increased in experimental models of acute and chronic liver injuries. To investigate the function of LCN2 in normal liver homeostasis and under conditions of inflammatory liver injury, we comparatively analyzed hepatic extracts taken from Lcn2-deficient and wild type mice under basal conditions and after stimulation with lipopolysaccharides. Liver was chemically and mechanically lysed and extracts were subjected to 2-D-DIGE after minimal labeling (G200 and G300 dyes) using an appropriate internal standard (G100). Afterwards MALDI TOF MS and MS/MS were used to identify differentially expressed proteins. Proteins that were identified to be differentially expressed include for example the chloride intracellular channel protein 4 (CLIC4), aminoacylase 1 and transketolase. The altered expression of respective genes was confirmed by Western blot analysis and further validated by quantitative real time PCR. Altogether, the complex expression alterations in mice lacking LCN2 under normal conditions and after exposure to inflammatory stimuli reveal that LCN2 has essential function in liver homeostasis and in the onset of inflammatory responses in which LCN2 expression dramatically increases.
Collapse
Affiliation(s)
- Kirsten Labbus
- Institute of Pathology, RWTH University Hospital Aachen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
225
|
Jang E, Lee S, Kim J, Kim J, Seo J, Lee W, Mori K, Nakao K, Suk K. Secreted protein lipocalin‐2 promotes microglial M1 polarization. FASEB J 2012. [DOI: 10.1096/fj.12-222257] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Eunha Jang
- Department of PharmacologyBrain Science and Engineering InstituteCell and Matrix Research Institute (CMRI)Kyungpook National University School of MedicineDaeguKorea
| | - Shinrye Lee
- Department of PharmacologyBrain Science and Engineering InstituteCell and Matrix Research Institute (CMRI)Kyungpook National University School of MedicineDaeguKorea
| | - Jong‐Heon Kim
- Department of PharmacologyBrain Science and Engineering InstituteCell and Matrix Research Institute (CMRI)Kyungpook National University School of MedicineDaeguKorea
| | - Jae‐Hong Kim
- Department of PharmacologyBrain Science and Engineering InstituteCell and Matrix Research Institute (CMRI)Kyungpook National University School of MedicineDaeguKorea
| | - Jung‐Wan Seo
- Department of PharmacologyBrain Science and Engineering InstituteCell and Matrix Research Institute (CMRI)Kyungpook National University School of MedicineDaeguKorea
| | - Won‐Ha Lee
- School of Life Sciences and BiotechnologyKyungpook National UniversityDaeguKorea
| | - Kiyoshi Mori
- Department of Medicine and Clinical ScienceKyoto University Graduate School of MedicineKyotoJapan
| | - Kazuwa Nakao
- Department of Medicine and Clinical ScienceKyoto University Graduate School of MedicineKyotoJapan
| | - Kyoungho Suk
- Department of PharmacologyBrain Science and Engineering InstituteCell and Matrix Research Institute (CMRI)Kyungpook National University School of MedicineDaeguKorea
| |
Collapse
|
226
|
Chappell WH, Abrams SL, Franklin RA, LaHair MM, Montalto G, Cervello M, Martelli AM, Nicoletti F, Candido S, Libra M, Polesel J, Talamini R, Milella M, Tafuri A, Steelman LS, McCubrey JA. Ectopic NGAL expression can alter sensitivity of breast cancer cells to EGFR, Bcl-2, CaM-K inhibitors and the plant natural product berberine. Cell Cycle 2012; 11:4447-61. [PMID: 23159854 PMCID: PMC3552927 DOI: 10.4161/cc.22786] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL, a.k.a Lnc2) is a member of the lipocalin family and has diverse roles. NGAL can stabilize matrix metalloproteinase-9 from autodegradation. NGAL is considered as a siderocalin that is important in the transport of iron. NGAL expression has also been associated with certain neoplasias and is implicated in the metastasis of breast cancer. In a previous study, we examined whether ectopic NGAL expression would alter the sensitivity of breast epithelial, breast and colorectal cancer cells to the effects of the chemotherapeutic drug doxorubicin. While abundant NGAL expression was detected in all the cells infected with a retrovirus encoding NGAL, this expression did not alter the sensitivity of these cells to doxorubicin as compared with empty vector-transduced cells. We were also interested in determining the effects of ectopic NGAL expression on the sensitivity to small-molecule inhibitors targeting key signaling molecules. Ectopic NGAL expression increased the sensitivity of MCF-7 breast cancer cells to EGFR, Bcl-2 and calmodulin kinase inhibitors as well as the natural plant product berberine. Furthermore, when suboptimal concentrations of certain inhibitors were combined with doxorubicin, a reduction in the doxorubicin IC 50 was frequently observed. An exception was observed when doxorubicin was combined with rapamycin, as doxorubicin suppressed the sensitivity of the NGAL-transduced MCF-7 cells to rapamycin when compared with the empty vector controls. In contrast, changes in the sensitivities of the NGAL-transduced HT-29 colorectal cancer cell line and the breast epithelial MCF-10A cell line were not detected compared with empty vector-transduced cells. Doxorubicin-resistant MCF-7/Dox (R) cells were examined in these experiments as a control drug-resistant line; it displayed increased sensitivity to EGFR and Bcl-2 inhibitors compared with empty vector transduced MCF-7 cells. These results indicate that NGAL expression can alter the sensitivity of certain cancer cells to small-molecule inhibitors, suggesting that patients whose tumors exhibit elevated NGAL expression or have become drug-resistant may display altered responses to certain small-molecule inhibitors.
Collapse
Affiliation(s)
- William H. Chappell
- Department of Microbiology & Immunology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| | - Stephen L. Abrams
- Department of Microbiology & Immunology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| | - Richard A. Franklin
- Department of Microbiology & Immunology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| | - Michelle M. LaHair
- Department of Microbiology & Immunology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| | - Giuseppe Montalto
- Department of Internal Medicine and Specialties; University of Palermo; Palermo, Italy
- Consiglio Nazionale delle Ricerche; Istituto di Biomedicina e Immunologia Molecolare “Alberto Monroy”; Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche; Istituto di Biomedicina e Immunologia Molecolare “Alberto Monroy”; Palermo, Italy
| | - Alberto M. Martelli
- Department of Biomedical and Neuromotor Sciences; Università di Bologna; Bologna, Italy
- Institute of Molecular Genetics; National Research Council-Rizzoli Orthopedic Institute; Bologna, Italy
| | | | - Saverio Candido
- Department of Bio-Medical Sciences; University of Catania; Catania, Italy
| | - Massimo Libra
- Department of Bio-Medical Sciences; University of Catania; Catania, Italy
| | - Jerry Polesel
- Unit of Epidemiology and Biostatistics; Centro di Riferimento Oncologico; IRCCS; Aviano, Italy
| | - Renato Talamini
- Unit of Epidemiology and Biostatistics; Centro di Riferimento Oncologico; IRCCS; Aviano, Italy
| | | | - Agostino Tafuri
- Department of Cellular Biotechnology and Hematology; University of Rome, Sapienza; Rome, Italy
| | - Linda S. Steelman
- Department of Microbiology & Immunology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| | - James A. McCubrey
- Department of Microbiology & Immunology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| |
Collapse
|
227
|
Activation of the unfolded protein response bypasses trastuzumab-mediated inhibition of the PI-3K pathway. Cancer Lett 2012. [PMID: 23200669 DOI: 10.1016/j.canlet.2012.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
HER2-positive breast cancer initially responds to trastuzumab treatment, but over time, resistance develops and rapid cancer progression occurs, for which various explanations have been proposed. Here we tested the hypothesis that induction of the unfolded protein response (UPR) could override HER2 inhibition by trastuzumab, leading to the re-activation of growth signaling and the activation of the downstream target Lipocalin 2 (LCN2). Trastuzumab significantly inhibited the basal expression of LCN2 in HER2 (+) SKBr3 human breast cancer cells. The induction of the UPR completely abrogated trastuzumab-mediated LCN2 downregulation, and, in fact caused an increase in transcription and secretion of LCN2 over baseline. Reduction of the UPR using 4-phenyl butyric acid (PBA) a chemical chaperone that ameliorates ER stress, restored trastuzumab-mediated inhibition. Inhibition of the PI3K/AKT signaling pathway in trastuzumab-treated/UPR-induced SKBr3 cells partially reduced the upregulation of LCN2. These results suggest that the UPR is a possible way to override the effect of trastuzumab in HER2(+) cancer cells.
Collapse
|
228
|
CCL2 is critical for immunosuppression to promote cancer metastasis. Clin Exp Metastasis 2012; 30:393-405. [DOI: 10.1007/s10585-012-9545-6] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 10/24/2012] [Indexed: 10/27/2022]
|
229
|
Germann S, Gratadou L, Zonta E, Dardenne E, Gaudineau B, Fougère M, Samaan S, Dutertre M, Jauliac S, Auboeuf D. Dual role of the ddx5/ddx17 RNA helicases in the control of the pro-migratory NFAT5 transcription factor. Oncogene 2012; 31:4536-4549. [PMID: 22266867 DOI: 10.1038/onc.2011.618] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 11/01/2011] [Accepted: 11/28/2011] [Indexed: 02/07/2023]
Abstract
Ddx5 and ddx17 are two highly related RNA helicases involved in both transcription and splicing. These proteins coactivate transcription factors involved in cancer such as the estrogen receptor alpha, p53 and beta-catenin. Ddx5 and ddx17 are part of the splicing machinery and can modulate alternative splicing, the main mechanism increasing the proteome diversity. Alternative splicing also has a role in gene expression level regulation when it is coupled to the nonsense-mediated mRNA decay (NMD) pathway. In this work, we report that ddx5 and ddx17 have a dual role in the control of the pro-migratory NFAT5 transcription factor. First, ddx5 and ddx17 act as transcriptional coactivators of NFAT5 and are required for activating NFAT5 target genes involved in tumor cell migration. Second, at the splicing level, ddx5 and ddx17 increase the inclusion of NFAT5 exon 5. As exon 5 contains a pre-mature translation termination codon, its inclusion leads to the regulation of NFAT5 mRNAs by the NMD pathway and to a decrease in NFAT5 protein level. Therefore, we demonstrated for the first time that a transcriptional coregulator can simultaneously regulate the transcriptional activity and alternative splicing of a transcription factor. This dual regulation, where ddx5 and ddx17 enhance the transcriptional activity of NFAT5 although reducing its protein expression level, suggests a critical role for ddx5 and ddx17 in tumor cell migration through the fine regulation of NFAT5 pathway.
Collapse
|
230
|
Leung L, Radulovich N, Zhu CQ, Organ S, Bandarchi B, Pintilie M, To C, Panchal D, Tsao MS. Lipocalin2 promotes invasion, tumorigenicity and gemcitabine resistance in pancreatic ductal adenocarcinoma. PLoS One 2012; 7:e46677. [PMID: 23056397 PMCID: PMC3464270 DOI: 10.1371/journal.pone.0046677] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 09/03/2012] [Indexed: 01/05/2023] Open
Abstract
Lipocalin 2 (LCN2) is a small secreted protein and its elevated expression has been observed in pancreatic as well as other cancer types. LCN2 has been reported to promote resistance to drug-induced apoptosis, enhance invasion through its physical association with matrix metalloproteinase-9, and promote in vivo tumor growth. LCN2 was found to be commonly expressed in patient PDAC samples and its pattern of immunohistochemical staining intensified with increasing severity in high-grade precursor lesions. Downregulation of LCN2 in two pancreatic ductal adenocarcinoma cell lines (BxPC3 and HPAF-II) with high LCN2 expression significantly reduced attachment, invasion, and tumour growth in vivo, but not proliferation or motility. Downregulation of LCN2 in two pancreatic ductal adenocarcinoma cell lines (BxPC3 and HPAF-II) with high expression significantly reduced attachment, invasion, and tumour growth in vivo. In contrast, LCN2 overexpression in PANC1, with low endogenous expression, significantly increased invasion, attachment, and enhanced tumor growth. Suppression of LCN2 in BxPC3 and HPAF-II cells increased their sensitivity to gemcitabine in vitro, and in vivo when BxPC3 was tested. Furthermore, LCN2 promotes expression of VEGF and HIF1A which contribute to enhanced vascularity. These overall results demonstrate that LCN2 plays an important role in the malignant progression of pancreatic ductal carcinoma and is a potential therapeutic target for this disease.
Collapse
Affiliation(s)
- Lisa Leung
- Ontario Cancer Institute/Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Nikolina Radulovich
- Ontario Cancer Institute/Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Chang-Qi Zhu
- Ontario Cancer Institute/Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Shawna Organ
- Ontario Cancer Institute/Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Bizhan Bandarchi
- Ontario Cancer Institute/Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Melania Pintilie
- Ontario Cancer Institute/Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Christine To
- Ontario Cancer Institute/Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Devang Panchal
- Ontario Cancer Institute/Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Ming Sound Tsao
- Ontario Cancer Institute/Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
231
|
Gaudineau B, Fougère M, Guaddachi F, Lemoine F, de la Grange P, Jauliac S. Lipocalin 2, the TNF-like receptor TWEAKR and its ligand TWEAK act downstream of NFAT1 to regulate breast cancer cell invasion. J Cell Sci 2012; 125:4475-4486. [PMID: 22767506 DOI: 10.1242/jcs.099879] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
NFAT1 is a transcription factor that elicits breast carcinoma cells to become invasive, thus contributing to metastasis. The molecular mechanisms by which NFAT1 operates in this respect are still poorly known. Here, we report that NFAT1 increases lipocalin 2 (LCN2) mRNA and protein expression by binding to specific sites in the LCN2 gene promoter region. We show that the LCN2 protein is required downstream of NFAT1 to increase breast cancer cell invasion. We demonstrate that the NFAT1-LCN2 axis is sufficient to regulate expression of the TNF-like receptor TWEAKR at the RNA level and of its ligand, TWEAK, at the protein level. We show, however, that TWEAKR mediates an anti-invasive effect in breast cancer cells whereas, depending on LCN2 expression, TWEAK has either anti- or pro-invasive capacities. Thus, we identify LCN2 and TWEAKR-TWEAK as crucial downstream effectors of NFAT1 that regulate breast cancer cell motility and invasive capacity.
Collapse
Affiliation(s)
- Benoît Gaudineau
- CNRS UMR7212, INSERM U944, Université Paris Diderot, Institut d'Hématologie, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75475 Paris Cedex 10, France
| | | | | | | | | | | |
Collapse
|
232
|
Yang J, McNeish B, Butterfield C, Moses MA. Lipocalin 2 is a novel regulator of angiogenesis in human breast cancer. FASEB J 2012; 27:45-50. [PMID: 22982376 DOI: 10.1096/fj.12-211730] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Lipocalin 2 (Lcn2), a member of the lipocalin family, is up-regulated in a variety of epithelial cancers. We have previously reported that Lcn2 induces the epithelial to mesenchymal transition in breast cancer through the estrogen receptor α/Slug axis and that it is a potential noninvasive biomarker of this disease. Here, we report the novel finding that Lcn2 regulates breast cancer angiogenesis. Vascular endothelial growth factor (VEGF), a key angiogenic activator, was significantly increased with Lcn2 expression in MCF-7 human breast cancer cells as well as in an angiogenic line derived from MDA-MB-436 cells. Treatment with a VEGF-neutralizing antibody demonstrates that VEGF is essential for the angiogenic activity of Lcn2. We further demonstrate that Lcn2-induced VEGF is mediated through hypoxia-inducible factor 1α (HIF-1α) and that Lcn2 regulates HIF-1α through extracellular signal-regulated kinase (Erk). The regulation of HIF-1α and VEGF by Lcn2 was also demonstrated in the aggressive MDA-MB-231 cell line. Using the mouse corneal pocket assay, we found that Lcn2 significantly enhanced the angiogenesis induced by VEGF. Taken together, these results are the first to demonstrate that Lcn2 promotes angiogenesis in vitro and in vivo and suggest a novel mechanism through which Lcn2 may promote tumor progression.
Collapse
Affiliation(s)
- Jiang Yang
- Vascular Biology Program, Children's Hospital Boston, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
233
|
Obr A, Edwards DP. The biology of progesterone receptor in the normal mammary gland and in breast cancer. Mol Cell Endocrinol 2012; 357:4-17. [PMID: 22193050 PMCID: PMC3318965 DOI: 10.1016/j.mce.2011.10.030] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2011] [Revised: 09/23/2011] [Accepted: 10/26/2011] [Indexed: 11/21/2022]
Abstract
This paper reviews work on progesterone and the progesterone receptor (PR) in the mouse mammary gland that has been used extensively as an experimental model. Studies have led to the concept that progesterone controls proliferation and morphogenesis of the luminal epithelium in a tightly orchestrated manner at distinct stages of development by paracrine signaling pathways, including receptor activator of nuclear factor κB ligand (RANKL) as a major paracrine factor. Progesterone also drives expansion of stem cells by paracrine signals to generate progenitors required for alveologenesis. During mid-to-late pregnancy, progesterone has another role to suppress secretory activation until parturition mediated in part by crosstalk between PR and prolactin/Stat5 signaling to inhibit induction of milk protein gene expression, and by inhibiting tight junction closure. In models of hormone-dependent mouse mammary tumors, the progesterone/PR signaling axis enhances pre-neoplastic progression by a switch from a paracrine to an autocrine mode of proliferation and dysregulation of the RANKL signaling pathway. Limited experiments with normal human breast show that progesterone/PR signaling also stimulates epithelial cell proliferation by a paracrine mechanism; however, the signaling pathways and whether RANKL is a major mediator remains unknown. Work with human breast cancer cell lines, patient tumor samples and clinical studies indicates that progesterone is a risk factor for breast cancer and that alteration in progesterone/PR signaling pathways contributes to early stage human breast cancer progression. However, loss of PR expression in primary tumors is associated with a less differentiated more invasive phenotype and worse prognosis, suggesting that PR may limit later stages of tumor progression.
Collapse
Affiliation(s)
- Alison Obr
- Departments of Molecular & Cellular Biology and Pathology and Immunology, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Dean P. Edwards
- Departments of Molecular & Cellular Biology and Pathology and Immunology, Baylor College of Medicine, Houston, Texas, 77030, USA
| |
Collapse
|
234
|
Cramer EP, Glenthøj A, Häger M, Juncker-Jensen A, Engelholm LH, Santoni-Rugiu E, Lund LR, Laerum OD, Cowland JB, Borregaard N. No effect of NGAL/lipocalin-2 on aggressiveness of cancer in the MMTV-PyMT/FVB/N mouse model for breast cancer. PLoS One 2012; 7:e39646. [PMID: 22737251 PMCID: PMC3380857 DOI: 10.1371/journal.pone.0039646] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 05/24/2012] [Indexed: 11/22/2022] Open
Abstract
NGAL/lipocalin-2 is a siderophore-binding protein that is highly expressed in several cancers. It is suggested to confer a proliferative advantage to cancer cells. Its expression has been correlated with aggressiveness of breast cancer as determined both in patients and in mouse breast cancer models. This was recently confirmed in two mouse models of spontaneous breast cancer in wild-type and lipocalin-2-deficient mice. We used a similar strategy using a different mouse strain. Lipocalin-2-deficient mice and mouse mammary tumor virus-polyoma middle T antigen (MMTV-PyMT) mice were crossed into the same FVB/N background. All mice developed tumors by week 8. The mice were sacrificed on week 13 and tissue was processed for biochemical and histological analysis. The total tumor volume and number of metastases were quantitated in 26 lipocalin-2-deficient mice and 34 wild-type controls. Lipocalin-2 expression in tumors of MMTV-PyMT-positive and wild-type mice was assessed by quantitative real-time PCR and by immunohistochemistry. The expression of the lipocalin-2 receptors 24p3R and megalin and of Mmp-9, transferrin receptor, and Bdh2 (a producer of a mammalian siderophore) were quantitated by real-time PCR. No significant difference was observed between wild-type and lipocalin-2-deficient mice. Lipocalin-2 was highly expressed in tumors from wild-type mice, but the expression did not correlate with tumor size. No effect of lipocalin-2 was observed with respect to time to tumor appearance, total tumor volume, or to the number of metastases. Histology and gelatinolytic activity of the mammary tumors did not differ between wild-type and lipocalin-2-deficient mice. We conclude that NGAL/lipocalin-2 does not invariably affect the aggressiveness of breast cancers as assessed in mouse models, thus questioning the role of lipocalin-2 in cancer development.
Collapse
Affiliation(s)
- Elisabeth P. Cramer
- The Granulocyte Research Laboratory, Department of Hematology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Andreas Glenthøj
- The Granulocyte Research Laboratory, Department of Hematology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mattias Häger
- The Granulocyte Research Laboratory, Department of Hematology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Anna Juncker-Jensen
- Finsen Laboratory, National University Hospital, Rigshospitalet, University of Copenhagen, Denmark
| | - Lars H. Engelholm
- Finsen Laboratory, National University Hospital, Rigshospitalet, University of Copenhagen, Denmark
| | - Eric Santoni-Rugiu
- Department of Pathology, National University Hospital, Rigshospitalet, University of Copenhagen, Denmark
| | - Leif R. Lund
- Department of Cellular and Molecular Medicine, University of Copenhagen, Denmark
| | - Ole D. Laerum
- Finsen Laboratory, National University Hospital, Rigshospitalet, University of Copenhagen, Denmark
- The Gade Institute, Section of Pathology, University of Bergen, Norway
| | - Jack B. Cowland
- The Granulocyte Research Laboratory, Department of Hematology, Copenhagen University Hospital, Copenhagen, Denmark
- * E-mail: (JBC); (NB)
| | - Niels Borregaard
- The Granulocyte Research Laboratory, Department of Hematology, Copenhagen University Hospital, Copenhagen, Denmark
- * E-mail: (JBC); (NB)
| |
Collapse
|
235
|
Sung H, Choi JY, Lee SA, Lee KM, Han S, Jeon S, Song M, Lee Y, Park SK, Yoo KY, Noh DY, Ahn SH, Kang D. The association between the preoperative serum levels of lipocalin-2 and matrix metalloproteinase-9 (MMP-9) and prognosis of breast cancer. BMC Cancer 2012; 12:193. [PMID: 22640376 PMCID: PMC3479006 DOI: 10.1186/1471-2407-12-193] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2011] [Accepted: 05/14/2012] [Indexed: 12/11/2022] Open
Abstract
Background Although a number of experimental studies have suggested the role of lipocalin-2 (LCN2) and matrix metalloproteinase-9 (MMP-9) in breast cancer progression, limited numbers of epidemiological studies have examined the relationship between the levels of lipocalin-2 and MMP-9 and breast cancer survival. Methods Preoperative serum levels of lipocalin-2 and MMP-9 were measured in 303 breast cancer patients and 74 healthy controls recruited between 2004 and 2007. We examined the association between lipocalin-2 and MMP-9 levels and disease-free survival (DFS) using Cox proportional hazard regression model. Results The serum levels of lipocalin-2 and MMP-9 were not significantly different between patients and controls (P > 0.05). Elevated lipocalin-2 and MMP-9 levels were associated with reduced DFS of breast cancer ( Ptrend = 0.029 and Ptrend = 0.063, respectively). When lipocalin-2 and MMP-9 levels were categorized based on the combined risk score, patients with higher levels of both lipocalin-2 and MMP-9 exhibited poor DFS compared to patients with lower levels (Ptrend = 0.004). Furthermore, these effects were profound in patients with BMI less than 25 kg/m2 (adjusted hazard ratio (aHR), 3.17; 95% confidence intervals (CI), 1.66-6.06, Ptrend < 0.001) or lymph-node negative breast cancer (aHR, 5.36; 95% CI, 2.18-13.2, Ptrend < 0.001). Conclusions Our study suggests that the elevated levels of lipocalin-2 and MMP-9 are associated with reduced breast cancer survival, particularly in patients with lower BMI and lymph-node negative breast cancers.
Collapse
Affiliation(s)
- Hyuna Sung
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
236
|
Reilly PT, Teo WL, Low MJ, Amoyo-Brion AA, Dominguez-Brauer C, Elia AJ, Berger T, Greicius G, Pettersson S, Mak TW. Lipocalin 2 performs contrasting, location-dependent roles in APCmin tumor initiation and progression. Oncogene 2012; 32:1233-9. [PMID: 22614012 PMCID: PMC3594828 DOI: 10.1038/onc.2012.159] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Evidence that lipocalin 2 (LCN2) is oncogenic has grown in recent years and comes from both animal models and expression analysis from a variety of human cancers. In the intestine, LCN2 is overexpressed in colitis patients and its overexpression is a negative prognostic indicator in colorectal cancer. Functionally, LCN2 has a number of different activities that may contribute to its oncogenic potential, including increasing matrix metalloproteinase activity, control of iron availability and stimulating inflammation. In this report, we examined APCmin intestinal tumorigenesis in an LCN2-deficient background. We found that the loss of LCN2 increased tumor multiplicity specifically in the duodenum, suggesting a potential tumor-suppressive activity. Concurrently, however, LCN2 increased the average small intestinal tumor size particularly in the distal small intestine. We found that this increase was correlated to tumor iron(II) content, suggesting that an iron-scavenging role is important for LCN2 oncogenic activity in the intestine.
Collapse
Affiliation(s)
- P T Reilly
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore, Singapore
| | | | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Mannelqvist M, Stefansson IM, Wik E, Kusonmano K, Raeder MB, Øyan AM, Kalland KH, Moses MA, Salvesen HB, Akslen LA. Lipocalin 2 expression is associated with aggressive features of endometrial cancer. BMC Cancer 2012; 12:169. [PMID: 22559235 PMCID: PMC3493289 DOI: 10.1186/1471-2407-12-169] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 04/19/2012] [Indexed: 01/21/2023] Open
Abstract
Background Increased expression of lipocalin 2 (LCN2) has been observed in several cancers. The aim of the present study was to investigate LCN2 in endometrial cancer in relation to clinico-pathologic phenotype, angiogenesis, markers of epithelial-mesenchymal transition (EMT), and patient survival. Methods Immunohistochemical staining was performed using a human LCN2 antibody on a population-based series of endometrial cancer patients collected in Hordaland County (Norway) during 1981-1990 (n = 256). Patients were followed from the time of primary surgery until death or last follow-up in 2007. The median follow-up time for survivors was 17 years. Gene expression data from a prospectively collected endometrial cancer series (n = 76) and a publicly available endometrial cancer series (n = 111) was used for gene correlation studies. Results Expression of LCN2 protein, found in 49% of the cases, was associated with non-endometrioid histologic type (p = 0.001), nuclear grade 3 (p = 0.001), >50% solid tumor growth (p = 0.001), ER and PR negativity (p = 0.028 and 0.006), and positive EZH2 expression (p < 0.001). LCN2 expression was significantly associated with expression of VEGF-A (p = 0.021), although not with other angiogenesis markers examined (vascular proliferation index, glomeruloid microvascular proliferation, VEGF-C, VEGF-D or bFGF2 expression). Further, LCN2 was not associated with several EMT-related markers (E-cadherin, N-cadherin, P-cadherin, β-catenin), nor with vascular invasion (tumor cells invading lymphatic or blood vessels). Notably, LCN2 was significantly associated with distant tumor recurrences, as well as with the S100A family of metastasis related genes. Patients with tumors showing no LCN2 expression had the best outcome with 81% 5-year survival, compared to 73% for intermediate and 38% for the small subgroup with strong LCN2 staining (p = 0.007). In multivariate analysis, LCN2 expression was an independent prognostic factor in addition to histologic grade and FIGO stage. Conclusion Increased LCN2 expression is associated with aggressive features and poor prognosis in endometrial cancer.
Collapse
Affiliation(s)
- Monica Mannelqvist
- The Gade Institute, Section for Pathology, University of Bergen, Bergen, Norway
| | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Correnti C, Strong RK. Mammalian siderophores, siderophore-binding lipocalins, and the labile iron pool. J Biol Chem 2012; 287:13524-31. [PMID: 22389496 PMCID: PMC3340207 DOI: 10.1074/jbc.r111.311829] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Bacteria use tight-binding, ferric-specific chelators called siderophores to acquire iron from the environment and from the host during infection; animals use proteins such as transferrin and ferritin to transport and store iron. Recently, candidate compounds that could serve endogenously as mammalian siderophore equivalents have been identified and characterized through associations with siderocalin, the only mammalian siderophore-binding protein currently known. Siderocalin, an antibacterial protein, acts by sequestering iron away from infecting bacteria as siderophore complexes. Candidate endogenous siderophores include compounds that only effectively transport iron as ternary complexes with siderocalin, explaining pleiotropic activities in normal cellular processes and specific disease states.
Collapse
Affiliation(s)
- Colin Correnti
- From the Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Roland K. Strong
- From the Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| |
Collapse
|
239
|
Kim YS, Kim YJ, Lee JM, Kim EK, Park YJ, Choe SK, Ko HJ, Kang CY. Functional Changes in Myeloid-Derived Suppressor Cells (MDSCs) during Tumor Growth: FKBP51 Contributes to the Regulation of the Immunosuppressive Function of MDSCs. THE JOURNAL OF IMMUNOLOGY 2012; 188:4226-34. [DOI: 10.4049/jimmunol.1103040] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
240
|
Iron levels in polarized macrophages: regulation of immunity and autoimmunity. Autoimmun Rev 2012; 11:883-9. [PMID: 22449938 DOI: 10.1016/j.autrev.2012.03.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 03/08/2012] [Indexed: 02/06/2023]
Abstract
Although the hallmark of autoimmune diseases remains the generation of autoantigen-specific lynfocytic cell response, growing evidence is showing a key role for macrophages in a number of autoimmune diseases. Macrophages are characterized by phenotypical and functional heterogeneity. Different immunological signals, coming from systemic blood circulation or from microenvironment, polarize macrophages to classical (M1) or alternative (M2) phenotypes. Iron accumulation in M1 macrophages is a well known bacteriostatic mechanism and one of the mechanisms at the basis of anemia associated to chronic inflammation. Moreover, some recent data suggest that iron accumulation in macrophages can directly activate macrophages to pro-inflammatory M1 phenotype, highlighting a putative role of macrophage iron retention in the pathogenesis of chronic inflammatory and autoimmune diseases. Conversely, iron content is low in M2 macrophages, principally due to increased iron release, and increased availability of iron in the extracellular milieu supported by M2 macrophages could influence the growth rate of adjacent cell and thus play an important role in tumor growth and tissue remodeling. In this review we summarize the molecular mechanisms sustaining differential iron metabolism in polarized macrophages, discuss the relevance of this metabolic signature in chronic inflammatory and autoimmune diseases, and finally focus on potential therapeutic implications rising from a better understanding of underlying molecular mechanisms.
Collapse
|
241
|
Wang N, Zhou F, Xiong H, Du S, Ma J, Okai I, Wang J, Suo J, Hao L, Song Y, Hu J, Shao S. Screening and Identification of Distant Metastasis-Related Differentially Expressed Genes in Human Squamous Cell Lung Carcinoma. Anat Rec (Hoboken) 2012; 295:748-57. [DOI: 10.1002/ar.22441] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 02/07/2012] [Indexed: 11/12/2022]
|
242
|
Guo H, Zhang Y, Brockman DA, Hahn W, Bernlohr DA, Chen X. Lipocalin 2 deficiency alters estradiol production and estrogen receptor signaling in female mice. Endocrinology 2012; 153:1183-93. [PMID: 22234464 PMCID: PMC3281525 DOI: 10.1210/en.2011-1642] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
We have previously characterized lipocalin 2 (Lcn2) as a new adipokine having a critical role in energy and lipid metabolism in male mice. Previous studies by others have suggested that Lcn2 is a putative target gene of estrogens. In this study, we reported the effect of Lcn2 deficiency on estradiol biosynthesis and estrogen receptor signaling in female Lcn2-deficient (Lcn2-/-) mice. We found that Lcn2 expression in white adipose tissue is gender, depot, and age dependent. In female mice, Lcn2 is predominantly expressed in inguinal adipose tissue but at relatively very low levels in perigonadal depot and ovary. After 22 wk of high-fat diet (HFD) feeding or at old age, Lcn2-/- female mice had significantly reduced levels of serum 17β-estradiol and down-regulated expression of estrogen receptor α in multiple metabolic tissues. Consistently, the expression of estrogen-regulated genes involved in cholesterol homeostasis, such as liver X receptor β and low-density lipoprotein receptor was also down-regulated in the adipose tissue of Lcn2-/- mice. These changes were in line with the development of atherogenic dyslipidemia in response to HFD feeding; female Lcn2-/- mice had significantly elevated levels of total cholesterol and low-density lipoprotein cholesterol, whereas reduced high-density lipoprotein cholesterol levels compared with wild-type female mice. Interestingly, when compared with wild-type controls, HFD-fed female Lcn2-/- mice had significantly reduced expression levels of aromatase, a key enzyme regulating estradiol biosynthesis, in adipose tissue. Moreover, Lcn2 deficiency markedly blunted age-related increase in adipose aromatase expression but had no significant impact on age-related reduction in ovarian aromatase expression. Our findings suggest that Lcn2 has a tissue-specific role in adipose estradiol biosynthesis, which may link Lcn2 to obesity- and age-related estradiol production and metabolic complications in females.
Collapse
Affiliation(s)
- Hong Guo
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, St. Paul, Minnesota 55108-1038, USA
| | | | | | | | | | | |
Collapse
|
243
|
Kim H, Lee S, Park HC, Lee WH, Lee MS, Suk K. Modulation of glial and neuronal migration by lipocalin-2 in zebrafish. Immune Netw 2011; 11:342-7. [PMID: 22346773 PMCID: PMC3275702 DOI: 10.4110/in.2011.11.6.342] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 10/14/2011] [Accepted: 10/18/2011] [Indexed: 12/01/2022] Open
Abstract
Background Glial cells are involved in immune and inflammatory responses in the central nervous system (CNS). Glial cells such as microglia and astrocytes also provide structural and functional support for neurons. Migration and morphological changes of CNS cells are associated with their physiological as well as pathological functions. The secreted protein lipocalin-2 (LCN2) has been previously implicated in regulation of diverse cellular processes of glia and neurons, including cell migration and morphology. Methods Here, we employed a zebrafish model to analyze the role of LCN2 in CNS cell migration and morphology in vivo. In the first part of this study, we examined the indirect effect of LCN2 on cell migration and morphology of microglia, astrocytes, and neurons cultured in vitro. Results Conditioned media collected from LCN2-treated astrocytes augmented migration of glia and neurons in the Boyden chamber assay. The conditioned media also increased the number of neuronal processes. Next, in order to further understand the role of LCN2 in the CNS in vivo, LCN2 was ectopically expressed in the zebrafish spinal cord. Expression of exogenous LCN2 modulated neuronal cell migration in the spinal cord of zebrafish embryos, supporting the role of LCN2 as a cell migration regulator in the CNS. Conclusion Thus, LCN2 proteins secreted under diverse conditions may play an important role in CNS immune and inflammatory responses by controlling cell migration and morphology.
Collapse
Affiliation(s)
- Ho Kim
- Department of Medical Science, Korea University Ansan Hospital, Ansan 425-707, Korea
| | | | | | | | | | | |
Collapse
|
244
|
Bose D, Zimmerman LJ, Pierobon M, Petricoin E, Tozzi F, Parikh A, Fan F, Dallas N, Xia L, Gaur P, Samuel S, Liebler DC, Ellis LM. Chemoresistant colorectal cancer cells and cancer stem cells mediate growth and survival of bystander cells. Br J Cancer 2011; 105:1759-67. [PMID: 22045189 PMCID: PMC3242606 DOI: 10.1038/bjc.2011.449] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Revised: 09/27/2011] [Accepted: 10/04/2011] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Recent studies suggest that cancer stem cells (CSCs) mediate chemoresistance, but interestingly, only a small percentage of cells in a resistant tumour are CSCs; this suggests that non-CSCs survive by other means. We hypothesised that chemoresistant colorectal cancer (CRC) cells generate soluble factors that enhance survival of chemonaive tumour cells. METHODS Chemoresistant CRC cells were generated by serial passage in oxaliplatin (Ox cells). Conditioned media (CM) was collected from parental and oxaliplatin-resistant (OxR) cells. CRC cells were treated with CM and growth and survival were assessed. Tumour growth rates were determined in nude mice after cells were treated with CM. Mass spectrometry (MS) identified proteins in CM. Reverse phase protein microarray assays determined signalling effects of CM in parental cells. RESULTS Oxaliplatin-resistant CM increased survival of chemo-naive cells. CSC CM also increased growth of parental cells. Parental and OxR mixed tumours grew larger than tumours composed of parental or OxR cells alone. Mass spectrometry detected unique survival-promoting factors in OxR CM compared with parental CM. Cells treated with OxR CM demonstrated early phosphorylation of EGFR and MEK1, with later upregulation of total Akt .We identified progranulin as a potential mediator of chemoresistance. CONCLUSION Chemoresistant tumour cells and CSCs may promote resistance through soluble factors that mediate survival in otherwise chemosensitive tumour cells.
Collapse
Affiliation(s)
- D Bose
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - L J Zimmerman
- Jim Ayers Institute for Precancer Detection and Diagnosis, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - M Pierobon
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Fairfax, VA 22030, USA
| | - E Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Fairfax, VA 22030, USA
| | - F Tozzi
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - A Parikh
- Jim Ayers Institute for Precancer Detection and Diagnosis, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - F Fan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - N Dallas
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - L Xia
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - P Gaur
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - S Samuel
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - D C Liebler
- Jim Ayers Institute for Precancer Detection and Diagnosis, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - L M Ellis
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| |
Collapse
|
245
|
Rodvold JJ, Mahadevan NR, Zanetti M. Lipocalin 2 in cancer: when good immunity goes bad. Cancer Lett 2011; 316:132-8. [PMID: 22075378 DOI: 10.1016/j.canlet.2011.11.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 10/29/2011] [Accepted: 11/01/2011] [Indexed: 10/15/2022]
Abstract
The innate immune molecule Lipocalin 2 (LCN2) was initially shown to combat bacterial infection by binding bacterial siderophores, hence impairing microbial iron sequestration. In recent years, it has become apparent that LCN2 is over-expressed in cancers of diverse histological origin and that it facilitates tumorigenesis by promoting survival, growth, and metastasis. Herein, we discuss emerging evidence that substantiates two functional roles for LCN2 in cancer: promotion of the epithelial-to-mesenchymal transition (EMT) that facilitates an invasive phenotype and metastasis, and sequestration of iron that results in cell survival and tumorigenesis. Further, we present evidence that upregulated LCN2 expression in solid tumors is induced by hypoxia and pro-inflammation, microenvironmental noxae that converge to cause an endoplasmic reticulum (ER) stress response. Taken together, it appears that tumor cells exploit the beneficial innate immune function of LCN2 to support uncontrolled growth. This duplicity in function highlights LCN2 and its upstream driver, the ER stress response, as key targets for cancer therapy.
Collapse
Affiliation(s)
- Jeffrey J Rodvold
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093-0815, United States
| | | | | |
Collapse
|
246
|
Coticchia CM, Curatolo AS, Zurakowski D, Yang J, Daniels KE, Matulonis UA, Moses MA. Urinary MMP-2 and MMP-9 predict the presence of ovarian cancer in women with normal CA125 levels. Gynecol Oncol 2011; 123:295-300. [DOI: 10.1016/j.ygyno.2011.07.034] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 07/19/2011] [Accepted: 07/21/2011] [Indexed: 10/17/2022]
|
247
|
Lee S, Lee WH, Lee MS, Mori K, Suk K. Regulation by lipocalin-2 of neuronal cell death, migration, and morphology. J Neurosci Res 2011; 90:540-50. [PMID: 22038922 DOI: 10.1002/jnr.22779] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 06/21/2011] [Accepted: 07/27/2011] [Indexed: 11/11/2022]
Abstract
A secreted protein, lipocalin-2 (LCN2), has been previously shown to regulate a variety of cellular phenotypes such as cell death, migration, and morphology. The role of LCN2, however, appears to be different depending on the cellular context. Here, we investigated how LCN2 influences neuronal phenotypes by using primary cortical neuronal cell cultures and neuroblastoma cell lines as a model. When exposed to LCN2 protein, neurons and neuroblastoma cells were sensitized to cell death evoked by nitric oxide, oxidative stress, and tumor necrosis factor-α (TNF-α). A forced expression of lcn2 in glia enhanced neuronal cell death in cocultures of glia and neurons, indicating that both exogenous protein addition and endogenous expression of lcn2 give rise to similar results. Iron and BCL2-interacting mediator of cell death (BIM) protein were involved in LCN2-induced cell death sensitization, based on the studies using iron donor, chelator, siderophore, and short hairpin RNA (shRNA)-mediated knockdown of bim expression. Furthermore, cell migration assay and immunofluorescence microscopic observation revealed that LCN2 accelerated neuronal motility and process extension, suggesting multiple roles for LCN2 in the regulation of neuronal cell death, migration, and morphology.
Collapse
Affiliation(s)
- Shinrye Lee
- Department of Pharmacology, Brain Science and Engineering Institute, CMRI, Kyungpook National University School of Medicine, Daegu, Korea
| | | | | | | | | |
Collapse
|
248
|
|
249
|
Lee S, Kim JH, Kim JH, Seo JW, Han HS, Lee WH, Mori K, Nakao K, Barasch J, Suk K. Lipocalin-2 Is a chemokine inducer in the central nervous system: role of chemokine ligand 10 (CXCL10) in lipocalin-2-induced cell migration. J Biol Chem 2011; 286:43855-43870. [PMID: 22030398 DOI: 10.1074/jbc.m111.299248] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The secreted protein lipocalin-2 (LCN2) has been implicated in diverse cellular processes, including cell morphology and migration. Little is known, however, about the role of LCN2 in the CNS. Here, we show that LCN2 promotes cell migration through up-regulation of chemokines in brain. Studies using cultured glial cells, microvascular endothelial cells, and neuronal cells suggest that LCN2 may act as a chemokine inducer on the multiple cell types in the CNS. In particular, up-regulation of CXCL10 by JAK2/STAT3 and IKK/NF-κB pathways in astrocytes played a pivotal role in LCN2-induced cell migration. The cell migration-promoting activity of LCN2 in the CNS was verified in vivo using mouse models. The expression of LCN2 was notably increased in brain following LPS injection or focal injury. Mice lacking LCN2 showed the impaired migration of astrocytes to injury sites with a reduced CXCL10 expression in the neuroinflammation or injury models. Thus, the LCN2 proteins, secreted under inflammatory conditions, may amplify neuroinflammation by inducing CNS cells to secrete chemokines such as CXCL10, which recruit additional inflammatory cells.
Collapse
Affiliation(s)
- Shinrye Lee
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Jong-Heon Kim
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Jae-Hong Kim
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Jung-Wan Seo
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Hyung-Soo Han
- Department of Physiology, Brain Science & Engineering Institute, Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Won-Ha Lee
- Departments of School of Life Sciences and Biotechnology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Kiyoshi Mori
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Kazuwa Nakao
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Jonathan Barasch
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10027
| | - Kyoungho Suk
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea.
| |
Collapse
|
250
|
Coudevylle N, Hoetzinger M, Geist L, Kontaxis G, Hartl M, Bister K, Konrat R. Lipocalin Q83 Reveals a Dual Ligand Binding Mode with Potential Implications for the Functions of Siderocalins. Biochemistry 2011; 50:9192-9. [DOI: 10.1021/bi201115q] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Nicolas Coudevylle
- Department of Structural and
Computational Biology, Max F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5/1, 1030 Vienna,
Austria
| | - Matthias Hoetzinger
- Department of Structural and
Computational Biology, Max F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5/1, 1030 Vienna,
Austria
| | - Leonhard Geist
- Department of Structural and
Computational Biology, Max F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5/1, 1030 Vienna,
Austria
| | - Georg Kontaxis
- Department of Structural and
Computational Biology, Max F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5/1, 1030 Vienna,
Austria
| | - Markus Hartl
- Institute of Biochemistry,
Center
for Molecular Biosciences (CMBI), University of Innsbruck, Peter-Mayr-Strasse 1a, 6020 Innsbruck, Austria
| | - Klaus Bister
- Institute of Biochemistry,
Center
for Molecular Biosciences (CMBI), University of Innsbruck, Peter-Mayr-Strasse 1a, 6020 Innsbruck, Austria
| | - Robert Konrat
- Department of Structural and
Computational Biology, Max F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5/1, 1030 Vienna,
Austria
| |
Collapse
|