Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
3.
Oncogene ; 39(1): 64-78, 2020 01.
Article in English | MEDLINE | ID: mdl-31462709

ABSTRACT

Mechanisms for breast cancer metastasis remain unclear. Whether truncated glioma-associated oncogene homolog 1 (TGLI1), a transcription factor known to promote angiogenesis, migration and invasion, plays any role in metastasis of any tumor type has never been investigated. In this study, results of two mouse models of breast cancer metastasis showed that ectopic expression of TGLI1, but not GLI1, promoted preferential metastasis to the brain. Conversely, selective TGLI1 knockdown using antisense oligonucleotides led to decreased breast cancer brain metastasis (BCBM) in vivo. Immunohistochemical staining showed that TGLI1, but not GLI1, was increased in lymph node metastases compared to matched primary tumors, and that TGLI1 was expressed at higher levels in BCBM specimens compared to primary tumors. TGLI1 activation is associated with a shortened time to develop BCBM and enriched in HER2-enriched and triple-negative breast cancers. Radioresistant BCBM cell lines and specimens expressed higher levels of TGLI1, but not GLI1, than radiosensitive counterparts. Since cancer stem cells (CSCs) are radioresistant and metastasis-initiating cells, we examined TGLI1 for its involvement in breast CSCs and found TGLI1 to transcriptionally activate stemness genes CD44, Nanog, Sox2, and OCT4 leading to CSC renewal, and TGLI1 outcompetes with GLI1 for binding to target promoters. We next examined whether astrocyte-priming underlies TGLI1-mediated brain tropism and found that TGLI1-positive CSCs strongly activated and interacted with astrocytes in vitro and in vivo. These findings demonstrate, for the first time, that TGLI1 mediates breast cancer metastasis to the brain, in part, through promoting metastasis-initiating CSCs and activating astrocytes in BCBM microenvironment.


Subject(s)
Brain Neoplasms/genetics , Breast Neoplasms/genetics , Neoplastic Stem Cells/pathology , Transcription Factors/genetics , Zinc Finger Protein GLI1/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Hyaluronan Receptors/genetics , Lymphatic Metastasis , Mice , Nanog Homeobox Protein/genetics , Neoplastic Stem Cells/radiation effects , Octamer Transcription Factor-3/genetics , Receptor, ErbB-2/genetics , SOXB1 Transcription Factors/genetics , Tumor Microenvironment/genetics , Zinc Finger Protein GLI1/genetics
4.
Int J Biol Markers ; 34(1): 90-97, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30854931

ABSTRACT

PURPOSE/OBJECTIVES: We aimed to assess the predictive value of a lung cancer gene panel for the development of brain metastases. MATERIALS/METHODS: Between 2011 and 2015, 102 patients with lung cancer were prospectively enrolled in a clinical trial in which a diagnostic fine-needle aspirate was obtained. Gene expression was conducted on all samples that rendered a diagnosis of non-small cell lung cancer (NSCLC). Subsequent retrospective analysis of brain metastases-related outcomes was performed by reviewing patient electronic medical records. A competing risk multivariable regression was performed to estimate the adjusted hazard ratio for the development of brain metastases and non-brain metastases from NSCLC. RESULTS: A total of 49 of 102 patients had died by the last follow-up. Median time of follow-up was 13 months (range 0.23-67 months). A total of 17 patients developed brain metastases. Median survival time after diagnosis of brain metastases was 3.58 months (95% confidence interval (CI) 2.17, not available). A total of 30 patients developed metastases without any evidence of brain metastases until the time of death or last follow-up. Competing risk analysis identified three genes that were downregulated differentially in the patients with brain metastases versus non-brain metastatic disease: CD37 (0.017), cystatin A (0.022), and IL-23A (0.027). Other factors associated with brain metastases include: stage T ( P ⩽ 8.3e-6) and stage N ( P= 6.8e-4). CONCLUSIONS: We have identified three genes, CD37, cystatin A, and IL-23A, for which downregulation of gene expression was associated with a greater propensity for developing brain metastases. Validation of these biomarkers could have implications on surveillance patterns in patients with brain metastases from NSCLC.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/pathology , Cystatin A/metabolism , Interleukin-23 Subunit p19/metabolism , Lung Neoplasms/pathology , Tetraspanins/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Aged , Brain Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/secondary , Female , Follow-Up Studies , Humans , Lung Neoplasms/metabolism , Male , Middle Aged , Prognosis , Prospective Studies , Retrospective Studies , Survival Rate
5.
J Cancer Res Clin Oncol ; 145(2): 337-344, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30417218

ABSTRACT

INTRODUCTION: Radiation-induced cognitive decline (RICD) is a late effect of radiotherapy (RT) occurring in 30-50% of irradiated brain tumor survivors. In preclinical models, pioglitazone prevents RICD but there are little safety data on its use in non-diabetic patients. We conducted a dose-escalation trial to determine the safety of pioglitazone taken during and after brain irradiation. METHODS: We enrolled patients > 18 years old with primary or metastatic brain tumors slated to receive at least 10 treatments of RT (≤ 3 Gy per fraction). We evaluated the safety of pioglitazone at 22.5 mg and 45 mg with a dose-escalation phase and dose-expansion phase. Pioglitazone was taken daily during RT and for 6 months after. RESULTS: 18 patients with a mean age of 54 were enrolled between 2010 and 2014. 14 patients had metastatic brain tumors and were treated with whole brain RT. Four patients had primary brain tumors and received partial brain RT and concurrent chemotherapy. No DLTs were identified. In the dose-escalation phase, there were only three instances of grade ≥ 3 toxicity: one instance of neuropathy in a patient receiving 22.5 mg, one instance of fatigue in a patient receiving 22.5 mg and one instance of dizziness in a patient receiving 45 mg. The attribution in each of these cases was considered "possible." In the dose-expansion phase, nine patients received 45 mg and there was only one grade 3 toxicity (fatigue) possibly attributable to pioglitazone. CONCLUSION: Pioglitazone was well tolerated by brain tumor patients undergoing RT. 45 mg is a safe dose to use in future efficacy trials.


Subject(s)
Brain Neoplasms/drug therapy , Hypoglycemic Agents/therapeutic use , Pioglitazone/therapeutic use , Adult , Aged , Aged, 80 and over , Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Female , Follow-Up Studies , Humans , Lymphatic Metastasis , Male , Maximum Tolerated Dose , Middle Aged , Patient Safety , Prognosis , Radiotherapy, Conformal , Survival Rate
6.
Radiat Res ; 190(4): 361-373, 2018 10.
Article in English | MEDLINE | ID: mdl-30016219

ABSTRACT

Late-delayed radiation-induced brain injury (RIBI) is a major adverse effect of fractionated whole-brain irradiation (fWBI). Characterized by progressive cognitive dysfunction, and associated cerebrovascular and white matter injury, RIBI deleteriously affects quality of life for cancer patients. Despite extensive morphological characterization of the injury, the pathogenesis is unclear, thus limiting the development of effective therapeutics. We previously reported that RIBI is associated with increased gene expression of the extracellular matrix (ECM) protein fibronectin (FN1). We hypothesized that fibronectin contributes to perivascular ECM, which may impair diffusion to the dependent parenchyma, thus contributing to the observed cognitive decline. The goal of this study was to determine the localization of fibronectin in RIBI and further characterize the composition of perivascular ECM, as well as identify the cell of origin for FN1 by in situ hybridization. Briefly, fibronectin localized to the vascular basement membrane of morphologically normal blood vessels from control comparators and animals receiving fWBI, and to the perivascular space of edematous and fibrotic vascular phenotypes of animals receiving fWBI. Additional mild diffuse parenchymal staining in areas of vascular injury suggested blood-brain-barrier disruption and plasma fibronectin extravasation. Perivascular ECM lacked amyloid and contained lesser amounts of collagens I and IV, which localized to the basement membrane. These changes occurred in the absence of alterations in microvascular area fraction or microvessel density. Fibronectin transcripts were rarely expressed in control comparators, and were most strongly induced within cerebrovascular endothelial and vascular smooth muscle cells after fWBI. Our results demonstrate that fibronectin is produced by cerebrovascular endothelial and smooth muscle cells in late-delayed RIBI and contributes to perivascular ECM, which we postulate may contribute to diffusion barrier formation. We propose that pathways that antagonize fibronectin deposition and matrix assembly or enhance degradation may serve as potential therapeutic targets in RIBI.


Subject(s)
Brain Injuries/metabolism , Cerebrovascular Circulation , Endothelium, Vascular/metabolism , Extracellular Matrix/metabolism , Fibronectins/physiology , Muscle, Smooth, Vascular/metabolism , Radiation Injuries, Experimental/metabolism , Animals , Brain/blood supply , Brain/radiation effects , Brain Injuries/etiology , Brain Injuries/pathology , Fibronectins/biosynthesis , Gene Expression , Macaca mulatta , Male , Radiation Injuries, Experimental/pathology
7.
Cancer Res ; 78(15): 4316-4330, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30026327

ABSTRACT

Up to 30% of patients with metastatic breast cancer eventually develop brain metastasis, yet the pathologic mechanism behind this development remains poorly understood. Here, we profiled long noncoding RNAs in brain metastatic tumors from patients with breast cancer and found that the X-inactive-specific transcript (XIST) was significantly downregulated in these tissues. XIST expression levels inversely correlated with brain metastasis, but not with bone metastasis in patients. Silencing of XIST preferentially promoted brain metastatic growth of XISThigh cells in our xenograft models. Moreover, knockout of XIST in mice mammary glands accelerated primary tumor growth as well as metastases in the brain. Decreased expression of XIST stimulated epithelial-mesenchymal transition and activated c-Met via MSN-mediated protein stabilization, which resulted in the promotion of stemness in the tumor cells. Loss of XIST also augmented secretion of exosomal miRNA-503, which triggered M1-M2 polarization of microglia. This M1-M2 conversion upregulated immune suppressive cytokines in microglia that suppressed T-cell proliferation. Furthermore, we screened an FDA-approved drug library and identified fludarabine as a synthetic lethal drug for XISTlow breast tumor cells and found that fludarabine blocked brain metastasis in our animal model. Our results indicate that XIST plays a critical role in brain metastasis in breast cancer by affecting both tumor cells and the tumor microenvironment and that the XIST-mediated pathway may serve as an effective target for treating brain metastasis.Significance: These findings describe mechanisms of how loss of the lncRNA XIST promotes brain metastasis in breast cancer and identify fludarabine as a potential therapeutic agent that specifically eliminates XISTlow tumor cells in the brain. Cancer Res; 78(15); 4316-30. ©2018 AACR.


Subject(s)
Bone Neoplasms/genetics , Exosomes/genetics , MicroRNAs/genetics , Microfilament Proteins/genetics , Microglia/pathology , Proto-Oncogene Proteins c-met/genetics , RNA, Long Noncoding/genetics , Animals , Bone Neoplasms/pathology , Brain/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , MCF-7 Cells , Mice , Mice, Nude , Tumor Microenvironment/genetics , Up-Regulation/genetics
8.
Radiat Res ; 187(5): 599-611, 2017 05.
Article in English | MEDLINE | ID: mdl-28398880

ABSTRACT

Fractionated whole-brain irradiation (fWBI) is a mainstay of treatment for patients with intracranial neoplasia; however late-delayed radiation-induced normal tissue injury remains a major adverse consequence of treatment, with deleterious effects on quality of life for affected patients. We hypothesize that cerebrovascular injury and remodeling after fWBI results in ischemic injury to dependent white matter, which contributes to the observed cognitive dysfunction. To evaluate molecular effectors of radiation-induced brain injury (RIBI), real-time quantitative polymerase chain reaction (RT-qPCR) was performed on the dorsolateral prefrontal cortex (DLPFC, Brodmann area 46), hippocampus and temporal white matter of 4 male Rhesus macaques (age 6-11 years), which had received 40 Gray (Gy) fWBI (8 fractions of 5 Gy each, twice per week), and 3 control comparators. All fWBI animals developed neurologic impairment; humane euthanasia was elected at a median of 6 months. Radiation-induced brain injury was confirmed histopathologically in all animals, characterized by white matter degeneration and necrosis, and multifocal cerebrovascular injury consisting of perivascular edema, abnormal angiogenesis and perivascular extracellular matrix deposition. Herein we demonstrate that RIBI is associated with white matter-specific up-regulation of hypoxia-associated lactate dehydrogenase A (LDHA) and that increased gene expression of fibronectin 1 (FN1), SERPINE1 and matrix metalloprotease 2 (MMP2) may contribute to cerebrovascular remodeling in late-delayed RIBI. Additionally, vascular stability and maturation associated tumor necrosis super family member 15 (TNFSF15) and vascular endothelial growth factor beta (VEGFB) mRNAs were increased within temporal white matter. We also demonstrate that radiation-induced brain injury is associated with decreases in white matter-specific expression of neurotransmitter receptors SYP, GRIN2A and GRIA4. We additionally provide evidence that macrophage/microglial mediated neuroinflammation may contribute to RIBI through increased gene expression of the macrophage chemoattractant CCL2 and macrophage/microglia associated CD68. Global patterns in cerebral gene expression varied significantly between regions examined (P < 0.0001, Friedman's test), with effects most prominent within cerebral white matter.


Subject(s)
Brain Injuries/physiopathology , Cerebrovascular Disorders/etiology , Cerebrovascular Disorders/physiopathology , Cranial Irradiation/adverse effects , Epilepsy/physiopathology , Radiation Injuries/physiopathology , Animals , Brain Injuries/etiology , Brain Injuries/pathology , Cerebrovascular Disorders/pathology , Epilepsy/etiology , Epilepsy/pathology , Humans , Macaca mulatta , Male , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/radiation effects , Radiation Injuries/etiology , Radiation Injuries/pathology , Radiotherapy Dosage , White Matter/pathology , White Matter/physiopathology , White Matter/radiation effects
9.
Cancer Res ; 76(17): 4970-80, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27364556

ABSTRACT

Brain metastasis is one of the chief causes of mortality in breast cancer patients, but the mechanisms that drive this process remain poorly understood. Here, we report that brain metastatic cells expressing high levels of c-Met promote the metastatic process via inflammatory cytokine upregulation and vascular reprogramming. Activated c-Met signaling promoted adhesion of tumor cells to brain endothelial cells and enhanced neovascularization by inducing the secretion of IL8 and CXCL1. Additionally, stimulation of IL1ß secretion by activation of c-Met induced tumor-associated astrocytes to secrete the c-Met ligand HGF. Thus, a feed-forward mechanism of cytokine release initiated and sustained by c-Met fed a vicious cycle that generated a favorable microenvironment for metastatic cells. Reinforcing our results, we found that pterostilbene, a compound that penetrates the blood-brain barrier, could suppress brain metastasis by targeting c-Met signaling. These findings suggest a potential utility of this natural compound for chemoprevention. Cancer Res; 76(17); 4970-80. ©2016 AACR.


Subject(s)
Brain Neoplasms/secondary , Breast Neoplasms/pathology , Neoplasm Invasiveness/pathology , Proto-Oncogene Proteins c-met/metabolism , Tumor Microenvironment/physiology , Animals , Antineoplastic Agents/pharmacology , Cell Adhesion/physiology , Cell Line, Tumor , Female , Heterografts , Humans , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Nude , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Signal Transduction/physiology , Stilbenes/pharmacology , Transcriptome
10.
BMC Cancer ; 15: 427, 2015 May 23.
Article in English | MEDLINE | ID: mdl-26002762

ABSTRACT

BACKGROUND: Proper functional association between mural cells and endothelial cells (EC) causes EC of blood vessels to become quiescent. Mural cells on tumor vessels exhibit decreased attachment to EC, which allows vessels to be unstable and proliferative. The mechanisms by which tumors prevent proper association between mural cells and EC are not well understood. Since gap junctions (GJ) play an important role in cell-cell contact and communication, we investigated whether loss of GJ plays a role in tumor-induced mural cell dissociation. METHODS: Mural cell regulation of endothelial proliferation was assessed by direct co-culture assays of fluorescently labeled cells quantified by flow cytometry or plate reader. Gap junction function was assessed by parachute assay. Connexin 43 (Cx43) protein in mural cells exposed to conditioned media from cancer cells was assessed by Western and confocal microscopy; mRNA levels were assessed by quantitative real-time PCR. Expression vectors or siRNA were utilized to overexpress or knock down Cx43. Tumor growth and angiogenesis was assessed in mouse hosts deficient for Cx43. RESULTS: Using parachute dye transfer assay, we demonstrate that media conditioned by MDA-MB-231 breast cancer cells diminishes GJ communication between mural cells (vascular smooth muscle cells, vSMC) and EC. Both protein and mRNA of the GJ component Connexin 43 (Cx43) are downregulated in mural cells by tumor-conditioned media; media from non-tumorigenic MCF10A cells had no effect. Loss of GJ communication by Cx43 siRNA knockdown, treatment with blocking peptide, or exposure to tumor-conditioned media diminishes the ability of mural cells to inhibit EC proliferation in co-culture assays, while overexpression of Cx43 in vSMC restores GJ and endothelial inhibition. Breast tumor cells implanted into mice heterozygous for Cx43 show no changes in tumor growth, but exhibit significantly increased tumor vascularization determined by CD31 staining, along with decreased mural cell support detected by NG2 staining. CONCLUSIONS: Our data indicate that i) functional Cx43 is required for mural cell-induced endothelial quiescence, and ii) downregulation of Cx43 GJ by tumors frees endothelium to respond to angiogenic cues. These data define a novel and important role for maintained Cx43 function in regulation of vessel quiescence, and suggest its loss may contribute to pathological tumor angiogenesis.


Subject(s)
Breast Neoplasms/metabolism , Connexin 43/metabolism , Endothelial Cells/physiology , Endothelium, Vascular/physiopathology , Gap Junctions/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Breast Neoplasms/blood supply , Cell Communication , Cell Line, Tumor , Cell Proliferation , Connexin 43/antagonists & inhibitors , Connexin 43/genetics , Culture Media, Conditioned , Humans , Mice , Neovascularization, Pathologic/physiopathology , RNA, Messenger/metabolism
11.
Radiat Res ; 182(5): 580-5, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25338095

ABSTRACT

Future space missions are expected to include increased extravehicular activities (EVAs) during which astronauts are exposed to high-energy space radiation while breathing 100% oxygen. Given that brain irradiation can lead to cognitive impairment, and that oxygen is a potent radiosensitizer, there is a concern that astronauts may be at greater risk of developing cognitive impairment when exposed to space radiation while breathing 100% O(2) during an EVA. To address this concern, unanesthetized, unrestrained, young adult male Fischer 344 × Brown Norway rats were allowed to breathe 100% O(2) for 30 min prior to, during and 2 h after whole-body irradiation with 0, 1, 3, 5 or 7 Gy doses of 18 MV X rays delivered from a medical linear accelerator at a dose rate of ~425 mGy/min. Irradiated and unirradiated rats breathing air (~21% O(2)) served as controls. Cognitive function was assessed 9 months postirradiation using the perirhinal cortex-dependent novel object recognition task. Cognitive function was not impaired until the rats breathing either air or 100% O(2) received a whole-body dose of 7 Gy. However, at all doses, cognitive function of the irradiated rats breathing 100% O(2) was improved over that of the irradiated rats breathing air. These data suggest that astronauts are not at greater risk of developing cognitive impairment when exposed to space radiation while breathing 100% O(2) during an EVA.


Subject(s)
Cognition Disorders/etiology , Cosmic Radiation/adverse effects , Oxygen/administration & dosage , Animals , Male , Rats , Rats, Inbred BN , Rats, Inbred F344 , Respiration , Space Flight
12.
Int J Radiat Biol ; 90(9): 790-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24937374

ABSTRACT

PURPOSE: To determine if the brain's response to single doses predicts its response to 'biologically equivalent' fractionated doses. METHODS: Young adult male Fischer 344 rats were whole-brain irradiated with either single 11, 14, or 16.5 Gy doses of (137)Cs γ rays or their 'biologically equivalent' 20, 30, or 40 Gy fractionated doses (fWBI) delivered in 5 Gy fractions, twice/week for 2, 3, or 4 weeks, respectively. At 2 months post-irradiation, cellular markers of inflammation (total, activated, and newborn microglia) and neurogenesis (newborn neurons) were measured in 40 µm sections of the dentate gyrus (DG). RESULTS: Although the total number of microglia in the DG/hilus was not significantly different (p > 0.7) in unirradiated, single dose, and fWBI rats, single doses produced a significant (p < 0.003) increase in the percent-activated microglia; fWBI did not (p > 0.1). Additionally, single doses produced a significant (p < 0.002) dose-dependent increase in surviving newborn microglia; fWBI did not (p < 0.8). Although total proliferation in the DG was reduced equally by single and fWBI doses, single doses produced a significant dose-dependent (p < 0.02) decrease in surviving newborn neurons; fWBI did not (p > 0.6). CONCLUSIONS: These data demonstrate that the rat brain's cellular response to single doses often does not predict its cellular response to 'biologically equivalent' fWBI doses.


Subject(s)
Brain/diagnostic imaging , Cesium Radioisotopes/chemistry , Animals , Cell Proliferation/radiation effects , Dentate Gyrus/radiation effects , Dose Fractionation, Radiation , Dose-Response Relationship, Radiation , Gamma Rays , Hippocampus/radiation effects , Inflammation/radiotherapy , Male , Microglia/pathology , Neurogenesis/radiation effects , Neurons/radiation effects , Radionuclide Imaging , Rats , Rats, Inbred F344
13.
Radiat Res ; 181(1): 21-32, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24377717

ABSTRACT

Fractionated partial or whole-brain irradiation is the primary treatment for metastatic brain tumors. Despite reducing tumor burden and increasing lifespan, progressive, irreversible cognitive impairment occurs in >50% of the patients who survive >6 months after fractionated whole-brain irradiation. The exact mechanism(s) responsible for this radiation-induced brain injury are unknown; however, preclinical studies suggest that radiation modulates the extracellular receptor kinase signaling pathway, which is associated with cognitive impairment in many neurological diseases. In the study reported here, we demonstrated that the extracellular receptor kinase transcriptionally-regulated early response gene, Homer1a, was up-regulated transiently in the hippocampus and down-regulated in the cortex of young adult male Fischer 344 X Brown Norway rats at 48 h after 40 Gy of fractionated whole-brain irradiation. Two months after fractionated whole-brain irradiation, these changes in Homer1a expression correlated with a down-regulation of the hippocampal glutamate receptor 1 and protein kinase Cγ, and an up-regulation of cortical glutamate receptor 1 and protein kinase Cγ. Two drugs that prevent radiation-induced cognitive impairment in rats, the angiotensin type-1 receptor blocker, L-158,809, and the angiotensin converting enzyme inhibitor, ramipril, reversed the fractionated whole-brain irradiation-induced Homer1a expression at 48 h in the hippocampus and cortex and restored glutamate receptor 1 and protein kinase Cγ to the levels in sham-irradiated controls at 2 months after fractionated whole-brain irradiation. These data indicate that Homer1a is, (1) a brain region specific regulator of radiation-induced brain injury, including cognitive impairment and (2) potentially a druggable target for preventing it.


Subject(s)
Brain Injuries/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Gene Expression Regulation/radiation effects , Hippocampus/metabolism , Hippocampus/radiation effects , Radiation Injuries/metabolism , Animals , Brain Injuries/genetics , Cognition/drug effects , Cognition/radiation effects , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Hippocampus/physiopathology , Homer Scaffolding Proteins , Imidazoles/pharmacology , Male , Protein Kinase C/metabolism , Radiation Injuries/genetics , Ramipril/pharmacology , Rats , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , Tetrazoles/pharmacology , Time Factors
14.
Free Radic Biol Med ; 65: 1060-1068, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24012919

ABSTRACT

About 500,000 new cancer patients will develop brain metastases in 2013. The primary treatment modality for these patients is partial or whole brain irradiation which leads to a progressive, irreversible cognitive impairment. Although the exact mechanisms behind this radiation-induced brain injury are unknown, neuroinflammation in glial populations is hypothesized to play a role. Blockers of the renin-angiotensin system (RAS) prevent radiation-induced cognitive impairment and modulate radiation-induced neuroinflammation. Recent studies suggest that RAS blockers may reduce inflammation by increasing endogenous concentrations of the anti-inflammatory heptapeptide angiotensin-(1-7) [Ang-(1-7)]. Ang-(1-7) binds to the AT(1-7) receptor and inhibits MAP kinase activity to prevent inflammation. This study describes the inflammatory response to radiation in astrocytes characterized by radiation-induced increases in (i) IL-1ß and IL-6 gene expression; (ii) COX-2 and GFAP immunoreactivity; (iii) activation of AP-1 and NF-κB transcription factors; and (iv) PKCα, MEK, and ERK (MAP kinase) activation. Treatment with U-0126, a MEK inhibitor, demonstrates that this radiation-induced inflammation in astrocytes is mediated through the MAP kinase pathway. Ang-(1-7) inhibits radiation-induced inflammation, increases in PKCα, and MAP kinase pathway activation (phosphorylation of MEK and ERK). Additionally Ang-(1-7) treatment leads to an increase in dual specificity phosphatase 1 (DUSP1). Furthermore, treatment with sodium vanadate (Na3VO4), a phosphatase inhibitor, blocks Ang-(1-7) inhibition of radiation-induced inflammation and MAP kinase activation, suggesting that Ang-(1-7) alters phosphatase activity to inhibit radiation-induced inflammation. These data suggest that RAS blockers inhibit radiation-induced inflammation and prevent radiation-induced cognitive impairment not only by reducing Ang II but also by increasing Ang-(1-7) levels.


Subject(s)
Angiotensin I/pharmacology , Astrocytes/immunology , MAP Kinase Signaling System , Peptide Fragments/pharmacology , Radiation-Protective Agents/pharmacology , Animals , Astrocytes/radiation effects , Cells, Cultured , Drug Evaluation, Preclinical , Dual Specificity Phosphatase 1/metabolism , Inflammation/metabolism , Primary Cell Culture , Rats
15.
Cancer Microenviron ; 6(1): 1-17, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22467426

ABSTRACT

The induction of tumor vasculature, known as the 'angiogenic switch', is a rate-limiting step in tumor progression. Normal blood vessels are composed of two distinct cell types: endothelial cells which form the channel through which blood flows, and mural cells, the pericytes and smooth muscle cells which serve to support and stabilize the endothelium. Most functional studies have focused on the responses of endothelial cells to pro-angiogenic stimuli; however, there is mounting evidence that the supporting mural cells, particularly pericytes, may play key regulatory roles in both promoting vessel growth as well as terminating vessel growth to generate a mature, quiescent vasculature. Tumor vessels are characterized by numerous structural and functional abnormalities, including altered association between endothelial cells and pericytes. These dysfunctional, unstable vessels contribute to hypoxia, interstitial fluid pressure, and enhanced susceptibility to metastatic invasion. Increasing evidence points to the pericyte as a critical regulator of endothelial activation and subsequent vessel development, stability, and function. Here we discuss both the stimulatory and inhibitory effects of pericytes on the vasculature and the possible utilization of vessel normalization as a therapeutic strategy to combat cancer.

16.
J Neurooncol ; 110(3): 381-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23001361

ABSTRACT

We investigate the variance in patterns of failure after Gamma Knife™ radiosurgery (GKRS) for patients with brain metastases based on the subtype of the primary breast cancer. Between 2000 and 2010, 154 breast cancer patients were treated with GKRS for brain metastases. Tumor subtypes were approximated based on hormone receptor (HR) and HER2 status of the primary cancer: Luminal A/B (HR+/HER2(-)); HER2 (HER2+/HR(-)); Luminal HER2 (HR+/HER2+), Basal (HR(-)/HER2(-)), and then based on HER2 status alone. The median follow-up period was 54 months. Kaplan-Meier method was used to estimate survival times. Multivariable analysis was performed using Cox regression models. Median number of lesions treated was two (range 1-15) with a median dose of 20 Gy (range 9-24 Gy). Median overall survival (OS) was 7, 9, 11 and 22 months for Basal, Luminal A/B, HER2, and Luminal HER2, respectively (p = 0.001), and was 17 and 8 months for HER2+ and HER(-) patients, respectively (p < 0.001). Breast cancer subtype did not predict time to local failure (p = 0.554), but did predict distant brain failure rate (76, 47, 47, 36 % at 1 year for Basal, Luminal A/B, HER2, and Luminal HER2 respectively, p < 0.001). An increased proportion of HER2+ patients experienced neurologic death (46 vs 31 %, p = 0.066). Multivariate analysis revealed that HER2+ patients (p = 0.007) independently predicted for improved survival. Women with basal subtype have high rates of distant brain failure and worsened survival. Our data suggest that differences in biologic behavior of brain metastasis occur across breast cancer subtypes.


Subject(s)
Brain Neoplasms/mortality , Breast Neoplasms/mortality , Carcinoma, Basal Cell/mortality , Radiosurgery , Adult , Aged , Brain Neoplasms/classification , Brain Neoplasms/secondary , Brain Neoplasms/surgery , Breast Neoplasms/classification , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Carcinoma, Basal Cell/classification , Carcinoma, Basal Cell/pathology , Carcinoma, Basal Cell/surgery , Female , Follow-Up Studies , Humans , Middle Aged , Neoplasm Staging , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Salvage Therapy , Survival Rate , Treatment Failure , Young Adult
17.
J Cell Biochem ; 113(4): 1142-51, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22095586

ABSTRACT

Reciprocal growth factor exchange between endothelial and malignant cells within the tumor microenvironment may directly stimulate neovascularization; however, the role of host vasculature in regulating tumor cell activity is not well understood. While previous studies have examined the angiogenic response of endothelial cells to tumor-secreted factors, few have explored tumor response to endothelial cells. Using an in vitro co-culture system, we investigated the influence of endothelial cells on the angiogenic phenotype of breast cancer cells. Specifically, VEGF, ANG1, and ANG2 gene and protein expression were assessed. When co-cultured with microvascular endothelial cells (HMEC-1), breast cancer cells (MDA-MB-231) significantly increased expression of ANG2 mRNA (20-fold relative to MDA-MB-231 monoculture). Moreover, MDA-MB-231/HMEC-1 co-cultures produced significantly increased levels of ANG2 (up to 580 pg/ml) and VEGF protein (up to 38,400 pg/ml) while ANG1 protein expression was decreased relative to MDA-MB-231 monocultures. Thus, the ratio of ANG1:ANG2 protein, a critical indicator of neovascularization, shifted in favor of ANG2, a phenomenon known to correlate with vessel destabilization and sprouting in vivo. This angiogenic response was not observed in nonmalignant breast epithelial cells (MCF-10A), where absolute protein levels of MCF-10A/HMEC-1 co-cultures were an order of magnitude less than that of the MDA-MB-231/HMEC-1 co-cultures. Results were further verified with a functional angiogenesis assay demonstrating well-defined microvascular endothelial cell (TIME) tube formation when cultured in media collected from MDA-MB-231/HMEC-1 co-cultures. This study demonstrates that the angiogenic activity of malignant mammary epithelial cells is significantly enhanced by the presence of endothelial cells.


Subject(s)
Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Breast Neoplasms/pathology , Endothelium, Vascular/cytology , Vascular Endothelial Growth Factor A/metabolism , Angiopoietin-1/genetics , Angiopoietin-2/genetics , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics
18.
Cancer Res ; 70(21): 8319-28, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20837666

ABSTRACT

Angiotensin-(1-7) [Ang-(1-7)] is an endogenous 7-amino acid peptide hormone of the renin-angiotensin system that has antiproliferative properties. In this study, Ang-(1-7) inhibited the growth of cancer-associated fibroblasts (CAF) and reduced fibrosis in the tumor microenvironment. A marked decrease in tumor volume and weight was observed in orthotopic human breast tumors positive for the estrogen receptor (BT-474 or ZR-75-1) and HER2 (BT-474) following Ang-(1-7) administration to athymic mice. Ang-(1-7) concomitantly reduced interstitial fibrosis in association with a significant decrease in collagen I deposition, along with a similar reduction in perivascular fibrosis. In CAFs isolated from orthotopic breast tumors, the heptapeptide markedly attenuated in vitro growth as well as reduced fibronectin, transforming growth factor-ß (TGF-ß), and extracellular signal-regulated kinase 1/2 kinase activity. An associated increase in the mitogen-activated protein kinase (MAPK) phosphatase DUSP1 following treatment with Ang-(1-7) suggested a potential mechanism by which the heptapeptide reduced MAPK signaling. Consistent with these in vitro observations, immunohistochemical analysis of Ang-(1-7)-treated orthotopic breast tumors revealed reduced TGF-ß and increased DUSP1. Together, our findings indicate that Ang-(1-7) targets the tumor microenvironment to inhibit CAF growth and tumor fibrosis.


Subject(s)
Angiotensin I/pharmacology , Antihypertensive Agents/pharmacology , Breast Neoplasms/prevention & control , Carcinoma, Ductal, Breast/prevention & control , Lung Diseases, Interstitial/prevention & control , Peptide Fragments/pharmacology , Animals , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Dual Specificity Phosphatase 1/metabolism , Female , Fibronectins/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Fibrosis/prevention & control , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Lung Diseases, Interstitial/metabolism , Lung Diseases, Interstitial/pathology , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured
19.
Semin Ophthalmol ; 21(1): 49-58, 2006.
Article in English | MEDLINE | ID: mdl-16517446

ABSTRACT

Vascular endothelial growth inhibitor (VEGI; TNFSF-15) is a new member of the tumor necrosis factor family. VEGI is predominantly an endothelial cell-specific gene, and recombinant VEGI is a potent inhibitor of endothelial cell proliferation, angiogenesis and tumor growth. VEGI exerts two activities on endothelial cells: early G1 arrest of G0/G1-cells responding to growth stimuli, and programmed death of proliferating cells. These activities are highly specific to endothelial cells. There are three VEGI isoforms identified thus far. One of the isoforms, VEGI-251, is a secreted protein. The gene products apparently play a role in normal vasculature, as the transcripts are found in normal adult tissues and some fetal tissues. VEGI gene expression is subject to regulation by inflammatory cytokines. VEGI is also able to regulate the expression of several important genes involved in angiogenesis. These findings are consistent with the view that VEGI functions as an autocrine cytokine to inhibit angiogenesis and stabilize the vasculature.


Subject(s)
Gene Expression , Neovascularization, Physiologic/physiology , Tumor Necrosis Factor-alpha/genetics , Animals , Cell Proliferation , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Tumor Necrosis Factor Ligand Superfamily Member 15
20.
Mol Cancer Ther ; 4(4): 537-46, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15827326

ABSTRACT

A naturally occurring, cocoa-derived pentameric procyanidin (pentamer) was previously shown to cause G0/G1 cell cycle arrest in human breast cancer cells by an unknown molecular mechanism. Here, we show that pentamer selectively inhibits the proliferation of human breast cancer cells (MDA MB-231, MDA MB-436, MDA MB-468, SKBR-3, and MCF-7) and benzo(a)pyrene-immortalized 184A1N4 and 184B5 cells. In contrast, normal human mammary epithelial cells in primary culture and spontaneously immortalized MCF-10A cells were significantly resistant. We evaluated whether this differential response to pentamer may involve depolarization of the mitochondrial membrane. Pentamer caused significant depolarization of mitochondrial membrane in MDA MB231 cells but not the more normal MCF-10A cells, whereas other normal and tumor cell lines tested gave variable results. Further investigations, using a proteomics approach with pentamer-treated MDA MB-231, revealed a specific dephosphorylation, without changes in protein expression, of several G1-modulatory proteins: Cdc2 (at Tyr15), forkhead transcription factor (at Ser256, the Akt phosphorylation site) and p53 (Ser392). Dephosphorylation of p53 (at Ser392) by pentamer was confirmed in MDA MB-468 cells. However, both expression and phosphorylation of retinoblastoma protein were decreased after pentamer treatment. Our results show that breast cancer cells are selectively susceptible to the cytotoxic effects of pentameric procyanidin, and suggest that inhibition of cellular proliferation by this compound is associated with the site-specific dephosphorylation or down-regulation of several cell cycle regulatory proteins.


Subject(s)
Biflavonoids/chemistry , Biflavonoids/therapeutic use , Breast Neoplasms/drug therapy , Catechin/chemistry , Catechin/therapeutic use , Malvaceae/metabolism , Plant Extracts/metabolism , Proanthocyanidins/chemistry , Proanthocyanidins/therapeutic use , Antioxidants/pharmacology , Benzo(a)pyrene/pharmacology , Breast/cytology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Epithelial Cells/cytology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoblotting , Membrane Potentials , Mitochondria/metabolism , Phosphorylation , Plant Extracts/therapeutic use , Receptors, Estrogen/metabolism , Retinoblastoma Protein/metabolism , Serine/chemistry , Time Factors , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...