Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 13(4)2021 Feb 20.
Article in English | MEDLINE | ID: mdl-33672628

ABSTRACT

Breast cancer (BrCa) relies on specific microRNAs to drive disease progression. Oncogenic miR-21 is upregulated in many cancers, including BrCa, and is associated with poor survival and treatment resistance. We sought to determine the role of miR-21 in BrCa tumor initiation, progression and treatment response. In a triple-negative BrCa model, radiation exposure increased miR-21 in both primary tumor and metastases. In vitro, miR-21 knockdown decreased survival in all BrCa subtypes in the presence of radiation. The role of miR-21 in BrCa initiation was evaluated by implanting wild-type miR-21 BrCa cells into genetically engineered mouse models where miR-21 was intact, heterozygous or globally ablated. Tumors were unable to grow in the mammary fat pads of miR-21-/- mice, and grew in ~50% of miR-21+/- and 100% in miR-21+/+ mice. The contribution of miR-21 to progression and metastases was tested by crossing miR-21-/- mice with mice that spontaneously develop BrCa. The global ablation of miR-21 significantly decreased the tumorigenesis and metastases of BrCa, while sensitizing tumors to radio- and chemotherapeutic agents via Fas/FasL-dependent apoptosis. Therefore, targeting miR-21 alone or in combination with various radio or cytotoxic therapies may represent novel and efficacious therapeutic modalities for the future treatment of BrCa patients.

2.
Int J Radiat Oncol Biol Phys ; 110(5): 1341-1349, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33647370

ABSTRACT

Outcomes for triple negative breast cancer (TNBC) are poor and may be improved by increasing CD8+ tumor infiltrating lymphocytes (TIL) to augment antitumor immunity. Radiation (RT) can promote immunogenic cell death with increased antitumor T cell activity but also stimulates suppressive regulatory T cells (Tregs). Because metabolic alterations affect immune homeostasis and prior studies show caloric restriction (CR) combined with RT improves preclinical TNBC outcomes, we hypothesized that CR augments RT, in part, by altering intratumoral immunity. Using an in vivo model of TNBC, we treated mice with ad libitum (AL) diet, radiation, a CR diet, or CR + RT, and demonstrated an immune suppressive environment with a significant increase in CD4+ CD25+Foxp3+ Tregs after RT but not in CR-fed mice. CD8:Treg ratio in CR + RT TIL increased 4-fold compared with AL + RT mice. In vivo CD8 depletion was performed to assess the role of effector T cells in mitigating the effects of CR, and it was found that in mice undergoing CR, depletion of CD8 T cells resulted in increased tumor progression and decreased median survival compared with isotype control-treated mice. In addition, PD-1 expression on CD3+CD8+ T cells within the tumor microenvironment was significantly increased in CR + RT versus AL + RT treated mice as per immunofluorescence. Serum from breast cancer patients undergoing RT alone or CR and RT was collected pre- and postintervention, and a cytokine array demonstrated that patients treated with CR + RT had notable decreases in immunosuppressive cytokines such as IL-2Rγ, IL-10Rß, and TGF-ß2 and 3 compared with patients receiving RT alone. In conclusion, combining CR with RT decreases intratumoral Tregs, increases CD8:Treg, and increases PD-1 expression via a process dependent on CD8 T cells in a TNBC model. Breast cancer patients undergoing CR concurrently with RT also had significant reduction in immunosuppressive cytokine levels compared with those receiving RT alone.


Subject(s)
Caloric Restriction , Lymphocytes, Tumor-Infiltrating/radiation effects , T-Lymphocytes, Regulatory/radiation effects , Triple Negative Breast Neoplasms/radiotherapy , Tumor Microenvironment/radiation effects , Adult , Aged , Animals , CD4-Positive T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/radiation effects , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/radiation effects , Combined Modality Therapy/methods , Disease Progression , Female , Flow Cytometry , Forkhead Transcription Factors , Humans , Interleukin Receptor Common gamma Subunit/blood , Interleukin-10 Receptor beta Subunit/blood , Interleukin-2 Receptor alpha Subunit , Lymphocyte Depletion/methods , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred BALB C , Middle Aged , Programmed Cell Death 1 Receptor/metabolism , Random Allocation , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta2/blood , Transforming Growth Factor beta3/blood , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/mortality , Tumor Microenvironment/immunology
3.
Int J Mol Sci ; 23(1)2021 Dec 24.
Article in English | MEDLINE | ID: mdl-35008602

ABSTRACT

Understanding metabolic and immune regulation inherent to patient populations is key to improving the radiation response for our patients. To date, radiation therapy regimens are prescribed based on tumor type and stage. Patient populations who are noted to have a poor response to radiation such as those of African American descent, those who have obesity or metabolic syndrome, or senior adult oncology patients, should be considered for concurrent therapies with radiation that will improve response. Here, we explore these populations of breast cancer patients, who frequently display radiation resistance and increased mortality rates, and identify the molecular underpinnings that are, in part, responsible for the radiation response and that result in an immune-suppressive tumor microenvironment. The resulting immune phenotype is discussed to understand how antitumor immunity could be improved. Correcting nutrient deficiencies observed in these populations should be considered as a means to improve the therapeutic index of radiation therapy.


Subject(s)
Breast Neoplasms/radiotherapy , Diet , Nutrients , Black or African American , Female , Humans , Metabolic Syndrome , Obesity , Treatment Outcome
4.
Nutr Metab (Lond) ; 17: 23, 2020.
Article in English | MEDLINE | ID: mdl-32211051

ABSTRACT

BACKGROUND: Metastatic cancer is incurable and understanding the molecular underpinnings is crucial to improving survival for our patients. The IGF-1/Akt signaling pathway is often impaired in cancer leading to its progression and metastases. Diet modification is known to alter the IGF-1/Akt pathway and affect the expression of microRNA involved in tumor initiation, growth and metastases. Liver metastases are one of the most common type of metastases in breast and colon cancer. In the present study, we looked at the effect of diet modification on the expression of microRNA in normal liver and liver with breast cancer metastases using in vivo model. METHODOLOGY: 6-month-old C57BL/6 J mice were put on either an ad libitum (AL) diet, or 40% calorie restricted (CR) diet or were fasted for 24 h (FA) before sacrifice. MicroRNA array analysis, western blot and qRT-PCR were performed using liver tissue to compare the treatment groups. A breast cancer model was also used to study the changes in microRNA expression in liver of a group of BALB/c mice orthotopically injected with 4 T1 cells in the mammary fat pad, put on either an AL or 30% CR diet. Liver and primary tumor tissues were used to perform qRT-PCR to compare the treatment groups. RESULTS: MicroRNA array analysis showed significant changes in miRNA expression in both CR and FA conditions in normal liver. Expression of miR-29 and miR-30 family members was increased in both CR and FA. Western blot analysis of the normal liver tissue showed that CR and FA downregulated the IGF-1/Akt pathway and qRT-PCR showed that the expression of miR-29b, miR-29c, miR-30a and miR-30b were increased with CR and FA. Liver tissue collected from mice in the breast cancer model showed an increase in expression of miR-29b, miR-29c and miR-30b while tumor tissue showed increased expression of miR-29c, miR-30a and miR-30b. DISCUSSION: Members of the miR-29 family are known to target and suppress IGF-1, while members of the miR-30 family are known to target and suppress both IGF-1 and IGF-1R. In the present study, we observe that calorie restriction increased the expression of miR-29 and miR-30 in both the normal liver as well as the liver with breast cancer metastases. These findings suggest that dietary alterations may play a role in the treatment of liver metastasis, which should be evaluated further.

5.
Int J Radiat Oncol Biol Phys ; 104(5): 1165-1174, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31039423

ABSTRACT

PURPOSE: Radiation therapy is an essential intervention used in the treatment of more than half of cancer patients. With the increasing use of hypofractionated radiation regimens, concurrent use of radiation and chemotherapy, targeted agents and immunotherapy, the risk of radiation-induced toxicities is increased. However, much remains unknown about the molecular underpinnings responsible for radiation-induced toxicity. MicroRNA (miRNA) are small, non-coding RNA involved in post-transcriptional regulation of gene expression. miR-21 is an oncomiR that is dysregulated in a significant fraction of human malignancies, and its overexpression is linked to poor overall survival, chemoresistance, and radioresistance in several human cancers. However, the contribution of miR-21 in governing radiation sensitivity in normal, untransformed cells, and the impact of silencing this miRNA in normal tissues remains largely unexplored. MATERIALS AND METHODS: miR-21 levels were evaluated in tissues by qRT-PCR without and after total body irradiation (TBI). Mice lacking miR-21 were genetically engineered, subjected to TBI, and monitored for survival. Hematopoietic stem and progenitor cell (HSPC) numbers and function were assessed using flow cytometry, histology, complete blood cell counts, and bone marrow transplantation. RESULTS: miR-21 expression was increased in radiosensitive tissues, but not in radioinsensitive tissues following TBI in wild-type mice, suggesting it may have a critical function in the normal tissue response to irradiation. Compared to wild-type mice, mice lacking one or both alleles of miR-21 showed reduced numbers of HSPCs and increased sensitivity to an LD50/30 dose of TBI with evidence of bone marrow failure. Transplantation of wild-type bone marrow into irradiated miR-21-deficient mice rescued the mice from death. CONCLUSIONS: Our data identify miR-21 as a critical component of HSPC viability and essential for bone marrow recovery following irradiation. Further investigation is warranted to determine whether miR-21 can be used to stratify patients at risk for hematopoietic toxicity following irradiation.


Subject(s)
Cell Survival/radiation effects , Hematopoietic Stem Cells/radiation effects , MicroRNAs/metabolism , Radiation Tolerance , Animals , Cause of Death , Female , Genetic Engineering , Hematopoietic Stem Cell Transplantation , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Radiation Exposure , Up-Regulation , Whole-Body Irradiation
6.
Pigment Cell Melanoma Res ; 27(2): 297-308, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24354797

ABSTRACT

We investigated the importance of the insulin-like growth factor-1 receptor (IGF-1R) in hepatic metastases of uveal melanoma. The expression pattern of IGF-1R in archival tissue samples of hepatic metastasis from 24 patients was analyzed by immunohistochemistry. All the samples of hepatic metastases stained positive for IGF-1R. To investigate the biological role of IGF-1R on the growth of metastatic uveal melanoma, a long-term cell line obtained from a hepatic metastasis (TJU-UM001) was evaluated. TJU-UM001 expressed cell surface IGF-1R (>90%) and proliferated in response to exogenous and endogenous insulin-like growth factor-1 (IGF-1). Correlatively, anti-IGF-1R antibody completely blocked IGF-1-induced growth of TJU-UM001 cells. IGF-1 preferentially induced phosphorylation of Akt (S473) in quiescent TJU-UM001 cells, and this was blocked by anti-IGF-1R antibody. This study suggests that autocrine and paracrine mechanisms underlie IGF-1-induced growth of metastatic uveal melanoma and underscore the potential benefit of IGF-1 or IGF-1R antagonism in treatment for metastatic uveal melanoma.


Subject(s)
Autocrine Communication , Melanoma/metabolism , Melanoma/pathology , Paracrine Communication , Receptor, IGF Type 1/metabolism , Uveal Neoplasms/metabolism , Uveal Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Antibodies/pharmacology , Autocrine Communication/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Insulin-Like Growth Factor I/pharmacology , Male , Melanoma/enzymology , Middle Aged , Neoplasm Metastasis , Paracrine Communication/drug effects , Signal Transduction/drug effects , Uveal Neoplasms/enzymology
7.
J Cell Physiol ; 228(7): 1482-6, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23254450

ABSTRACT

The v-src oncogene is one of only two oncogenes capable of transforming mouse embryo fibroblasts (MEFs) lacking the IGF-IR gene (R-cells). R-/v-src cells grow robustly in the absence of serum, suggesting the hypothesis that they may produce one or more growth factors that would sustain their ability to proliferate in serum-free condition. Using proteomic approaches on serum-free conditioned media derived from v-src-transformed cells, we have identified two growth promoting factors: ostepontin and proliferin. Subsequent experiments have indicated that osteopontin plays a prevalent role in promoting growth of v-src-transformed cells in serum-deprived condition.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Genes, src , Glycoproteins/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Osteopontin/biosynthesis , Animals , Cell Line , Culture Media, Serum-Free , Gene Knockdown Techniques , Glycoproteins/antagonists & inhibitors , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Mice , Osteopontin/antagonists & inhibitors , Osteopontin/genetics , Prolactin , Proteomics , Receptor, IGF Type 1/deficiency , Receptor, IGF Type 1/genetics , Signal Transduction
8.
Cell Cycle ; 10(12): 1956-9, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21558809

ABSTRACT

The mammalian homolog of the Drosophila dachshund gene (DACH1) has been reported as a tumor suppressor in human breast and prostate cancers. It downregulates the epidermal growth factor receptor (EGFR) and cyclin D1. The signaling pathway of the type 1 insulin-like growth factor receptor (IGF-IR) is known to be responsible for the development of resistance to treatment of human cancer with antibodies to the EGFR. We have asked whether DACH1 still exerts its tumor suppressor activity in cells dependent on the IGF-IR for growth. We find that in cells growing in IGF-1 (and unresponsive to EGF), DACH1 is devoid of tumor suppressor activity.


Subject(s)
Drug Resistance, Neoplasm , Eye Proteins/physiology , Receptor, IGF Type 1/physiology , Transcription Factors/physiology , Animals , Cell Line , Cell Line, Tumor , Humans , Insulin-Like Growth Factor I/pharmacology , Mice , Signal Transduction/physiology
9.
J Cell Physiol ; 224(3): 658-63, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20578241

ABSTRACT

The insulin receptor substrate-1 (IRS-1) and c-met, the receptor for the hepatocyte growth factor (HGF) co-immuno-precipitate from lysates treated with the respective antibodies. The interaction between IRS-1 and c-met requires a tyrosyl phosphorylated IRS-1 and results in reciprocal down-regulation. IRS-1 inhibits cell motility, while the activated c-met promotes it. These and other results suggest an explanation for reports in the literature indicating that c-met levels are high and IRS-1 levels are low in human cancer metastases.


Subject(s)
Down-Regulation , Insulin Receptor Substrate Proteins/metabolism , Proto-Oncogene Proteins c-met/metabolism , Animals , Cell Line , Cell Movement/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Hepatocyte Growth Factor/metabolism , Humans , Insulin Receptor Substrate Proteins/genetics , Mice , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/genetics , Signal Transduction/physiology
10.
J Cell Physiol ; 220(2): 485-91, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19391107

ABSTRACT

MicroRNA 145 (miR145) has been proposed as a tumor suppressor. It was previously shown that miR145 targets the 3' UTR of the insulin receptor substrate-1 (IRS-1) and dramatically inhibits the growth of colon cancer cells. miR145 also targets the type 1 insulin-like growth factor receptor (IGF-IR). We show here that an IRS-1 lacking its 3' UTR is no longer down-regulated by miR145 and rescues colon cancer cells from miR145-induced inhibition of growth. An IGF-IR resistant to miR145 (again by elimination of its 3' UTR) is not down-regulated by miR145 but fails to rescue colon cancer cells from growth inhibition. These and other results, taken together, indicate that down-regulation of IRS-1 plays a significant role in the tumor suppressor activity of miR145.


Subject(s)
Insulin Receptor Substrate Proteins/metabolism , MicroRNAs/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction/physiology , 3' Untranslated Regions , 3T3 Cells , Active Transport, Cell Nucleus/physiology , Animals , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Insulin Receptor Substrate Proteins/genetics , Mice , MicroRNAs/genetics , Receptor, IGF Type 1/genetics
11.
J Cell Physiol ; 217(1): 281-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18506777

ABSTRACT

The insulin receptor substrate-1 (IRS-1), a docking protein of the type 1 insulin-like growth factor receptor (IGF-IR) plays a significant role in cell proliferation and differentiation. The expression of IRS-1 is down-regulated in mouse embryo fibroblasts (MEFs) with a deletion of caveolin-1 (cav1) genes (KO cells). Levels of IRS-1 mRNA are not affected. Re-introduction of cav1 into KO cells rescues IRS-1 expression. Stabilization of protein levels is reciprocal and a strict correlation between IRS-1 and cav1 levels was confirmed in five cell lines, and in mouse tissues. IRS-1 binds through its phosphotyrosine binding (PTB) domain to tyrosine 14 (Y14) of cav1, the residue phosphorylated by IGF-1 stimulation and by v-src. The down-regulation of IRS-1 in cav-/- cells occurs via the proteasome pathway. These results indicate a novel mechanism for the regulation of IRS-1 expression levels, an important finding in view of IRS-1 role in cell proliferation and transformation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Caveolin 1/metabolism , Fibroblasts/metabolism , Gene Expression Regulation , Adaptor Proteins, Signal Transducing/genetics , Animals , Blotting, Western , Cell Differentiation/physiology , Cell Proliferation , Down-Regulation , Embryo, Mammalian , Fibroblasts/cytology , Gene Expression , Immunoprecipitation , Insulin Receptor Substrate Proteins , Mice , Mice, Knockout , RNA Interference , RNA, Messenger/analysis , RNA, Small Interfering , Transfection
12.
J Biol Chem ; 282(45): 32582-90, 2007 Nov 09.
Article in English | MEDLINE | ID: mdl-17827156

ABSTRACT

The insulin receptor substrate-1 (IRS-1), a docking protein for both the type 1 insulin-like growth factor receptor (IGF-IR) and the insulin receptor, is known to send a mitogenic, anti-apoptotic, and anti-differentiation signal. Several micro RNAs (miRs) are suggested by the data base as possible candidates for targeting IRS-1. We show here that one of the miRs predicted by the data base, miR145, whether transfected as a synthetic oligonucleotide or expressed from a plasmid, causes down-regulation of IRS-1 in human colon cancer cells. IRS-1 mRNA is not decreased by miR145, while it is down-regulated by an siRNA targeting IRS-1. Targeting of the IRS-1 3'-untranslated region (UTR) by miR145 was confirmed using a reporter gene (luciferase) expressing the miR145 binding sites of the IRS-1 3'-UTR. In agreement with the role of IRS-1 in cell proliferation, we show that treatment of human colon cancer cells with miR145 causes growth arrest comparable to the use of an siRNA against IRS-1. Taken together, these results identify miR145 as a micro RNA that down-regulates the IRS-1 protein, and inhibits the growth of human cancer cells.


Subject(s)
Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , MicroRNAs/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Base Sequence , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/genetics , Down-Regulation , Gene Expression Regulation, Neoplastic , Genes, Reporter/genetics , Humans , Insulin Receptor Substrate Proteins , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...