Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-29264372

ABSTRACT

Programmed cell death, or apoptosis, occurs in nearly all tissues of all multi-cellular organisms. In order to avoid leakage of intracellular contents, which could generate tissue damaging inflammation, apoptotic cells are cleared from tissues by phagocytes, which then dispatch the engulfed dying cell through the lysosomal pathway. Phagocytic clearance of apoptotic cells is referred to as efferocytosis. One key feature of efferocytosis is the production and release of wound healing cytokines by the phagocyte, which acts to resolve inflammation, and promote tissue repair. Phagocytic engulfment of apoptotic cells coupled with cytokine modulation aimed at immune suppression ensures that physiological programmed cell death does not induce inflammation and tissue damage. However, cytokines involved in wound healing and immune suppression are notorious for their role in the tumor microenvironment, increasing tumor cell motility and promoting evasion of anti-tumor immunity. Therefore, current and future studies aimed at targeting important players of efferocytosis should reveal new and efficacious therapeutic approaches for limiting cancer progression and relapse.

2.
J Clin Invest ; 124(11): 4737-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25250573

ABSTRACT

Breast cancers that occur in women 2-5 years postpartum are more frequently diagnosed at metastatic stages and correlate with poorer outcomes compared with breast cancers diagnosed in young, premenopausal women. The molecular mechanisms underlying the malignant severity associated with postpartum breast cancers (ppBCs) are unclear but relate to stromal wound-healing events during postpartum involution, a dynamic process characterized by widespread cell death in milk-producing mammary epithelial cells (MECs). Using both spontaneous and allografted mammary tumors in fully immune-competent mice, we discovered that postpartum involution increases mammary tumor metastasis. Cell death was widespread, not only occurring in MECs but also in tumor epithelium. Dying tumor cells were cleared through receptor tyrosine kinase MerTK-dependent efferocytosis, which robustly induced the transcription of genes encoding wound-healing cytokines, including IL-4, IL-10, IL-13, and TGF-ß. Animals lacking MerTK and animals treated with a MerTK inhibitor exhibited impaired efferocytosis in postpartum tumors, a reduction of M2-like macrophages but no change in total macrophage levels, decreased TGF-ß expression, and a reduction of postpartum tumor metastasis that was similar to the metastasis frequencies observed in nulliparous mice. Moreover, TGF-ß blockade reduced postpartum tumor metastasis. These data suggest that widespread cell death during postpartum involution triggers efferocytosis-induced wound-healing cytokines in the tumor microenvironment that promote metastatic tumor progression.


Subject(s)
Lung Neoplasms/secondary , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/pathology , Animals , Apoptosis , Coculture Techniques , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , MCF-7 Cells , Male , Mammary Glands, Animal/physiopathology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mice, Transgenic , Neoplasm Transplantation , Phagocytosis , Postpartum Period , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Tumor Burden , Up-Regulation , c-Mer Tyrosine Kinase
3.
J Clin Invest ; 123(10): 4329-43, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23999432

ABSTRACT

Aberrant regulation of the erythroblastosis oncogene B (ErbB) family of receptor tyrosine kinases (RTKs) and their ligands is common in human cancers. ErbB3 is required in luminal mammary epithelial cells (MECs) for growth and survival. Since breast cancer phenotypes may reflect biological traits of the MECs from which they originate, we tested the hypothesis that ErbB3 drives luminal breast cancer growth. We found higher ERBB3 expression and more frequent ERBB3 gene copy gains in luminal A/B breast cancers compared with other breast cancer subtypes. In cell culture, ErbB3 increased growth of luminal breast cancer cells. Targeted depletion of ErbB3 with an anti-ErbB3 antibody decreased 3D colony growth, increased apoptosis, and decreased tumor growth in vivo. Treatment of clinical breast tumors with the antiendocrine drug fulvestrant resulted in increased ErbB3 expression and PI3K/mTOR signaling. Depletion of ErbB3 in fulvestrant-treated tumor cells reduced PI3K/mTOR signaling, thus decreasing tumor cell survival and tumor growth. Fulvestrant treatment increased phosphorylation of all ErbB family RTKs; however, phospho-RTK upregulation was not seen in tumors treated with both fulvestrant and anti-ErbB3. These data indicate that upregulation of ErbB3 in luminal breast cancer cells promotes growth, survival, and resistance to fulvestrant, thus suggesting ErbB3 as a target for breast cancer treatment.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/metabolism , Estradiol/analogs & derivatives , Estrogen Receptor Modulators/pharmacology , Receptor, ErbB-3/genetics , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Cell Proliferation , Cell Survival , Down-Regulation/drug effects , Drug Resistance, Neoplasm , Drug Synergism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Estradiol/pharmacology , Female , Fulvestrant , Gene Dosage , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Oligonucleotide Array Sequence Analysis , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/immunology , Receptor, ErbB-3/metabolism , Signal Transduction , Survival Analysis , Transcriptome , Xenograft Model Antitumor Assays
4.
J Clin Invest ; 123(8): 3231-42, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23867499

ABSTRACT

MerTK, a receptor tyrosine kinase (RTK) of the TYRO3/AXL/MerTK family, is expressed in myeloid lineage cells in which it acts to suppress proinflammatory cytokines following ingestion of apoptotic material. Using syngeneic mouse models of breast cancer, melanoma, and colon cancer, we found that tumors grew slowly and were poorly metastatic in MerTK-/- mice. Transplantation of MerTK-/- bone marrow, but not wild-type bone marrow, into lethally irradiated MMTV-PyVmT mice (a model of metastatic breast cancer) decreased tumor growth and altered cytokine production by tumor CD11b+ cells. Although MerTK expression was not required for tumor infiltration by leukocytes, MerTK-/- leukocytes exhibited lower tumor cell-induced expression of wound healing cytokines, e.g., IL-10 and growth arrest-specific 6 (GAS6), and enhanced expression of acute inflammatory cytokines, e.g., IL-12 and IL-6. Intratumoral CD8+ T lymphocyte numbers were higher and lymphocyte proliferation was increased in tumor-bearing MerTK-/- mice compared with tumor-bearing wild-type mice. Antibody-mediated CD8+ T lymphocyte depletion restored tumor growth in MerTK-/- mice. These data demonstrate that MerTK signaling in tumor-associated CD11b+ leukocytes promotes tumor growth by dampening acute inflammatory cytokines while inducing wound healing cytokines. These results suggest that inhibition of MerTK in the tumor microenvironment may have clinical benefit, stimulating antitumor immune responses or enhancing immunotherapeutic strategies.


Subject(s)
Colonic Neoplasms/enzymology , Leukocytes/enzymology , Mammary Neoplasms, Experimental/enzymology , Melanoma, Experimental/enzymology , Animals , CD8-Positive T-Lymphocytes/enzymology , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Resistance/immunology , Female , Gene Expression Regulation, Neoplastic , Male , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases , Transcriptome , Tumor Burden , Tumor Microenvironment , c-Mer Tyrosine Kinase
5.
J Clin Invest ; 123(3): 1348-58, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23391723

ABSTRACT

After an initial response to chemotherapy, many patients with triple-negative breast cancer (TNBC) have recurrence of drug-resistant metastatic disease. Studies with TNBC cells suggest that chemotherapy-resistant populations of cancer stem-like cells (CSCs) with self-renewing and tumor-initiating capacities are responsible for these relapses. TGF-ß has been shown to increase stem-like properties in human breast cancer cells. We analyzed RNA expression in matched pairs of primary breast cancer biopsies before and after chemotherapy. Biopsies after chemotherapy displayed increased RNA transcripts of genes associated with CSCs and TGF-ß signaling. In TNBC cell lines and mouse xenografts, the chemotherapeutic drug paclitaxel increased autocrine TGF-ß signaling and IL-8 expression and enriched for CSCs, as indicated by mammosphere formation and CSC markers. The TGF-ß type I receptor kinase inhibitor LY2157299, a neutralizing TGF-ß type II receptor antibody, and SMAD4 siRNA all blocked paclitaxel-induced IL8 transcription and CSC expansion. Moreover, treatment of TNBC xenografts with LY2157299 prevented reestablishment of tumors after paclitaxel treatment. These data suggest that chemotherapy-induced TGF-ß signaling enhances tumor recurrence through IL-8-dependent expansion of CSCs and that TGF-ß pathway inhibitors prevent the development of drug-resistant CSCs. These findings support testing a combination of TGF-ß inhibitors and anticancer chemotherapy in patients with TNBC.


Subject(s)
Breast Neoplasms/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Quinolines/pharmacology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Gene Expression/drug effects , Gene Knockdown Techniques , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Paclitaxel/pharmacology , RNA, Small Interfering/genetics , Receptor, ErbB-2/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Signal Transduction , Smad4 Protein/genetics , Smad4 Protein/metabolism , Spheroids, Cellular/metabolism , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/physiology , Xenograft Model Antitumor Assays
6.
Cancer Res ; 72(10): 2672-82, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22461506

ABSTRACT

Increasing evidence suggests that HER2-amplified breast cancer cells use HER3/ErbB3 to drive therapeutic resistance to HER2 inhibitors. However, the role of ErbB3 in the earliest events of breast epithelial transformation remains unknown. Using mouse mammary specific models of Cre-mediated ErbB3 ablation, we show that ErbB3 loss prevents the progressive transformation of HER2-overexpressing mammary epithelium. Decreased proliferation and increased apoptosis were seen in MMTV-HER2 and MMTV-Neu mammary glands lacking ErbB3, thus inhibiting premalignant HER2-induced hyperplasia. Using a transgenic model in which HER2 and Cre are expressed from a single polycistronic transcript, we showed that palpable tumor penetrance decreased from 93.3% to 6.7% upon ErbB3 ablation. Penetrance of ductal carcinomas in situ was also decreased. In addition, loss of ErbB3 impaired Akt and p44/42 phosphorylation in preneoplastic HER2-overexpressing mammary glands and in tumors, decreased growth of preexisting HER2-overexpressing tumors, and improved tumor response to the HER2 tyrosine kinase inhibitor lapatinib. These events were rescued by reexpression of ErbB3, but were only partially rescued by ErbB36F, an ErbB3 mutant harboring six tyrosine-to-phenylalanine mutations that block its interaction with phosphatidyl inositol 3-kinase. Taken together, our findings suggest that ErbB3 promotes HER2-induced changes in the breast epithelium before, during, and after tumor formation. These results may have important translational implications for the treatment and prevention of HER2-amplified breast tumors through ErbB3 inhibition.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic , Receptor, ErbB-2/physiology , Receptor, ErbB-3/physiology , Adenocarcinoma/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Epithelium/metabolism , Female , Humans , Hyperplasia/metabolism , Mice , Mice, Nude , Mice, Transgenic , Phosphatidylinositol 3-Kinases/metabolism
7.
J Biol Chem ; 287(16): 13457-64, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22389505

ABSTRACT

Recent studies suggest that sphingolipid metabolism is altered during type 2 diabetes. Increased levels of the sphingolipid ceramide are associated with insulin resistance. However, a role for sphingolipids in pancreatic beta cell function, or insulin production, and release remains to be established. Our studies in MIN6 cells and mouse pancreatic islets demonstrate that glucose stimulates an intracellular rise in the sphingolipid, sphingosine 1-phosphate (S1P), whereas the levels of ceramide and sphingomyelin remain unchanged. The increase in S1P levels by glucose is due to activation of sphingosine kinase 2 (SphK2). Interestingly, rises in S1P correlate with increased glucose-stimulated insulin secretion (GSIS). Decreasing S1P levels by treatment of MIN6 cells or primary islets with the sphingosine kinase inhibitor reduces GSIS. Moreover, knockdown of SphK2 alone results in decreased GSIS, whereas knockdown of the S1P phosphatase, Sgpp1, leads to a rise in GSIS. Treatment of mice with the sphingosine kinase inhibitor impairs glucose disposal due to decreased plasma insulin levels. Altogether, our data suggest that glucose activates SphK2 in pancreatic beta cells leading to a rise in S1P levels, which is important for GSIS.


Subject(s)
Glucose Intolerance/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Cell Line, Tumor , Glucose/pharmacology , Glucose Tolerance Test , Injections, Intraperitoneal , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulinoma , Lysophospholipids/pharmacology , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RNA, Small Interfering/pharmacology , Sphingosine/metabolism , Sphingosine/pharmacology
8.
Cancer Res ; 71(11): 3941-51, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21482676

ABSTRACT

The ErbB receptor family member ErbB3 has been implicated in breast cancer growth, but it has yet to be determined whether its disruption is therapeutically valuable. In a mouse model of mammary carcinoma driven by the polyomavirus middle T (PyVmT) oncogene, the ErbB2 tyrosine kinase inhibitor lapatinib reduced the activation of ErbB3 and Akt as well as tumor cell growth. In this phosphatidylinositol-3 kinase (PI3K)-dependent tumor model, ErbB2 is part of a complex containing PyVmT, p85 (PI3K), and ErbB3, that is disrupted by treatment with lapatinib. Thus, full engagement of PI3K/Akt by ErbB2 in this oncogene-induced mouse tumor model may involve its ability to dimerize with and phosphorylate ErbB3, which itself directly binds PI3K. In this article, we report that ErbB3 is critical for PI3K/Akt-driven tumor formation triggered by the PyVmT oncogene. Tissue-specific, Cre-mediated deletion of ErbB3 reduced Akt phosphorylation, primary tumor growth, and pulmonary metastasis. Furthermore, EZN-3920, a chemically stabilized antisense oligonucleotide that targets the ErbB3 mRNA in vivo, produced similar effects while causing no toxicity in the mouse model. Our findings offer further preclinical evidence that ErbB3 ablation may be therapeutically effective in tumors where ErbB3 engages PI3K/Akt signaling.


Subject(s)
Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/prevention & control , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-3/deficiency , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/prevention & control , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Female , Humans , Mammary Glands, Animal/enzymology , Mammary Neoplasms, Experimental/genetics , Mammary Tumor Virus, Mouse , Mice , Mice, Transgenic , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...