Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Cancers (Basel) ; 14(9)2022 Apr 29.
Article in English | MEDLINE | ID: mdl-35565370

ABSTRACT

Endometriosis, a painful gynecological condition accompanied by inflammation in women of reproductive age, is associated with an increased risk of ovarian cancer. We evaluated the role of peritoneal heme accumulated during menstrual cycling, as well as peritoneal and lesional macrophage phenotype, in promoting an oncogenic microenvironment. We quantified the heme-degrading enzyme, heme oxygenase-1 (HO-1, encoded by Hmox1) in normal peritoneum, endometriotic lesions and endometriosis-associated ovarian cancer (EAOC) of clear cell type (OCCC). HO-1 was expressed primarily in macrophages and increased in endometrioma and OCCC tissues relative to endometriosis and controls. Further, we compared cytokine expression profiles in peritoneal macrophages (PM) and peripheral blood mononuclear cells (PBMC) in women with endometriosis versus controls as a measure of a tumor-promoting environment in the peritoneum. We found elevated levels of HO-1 along with IL-10 and the pro-inflammatory cytokines (IL-1ß, IL-16, IFNγ) in PM but not in PBMC from endometriosis patients. Using LysM-Cre:Hmox1flfl conditional knockout mice, we show that a deficiency of HO-1 in macrophages led to the suppression of growth of ID8 ovarian tumors implanted into the peritoneum. The restriction of ID8 ovarian tumor growth was associated with an increased number of Mac3+ macrophage and B cells in LysM-Cre:Hmox1flfl mice compared to controls. Functional experiments in ovarian cancer cell lines show that HO-1 is induced by heme. Low levels of exogenous heme promoted ovarian cancer colony growth in soft agar. Higher doses of heme led to slower cancer cell colony growth in soft agar and the induction of HO-1. These data suggest that perturbation of heme metabolism within the endometriotic niche and in cancer cells themselves may be an important factor that influences tumor initiation and growth.

2.
Cell Rep ; 32(12): 108181, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32966797

ABSTRACT

Hemopexin (Hx) is a scavenger of labile heme. Herein, we present data defining the role of tumor stroma-expressed Hx in suppressing cancer progression. Labile heme and Hx levels are inversely correlated in the plasma of patients with prostate cancer (PCa). Further, low expression of Hx in PCa biopsies characterizes poorly differentiated tumors and correlates with earlier time to relapse. Significantly, heme promotes tumor growth and metastases in an orthotopic murine model of PCa, with the most aggressive phenotype detected in mice lacking Hx. Mechanistically, labile heme accumulates in the nucleus and modulates specific gene expression via interacting with guanine quadruplex (G4) DNA structures to promote PCa growth. We identify c-MYC as a heme:G4-regulated gene and a major player in heme-driven cancer progression. Collectively, these results reveal that sequestration of labile heme by Hx may block heme-driven tumor growth and metastases, suggesting a potential strategy to prevent and/or arrest cancer dissemination.


Subject(s)
Heme/metabolism , Hemopexin/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , DNA/genetics , Disease Progression , G-Quadruplexes , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Metastasis , Phenotype , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/blood , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/metabolism , Treatment Outcome , Urokinase-Type Plasminogen Activator/metabolism
3.
Cell Death Dis ; 10(2): 72, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30683864

ABSTRACT

Phenotypic changes of myeloid cells are critical to the regulation of premature aging, development of cancer, and responses to infection. Heme metabolism has a fundamental role in the regulation of myeloid cell function and activity. Here, we show that deletion of heme oxygenase-1 (HO-1), an enzyme that removes heme, results in an impaired DNA damage response (DDR), reduced cell proliferation, and increased cellular senescence. We detected increased levels of p16INK4a, H2AXγ, and senescence-associated-ß-galactosidase (SA-ß-Gal) in cells and tissues isolated from HO-1-deficient mice. Importantly, deficiency of HO-1 in residential macrophages in chimeric mice results in elevated DNA damage and senescence upon radiation-induced injury. Mechanistically, we found that mammalian target of rapamycin (mTOR)/S6 protein signaling is critical for heme and HO-1-regulated phenotype of macrophages. Collectively, our data indicate that HO-1, by detoxifying heme, blocks p16INK4a expression in macrophages, preventing DNA damage and cellular senescence.


Subject(s)
Cell Proliferation/genetics , Heme Oxygenase-1/metabolism , Heme/metabolism , Macrophages/metabolism , Membrane Proteins/metabolism , Radiation Injuries, Experimental/immunology , Radiation Injuries, Experimental/metabolism , Animals , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Damage/genetics , Gene Knockdown Techniques , Heme Oxygenase-1/genetics , Membrane Proteins/genetics , Mice , Mice, Transgenic , Phenotype , RAW 264.7 Cells , TOR Serine-Threonine Kinases/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...