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.
Cancers (Basel) ; 13(22)2021 Nov 11.
Article in English | MEDLINE | ID: mdl-34830790

ABSTRACT

In previous studies, we reported that progesterone receptor membrane component 1 (PGRMC1) is implicated in progestin signaling and possibly associated with increased breast cancer risk upon combined hormone replacement therapy. To gain mechanistic insight, we searched for potential PGRMC1 interaction partners upon progestin treatment by co-immunoprecipitation and mass spectrometry. The interactions with the identified partners were further characterized with respect to PGRMC1 phosphorylation status and with emphasis on the crosstalk between PGRMC1 and estrogen receptor α (ERα). We report that PGRMC1 overexpression resulted in increased proliferation of hormone receptor positive breast cancer cell lines upon treatment with a subgroup of progestins including norethisterone and dydrogesterone that promote PGRMC1-phosphorylation on S181. The ERα modulators prohibitin-1 (PHB1) and prohibitin-2 (PHB2) interact with PGRMC1 in dependency on S181-phosphorylation upon treatment with the same progestins. Moreover, increased interaction between PGRMC1 and PHBs correlated with decreased binding of PHBs to ERα and subsequent ERα activation. Inhibition of either PGRMC1 or ERα abolished this effect. In summary, we provide strong evidence that activated PGRMC1 associates with PHBs, competitively removing them from ERα, which then can develop its transcriptional activities on target genes. This study emphasizes the role of PGRMC1 in a key breast cancer signaling pathway which may provide a new avenue to target hormone-dependent breast cancer.

3.
Breast Cancer Res ; 22(1): 75, 2020 07 13.
Article in English | MEDLINE | ID: mdl-32660617

ABSTRACT

BACKGROUND: PGRMC1 (progesterone receptor membrane component 1) is a highly conserved heme binding protein, which is overexpressed especially in hormone receptor-positive breast cancer and plays an important role in breast carcinogenesis. Nevertheless, little is known about the mechanisms by which PGRMC1 drives tumor progression. The aim of our study was to investigate the involvement of PGRMC1 in cholesterol metabolism to detect new mechanisms by which PGRMC1 can increase lipid metabolism and alter cancer-related signaling pathways leading to breast cancer progression. METHODS: The effect of PGRMC1 overexpression and silencing on cellular proliferation was examined in vitro and in a xenograft mouse model. Next, we investigated the interaction of PGRMC1 with enzymes involved in the cholesterol synthesis pathway such as CYP51, FDFT1, and SCD1. Further, the impact of PGRMC1 expression on lipid levels and expression of enzymes involved in lipid homeostasis was examined. Additionally, we assessed the role of PGRMC1 in key cancer-related signaling pathways including EGFR/HER2 and ERα signaling. RESULTS: Overexpression of PGRMC1 resulted in significantly enhanced proliferation. PGRMC1 interacted with key enzymes of the cholesterol synthesis pathway, alters the expression of proteins, and results in increased lipid levels. PGRMC1 also influenced lipid raft formation leading to altered expression of growth receptors in membranes of breast cancer cells. Analysis of activation of proteins revealed facilitated ERα and EGFR activation and downstream signaling dependent on PGRMC1 overexpression in hormone receptor-positive breast cancer cells. Depletion of cholesterol and fatty acids induced by statins reversed this growth benefit. CONCLUSION: PGRMC1 may mediate proliferation and progression of breast cancer cells potentially by altering lipid metabolism and by activating key oncogenic signaling pathways, such as ERα expression and activation, as well as EGFR signaling. Our present study underlines the potential of PGRMC1 as a target for anti-cancer therapy.


Subject(s)
Breast Neoplasms/metabolism , Membrane Proteins/metabolism , Receptors, Progesterone/metabolism , Animals , Apoptosis/physiology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis , Cell Proliferation/physiology , Disease Progression , Female , Heterografts , Homeostasis , Humans , Lipid Metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/genetics , Tumor Cells, Cultured
4.
BMC Mol Cell Biol ; 21(1): 26, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32293262

ABSTRACT

BACKGROUND: Progesterone receptor membrane component 1 (PGRMC1) is often elevated in cancers, and exists in alternative states of phosphorylation. A motif centered on PGRMC1 Y180 was evolutionarily acquired concurrently with the embryological gastrulation organizer that orchestrates vertebrate tissue differentiation. RESULTS: Here, we show that mutagenic manipulation of PGRMC1 phosphorylation alters cell metabolism, genomic stability, and CpG methylation. Each of several mutants elicited distinct patterns of genomic CpG methylation. Mutation of S57A/Y180/S181A led to increased net hypermethylation, reminiscent of embryonic stem cells. Pathways enrichment analysis suggested modulation of processes related to animal cell differentiation status and tissue identity, as well as cell cycle control and ATM/ATR DNA damage repair regulation. We detected different genomic mutation rates in culture. CONCLUSIONS: A companion manuscript shows that these cell states dramatically affect protein abundances, cell and mitochondrial morphology, and glycolytic metabolism. We propose that PGRMC1 phosphorylation status modulates cellular plasticity mechanisms relevant to early embryological tissue differentiation.


Subject(s)
Phosphorylation , Receptors, Progesterone , Animals , Cell Differentiation , Cell Line , DNA Methylation , Disease , Embryology , Epigenomics , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Mice , Mutation , Mutation Rate , Protein Processing, Post-Translational , Receptors, Progesterone/biosynthesis , Receptors, Progesterone/metabolism
5.
BMC Mol Cell Biol ; 21(1): 24, 2020 Apr 03.
Article in English | MEDLINE | ID: mdl-32245408

ABSTRACT

BACKGROUND: Progesterone Receptor Membrane Component 1 (PGRMC1) is expressed in many cancer cells, where it is associated with detrimental patient outcomes. It contains phosphorylated tyrosines which evolutionarily preceded deuterostome gastrulation and tissue differentiation mechanisms. RESULTS: We demonstrate that manipulating PGRMC1 phosphorylation status in MIA PaCa-2 (MP) cells imposes broad pleiotropic effects. Relative to parental cells over-expressing hemagglutinin-tagged wild-type (WT) PGRMC1-HA, cells expressing a PGRMC1-HA-S57A/S181A double mutant (DM) exhibited reduced levels of proteins involved in energy metabolism and mitochondrial function, and altered glucose metabolism suggesting modulation of the Warburg effect. This was associated with increased PI3K/AKT activity, altered cell shape, actin cytoskeleton, motility, and mitochondrial properties. An S57A/Y180F/S181A triple mutant (TM) indicated the involvement of Y180 in PI3K/AKT activation. Mutation of Y180F strongly attenuated subcutaneous xenograft tumor growth in NOD-SCID gamma mice. Elsewhere we demonstrate altered metabolism, mutation incidence, and epigenetic status in these cells. CONCLUSIONS: Altogether, these results indicate that mutational manipulation of PGRMC1 phosphorylation status exerts broad pleiotropic effects relevant to cancer and other cell biology.


Subject(s)
Phosphorylation , Receptors, Progesterone , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Shape , Energy Metabolism , Glycolysis , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/metabolism , Neoplasms , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Progesterone/biosynthesis , Receptors, Progesterone/metabolism
6.
Biochem Biophys Res Commun ; 524(1): 64-69, 2020 03 26.
Article in English | MEDLINE | ID: mdl-31980178

ABSTRACT

PGRMC1 is a protein from the MAPR family with a range of cellular functions. PGRMC1 has been described to play a role in fertility, neuroprotection, steroidogenesis, membrane trafficking and in cancer cell biology. PGRMC1 represents a likely key regulator of cell metabolism and proliferation, as well as a potential target for anti-cancer therapies. To further understand the functions of PGRMC1 and the mechanism of the small molecule inhibitor of PGRMC1, AG-205, proteins differentially bound to PGRMC1 were identified following AG-205 treatment of MIA PaCa-2 cells. Our results suggest that AG-205 influences PGRMC1 interactions with the actin cytoskeleton. The binding of two PGRMC1-associated proteins that support this, RACK1 and alpha-Actinin-1, was reduced following AG-205 treatment. The biology associated with PGRMC1 binding partners identified here merits further investigation.


Subject(s)
Actins/metabolism , Indoles/metabolism , Membrane Proteins/antagonists & inhibitors , Receptors, Progesterone/antagonists & inhibitors , Actin Cytoskeleton/metabolism , Cell Line, Tumor , Humans , Mass Spectrometry , Protein Binding , Receptors for Activated C Kinase/metabolism
7.
EJNMMI Res ; 9(1): 75, 2019 Aug 13.
Article in English | MEDLINE | ID: mdl-31410585

ABSTRACT

BACKGROUND: The development and clinical translation of [68Ga] Pentixafor has substantially promoted the relevance of non-invasive PET imaging of CXCR4 expression in a broad spectrum of diseases, including cancer and inflammation. Its pronounced selectivity for the human receptor (hCXCR4), however, precludes the use of [68Ga] Pentixafor for imaging receptor expression and dynamics in CXCR4-related diseases in endogenous mouse models. To overcome this restriction, [125I]CPCR4.3, a structurally related pentapeptide ligand, has been evaluated as a preclinical tool for efficient in vitro and in vivo targeting of hCXCR4 and mCXCR4. RESULTS: Compared to the reference [68Ga] Pentixafor, [125I]CPCR4.3 showed 2.4- to 11-fold increased specific binding to human cancer cell lines with different hCXCR4 expression levels (Jurkat, Daudi, HT-29, SH-5YSY, MCF-7, LNCaP) as well as strong and highly specific binding to mCXCR4 expressing cells (mCXCR4-transfected CHO cells, Eµ-myc 1080, 4 T1), which was not detectable for [68Ga]Pentixafor. This is the consequence of the equally high affinity of iodo-CPCR4 to hCXCR4 and mCXCR4 (IC50 = 5.4 ± 1.5 and 4.9 ± 1.7 nM, respectively) as opposed to [natGa] Pentixafor (hCXCR4: 42.4 ± 11.6 nM, mCXCR4: > 1000 nM). Additionally, [125I]CPCR4.3 showed enhanced tracer internalization (factor of 1.5-2 compared to the reference). In vivo biodistribution studies in immunocompetent Black Six and immunocompromised CD-1 nude mice showed predominant hepatobiliary excretion of [125I]CPCR4.3 (logP = 0.51), leading to high activity levels in liver and intestines. However, [125I]CPCR4.3 also showed high and specific accumulation in organs with endogenous mCXCR4 expression (spleen, lung, adrenals), even at low receptor expression levels. CONCLUSIONS: Due to its excellent hCXCR4 and mCXCR4 targeting efficiency, both in vitro and in vivo, [125I]CPCR4.3 represents a sensitive and reliable tool for the species-independent quantification of CXCR4 expression. Its suboptimal clearance properties will certainly restrict its use for in vivo imaging applications using 123I (for SPECT) or 124I (for PET), but due to its high and specific accumulation in mCXCR4 expressing tissues, [125I]CPCR4.3 holds promise as a powerful preclinical tool for the investigation and quantification of CXCR4 involvement and kinetics in various murine disease models via, e.g., biodistribution and autoradiography studies.

8.
Nanotechnology ; 30(39): 395101, 2019 Sep 27.
Article in English | MEDLINE | ID: mdl-31239418

ABSTRACT

Among various nanoparticles tested for pharmacological applications over the recent years, graphene quantum dots (GQDs) seem to be promising candidates for the construction of drug delivery systems due to their superior biophysical and biochemical properties. The subcellular fate of incorporated nanomaterial is decisive for transporting pharmaceuticals into target cells. Therefore a detailed characterization of the uptake of GQDs into different breast cancer models was performed. The demonstrated accumulation inside the endolysosomal system might be the reason for the particles' low toxicity, but has to be overcome for cytosolic or nuclear drug delivery. Furthermore, the penetration of GQDs into precision-cut mammary tumor slices was studied. These constitute a far closer to reality model system than monoclonal cell lines. The constant uptake into the depth of the tissue slices underlines the systems' potential for drug delivery into solid tumors.


Subject(s)
Breast Neoplasms/metabolism , Graphite/metabolism , Quantum Dots/metabolism , Breast Neoplasms/pathology , Epithelial Cells/metabolism , Graphite/chemistry , Humans , Nanostructures/chemistry , Particle Size , Quantum Dots/chemistry , Subcellular Fractions/metabolism , Tissue Culture Techniques , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...