Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Curr Res Transl Med ; 71(3): 103401, 2023.
Article in English | MEDLINE | ID: mdl-37364351

ABSTRACT

Breast implant-associated anaplastic large cell lymphoma (BIA-ALCL) is an uncommon T-cell lymphoma type with distinct clinical, molecular and genetic features. Establishment of BIA-ALCL cell lines and patient-derived xenograft (PDX) models are essential experimental tools to investigate the molecular pathogenesis of the disease. We characterized a novel BIA-ALCL cell line and PDX model, named BIA-XR1, derived from a patient with textured breast implant who developed lymphoma. Next-generation sequencing revealed a STAT3 mutation, commonly detected in BIA-ALCL, and a unique KRAS mutation reported for the first time in this lymphoma type. Both JAK/STAT3 and RAS/MEK/ERK oncogenic pathways were activated in BIA-XR1, which are targetable with clinically available agents.

2.
Cancers (Basel) ; 12(8)2020 Jul 22.
Article in English | MEDLINE | ID: mdl-32707827

ABSTRACT

Background: Cathepsin L (Ctsl) is a cysteine protease mainly located within the endosomal/lysosomal cell compartment. High expression of Ctsl indicates poor prognosis in human breast cancer. However, the cell type-specific Ctsl functions responsible for this association remain elusive. Methods: Because constitutive Ctsl-/- mice develop a complex phenotype, we developed a conditional model allowing for cell type-specific inactivation of Ctsl in mammary epithelium or myeloid cells in the transgenic mouse mammary tumor virus (MMTV)-polyoma middle T (PyMT) breast cancer model. Results: Ctsl ablation in mammary epithelial cells resulted in delayed initiation and end-stage of cancers. The latter displayed large dead cell areas. Inducible in vitro deletion of Ctsl in MMTV-PyMT-derived breast cancer cells revealed expansion of the acidic cell compartment, alteration of intracellular amino acid levels, and impaired mTOR signaling. In consequence, Ctsl-deficient cells exhibited slow growth rates and high apoptosis susceptibility. In contrast to Ctsl-deficient mammary epithelium, selective knockout of Ctsl in myeloid cells had no effects on primary tumors, but promoted lung metastasis formation. Conclusions: Our cell type-specific in vivo analysis provides strong evidence for a cancer cell-intrinsic, tumor-promoting role of Ctsl in primary breast cancer, whereas metastasis is negatively regulated by Ctsl expressed by bone marrow-derived cells.

3.
Nat Commun ; 11(1): 5133, 2020 10 12.
Article in English | MEDLINE | ID: mdl-33046706

ABSTRACT

Cathepsin D (CTSD) is a lysosomal protease and a marker of poor prognosis in breast cancer. However, the cells responsible for this association and the function of CTSD in cancer are still incompletely understood. By using a conditional CTSD knockout mouse crossed to the transgenic MMTV-PyMT breast cancer model we demonstrate that CTSD deficiency in the mammary epithelium, but not in myeloid cells, blocked tumor development in a cell-autonomous manner. We show that lack of CTSD impaired mechanistic Target of Rapamycin Complex 1 (mTORC1) signaling and induced reversible cellular quiescence. In line, CTSD-deficient tumors started to grow with a two-month delay and quiescent Ctsd-/- tumor cells re-started proliferation upon long-term culture. This was accompanied by rewiring of oncogenic gene expression and signaling pathways, while mTORC1 signaling remained permanently disabled in CTSD-deficient cells. Together, these studies reveal a tumor cell-autonomous effect of CTSD deficiency, and establish a pivotal role of this protease in the cellular response to oncogenic stimuli.


Subject(s)
Breast Neoplasms/metabolism , Cathepsin D/genetics , Epithelium/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Animals , Breast Neoplasms/genetics , Cathepsin D/deficiency , Female , Humans , Mammary Glands, Animal/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
4.
FEBS J ; 284(10): 1437-1454, 2017 05.
Article in English | MEDLINE | ID: mdl-27926992

ABSTRACT

Lysosomal cathepsins are proteolytic enzymes increasingly recognized as prognostic markers and potential therapeutic targets in a variety of diseases. In those conditions, the cathepsins are mostly overexpressed, thereby driving the respective pathogenic processes. Although less known, there are also diseases with a genetic deficiency of cathepsins. In fact, nowadays 6 of the 15 human proteases called 'cathepsins' have been linked to inherited syndromes. However, only three of these syndromes are typical lysosomal storage diseases, while the others are apparently caused by defective cleavage of specific protein substrates. Here, we will provide an introduction on lysosomal cathepsins, followed by a brief description of the clinical symptoms of the various genetic diseases. For each disease, we focus on the known mutations of which many have been only recently identified by modern genome sequencing approaches. We further discuss the effect of the respective mutation on protease structure and activity, the resulting pathogenesis, and possible therapeutic strategies.


Subject(s)
Cathepsins/metabolism , Animals , Cathepsins/genetics , Humans , Mutation/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/metabolism , Papillon-Lefevre Disease/genetics , Papillon-Lefevre Disease/metabolism , Pycnodysostosis/genetics , Pycnodysostosis/metabolism
5.
Biochimie ; 122: 219-26, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26232697

ABSTRACT

Cathepsin D (Ctsd) is a ubiquitously expressed aspartic protease functioning primarily in the acidic endosomal/lysosomal cell compartment. At an age of 26 ± 1 days, mice with constitutive Ctsd deficiency (Ctsd(-/-)) die from a neurodegenerative lysosomal storage disease equivalent to the congenital neuronal ceroid lipofuscinosis (NCL) type 10 in humans. In addition to neurodegeneration, Ctsd(-/-) mice exhibit a loss of CD4(+)/CD8(+)-double-positive thymocytes and an atrophy of the intestinal mucosa. To date, it is not understood if and how these phenotypes are triggering each other. In addition, the cell type causing initiation of NCL in Ctsd(-/-) mice has not been identified yet. To investigate the tissue- and cell type-specific functions of Ctsd, we generated a novel conditional Ctsd allele by flanking the second exon with loxP sites. We compared a ubiquitous Ctsd deletion with a deletion of the protease by a Nestin-promoter controlled Cre-recombinase expression in cells of neuroectodermal origin, e.g. in neurons and astroglia, but not in microglia. First, we confirmed absence of Ctsd in the respective cell- and tissue types. The neuroectoderm specific knock-out mice survived about 5.5 days longer than the mice with ubiquitous Ctsd deletion, which was in line with the progress in brain histopathology. Atrophies of thymus and small intestine were delayed to similar extend. The conditional Ctsd knock-out mouse model established in this study not only demonstrates that this type of NCL is initiated by cells of neuroectodermal origin, but will also help to further study tissue-specific functions of Ctsd in vivo.


Subject(s)
Cathepsin D/deficiency , Disease Models, Animal , Ectoderm/metabolism , Neuronal Ceroid-Lipofuscinoses/enzymology , Animals , Astrocytes/enzymology , Astrocytes/metabolism , Atrophy/genetics , Blotting, Western , Cathepsin D/genetics , Gene Deletion , Gene Expression Regulation, Enzymologic , Humans , Immunohistochemistry , Intestine, Small/enzymology , Intestine, Small/metabolism , Intestine, Small/pathology , Mice, Knockout , Neural Tube/metabolism , Neuronal Ceroid-Lipofuscinoses/genetics , Neurons/enzymology , Neurons/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Thymus Gland/enzymology , Thymus Gland/metabolism , Thymus Gland/pathology , Time Factors
6.
Oncotarget ; 5(24): 12646-64, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25504435

ABSTRACT

The major threat in prostate cancer is the occurrence of metastases in androgen-independent tumor stage, for which no causative cure is available. Here we show that metastatic behavior of androgen-independent prostate tumor cells requires the protein-kinase-C-related kinase (PRK1/PKN1) in vitro and in vivo. PRK1 regulates cell migration and gene expression through its kinase activity, but does not affect cell proliferation. Transcriptome and interactome analyses uncover that PRK1 regulates expression of migration-relevant genes by interacting with the scaffold protein sperm-associated antigen 9 (SPAG9/JIP4). SPAG9 and PRK1 colocalize in human cancer tissue and are required for p38-phosphorylation and cell migration. Accordingly, depletion of either ETS domain-containing protein Elk-1 (ELK1), an effector of p38-signalling or p38 depletion hinders cell migration and changes expression of migration-relevant genes as observed upon PRK1-depletion. Importantly, a PRK1 inhibitor prevents metastases in mice, showing that the PRK1-pathway is a promising target to hamper prostate cancer metastases in vivo. Here we describe a novel mechanism controlling the metastatic behavior of PCa cells and identify PRK1 as a promising therapeutic target to treat androgen-independent metastatic prostate cancer.


Subject(s)
Androgens/metabolism , Cell Movement/physiology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Kinase C/metabolism , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Proliferation/physiology , Humans , Male , Mice , Neoplasm Metastasis , Phosphorylation , Prostatic Neoplasms/genetics , Protein Kinase C/genetics , Transcriptome , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL