Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Int J Biol Macromol ; 254(Pt 1): 127596, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37898250

ABSTRACT

Philadelphia-like acute lymphoblastic leukemia (Ph-like ALL) is an aggressive B-ALL malignancy associated with high rates of relapse and inferior survival rate. While targeted treatments against the cell surface proteins CD22 or CD19 have been transformative in the treatment of refractory B-ALL, patients may relapse due to antigen loss, necessitating targeting alternative antigens. Cytokine receptor-like factor 2 (CRLF2) is overexpressed in half of Ph-like ALL cases conferring chemoresistance and enhancement of leukemia cell survival. Therefore, targeting CRLF2 may reduce the likelihood of relapse associated with antigen loss. We developed a CRLF2-targeting single-chain variable fragment modified by the fragment crystallizable region (CRLF2 scFv-Fc) conjugated to a drug maytansinoid 1 (DM1)-DOPC liposomal conjugate, creating homogeneous CRLF2-targeted liposomes (CRLF2-DM1 LIP). Cellular association and internalization studies in a Ph-like ALL cell line, MHH-CALL-4, compared to its lentivirally transduced CRLF2-knockdown counterpart (KD-CALL-4) revealed excellent CRLF2-targeting efficiency of CRLF2-DM1 LIP. Moreover, CRLF2-DM1 LIP showed selective association and internalization ex vivo using Ph-like ALL patient-derived xenograft (PDX) cells with minimal reactivity with non-target cells. Cell apoptosis assays demonstrated the CRLF2-dependent potency of CRLF2-DM1 LIP in Ph-like ALL cell lines. This study is the first to highlight the therapeutic potential of a CRLF2-directed scFv-Fc-liposomal conjugate for targeting Ph-like ALL.


Subject(s)
Immunoconjugates , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Animals , Humans , Immunoglobulin Fragments , Liposomes/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Disease Models, Animal , Immunoconjugates/pharmacology , Recurrence
2.
Biol Chem ; 403(2): 243-249, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34699696

ABSTRACT

Thymic stromal lymphopoietin (TSLP) is a pro-inflammatory cytokine with important pathological roles in Asthma bronchiale, malignant tumours and other diseases. The heterodimeric human TSLP receptor (hTSLPR) consists of the TSLP-binding subunit (TSLPRα) and the IL-7Rα-subunit. We studied the properties of hTSLP variants with mutations in their bipartite interaction interface towards IL-7Rα. One mutant (T46D/K101D) showed only mild impairment in receptor affinity but a massive reduction in biological activity. To facilitate the future development of hTSLP mutants with drug properties, we have devised a eukaryontic cytokine display assay with activity read-out and intrinsic genotype-phenotype coupling.


Subject(s)
Cytokines , Receptors, Cytokine , Cytokines/chemistry , Humans , Interleukin-7 Receptor alpha Subunit , Protein Domains , Receptors, Cytokine/genetics , Thymic Stromal Lymphopoietin
3.
Int J Biol Macromol ; 190: 214-223, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34481852

ABSTRACT

Antibody fragments are promising building blocks for developing targeted therapeutics, thus improving treatment efficacy while minimising off-target toxicity. Despite recent advances in targeted therapeutics, patients with Philadelphia-like acute lymphoblastic leukemia (Ph-like ALL), a high-risk malignancy, lack specific and effective targeted treatments. Cytokine receptor-like factor 2 (CRLF2) is overexpressed in 50% of Ph-like ALL cases, conferring the survival of leukemia blasts through activation of the JAK/STAT signalling pathway. Targeting such a vital cell-surface protein could result in potent anti-leukaemic efficacy and reduce the likelihood of relapse associated with antigen loss. Herein, we developed a novel single-chain variable fragment (scFv) against CRLF2 based on a monoclonal antibody raised against the recombinant extracellular domain of human TSLPRα chain. The scFv fragment demonstrated excellent binding affinity with CRLF2 protein in the nanomolar range. Cellular association studies in vitro using an inducible CRLF2 knockdown cell line and ex vivo using patient-derived xenografts revealed the selective association of the scFv with CRLF2. The fragment exhibited significant receptor antagonistic effects on STAT5 signalling, suggesting possible therapeutic implications in vivo. This study is the first to describe the potential use of a novel scFv for targeting Ph-like ALL.


Subject(s)
Immunoglobulin Fragments/metabolism , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Cytokine/metabolism , Recombinant Proteins/metabolism , Animals , Cell Line, Tumor , Child , Endocytosis , HEK293 Cells , Humans , Mice , Phosphorylation , STAT5 Transcription Factor/metabolism , Signal Transduction , Single-Chain Antibodies/isolation & purification
4.
Oncol Rep ; 46(2)2021 Aug.
Article in English | MEDLINE | ID: mdl-34261292

ABSTRACT

Following the publication of the above article, the authors have requested a change in the authorship on the paper, and the revised list of authors is presented above; essentially, the ninth intended author, Giuseppe Salvo (G.S.), was inadverently omitted from the author list. G.S. contributed towards the T design and the preparation of the tagged ScFv. Therefore, the revised authors' names and affiliations, as they should have been presented in the original version of this paper, are as follows: Elisa Tremante1, Leonardo Sibilio2,7, Fabio Centola2,8, Nadine Knutti3,9, Gerd Holzapfel4, Isabella Manni5, Matteo Allegretti1, Paolo Lombardi6, Giuseppe Salvo2,10, Loredana Cecchetelli2, Karlheinz Friedrich3, Joachim BertraM4 and Patrizio Giacomini1. 1Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, Rome; 2Ibi Lorenzini, Aprilia, Italy; 3University Hospital Jena, Institute of Biochemistry II, Jena; 4IBA GmbH, Göttingen, Germany; 5SAFU, IRCCS Regina Elena National Cancer Institute, Rome; 6NaxosPharma, Novate Milanese, Milan, Italy. Correspondence to: Dr Patrizio Giacomini. Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy. E­mail: patrizio.giacomini@ifo.gov.it. Present address: 7Menarini Biotech, Pomezia, Rome, Italy. Present address: 8Merck Serono Spa, Global Analytical Department, Guidonia Montecelio, Rome, Italy. Present address: 9University Hospital Jena Institute for Clinical Chemistry and Laboratory Diagnostics, Jena, Germany. Present address: 10External Quality Assurance (ExM), MSD Italia S.r.l., Via Vitorchiano 151, 00189 Rome.Italy. Furthermore, the Authors' contributions section should be amended to read as follows: Authors' contributions: ET and MA tested the TOOLBOX concept and performed the flow cytometry experiments. LS, FC, GS and LC designed and prepared the tagged ScFv. NK and KF designed and prepared the GFP promoter­reporter construct. GH and JB designed and manufactured Strep­Tactins. ET and IM performed animal studies. PL designed and manufactured NAX and NAXT compounds. PG conceptualized TOOLBOX and wrote the manuscript with the contribution of all authors. All authors have approved the final version of the manuscript. All the authors agree with the inclusion of Giuseppe Salvo as an author on this paper, and are grateful to the Editor for allowing them the opportunity to publish this Corrigendum. Furthermore, they apologize to the readership of the Journal for any inconvenience caused. [the original article was published in Oncology Reports 45: 77, 2021; DOI: 10.3892/or.2021.8028].

5.
Oncol Rep ; 45(5)2021 05.
Article in English | MEDLINE | ID: mdl-34105341

ABSTRACT

Herein, we describe TOOLBOX, a 3­step modular nano­assembly targeting system that permits the combinatorial exchange of antibody specificities and toxic payloads, introducing modularity in antibody­drug conjugate (ADC) manufacturing. TOOLBOX integrates 3 building blocks: i) a recombinant antibody fragment (that in the selected setting targets the proto­oncogene ERBB2) genetically fused to an 8 amino acid Strep­Tag®; ii) a multivalent protein adapter, called Strep­Tactin®; iii) two anticancer agents, e.g. DNA nanobinders and the maytansinoid DM1, both carrying a chemically attached Strep­Tag that reversibly turns them into inactive prodrugs. Stoichiometrically optimized complexes of Strep­Tagged antibody fragments and drugs, bridged by Strep­Tactin, were specifically uptaken by breast cancer cells expressing ERBB2, and this unexpectedly resulted in conditional prodrug reactivation. A promoter­reporter system showed that TOOLBOX inhibited downstream ERBB2 signaling not only in ERBB2­overexpressing/­amplified SK­BR­3 cells grown in vitro, but also in ERBB2­low/non­amplified BRC230 triple­negative breast carcinoma cells xenotransplanted in nude mice. Thus, TOOLBOX is a modular ADC­like nano­assembly platform for precision oncology.


Subject(s)
Breast Neoplasms/drug therapy , Immunoconjugates/administration & dosage , Nanostructures/administration & dosage , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Breast Neoplasms/immunology , Cell Line, Tumor , Humans , Immunoconjugates/chemistry , Mice , Mice, Nude , Nanostructures/chemistry , Receptor, ErbB-2/immunology , Xenograft Model Antitumor Assays
6.
Sci Rep ; 10(1): 10182, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32576855

ABSTRACT

In this study, we present a straightforward approach for functional cell-based screening by co-encapsulation of secretor yeast cells and reporter mammalian cells in millions of individual agarose-containing microdroplets. Our system is compatible with ultra-high-throughput selection utilizing standard fluorescence-activated cell sorters (FACS) without need of extensive adaptation and optimization. In a model study we co-encapsulated murine interleukin 3 (mIL-3)-secreting S. cerevisiae cells with murine Ba/F3 reporter cells, which express green fluorescent protein (GFP) upon stimulation with mIL-3, and could observe specific and robust induction of fluorescence signal compared to a control with yeast cells secreting a non-functional mIL-3 mutant. We demonstrate the successful enrichment of activating mIL-3 wt-secreting yeast cells from a 1:10,000 dilution in cells expressing the inactive cytokine variant by two consecutive cycles of co-encapsulation and FACS. This indicates the suitability of the presented strategy for functional screening of high-diversity yeast-based libraries and demonstrates its potential for the efficient isolation of clones secreting bioactive recombinant proteins.


Subject(s)
Flow Cytometry/methods , High-Throughput Screening Assays/methods , Mammals/metabolism , Mammals/physiology , Saccharomyces cerevisiae/metabolism , Animals , Cell Line , Fluorescence , Green Fluorescent Proteins/metabolism , Interleukin-3/metabolism , Mice , Microfluidics/methods
7.
Int J Clin Exp Pathol ; 12(5): 1731-1737, 2019.
Article in English | MEDLINE | ID: mdl-31933991

ABSTRACT

Alterations in colonic mucus secretion are linked to the induction and maintenance of inflammation during inflammatory bowel disease (IBD) and its progression to colorectal cancer (CRC). MUC1, a multifunctional glycoprotein, is the best studied cell surface mucin in mouse models of IBD and CRC. However, little information on MUC1 expression and localization in different types of pathologic human colon mucosa is available. In this work, expression and subcellular localization of MUC1 in different types of diseased human colon mucosa from a cohort of Tunisian patients is analyzed and correlated with the type of disorder. Colon tissue samples were obtained from 39 cases of CRC and 18 cases of IBD. 13 cases of normal adjacent colon mucosa tissues served as controls. Biopsies were subjected to immunohistochemical analysis of MUC1 expression. Signals were quantified densitometrically and characterized with regard to tissue and intracellular distribution. Results were then correlated with the different types of colon disorder. Immunohistochemical investigation of MUC1 in a cohort of inflammatory bowel diseases and colorectal cancer showed a significant divergence in the expression of MUC1 in terms intensity (18.96% ± 0.55 vs 27.26% ± 1.24 respectively; P=0.005) and localization between the two types of lesions (30.76% vs 70.96% respectively; P=0.0199). Our findings show divergent characteristic patterns for MUC1 expression and localization in different types of pathologic alterations of the colon mucosa. These results are of potential diagnostic and predictive clinical value.

8.
Cytokine ; 113: 228-237, 2019 01.
Article in English | MEDLINE | ID: mdl-30033138

ABSTRACT

Cytokines and growth factors are signaling proteins involved in communication processes between cells. They are involved in the control of numerous essential physiological processes such as cell proliferation, gene transcription and differentiation; therefore being in the focus of basic and applied research. Many of them are also of relevance for human diseases. When observed as potential targets for pharmacological intervention and objects of structure/function studies, it is important to measure their biological activities, optionally along with potential inhibitors, in a convenient and rational manner. Such tests are frequently laborious to set up and their establishment is complicated by the necessity to employ problematic cell types and sophisticated assays. Here we present a robust and modular activity assay system which can be adapted to virtually all ligands that signal through dimerization of membrane receptors from different families. The technique rests on fusing ligand-binding domains of specific receptors to the transmembrane and intracellular components of the thymic stromal lymphopoietin (TSLP) receptor which translates signals into readily quantifiable luciferase expression in reporter cells. We show that the activation of various hematopoietic cytokine receptors, of receptor tyrosine kinases as well as of receptors bearing serine/threonine kinase domains by their respective ligands was faithfully reflected both upon transient and stable introduction of hybrid receptor and reporter gene constructs into the murine pro-B cell line Ba/F3. Moreover, we demonstrate the suitability of this platform for the functional characterization of cytokine/growth factor receptor inhibitors.


Subject(s)
Biological Assay , Cytokines/analysis , Immunoglobulins/metabolism , Protein Multimerization , Receptors, Cytokine/metabolism , Animals , HEK293 Cells , Humans , Immunoglobulins/genetics , Mice , Receptors, Cytokine/genetics
9.
Mol Cell Biochem ; 451(1-2): 197-209, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30022447

ABSTRACT

EMMPRIN (extracellular matrix metalloproteinase inducer, EMN, CD147) is a member of the immunoglobulin superfamily expressed in numerous cell types both as a soluble and a membrane-spanning glycoprotein. It is involved in many physiological processes, as well as in cancer. This study addresses mechanisms of crosstalk between EMN-driven cancer-related cellular responses and the canonical Wnt-pathway in MCF-7 breast carcinoma cells. Genetic knockdown of EMN in MCF-7 resulted in characteristic changes in cellular shape, organization of the actin cytoskeleton and malignancy profile, indicating that EMN expression represses cell motility, but, in contrast, exerts a stimulatory effect on cell proliferation and invasive properties. Increased invasiveness coincided with elevated expression of Wnt-target genes and established invasion driver matrix metalloproteinase MMP14. Activation of the downstream Wnt-pathway by means of heterologous ß-catenin and/or TCF-4 expression, through inhibition of GSK-3ß by LiCl treatment, or by cell stimulation with insulin-like growth factor-1 (IGF-1) resulted in increased EMN expression. EMN over-expression raised the ratio of the two opposing Wnt pathway-driven transcription factors Sp1 and Sp5, leading to stimulation of the EMN promoter. Furthermore, the EMN promoter was activated by a feed-forward circuit involving an EMN-dependent drop in expression of the repressive signal transducer and activator of transcription 1 (STAT1). Taken together, we show that the influence of EMMPRIN on malignancy-related properties of breast cancer cells is functionally connected to both Wnt- and JAK/STAT pathways.


Subject(s)
Basigin/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Janus Kinase 1/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Wnt1 Protein/metabolism , Apoptosis , Breast Neoplasms/metabolism , Cell Movement , Cell Proliferation , Female , Humans , Janus Kinase 1/genetics , Neoplasm Invasiveness , STAT1 Transcription Factor/genetics , Tumor Cells, Cultured , Wnt1 Protein/genetics
11.
Mol Oncol ; 12(4): 514-528, 2018 04.
Article in English | MEDLINE | ID: mdl-29419930

ABSTRACT

The interferon-inducible transcription factor STAT1 is a tumor suppressor in various malignancies. We investigated sex-specific STAT1 functions in colitis and colitis-associated colorectal cancer (CRC) using mice with specific STAT1 deletion in intestinal epithelial cells (STAT1∆IEC ). Male but not female STAT1∆IEC mice were more resistant to DSS-induced colitis than sex-matched STAT1flox/flox controls and displayed reduced intraepithelial infiltration of CD8+ TCRαß+ granzyme B+ T cells. Moreover, DSS treatment failed to induce expression of T-cell-attracting chemokines in intestinal epithelial cells of male but not of female STAT1∆IEC mice. Application of the AOM-DSS protocol for induction of colitis-associated CRC resulted in increased intestinal tumor load in male but not in female STAT1∆IEC mice. A sex-specific stratification of human CRC patients corroborated the data obtained in mice and revealed that reduced tumor cell-intrinsic nuclear STAT1 protein expression is a poor prognostic factor in men but not in women. These data demonstrate that epithelial STAT1 is a male-specific tumor suppressor in CRC of mice and humans.


Subject(s)
Colitis/metabolism , Colorectal Neoplasms/metabolism , STAT1 Transcription Factor/metabolism , Sex Characteristics , Tumor Suppressor Proteins/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Chemokines/biosynthesis , Colitis/chemically induced , Colitis/pathology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Dextran Sulfate/toxicity , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/metabolism , STAT1 Transcription Factor/genetics , Tumor Suppressor Proteins/genetics
12.
Ann N Y Acad Sci ; 1397(1): 157-168, 2017 06.
Article in English | MEDLINE | ID: mdl-28436082

ABSTRACT

Tricellulin, a member of the tight junction-associated MAGUK protein family, preferentially localizes to tricellular junctions in confluent polarized epithelial cell layers and is downregulated during the epithelial-mesenchymal transition. Posttranslational modifications are assumed to play critical roles in the process of downregulation of tricellulin at the protein level. Here, we report that the E3 ubiquitin ligase Itch forms a complex with tricellulin and thereby enhances its ubiquitination. Pull-down assays confirmed a direct interaction between tricellulin and Itch, which is mediated by the Itch WW domain and the N-terminus of tricellulin. Experiments in the presence of the proteasome inhibitor MG-132 did not show major changes in the levels of ubiquitinated tricellulin in epithelial cells, suggesting that ubiquitination is not primarily involved in proteasomal degradation of tricellulin, but it appears to be important for endocytosis or recycling. In contrast, in HEK-293 cells, MG-132 caused polyubiquitination. Moreover, we observed that well-differentiated RT-112 and de-differentiated Cal-29 bladder cancer cells show an inverse expression of tricellulin and Itch. We postulate that ubiquitination is an important posttranslational modification involved in the determination of the intracellular fate of tricellulin deserving of more detailed further investigations into the underlying molecular mechanisms and their regulation.


Subject(s)
MARVEL Domain Containing 2 Protein/metabolism , Protein Processing, Post-Translational , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , Binding Sites/genetics , Blotting, Western , Caco-2 Cells , Cell Line, Tumor , Dogs , Epithelial Cells/drug effects , Epithelial Cells/metabolism , HEK293 Cells , Humans , Leupeptins/pharmacology , MARVEL Domain Containing 2 Protein/genetics , Madin Darby Canine Kidney Cells , Protein Binding , Repressor Proteins/genetics , Tight Junctions/drug effects , Tight Junctions/metabolism , Ubiquitin-Protein Ligases/genetics
13.
Biosci Trends ; 11(1): 1-8, 2017 Mar 22.
Article in English | MEDLINE | ID: mdl-28154246

ABSTRACT

STAT1/STAT3 transcription factors are important regulators for development of normal, infected or inflammed cells. They are also critically involved in the progression of various malignant tumours, including epithelial-derived carcinomas. Here, we focus on colorectal cancer (CRC) insights for STAT1/3, where controversial functions for STAT3 were reported. For a long time STAT3 has been regarded as a driver of tumour malignancy and its activation was associated with negative clinical outcome. In contrast, STAT1 was generally viewed as an independent tumour suppressor and positive prognostic marker. Here we discuss the experimental evidence for the tight association and regulation of oncogenic STAT3 transcription kept at bay by nuclear STAT1. We summarise current research and describe cellular models of different STAT1/STAT3 expression ratios. STAT1/3 expression levels are influenced by the mutational status of carcinoma cells associated with nuclear unphosphorylated STAT1. Animal tumour models and results from in vitro experiments allow for the conclusion that both proteins interact as antagonistic transcription factors in CRC cells. These STATs steer also important processes during infection and inflammation that influence development and progression of CRC. The STAT1/3 interplay is important to understand gene regulation and we describe it here similar like the YIN/YANG dualism. Thus, we propose to evaluate both STAT1 and STAT3 expression patterns in cancers in a dual manner instead of regarding them as independent transcription factors. This conceptual dualistic view could advance diagnostic predictions in the future.


Subject(s)
Carcinoma/metabolism , Carcinoma/pathology , Disease Progression , STAT Transcription Factors/metabolism , Signal Transduction , Animals , Humans , Models, Biological , Protein Binding
14.
Cell Commun Signal ; 15(1): 10, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28212658

ABSTRACT

C-Met is a receptor tyrosine kinase with multiple functions throughout embryonic development, organogenesis and wound healing and is expressed in various epithelia. The ligand of c-Met is Hepatocyte Growth Factor (HGF) which is secreted among others by mesenchymal stroma/stem (MSC) cells.Physiological c-Met functions are centred around processes that underly cellular motility and invasive growth. Aberrant c-Met expression and activity is observed in numerous cancers and makes major contributions to cell malignancy. Importantly, HGF/c-Met signaling is crucial in the context of communication between cancer cells and the the tumor stroma.Here, we review recent findings on roles of dysregulated c-Met in urogenital tumors such as cancers of the urinary bladder, prostate, and ovary. We put emphasis on novel aspects of cancer-associated c-Met expression regulation on both, HGF-dependent and HGF-independent non-canonical mechanisms. Moreover, this review focusses on c-Met-triggered signalling with potential relevance for urogenital oncogenesis, and on strategies to specifically inhibit c-Met activity.


Subject(s)
Signal Transduction , Urogenital Neoplasms/metabolism , Animals , Humans , Models, Biological , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/metabolism , Urogenital Neoplasms/pathology
15.
Hum Mol Genet ; 25(13): 2838-2852, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27170314

ABSTRACT

Small Kinetochore-Associated Protein (SKAP)/Kinastrin is a multifunctional protein with proposed roles in mitosis, apoptosis and cell migration. Exact mechanisms underlying its activities in these cellular processes are not completely understood. SKAP is predicted to have different isoforms, however, previous studies did not differentiate between them. Since distinct molecular architectures of protein isoforms often influence their localization and functions, this study aimed to examine the expression profile and functional differences between SKAP isoforms in human and mouse. Analyses of various human tissues and cells of different origin by RT-PCR, and by Western blotting and immunocytochemistry applying newly generated anti-SKAP monoclonal antibodies revealed that human SKAP exists in two protein isoforms: ubiquitously expressed SKAP16 and testis/sperm-specific SKAP1. In mouse, SKAP1 expression is detectable in testis at 4 weeks postnatally, when the first wave of spermatogenesis in mice is complete and the elongated spermatids are present in the testes. Furthermore, we identified Pontin as a new SKAP1 interaction partner. SKAP1 and Pontin co-localized in the flagellar region of human sperm suggesting a functional relevance for SKAP1-Pontin interaction in sperm motility. Since most previous studies on SKAP were performed with the testis-specific isoform SKAP1, our findings provide a new basis for future studies on the role of SKAP in both human somatic cells and male germ cells, including studies on male fertility.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Animals , Apoptosis , Humans , Male , Mice , Mitosis , Organ Specificity/genetics , Protein Domains/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sperm Motility , Spermatogenesis , Spermatozoa/metabolism , Testis/metabolism
16.
Oncotarget ; 7(32): 51096-51106, 2016 Aug 09.
Article in English | MEDLINE | ID: mdl-27191495

ABSTRACT

The role of STAT1 and STAT3 for colorectal carcinoma (CRC) development and progression is controversial. We evaluated 414 CRC patient samples on tissue microarrays for differential expression of STAT1 and STAT3 protein levels and correlated ratios with clinical parameters. Concomitant absence of nuclear STAT1 and STAT3 expression was associated with significantly reduced median survival by ≥33 months (p=0.003). To gain insight into underlying mechanisms, we generated four CRC cell lines with STAT3 knockdown. The cell lines harbor different known mutational drivers and were xenografted into SCID mice to analyze the influence of STAT3 on their tumor growth behavior. Experimental downregulation of STAT3 expression had differential, cell-line specific effects on STAT1 expression levels. STAT1 consistently showed nuclear localization irrespective of its tyrosine phosphorylation status. Two characteristic STAT1/3 expression patterns with opposite growth behavior could be distinguished: cell lines with a low STAT1/high STAT3 ratio showed faster tumor growth in xenografts. In contrast, xenografts of cell lines showing high STAT1 and low STAT3 levels grew slower. Importantly, these ratios reflected clinical outcome in CRC patients as well. We conclude that the ratio of STAT1 to STAT3 expression is a key determinant of CRC progression and that STAT1 counteracts pro-tumorigenic STAT3 signaling. Thus, we suggest that the STAT3/STAT1 ratios are better clinical predictors in CRC as compared to STAT3 or STAT1 levels alone.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , Animals , Biomarkers, Tumor/genetics , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/genetics , Colorectal Neoplasms/diagnosis , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, SCID , Prognosis
17.
Leuk Res ; 40: 38-43, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26652578

ABSTRACT

PURPOSE: The cytokine thymic stromal lymphopoietin (TSLP) and its receptor TSLPR are involved in intercellular communication in the course of allergic inflammation and have recently been implicated in the development of various malignancies including B cell precursor acute lymphoblastic leukemia (BCP-ALL). We studied TSLPR expression, TSLP-induced signal transduction and its antibody-mediated inhibition in long-term cultures of primary cells derived from B-precursor ALL patients. METHODS: TSLPR expression was determined by flow cytometry and Western blot analysis, cell proliferation, signal transduction via the JAK/STAT pathway was analysed by Western blot detection of STAT tyrosine phosphorylation and by measuring TSLP-dependent activation of a STAT-specific reporter gene construct. For inhibition studies a recently introduced antagonistic antibody to the TSLPRα-subunit was used. RESULTS: TSLPR surface expression was observed in leukemic lymphoblasts from two out of ten patients with BCP-ALL. Upon TSLP stimulation, the cells with the highest TSLPR expression level showed enhanced proliferation and JAK/STAT-mediated gene regulation in a dose-dependent manner. By employment of an inhibitory antibody to the TSLPR, both TSLP-triggered cell proliferation and STAT transcription factor activation were specifically inhibited. CONCLUSIONS: These results suggest that blockade of the TSLPR might be a therapeutic option for a subset of BCP-ALL patients.


Subject(s)
Cell Proliferation/physiology , Cytokines/physiology , Leukemia, B-Cell/pathology , Receptors, Cytokine/antagonists & inhibitors , Signal Transduction , Humans , Leukemia, B-Cell/metabolism , Thymic Stromal Lymphopoietin
18.
FEBS J ; 282(21): 4187-200, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26277583

ABSTRACT

EMMPRIN (extracellular matrix metalloproteinase inducer) is a widely expressed glycoprotein and a member of the immunoglobulin superfamily which exists in both a membrane-spanning and a soluble form. Homotypic interactions of EMMPRIN underlie its multiple roles in normal development and pathological situations such as viral infections, Alzheimer's disease and cancer. This study employed a recombinant soluble, fully glycosylated EMMPRIN domain (rhsEMN) as a tool to characterize the structural basis of EMMPRIN-EMMPRIN receptor (EMNR) contacts and their functional effects on MCF-7 breast carcinoma cells. rhsEMN did not form dimers in solution but bound to surface EMMPRIN (EMN) on MCF-7 cells with high affinity and was readily internalized. The interaction interface for the homotypic contact was localized to the N-terminal Ig domain. rhsEMN exerted a stimulatory effect on proliferation of MCF-7 cells whereas it reduced cell migration in a dose-dependent manner. These effects were accompanied by an upregulation of endogenous EMMPRIN as well as of matrix metalloproteinase-14 (MMP-14), a membrane-bound protease involved in the extracellular release of soluble EMMPRIN, indicating a regulatory feedback mechanism. The proliferation-promoting activity of rhsEMN was mimicked by a novel functional antibody directed to EMMPRIN, underscoring that crosslinking of cell surface EMMPRIN (EMNR) is crucial for eliciting intracellular signalling. Addressing malignancy-related signal transduction in HEK-293 cells, we could show that rhsEMN triggers the oncogenic Wnt pathway.


Subject(s)
Basigin/metabolism , Basigin/chemistry , Basigin/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement/physiology , Cell Proliferation/physiology , Female , Glycosylation , HEK293 Cells , Humans , MCF-7 Cells , Matrix Metalloproteinase 14/biosynthesis , Matrix Metalloproteinase 14/genetics , Protein Structure, Tertiary , Receptors, Antigen/chemistry , Receptors, Antigen/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Solubility , Wnt Signaling Pathway , beta Catenin/metabolism
19.
Transl Oncol ; 8(2): 97-105, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25926075

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is constitutively activated in the majority of lung cancer. This study aims at defining connections between STAT3 function and the malignant properties of non-small cell lung carcinoma (NSCLC) cells. To address possible mechanisms by which STAT3 influences invasiveness, the expression of matrix metalloproteinase-1 (MMP-1) was analyzed and correlated with the STAT3 activity status. Studies on both surgical biopsies and on lung cancer cell lines revealed a coincidence of STAT3 activation and strong expression of MMP-1. MMP-1 and tyrosine-phosphorylated activated STAT3 were found co-localized in cancer tissues, most pronounced in tumor fronts, and in particular in adenocarcinomas. STAT3 activity was constitutive, although to different degrees, in the lung cancer cell lines investigated. Three cell lines (BEN, KNS62, and A549) were identified in which STAT3 activitation was inducible by Interleukin-6 (IL-6). In A549 cells, STAT3 activity enhanced the level of MMP-1 mRNA and stimulated transcription from the MMP-1 promoter in IL-6-stimulated A549 cells. STAT3 specificity of this effect was confirmed by STAT3 knockdown through RNA interference. Our results link aberrant activity of STAT3 in lung cancer cells to malignant tumor progression through up-regulation of expression of invasiveness-associated MMPs.

20.
Exp Cell Res ; 335(1): 1-11, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25911129

ABSTRACT

The microenvironment of tumor cells is critically involved in tumor development and progression. Tumor-associated fibroblasts (TAFs) represent a major constituent of the tumor stroma. Tumor cells are operative in the activation of TAFs, whereas TAFs in turn contribute to tumor cell malignancy. This report describes mechanisms of communication between fibroblasts and urinary bladder cancer (UBC) cells. Migration of bladder cancer cell lines RT112 and Cal-29, representing two different grades of dedifferentiation, was enhanced by cocultivation with TAFs. Conditioned medium from tumor cells induced the release of interleukin (IL)-8, hepatocyte growth factor (HGF), matrix metalloproteinase-2, granulocyte macrophage colony-stimulating factor, and monocyte chemotactic protein (MCP)-1 by TAFs. Tumor cell-derived IL-1α was identified as a major mediator of these stimulatory effects. Fibroblasts, on the other hand, exerted a migration and invasion stimulating influence on UBC cells. MCP-1 and HGF were shown to promote cell migration of both bladder cancer cell lines.


Subject(s)
Chemokines/metabolism , Fibroblasts/pathology , Urinary Bladder Neoplasms/pathology , Urinary Bladder/pathology , Cell Communication , Cell Line, Tumor , Cell Movement , Chemokine CCL2/metabolism , Coculture Techniques , Culture Media, Conditioned/pharmacology , Epithelial-Mesenchymal Transition/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hepatocyte Growth Factor/metabolism , Humans , Interleukin-1alpha/metabolism , Interleukin-8/metabolism , Matrix Metalloproteinase 2/metabolism , Neoplasm Invasiveness , Stromal Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...