1
|
Hass R, von der Ohe J, Luo T. Human mesenchymal stroma/stem-like cell-derived taxol-loaded EVs/exosomes transfer anti-tumor microRNA signatures and express enhanced SDF-1-mediated tumor tropism. Cell Commun Signal 2024; 22:506. [PMID: 39420354 PMCID: PMC11488203 DOI: 10.1186/s12964-024-01886-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND The release of extracellular vesicles (EVs) including exosomes from human mesenchymal stroma/stem-like cells (MSC) represents valuable cell-free carriers for the delivery of regenerative and medicinal compounds. METHODS EVs/exosomes were isolated by differential centrifugation from four individual MSC as controls and after treatment with a sub-lethal concentration of 10 mM taxol for 24 h, respectively. The isolated EVs/exosomes were characterized and quantified by nano-tracking-analysis and by Western blots. MicroRNAs (miRs) were isolated from the different EVs/exosome populations and expression levels were quantified by qPCR using 1246 miR templates. Cytotoxic effects of the different MSC-derived taxol-loaded EVs/exosomes were determined in five different GFP-transduced cancer cell lines and quantified by a fluoroscan assay with a GFP-detecting fluorimeter. The presence of stroma cell-derived factor 1 (SDF-1) in MSC-derived EVs/exosomes and its enhanced expression in the vesicles after taxol treatment of MSC was quantified by a specific ELISA. RESULTS EVs/exosomes isolated from four individual taxol-treated MSC displayed a larger size and higher yields as the control EVs/exosomes and were used as anti-tumor therapeutic vehicles. Application of each of the four MSC-derived taxol-loaded EVs/exosome populations revealed significant cytotoxic effects in cell lines of five different tumor entities (carcinomas of lung, breast, ovar, colon, astrocytoma) in a concentration-dependent manner. Expression analysis of 1246 miRs in these taxol-loaded EVs/exosomes as compared to the corresponding MSC-derived control EVs/exosomes unraveled a taxol-mediated up-regulation of 11 miRs with predominantly anti-tumorigenic properties. Moreover, various constitutively expressed protein levels were unanimously altered in the MSC cultures. Taxol treatment of the different MSC revealed an up-regulation of tetraspanins and a 2.2-fold to 5.4-fold increased expression of SDF-1 among others. Treatment of cancer cells with MSC-derived taxol-loaded EVs/exosomes in the presence of a neutralizing SDF-1 antibody significantly abolished the cytotoxic effects between 20.3% and 27%. CONCLUSIONS These findings suggested a taxol-mediated increase of anti-cancer properties in MSC that enhance the tropism of derived EVs/exosomes to tumors, thereby specifically focusing the therapeutic effects of the delivered products.
Collapse
Affiliation(s)
- Ralf Hass
- Department of Obstetrics and Gynecology, Biochemistry and Tumor Biology Laboratory, Hannover Medical School, 30625, Hannover, Germany.
| | - Juliane von der Ohe
- Department of Obstetrics and Gynecology, Biochemistry and Tumor Biology Laboratory, Hannover Medical School, 30625, Hannover, Germany
| | - Tianjiao Luo
- Department of Obstetrics and Gynecology, Biochemistry and Tumor Biology Laboratory, Hannover Medical School, 30625, Hannover, Germany
| |
Collapse
|
2
|
Antmen E, Ermis M, Kuren O, Beksac K, Irkkan C, Hasirci V. Nuclear Deformability of Breast Cells Analyzed from Patients with Malignant and Benign Breast Diseases. ACS Biomater Sci Eng 2023; 9:1629-1643. [PMID: 36706038 DOI: 10.1021/acsbiomaterials.2c01059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Breast cancer is a heterogeneous and dynamic disease, in which cancer cells are highly responsive to alterations in the microenvironment. Today, conventional methods of detecting cancer give a rather static image of the condition of the disease, so dynamic properties such as invasiveness and metastasis are difficult to capture. In this study, conventional molecular-level evaluations of the patients with breast adenocarcinoma were combined with in vitro methods on micropatterned poly(methyl methacrylate) (PMMA) biomaterial surfaces that deform cells. A correlation between deformability of the nuclei and cancer stemness, invasiveness, and metastasis was sought. Clinical patient samples were from regions of the breast with different proximities to the tumor. Responses at the single-cell level toward the micropatterned surfaces were studied using CD44/24, epithelial cell adhesion marker (EpCAM), MUC1, and PCK. Results showed that molecular markers and shape descriptors can discriminate the cells from different proximities to the tumor center and from different patients. The cells with the most metastatic and invasive properties showed both the highest deformability and the highest level of metastatic markers. In conclusion, by using a combination of molecular markers together with nuclear deformation, it is possible to improve detection and separation of subpopulations in heterogenous breast cancer specimens at the single-cell level.
Collapse
Affiliation(s)
- Ezgi Antmen
- BIOMATEN, Middle East Technical University (METU) Center of Excellence in Biomaterials and Tissue Engineering, Ankara06800, Turkey
| | - Menekse Ermis
- BIOMATEN, Middle East Technical University (METU) Center of Excellence in Biomaterials and Tissue Engineering, Ankara06800, Turkey
| | - Ozgur Kuren
- BIOMATEN, Middle East Technical University (METU) Center of Excellence in Biomaterials and Tissue Engineering, Ankara06800, Turkey
| | - Kemal Beksac
- Department of General Surgery, Ankara Oncology Hospital, Yenimahalle, Ankara06800, Turkey
| | - Cigdem Irkkan
- Department of Pathology, Ankara Oncology Hospital, Yenimahalle, Ankara06800, Turkey
| | - Vasif Hasirci
- BIOMATEN, Middle East Technical University (METU) Center of Excellence in Biomaterials and Tissue Engineering, Ankara06800, Turkey
- Department of Biomedical Engineering, Acibadem Mehmet Ali Aydinlar University (ACU), Istanbul34752, Turkey
- ACU Biomaterials Center, Acibadem Mehmet Ali Aydinlar University (ACU), Atasehir, Istanbul34752, Turkey
| |
Collapse
|
3
|
Skok K, Gradišnik L, Čelešnik H, Milojević M, Potočnik U, Jezernik G, Gorenjak M, Sobočan M, Takač I, Kavalar R, Maver U. MFUM-BrTNBC-1, a Newly Established Patient-Derived Triple-Negative Breast Cancer Cell Line: Molecular Characterisation, Genetic Stability, and Comprehensive Comparison with Commercial Breast Cancer Cell Lines. Cells 2021; 11:cells11010117. [PMID: 35011679 PMCID: PMC8749978 DOI: 10.3390/cells11010117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/18/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a breast cancer (BC) subtype that accounts for approximately 15–20% of all BC cases. Cancer cell lines (CLs) provide an efficient way to model the disease. We have recently isolated a patient-derived triple-negative BC CL MFUM-BrTNBC-1 and performed a detailed morphological and molecular characterisation and a comprehensive comparison with three commercial BC CLs (MCF-7, MDA-MB-231, MDA-MB-453). Light and fluorescence microscopy were used for morphological studies; immunocytochemical staining for hormone receptor, p53 and Ki67 status; RNA sequencing, qRT-PCR and STR analysis for molecular characterisation; and biomedical image analysis for comparative phenotypical analysis. The patient tissue-derived MFUM-BrTNBC-1 maintained the primary triple-negative receptor status. STR analysis showed a stable and unique STR profile up to the 6th passage. MFUM-BrTNBC-1 expressed EMT transition markers and displayed changes in several cancer-related pathways (MAPK, Wnt and PI3K signalling; nucleotide excision repair; and SWI/SNF chromatin remodelling). Morphologically, MFUM-BrTNBC-1 differed from the commercial TNBC CL MDA-MB-231. The advantages of MFUM-BrTNBC-1 are its isolation from a primary tumour, rather than a metastatic site; good growth characteristics; phenotype identical to primary tissue; complete records of origin; a unique identifier; complete, unique STR profile; quantifiable morphological properties; and genetic stability up to (at least) the 6th passage.
Collapse
Affiliation(s)
- Kristijan Skok
- Department of Pathology, Hospital Graz II, Location West, Göstinger Straße 22, 8020 Graz, Austria
- Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia; (L.G.); (H.Č.); (M.M.); (U.P.); (G.J.); (M.G.); (M.S.); (I.T.); (R.K.)
- Correspondence: (K.S.); (U.M.); Tel.: +43-316-5466-5541 (K.S.); +386-2-234-5823 (U.M.)
| | - Lidija Gradišnik
- Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia; (L.G.); (H.Č.); (M.M.); (U.P.); (G.J.); (M.G.); (M.S.); (I.T.); (R.K.)
| | - Helena Čelešnik
- Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia; (L.G.); (H.Č.); (M.M.); (U.P.); (G.J.); (M.G.); (M.S.); (I.T.); (R.K.)
- Faculty of Chemistry & Chemical Engineering, University of Maribor, Smetanova Ulica 17, 2000 Maribor, Slovenia
| | - Marko Milojević
- Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia; (L.G.); (H.Č.); (M.M.); (U.P.); (G.J.); (M.G.); (M.S.); (I.T.); (R.K.)
| | - Uroš Potočnik
- Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia; (L.G.); (H.Č.); (M.M.); (U.P.); (G.J.); (M.G.); (M.S.); (I.T.); (R.K.)
- Faculty of Chemistry & Chemical Engineering, University of Maribor, Smetanova Ulica 17, 2000 Maribor, Slovenia
| | - Gregor Jezernik
- Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia; (L.G.); (H.Č.); (M.M.); (U.P.); (G.J.); (M.G.); (M.S.); (I.T.); (R.K.)
| | - Mario Gorenjak
- Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia; (L.G.); (H.Č.); (M.M.); (U.P.); (G.J.); (M.G.); (M.S.); (I.T.); (R.K.)
| | - Monika Sobočan
- Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia; (L.G.); (H.Č.); (M.M.); (U.P.); (G.J.); (M.G.); (M.S.); (I.T.); (R.K.)
- Division for Gynecology and Perinatology, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia
| | - Iztok Takač
- Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia; (L.G.); (H.Č.); (M.M.); (U.P.); (G.J.); (M.G.); (M.S.); (I.T.); (R.K.)
- Division for Gynecology and Perinatology, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia
| | - Rajko Kavalar
- Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia; (L.G.); (H.Č.); (M.M.); (U.P.); (G.J.); (M.G.); (M.S.); (I.T.); (R.K.)
- Department of Pathology, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia
| | - Uroš Maver
- Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia; (L.G.); (H.Č.); (M.M.); (U.P.); (G.J.); (M.G.); (M.S.); (I.T.); (R.K.)
- Correspondence: (K.S.); (U.M.); Tel.: +43-316-5466-5541 (K.S.); +386-2-234-5823 (U.M.)
| |
Collapse
|
4
|
Liao CM, Luo T, von der Ohe J, de Juan Mora B, Schmitt R, Hass R. Human MSC-Derived Exosomes Reduce Cellular Senescence in Renal Epithelial Cells. Int J Mol Sci 2021; 22:13562. [PMID: 34948355 PMCID: PMC8709122 DOI: 10.3390/ijms222413562] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/10/2021] [Accepted: 12/14/2021] [Indexed: 12/23/2022] Open
Abstract
Cellular senescence of renal tubular cells is associated with chronic diseases and age-related kidney disorders. Therapies to antagonize senescence are, therefore, explored as novel approaches in nephropathy. Exosomes derived from human mesenchymal stroma-/stem-like cells (MSC) entail the transfer of multiple bioactive molecules, exhibiting profound regenerative potential in various tissues, including therapeutic effects in kidney diseases. Here, we first demonstrate that exosomes promote proliferation and reduce senescence in aged MSC cultures. For potential therapeutic perspectives in organ rejuvenation, we used MSC-derived exosomes to antagonize senescence in murine kidney primary tubular epithelial cells (PTEC). Exosome treatment efficiently reduced senescence while diminishing the transcription of senescence markers and senescence-associated secretory phenotype (SASP) factors. Concomitantly, we observed less DNA damage foci and more proliferating cells. These data provide new information regarding the therapeutic property of MSC exosomes in the development of renal senescence, suggesting a contribution to a new chapter of regenerative vehicles in senotherapy.
Collapse
Affiliation(s)
- Chieh Ming Liao
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany; (C.M.L.); (B.d.J.M.); (R.S.)
| | - Tianjiao Luo
- Biochemistry and Tumor Biology Lab, Department of Gynecology and Obstetrics, Hannover Medical School, 30625 Hannover, Germany; (T.L.); (J.v.d.O.)
| | - Juliane von der Ohe
- Biochemistry and Tumor Biology Lab, Department of Gynecology and Obstetrics, Hannover Medical School, 30625 Hannover, Germany; (T.L.); (J.v.d.O.)
| | - Blanca de Juan Mora
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany; (C.M.L.); (B.d.J.M.); (R.S.)
| | - Roland Schmitt
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany; (C.M.L.); (B.d.J.M.); (R.S.)
| | - Ralf Hass
- Biochemistry and Tumor Biology Lab, Department of Gynecology and Obstetrics, Hannover Medical School, 30625 Hannover, Germany; (T.L.); (J.v.d.O.)
| |
Collapse
|
5
|
Li Z, Zhuo W, Chen L, Zhang X, Chen C, Hu D, Chen Y, Yang J, Zhou Y, Mao M, Xu L, Ju S, Shen J, Wang Q, Dong M, Xie S, Zhou J, Wang L. Establishment and Characterization of a HER2-Positive Cell Line Derived From the Pleural Effusion of a Drug-Resistant Breast Cancer Patient. Front Cell Dev Biol 2021; 9:680968. [PMID: 34141711 PMCID: PMC8204809 DOI: 10.3389/fcell.2021.680968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/03/2021] [Indexed: 11/30/2022] Open
Abstract
Drug resistance is a daunting challenge in the treatment of breast cancer, making it an urgent problem to solve in studies. Cell lines are important tools in basic and preclinical studies; however, few breast cell lines from drug-resistant patients are available. Herein, we established a novel HER2-positive breast cancer cell line from the pleural effusion of a drug-resistant metastatic breast cancer patient. This cell line has potent proliferative capability and tumorigenicity in nude mice but weak invasive and colony-forming capability. The molecular subtype of the cell line and its sensitivity to chemotherapeutics and HER2-targeting agents are different from those of its origin, suggesting that the phenotype changes between the primary and metastatic forms of breast cancer.
Collapse
Affiliation(s)
- Zhaoqing Li
- Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, School of Medicine, Cancer Institute, China National Ministry of Education, Zhejiang University, Hangzhou, China.,Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| | - Wenying Zhuo
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China.,The Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, China
| | - Lini Chen
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| | - Xun Zhang
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| | - Cong Chen
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| | - Dengdi Hu
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China.,The Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, China
| | - Yongxia Chen
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| | - Jingjing Yang
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| | - Yulu Zhou
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| | - Misha Mao
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| | - Ling Xu
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| | - Siwei Ju
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| | - Jun Shen
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| | - Qinchuan Wang
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| | - Minjun Dong
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| | - Shuduo Xie
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| | - Jichun Zhou
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| | - Linbo Wang
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Biomedical Research Center, Hangzhou, China
| |
Collapse
|
6
|
Melzer C, von der Ohe J, Luo T, Hass R. Spontaneous Fusion of MSC with Breast Cancer Cells Can Generate Tumor Dormancy. Int J Mol Sci 2021; 22:ijms22115930. [PMID: 34072967 PMCID: PMC8198754 DOI: 10.3390/ijms22115930] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/02/2020] [Accepted: 05/26/2021] [Indexed: 12/22/2022] Open
Abstract
Direct cellular interactions of MDA-MB-231cherry breast cancer cells with GFP-transduced human mesenchymal stroma/stem-like cells (MSCGFP) in a co-culture model resulted in spontaneous cell fusion by the generation of MDA-MSC-hyb5cherry GFP breast cancer hybrid cells. The proliferative capacity of MDA-MSC-hyb5 cells was enhanced about 1.8-fold when compared to the parental MDA-MB-231cherry breast cancer cells. In contrast to a spontaneous MDA-MB-231cherry induced tumor development in vivo within 18.8 days, the MDA-MSC-hyb5 cells initially remained quiescent in a dormancy-like state. At distinct time points after injection, NODscid mice started to develop MDA-MSC-hyb5 cell-induced tumors up to about a half year later. Following tumor initiation, however, tumor growth and formation of metastases in various different organs occurred rapidly within about 10.5 days. Changes in gene expression levels were evaluated by RNA-microarray analysis and revealed certain increase in dormancy-associated transcripts in MDA-MSC-hyb5. Chemotherapeutic responsiveness of MDA-MSC-hyb5 cells was partially enhanced when compared to MDA-MB-231 cells. However, some resistance, e.g., for taxol was detectable in cancer hybrid cells. Moreover, drug response partially changed during the tumor development of MDA-MSC-hyb5 cells; this suggests the presence of unstable in vivo phenotypes of MDA-hyb5 cells with increased tumor heterogeneity.
Collapse
Affiliation(s)
| | | | | | - Ralf Hass
- Correspondence: ; Tel.: +49-511-532-6070
| |
Collapse
|
7
|
Melzer C, Jacobs R, Dittmar T, Pich A, von der Ohe J, Yang Y, Hass R. Reversible Growth-Arrest of a Spontaneously-Derived Human MSC-Like Cell Line. Int J Mol Sci 2020; 21:ijms21134752. [PMID: 32635395 PMCID: PMC7369918 DOI: 10.3390/ijms21134752] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022] Open
Abstract
Life cycle limitation hampers the production of high amounts of primary human mesenchymal stroma-/stem-like cells (MSC) and limits cell source reproducibility for clinical applications. The characterization of permanently growing MSC544 revealed some differentiation capacity and the simultaneous presence of known MSC markers CD73, CD90, and CD105 even after continuous long-term culture for more than one year and 32 passages. The expression of CD13, CD29, CD44, and CD166 were identified as further surface proteins, all of which were also simultaneously detectable in various other types of primary MSC populations derived from the umbilical cord, bone marrow, and placenta suggesting MSC-like properties in the cell line. Proliferating steady state MSC544 exhibited immune-modulatory activity similar to a subpopulation of long-term growth-inhibited MSC544 after 189d of continuous culture in confluency. This confluent connective cell layer with fibroblast-like morphology can spontaneously contract and the generated space is subsequently occupied by new cells with regained proliferative capacity. Accordingly, the confluent and senescence-associated beta-galactosidase-positive MSC544 culture with about 95% G0/G1 growth-arrest resumed re-entry into the proliferative cell cycle within 3d after sub-confluent culture. The MSC544 cells remained viable during confluency and throughout this transition which was accompanied by marked changes in the release of proteins. Thus, expression of proliferation-associated genes was down-modulated in confluent MSC544 and re-expressed following sub-confluent conditions whilst telomerase (hTERT) transcripts remained detectable at similar levels in both, confluent growth-arrested and proliferating MSC544. Together with the capability of connective cell layer formation for potential therapeutic approaches, MSC544 provide a long term reproducible human cell source with constant properties.
Collapse
Affiliation(s)
- Catharina Melzer
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany; (C.M.); (J.v.d.O.); (Y.Y.)
| | - Roland Jacobs
- Department of Rheumatology and Clinical Immunology, Hannover Medical School, 30625 Hannover, Germany;
| | - Thomas Dittmar
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58453 Witten, Germany;
| | - Andreas Pich
- Department of Toxicology, Hannover Medical School, 30625 Hannover, Germany;
| | - Juliane von der Ohe
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany; (C.M.); (J.v.d.O.); (Y.Y.)
| | - Yuanyuan Yang
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany; (C.M.); (J.v.d.O.); (Y.Y.)
| | - Ralf Hass
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany; (C.M.); (J.v.d.O.); (Y.Y.)
- Correspondence: ; Tel.: +49-511-532-6070
| |
Collapse
|
8
|
A Combination of an Antimitotic and a Bromodomain 4 Inhibitor Synergistically Inhibits the Metastatic MDA-MB-231 Breast Cancer Cell Line. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1850462. [PMID: 31886177 PMCID: PMC6927020 DOI: 10.1155/2019/1850462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/05/2019] [Accepted: 11/19/2019] [Indexed: 12/30/2022]
Abstract
Current chemotherapeutic agents have many side effects and are toxic to normal cells, providing impetus to identify agents that can effectively eliminate tumorigenic cells without damaging healthy cells. The aim of this study was to examine whether combining a novel BRD4 inhibitor, ITH-47, with the antimitotic estradiol analogue, ESE-15-ol, would have a synergistic effect on inhibiting the growth of two different breast cancer cell lines in vitro. Our docking and molecular dynamics studies showed that compared to JQ1, ITH-47 showed a similar binding mode with hydrogen bonds forming between the ligand nitrogens of the pyrazole, ASN99, and water of the BRD4 protein. Data from cell growth studies revealed that the GI50 of ITH-47 and ESE-15-ol after 48 hours of exposure was determined to be 15 μM and 70 nM, respectively, in metastatic MDA-MB-231 breast cancer cells. In tumorigenic MCF-7 breast cancer cells, the GI50 of ITH-47 and ESE-15-ol was 75 μM and 60 nM, respectively, after 48 hours of exposure. Furthermore, the combination of 7.5 μM and 14 nM of ITH-47 and ESE-15-ol, respectively, resulted in 50% growth inhibition of MDA-MB-231 cells resulting in a synergistic combination index (CI) of 0.7. Flow cytometry studies revealed that, compared to the control, combination-treated MDA-MB-231 cells had significantly more cells present in the sub-G1 phase and the combination treatment induced apoptosis in the MDA-MB-231 cells. Compared to vehicle-treated cells, the combination-treated cells showed decreased levels of the BRD4, as well as c-Myc protein after 48 hours of exposure. In combination, the selective BRD4 inhibitor, ITH-47, and ESE-15-ol synergistically inhibited the growth of MDA-MB-231 breast cancer cells, but not of the MCF-7 cell line. This study provides evidence that resistance to BRD4 inhibitors may be overcome by combining inhibitors with other compounds, which may have treatment potential for hormone-independent breast cancers.
Collapse
|
9
|
Hass R, von der Ohe J, Ungefroren H. Potential Role of MSC/Cancer Cell Fusion and EMT for Breast Cancer Stem Cell Formation. Cancers (Basel) 2019; 11:cancers11101432. [PMID: 31557960 PMCID: PMC6826868 DOI: 10.3390/cancers11101432] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/20/2019] [Accepted: 09/24/2019] [Indexed: 02/06/2023] Open
Abstract
Solid tumors comprise of maturated cancer cells and self-renewing cancer stem-like cells (CSCs), which are associated with various other nontumorigenic cell populations in the tumor microenvironment. In addition to immune cells, endothelial cells, fibroblasts, and further cell types, mesenchymal stroma/stem-like cells (MSC) represent an important cell population recruited to tumor sites and predominantly interacting with the different cancer cells. Breast cancer models were among the first to reveal distinct properties of CSCs, however, the cellular process(es) through which these cells are generated, maintained, and expanded within neoplastic tissues remains incompletely understood. Here, we discuss several possible scenarios that are not mutually exclusive but may even act synergistically: fusion of cancer cells with MSC to yield hybrid cells and/or the induction of epithelial-mesenchymal transition (EMT) in breast cancer cells by MSC, which can relay signals for retrodifferentiation and eventually, the generation of breast CSCs (BCSCs). In either case, the consequences may be promotion of self-renewal capacity, tumor cell plasticity and heterogeneity, an increase in the cancer cells’ invasive and metastatic potential, and the acquisition of resistance mechanisms towards chemo- or radiotherapy. While specific signaling mechanisms involved in each of these properties remain to be elucidated, the present review article focusses on a potential involvement of cancer cell fusion and EMT in the development of breast cancer stem cells.
Collapse
Affiliation(s)
- Ralf Hass
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany.
| | - Juliane von der Ohe
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany.
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany.
- Department of General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany.
| |
Collapse
|
10
|
Taxol-Loaded MSC-Derived Exosomes Provide a Therapeutic Vehicle to Target Metastatic Breast Cancer and Other Carcinoma Cells. Cancers (Basel) 2019; 11:cancers11060798. [PMID: 31181850 PMCID: PMC6627807 DOI: 10.3390/cancers11060798] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/01/2019] [Accepted: 06/04/2019] [Indexed: 02/06/2023] Open
Abstract
MSC-derived exosomes display, among others, an efficient biocompatibility and a reduced intrinsic immunogenicity, representing a valuable vehicle for drug delivery in a tumor-therapeutic approach. Following treatment of several human mesenchymal stroma/stem-like cell (MSC) populations with sub-lethal concentrations of taxol for 24 h, exosomes were isolated and applied to different human cancer populations including A549 lung cancer, SK-OV-3 ovarian cancer, and MDA-hyb1 breast cancer cells. While MSC control exosomes revealed little if any growth inhibition on the tumor cells, exposure to taxol-loaded MSC-derived exosomes was associated with 80–90% cytotoxicity. A similar application of taxol-loaded exosomes from HuVEC displayed much fewer effects. Quantification by LC-MS/MS analysis demonstrated a 7.6-fold reduced taxol concentration in MSC exosomes when compared to equivalent cytotoxic in vitro effects achieved with taxol substances, indicating a specific and more efficient tumor-targeting property. Consequently, MSC-derived taxol exosomes were tested in vivo. Highly metastatic MDA-hyb1 breast tumors were induced in NODscid mice, and systemic intravenous application of MSC-derived taxol exosomes revealed a more than 60% reduction of subcutaneous primary tumors. Moreover, the amount of distant organ metastases observed at least in lung, liver, spleen, and kidney was reduced by 50% with MSC taxol exosomes, similar to the effects observed with taxol, although the concentration of taxol in exosomes was about 1000-fold reduced. Together, these findings in different cancer cell populations and in vivo provide promising future perspectives for drug-loaded MSC-derived exosomes in efficiently targeting primary tumors and metastases by reducing side effects.
Collapse
|
11
|
Zhou F, Zhang Y, Xu X, Luo J, Yang F, Wang L, Xie S, Sun J, Yang X. Establishment and characterization of three stable Basal/HER2-positive breast cancer cell lines derived from Chinese breast carcinoma with identical missense mutations in the DNA-binding domain of TP53. Cancer Cell Int 2018; 18:118. [PMID: 30140169 PMCID: PMC6098622 DOI: 10.1186/s12935-018-0617-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/09/2018] [Indexed: 12/31/2022] Open
Abstract
Background Basal/human epidermal growth factor receptor (HER)2-positive (HER2+) breast cancer is resistant to monoclonal antibody (herceptin) treatment. There are currently only three basal/HER2+ breast cancer cell lines available, but they are not from Chinese populations. Methods Three immortalized cell lines (ZJU-0327, ZJU-0725, and ZJU-1127) were established from invasive ductal breast carcinoma tissue of two patients treated by surgical resection at our center. The cell lines were characterized in terms of histology, therapeutic response, and biomarker expression. Their tumorigenic potential was evaluated in an athymic nude (BALB/C nu) mouse xenograft model. Cell authentication testing by the techniques of short tandem repeat. Results ZJU-0327, ZJU-0725, and ZJU-1127 cell lines were maintained for more than 110 passages in vitro. The cells grew as monolayers; showed typical epithelial morphology and ultrastructure; were polyploid; had doubling times of 18, 57.5, and 18 h, respectively; had a near-tetraploid (ZJU-0327 and ZJU-1127) or aneuploid (ZJU-0725) karyotype with structural aberrations and tumor protein 53 mutation; insensitive to chemotherapeutic drugs and/or radiation; show high invasiveness and tumorigenicity in mice; and had no mycoplasma contamination. The cell lines were basal/HER2+, expressed cluster of differentiation, and were associated with poor prognosis. Cell authentication testing by the American Type Culture Collection confirmed the human origin of the cell lines, which did not match those in existing databases. Conclusions The three novel basal/HER2+ breast cancer cell lines recapitulating the malignant characteristics of the parent tumor's, and can be useful for clarifying the molecular pathogenesis of basal/HER2+ breast cancer.
Collapse
Affiliation(s)
- Fei Zhou
- 1Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| | - Yanhua Zhang
- 2Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| | - Xiufang Xu
- Department of Medical Imagine, Hangzhou Medical College, Hangzhou, Zhejiang China
| | - Jingfeng Luo
- 1Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| | - Fang Yang
- 2Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| | - Linbo Wang
- 4Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| | - Shuduo Xie
- 4Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| | - Jihong Sun
- 1Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| | - Xiaoming Yang
- 1Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China.,5Image-Guided Bio-Molecular Intervention Research, Department of Radiology, University of Washington School of Medicine, Seattle, WA USA
| |
Collapse
|
12
|
Melzer C, von der Ohe J, Hass R. In Vitro Fusion of Normal and Neoplastic Breast Epithelial Cells with Human Mesenchymal Stroma/Stem Cells Partially Involves Tumor Necrosis Factor Receptor Signaling. Stem Cells 2018; 36:977-989. [PMID: 29569804 DOI: 10.1002/stem.2819] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 02/05/2018] [Accepted: 02/24/2018] [Indexed: 12/12/2022]
Abstract
Formation of hybrid cells by "accidental cell fusion" of normal and neoplastic breast epithelial cells with local tissue-associated mesenchymal stroma/stem-like cells (MSC) in an inflammatory microenvironment can generate new cancer cell populations whereby molecular signaling mechanisms of this process remain unclear. Fusions of lentiviral enhanced green fluorescent protein-labeled MSC with mcherry-labeled breast epithelial cells were quantified and effects of tumor necrosis factor alpha (TNF-α) and receptor downstream signaling were investigated. Cocultures of MSC with normal human mammary epithelial cells, with neoplastic MCF10A, or with MDA-MB-231 or MCF7 breast cancer cells demonstrated hybrid cell formation between 0.1% and about 2% of the populations within 72 hours, whereby the fusion process occurred in less than 5 minutes. Addition of the pro-inflammatory cytokine TNF-α significantly enhanced MCF10A-MSC cell fusion. Small-interfering RNA (siRNA) knockdown experiments revealed an involvement of tumor necrosis factor (TNF) receptor-1 and -2 in this process. This was also substantiated by siRNA knockdown of tumor necrosis factor receptor type 1-associated death domain which abolished TNF-α-stimulated fusion. While TNF receptor signaling can be relayed via the Mitogen-activated protein kinase 8 (MAPK8), NF-κB or cell death pathways, examination of further downstream signaling exhibited little if any effects of MAPK8 or RelA (p65) on TNF-α-mediated cell fusion, respectively. These data suggested that cell fusion between MSC and MCF10A breast epithelial cells can be stimulated by TNF-α involving TNF receptor-activated cell death pathways or additional NF-κB signaling. Stem Cells 2018;36:977-989.
Collapse
Affiliation(s)
- Catharina Melzer
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Juliane von der Ohe
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Ralf Hass
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
13
|
Joshi PS, Modur V, Cheng J, Robinson K, Rao K. Characterization of immortalized human mammary epithelial cell line HMEC 2.6. Tumour Biol 2017; 39:1010428317724283. [PMID: 29022488 DOI: 10.1177/1010428317724283] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Primary human mammary epithelial cells have a limited life span which makes it difficult to study them in vitro for most purposes. To overcome this problem, we have developed a cell line that was immortalized using defined genetic elements, and we have characterized this immortalized non-tumorigenic human mammary epithelial cell line to establish it as a potential model system. human mammary epithelial cells were obtained from a healthy individual undergoing reduction mammoplasty at SIU School of Medicine. The cells were transduced with CDK4R24C followed by transduction with human telomerase reverse transcriptase. Post all manipulation, the cells displayed a normal cell cycle phase distribution and were near diploid in nature, which was confirmed by flow cytometry and karyotyping. In vitro studies showed that the cells were anchorage dependent and were non-invasive in nature. The cell line expressed basal epithelial markers such as cytokeratin 7, CD10, and p63 and was negative for the expression of estrogen receptor and progesterone receptor. Upon G-band karyotyping, the cell line displayed the presence of a few cytogenic abnormalities, including trisomy 20 and trisomy 7, which are also commonly present in other immortalized mammary cell lines. Furthermore, the benign nature of these cells was confirmed by multiple in vitro and in vivo experiments. Therefore, we think that this cell line could serve as a good model to understand the molecular mechanisms involved in the development and progression of breast cancer and to also assess the effect of novel therapeutics on human mammary epithelial cells.
Collapse
Affiliation(s)
- Pooja S Joshi
- 1 Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Vishnu Modur
- 2 Department of Pediatrics and Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, OH, USA
| | - JiMing Cheng
- 3 For You Dentistry, 477 Union Ave., Bridgewater, NJ
| | - Kathy Robinson
- 4 Division of Hematology/Oncology, Department of Internal Medicine, Southern Illinois University School of Medicine, USA.,5 Simmons Cancer Institute at Southern Illinois University, Springfield, IL, USA
| | - Krishna Rao
- 1 Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA.,4 Division of Hematology/Oncology, Department of Internal Medicine, Southern Illinois University School of Medicine, USA.,5 Simmons Cancer Institute at Southern Illinois University, Springfield, IL, USA
| |
Collapse
|
14
|
Winnard PT, Zhang C, Vesuna F, Kang JW, Garry J, Dasari RR, Barman I, Raman V. Organ-specific isogenic metastatic breast cancer cell lines exhibit distinct Raman spectral signatures and metabolomes. Oncotarget 2017; 8:20266-20287. [PMID: 28145887 PMCID: PMC5386761 DOI: 10.18632/oncotarget.14865] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/17/2017] [Indexed: 12/13/2022] Open
Abstract
Molecular characterization of organ-specific metastatic lesions, which distinguish them from the primary tumor, will provide a better understanding of tissue specific adaptations that regulate metastatic progression. Using an orthotopic xenograft model, we have isolated isogenic metastatic human breast cancer cell lines directly from organ explants that are phenotypically distinct from the primary tumor cell line. Label-free Raman spectroscopy was used and informative spectral bands were ascertained as differentiators of organ-specific metastases as opposed to the presence of a single universal marker. Decision algorithms derived from the Raman spectra unambiguously identified these isogenic cell lines as unique biological entities – a finding reinforced through metabolomic analyses that indicated tissue of origin metabolite distinctions between the cell lines. Notably, complementarity of the metabolomics and Raman datasets was found. Our findings provide evidence that metastatic spread generates tissue-specific adaptations at the molecular level within cancer cells, which can be differentiated with Raman spectroscopy.
Collapse
Affiliation(s)
- Paul T Winnard
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chi Zhang
- The Johns Hopkins University, Department of Mechanical Engineering, Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Farhad Vesuna
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jeon Woong Kang
- Laser Biomedical Research Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jonah Garry
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ramachandra Rao Dasari
- Laser Biomedical Research Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ishan Barman
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,The Johns Hopkins University, Department of Mechanical Engineering, Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Venu Raman
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Pathology, University Medical Center Utrecht Cancer Center, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
15
|
Roberts S, Speirs V. Advances in the development of improved animal-free models for use in breast cancer biomedical research. Biophys Rev 2017; 9:321-327. [PMID: 28748520 PMCID: PMC5578919 DOI: 10.1007/s12551-017-0276-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/03/2017] [Indexed: 02/08/2023] Open
Abstract
Through translational research, the outcomes for women (and men) diagnosed with breast cancer have improved significantly, with now over 80% of women surviving for at least 5 years post-diagnosis. Much of this success has been translated from the bench to the bedside using laboratory models. Here, we outline the types of laboratory models that have helped achieve this and discuss new approaches as we move towards animal-free disease modelling.
Collapse
Affiliation(s)
- Sophie Roberts
- Leeds Institute of Cancer & Pathology, University of Leeds, St James's University Hospital, Wellcome Trust Brenner Building, Leeds, LS9 7TF, UK
| | - Valerie Speirs
- Leeds Institute of Cancer & Pathology, University of Leeds, St James's University Hospital, Wellcome Trust Brenner Building, Leeds, LS9 7TF, UK.
| |
Collapse
|
16
|
TGF-β-Dependent Growth Arrest and Cell Migration in Benign and Malignant Breast Epithelial Cells Are Antagonistically Controlled by Rac1 and Rac1b. Int J Mol Sci 2017; 18:ijms18071574. [PMID: 28726720 PMCID: PMC5536062 DOI: 10.3390/ijms18071574] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 07/14/2017] [Accepted: 07/16/2017] [Indexed: 12/20/2022] Open
Abstract
Despite improvements in diagnosis and treatment, breast cancer is still the most common cancer type among non-smoking females. TGF-β can inhibit breast cancer development by inducing cell cycle arrest in both, cancer cells and, as part of a senescence program in normal human mammary epithelial cells (HMEC). Moreover, TGF-β also drives cell migration and invasion, in part through the small GTPases Rac1 and Rac1b. Depletion of Rac1b or Rac1 and Rac1b in MDA-MB-231 or MDA-MB-435s breast cancer cells by RNA interference enhanced or suppressed, respectively, TGF-β1-induced migration/invasion. Rac1b depletion in MDA-MB-231 cells also increased TGF-β-induced p21WAF1 expression and ERK1/2 phosphorylation. Senescent HMEC (P15/P16), when compared to their non-senescent counterparts (P11/P12), presented with dramatically increased migratory activity. These effects were paralleled by elevated expression of genes associated with TGF-β signaling and metastasis, downregulated Rac1b, and upregulated Rac1. Our data suggest that acquisition of a motile phenotype in HMEC resulted from enhanced autocrine TGF-β signaling, invasion/metastasis-associated gene expression, and a shift in the ratio of antimigratory Rac1b to promigratory Rac1. We conclude that although enhanced TGF-β signaling is considered antioncogenic in HMEC by suppressing oncogene-induced transformation, this occurs at the expense of a higher migration and invasion potential.
Collapse
|
17
|
Cancer stem cell niche models and contribution by mesenchymal stroma/stem cells. Mol Cancer 2017; 16:28. [PMID: 28148265 PMCID: PMC5286787 DOI: 10.1186/s12943-017-0595-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/18/2017] [Indexed: 02/08/2023] Open
Abstract
Background The initiation and progression of malignant tumors is driven by distinct subsets of tumor-initiating or cancer stem-like cells (CSCs) which develop therapy/apoptosis resistance and self-renewal capacity. In order to be able to eradicate these CSCs with novel classes of anti-cancer therapeutics, a better understanding of their biology and clinically-relevant traits is mandatory. Main body Several requirements and functions of a CSC niche physiology are combined with current concepts for CSC generation such as development in a hierarchical tumor model, by stochastic processes, or via a retrodifferentiation program. Moreover, progressive adaptation of endothelial cells and recruited immune and stromal cells to the tumor site substantially contribute to generate a tumor growth-permissive environment resembling a CSC niche. Particular emphasis is put on the pivotal role of multipotent mesenchymal stroma/stem cells (MSCs) in supporting CSC development by various kinds of interaction and cell fusion to form hybrid tumor cells. Conclusion A better knowledge of CSC niche physiology may increase the chances that cancer stemness-depleting interventions ultimately result in arrest of tumor growth and metastasis.
Collapse
|
18
|
Impact of Heat Stress on Cellular and Transcriptional Adaptation of Mammary Epithelial Cells in Riverine Buffalo (Bubalus Bubalis). PLoS One 2016; 11:e0157237. [PMID: 27682256 PMCID: PMC5040452 DOI: 10.1371/journal.pone.0157237] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 05/26/2016] [Indexed: 12/19/2022] Open
Abstract
The present study aims to identify the heat responsive genes and biological pathways in heat stressed buffalo mammary epithelial cells (MECs). The primary mammary epithelial cells of riverine buffalo were exposed to thermal stress at 42°C for one hour. The cells were subsequently allowed to recover at 37°C and harvested at different time intervals (30 min to 48 h) along with control samples (un-stressed). In order to assess the impact of heat stress in buffalo MECs, several in-vitro cellular parameters (lactate dehydrogenase activity, cell proliferation assay, cellular viability, cell death and apoptosis) and transcriptional studies were conducted. The heat stress resulted in overall decrease in cell viability and cell proliferation of MECs while induction of cellular apoptosis and necrosis. The transcriptomic profile of heat stressed MECs was generated using Agilent 44 K bovine oligonucleotide array and at cutoff criteria of ≥3-or ≤3 fold change, a total of 153 genes were observed to be upregulated while 8 genes were down regulated across all time points post heat stress. The genes that were specifically up-regulated or down-regulated were identified as heat responsive genes. The upregulated genes in heat stressed MECs belonged to heat shock family viz., HSPA6, HSPB8, DNAJB2, HSPA1A. Along with HSPs, genes like BOLA, MRPL55, PFKFB3, PSMC2, ENDODD1, ARID5A, and SENP3 were also upregulated. Microarray data revealed that the heat responsive genes belonged to different functional classes viz., chaperons; immune responsive; cell proliferation and metabolism related. Gene ontology analysis revealed enrichment of several biological processes like; cellular process, metabolic process, response to stimulus, biological regulation, immune system processes and signaling. The transcriptome analysis data was further validated by RT-qPCR studies. Several HSP (HSP40, HSP60, HSP70, HSP90, and HSPB1), apoptotic (Bax and Bcl2), immune (IL6, TNFα and NF-kβ) and oxidative stress (GPX1 and DUSP1) related genes showed differential expression profile at different time points post heat stress. The transcriptional data strongly indicated the induction of survival/apoptotic mechanism in heat stressed buffalo MECs. The overrepresented pathways across all time points were; electron transport chain, cytochrome P450, apoptosis, MAPK, FAS and stress induction of HSP regulation, delta Notch signaling, apoptosis modulation by HSP70, EGFR1 signaling, cytokines and inflammatory response, oxidative stress, TNF-alpha and NF- kB signaling pathway. The study thus identified several genes from different functional classes and biological pathways that could be termed as heat responsive in buffalo MEC. The responsiveness of buffalo MECs to heat stress in the present study clearly suggested its suitability as a model to understand the modulation of buffalo mammary gland expression signature in response to environmental heat load.
Collapse
|
19
|
Beck TN, Kaczmar J, Handorf E, Nikonova A, Dubyk C, Peri S, Lango M, Ridge JA, Serebriiskii IG, Burtness B, Golemis EA, Mehra R. Phospho-T356RB1 predicts survival in HPV-negative squamous cell carcinoma of the head and neck. Oncotarget 2016; 6:18863-74. [PMID: 26265441 PMCID: PMC4662460 DOI: 10.18632/oncotarget.4321] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/22/2015] [Indexed: 11/25/2022] Open
Abstract
Locally advanced squamous cell carcinoma of the head and neck (SCCHN) that is not associated with human papillomavirus (HPV) has a poor prognosis in contrast to HPV-positive disease. To better understand the importance of RB1 activity in HPV-negative SCCHN, we investigated the prognostic value of inhibitory CDK4/6 phosphorylation of RB1 on threonine 356 (T356) in archival HPV-negative tumor specimens from patients who underwent surgical resection and adjuvant radiation. We benchmarked pT356RB1 to total RB1, Ki67, pT202/Y204ERK1/2, and TP53, as quantified by automatic quantitative analysis (AQUA), and correlated protein expression with tumor stage and grade. High expression of pT356RB1 but not total RB1 predicted reduced overall survival (OS; P = 0.0295), indicating the potential relevance of post-translational phosphorylation. Paired analysis of The Cancer Genome Atlas (TCGA) data for regulators of this RB1 phosphorylation identified loss or truncating mutation of negative regulator CDKN2A (p16) and elevated expression of the CDK4/6 activator CCND1 (cyclin D) as also predicting poor survival. Given that CDK4/6 inhibitors have been most effective in the context of functional RB1 and low expression or deletion of p16 in other tumor types, these data suggest such agents may merit evaluation in HPV-negative SCCHN, specifically in cases associated with high pT356RB1.
Collapse
Affiliation(s)
- Tim N Beck
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA.,Molecular and Cell Biology and Genetics, Drexel University College of Medicine, Philadelphia, PA, USA
| | - John Kaczmar
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA.,Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Elizabeth Handorf
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Anna Nikonova
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Cara Dubyk
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Suraj Peri
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Miriam Lango
- Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - John A Ridge
- Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Ilya G Serebriiskii
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA.,Department of Biochemistry, Kazan Federal University, Kazan, Russia
| | - Barbara Burtness
- Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT, USA
| | - Erica A Golemis
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA.,Molecular and Cell Biology and Genetics, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Ranee Mehra
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA.,Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
20
|
Weigand A, Boos AM, Tasbihi K, Beier JP, Dalton PD, Schrauder M, Horch RE, Beckmann MW, Strissel PL, Strick R. Selective isolation and characterization of primary cells from normal breast and tumors reveal plasticity of adipose derived stem cells. Breast Cancer Res 2016; 18:32. [PMID: 26968831 PMCID: PMC4788819 DOI: 10.1186/s13058-016-0688-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 02/19/2016] [Indexed: 02/08/2023] Open
Abstract
Background There is a need to establish more cell lines from breast tumors in contrast to immortalized cell lines from metastatic effusions in order to represent the primary tumor and not principally metastatic biology of breast cancer. This investigation describes the simultaneous isolation, characterization, growth and function of primary mammary epithelial cells (MEC), mesenchymal cells (MES) and adipose derived stem cells (ADSC) from four normal breasts, one inflammatory and one triple-negative ductal breast tumors. Methods A total of 17 cell lines were established and gene expression was analyzed for MEC and MES (n = 42) and ADSC (n = 48) and MUC1, pan-KRT, CD90 and GATA-3 by immunofluorescence. DNA fingerprinting to track cell line identity was performed between original primary tissues and isolates. Functional studies included ADSC differentiation, tumor MES and MEC invasion co-cultured with ADSC-conditioned media (CM) and MES adhesion and growth on 3D-printed scaffolds. Results Comparative analysis showed higher gene expression of EPCAM, CD49f, CDH1 and KRTs for normal MEC lines; MES lines e.g. Vimentin, CD10, ACTA2 and MMP9; and ADSC lines e.g. CD105, CD90, CDH2 and CDH11. Compared to the mean of all four normal breast cell lines, both breast tumor cell lines demonstrated significantly lower ADSC marker gene expression, but higher expression of mesenchymal and invasion gene markers like SNAI1 and MMP2. When compared with four normal ADSC differentiated lineages, both tumor ADSC showed impaired osteogenic and chondrogenic but enhanced adipogenic differentiation and endothelial-like structures, possibly due to high PDGFRB and CD34. Addressing a functional role for overproduction of adipocytes, we initiated 3D-invasion studies including different cell types from the same patient. CM from ADSC differentiating into adipocytes induced tumor MEC 3D-invasion via EMT and amoeboid phenotypes. Normal MES breast cells adhered and proliferated on 3D-printed scaffolds containing 20 fibers, but not on 2.5D-printed scaffolds with single fiber layers, important for tissue engineering. Conclusion Expression analyses confirmed successful simultaneous cell isolations of three different phenotypes from normal and tumor primary breast tissues. Our cell culture studies support that breast-tumor environment differentially regulates tumor ADSC plasticity as well as cell invasion and demonstrates applications for regenerative medicine. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0688-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Annika Weigand
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstr. 12, Erlangen, D-91054, Germany.
| | - Anja M Boos
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstr. 12, Erlangen, D-91054, Germany
| | - Kereshmeh Tasbihi
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstr. 12, Erlangen, D-91054, Germany
| | - Justus P Beier
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstr. 12, Erlangen, D-91054, Germany
| | - Paul D Dalton
- Department of Functional Materials in Medicine and Dentistry, University of Würzburg, Pleicherwall 2, Würzburg, Germany
| | - Michael Schrauder
- Department of Obstetrics and Gynecology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raymund E Horch
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstr. 12, Erlangen, D-91054, Germany
| | - Matthias W Beckmann
- Department of Obstetrics and Gynecology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Pamela L Strissel
- Department of Obstetrics and Gynecology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Reiner Strick
- Department of Obstetrics and Gynecology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
21
|
Shandilya UK, Sharma A, Sodhi M, Kapila N, Kishore A, Mohanty A, Kataria R, Malakar D, Mukesh M. Matrix-based three-dimensional culture of buffalo mammary epithelial cells showed higher induction of genes related to milk protein and fatty acid metabolism. Cell Biol Int 2015; 40:232-8. [DOI: 10.1002/cbin.10555] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 10/23/2015] [Indexed: 11/08/2022]
Affiliation(s)
| | - Ankita Sharma
- National Bureau of Animal Genetic Resources; Karnal Haryana India
| | - Monika Sodhi
- National Bureau of Animal Genetic Resources; Karnal Haryana India
| | - Neha Kapila
- National Bureau of Animal Genetic Resources; Karnal Haryana India
| | - Amit Kishore
- National Bureau of Animal Genetic Resources; Karnal Haryana India
| | - Ashok Mohanty
- National Bureau of Animal Genetic Resources; Karnal Haryana India
| | - Ranjit Kataria
- National Bureau of Animal Genetic Resources; Karnal Haryana India
| | - Dhruva Malakar
- National Bureau of Animal Genetic Resources; Karnal Haryana India
| | - Manishi Mukesh
- National Bureau of Animal Genetic Resources; Karnal Haryana India
| |
Collapse
|
22
|
Al-Shammari AM, Alshami MA, Umran MA, Almukhtar AA, Yaseen NY, Raad K, Hussien AA. Establishment and characterization of a receptor-negative, hormone-nonresponsive breast cancer cell line from an Iraqi patient. BREAST CANCER (DOVE MEDICAL PRESS) 2015; 7:223-30. [PMID: 26300657 PMCID: PMC4536763 DOI: 10.2147/bctt.s74509] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A new breast cancer cell line (AMJ13) has been established from an Iraqi breast cancer patient. It is considered unique because it is the first for an Iraqi population, and is expected to be a useful tool in breast cancer research. The AMJ13 cell line was established from the primary tumor of a 70-year-old Iraqi woman with a histological diagnosis of infiltrating ductal carcinoma. The cells were morphologically characterized by light and scanning electron microscopy, and found to be elongated multipolar epithelial-like cells with a population doubling time of 22 hours. The anchorage-independent growth ability test showed that the cells were able to grow in semisolid agarose, confirming their transformed nature. Cytogenetic study of these cells showed chromosomal aberrations with many structural and numerical abnormalities, producing chromosomes of unknown origin called marker chromosomes. Immunocytochemistry showed that the estrogen receptor and the progesterone receptor were not expressed, and a weak positive result was found for HER2/neu gene expression. AMJ13 cells were positive for BRCA1 and BRCA2, as well as for vimentin. This cell line should be useful when testing new therapies for breast cancer in the Middle East.
Collapse
Affiliation(s)
- Ahmed Majeed Al-Shammari
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriya University, Baghdad, Iraq
| | - Mortadha A Alshami
- Biotechnology Department, Collage of Science, Baghdad University, Baghdad, Iraq
| | | | - Asmaa Amer Almukhtar
- Medical Genetics Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriya University, Baghdad, Iraq
| | - Nahi Y Yaseen
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriya University, Baghdad, Iraq
| | - Khansaa Raad
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriya University, Baghdad, Iraq
| | - Ayman A Hussien
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriya University, Baghdad, Iraq
| |
Collapse
|
23
|
Jakeman PG, Hills TE, Tedcastle AB, Di Y, Fisher KD, Seymour LW. Improved in vitro human tumor models for cancer gene therapy. Hum Gene Ther 2015; 26:249-56. [PMID: 25808057 DOI: 10.1089/hum.2015.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Developing effective anticancer treatments is a particular challenge, as agents must contend with not only the target cellular biology, but also with the complex tumor microenvironment. Here we discuss various in vitro strategies that have sought to address this issue, with a particular focus on new methodologies that utilize clinical samples in basic research and their application in gene therapy and virotherapy.
Collapse
Affiliation(s)
- Philip G Jakeman
- Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Thomas E Hills
- Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Alison B Tedcastle
- Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Ying Di
- Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Kerry D Fisher
- Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Leonard W Seymour
- Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| |
Collapse
|
24
|
Low WS, Wan Abas WAB. Benchtop technologies for circulating tumor cells separation based on biophysical properties. BIOMED RESEARCH INTERNATIONAL 2015; 2015:239362. [PMID: 25977918 PMCID: PMC4419234 DOI: 10.1155/2015/239362] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 02/26/2015] [Accepted: 02/26/2015] [Indexed: 12/11/2022]
Abstract
Circulating tumor cells (CTCs) are tumor cells that have detached from primary tumor site and are transported via the circulation system. The importance of CTCs as prognostic biomarker is leveraged when multiple studies found that patient with cutoff of 5 CTCs per 7.5 mL blood has poor survival rate. Despite its clinical relevance, the isolation and characterization of CTCs can be quite challenging due to their large morphological variability and the rare presence of CTCs within the blood. Numerous methods have been employed and discussed in the literature for CTCs separation. In this paper, we will focus on label free CTCs isolation methods, in which the biophysical and biomechanical properties of cells (e.g., size, deformability, and electricity) are exploited for CTCs detection. To assess the present state of various isolation methods, key performance metrics such as capture efficiency, cell viability, and throughput will be reported. Finally, we discuss the challenges and future perspectives of CTC isolation technologies.
Collapse
Affiliation(s)
- Wan Shi Low
- Department of Biomedical Engineering, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Wan Abu Bakar Wan Abas
- Department of Biomedical Engineering, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
25
|
Yang Y, Otte A, Hass R. Human mesenchymal stroma/stem cells exchange membrane proteins and alter functionality during interaction with different tumor cell lines. Stem Cells Dev 2015; 24:1205-22. [PMID: 25525832 DOI: 10.1089/scd.2014.0413] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
To analyze effects of cellular interaction between human mesenchymal stroma/stem cells (MSC) and different cancer cells, direct co-cultures were performed and revealed significant growth stimulation of the tumor populations and a variety of protein exchanges. More than 90% of MCF-7 and primary human HBCEC699 breast cancer cells as well as NIH:OVCAR-3 ovarian adenocarcinoma cells acquired CD90 proteins during MSC co-culture, respectively. Furthermore, SK-OV-3 ovarian cancer cells progressively elevated CD105 and CD90 proteins in co-culture with MSC. Primary small cell hypercalcemic ovarian carcinoma cells (SCCOHT-1) demonstrated undetectable levels of CD73 and CD105; however, both proteins were significantly increased in the presence of MSC. This co-culture-mediated protein induction was also observed at transcriptional levels and changed functionality of SCCOHT-1 cells by an acquired capability to metabolize 5'cAMP. Moreover, exchange between tumor cells and MSC worked bidirectional, as undetectable expression of epithelial cell adhesion molecule (EpCAM) in MSC significantly increased after co-culture with SK-OV-3 or NIH:OVCAR-3 cells. In addition, a small population of chimeric/hybrid cells appeared in each MSC/tumor cell co-culture by spontaneous cell fusion. Immune fluorescence demonstrated nanotube structures and exosomes between MSC and tumor cells, whereas cytochalasin-D partially abolished the intercellular protein transfer. More detailed functional analysis of FACS-separated MSC and NIH:OVCAR-3 cells after co-culture revealed the acquisition of epithelial cell-specific properties by MSC, including increased gene expression for cytokeratins and epithelial-like differentiation factors. Vice versa, a variety of transcriptional regulatory genes were down-modulated in NIH:OVCAR-3 cells after co-culture with MSC. Together, these mutual cellular interactions contributed to functional alterations in MSC and tumor cells.
Collapse
Affiliation(s)
- Yuanyuan Yang
- 1 Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School , Hannover, Germany
| | | | | |
Collapse
|
26
|
Dastagir N, Lazaridis A, Dastagir K, Reimers K, Vogt PM, Bucan V. Role of lifeguard β-isoform in the development of breast cancer. Oncol Rep 2014; 32:1335-40. [PMID: 25069766 PMCID: PMC4148365 DOI: 10.3892/or.2014.3363] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 06/19/2014] [Indexed: 12/30/2022] Open
Abstract
In the last century there has been great progress in the treatment of breast cancer by improving drug and radiation therapy as well as surgical techniques. Despite this development, breast cancer remains a major cause of death among women in Europe and the US. The cause of breast cancer at the cellular level is still not fully understood. In the present study, we investigated the expression of the Lifeguard β-isoform in breast cancer tissues. In contrast to Lifeguard, the β-isoform has one transmembrane domain less, which is the last of seven (99 bp), and due to this we suspect that the Lifeguard β-isoform exhibits a different function. We determined the expression and function of the β-isoform of Lifeguard in breast cancer cell lines (MCF-7 and MDA-MB-231), a human breast epithelial cell line (MCF10A) and in breast tumour tissue sections. Western blotting, PCR arrays and immunofluorescence were used to investigate the expression of Lifeguard and its β-isoform. Moreover, we investigated the ability of Lifeguard β-isoform expression to inhibit apoptosis induced by Fas. Our results indicated that Lifeguard β-isoform is strongly expressed in breast tumour tissues. More notably, we demonstrated that Fas sensitivity was reduced in the MCF10A breast cells expressing the Lifeguard β-isoform. Taken together, our findings indicate the role of the Lifeguard β-isoform as an anti-apoptotic protein and provide further evidence of the potential of the Lifeguard β-isoform as a target for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Nadjib Dastagir
- Department of Plastic, Hand and Reconstructive Surgery, Hannover Medical School, D-30659 Hannover, Germany
| | - Andrea Lazaridis
- Department of Plastic, Hand and Reconstructive Surgery, Hannover Medical School, D-30659 Hannover, Germany
| | - Khaled Dastagir
- Department of Plastic, Hand and Reconstructive Surgery, Hannover Medical School, D-30659 Hannover, Germany
| | - Kerstin Reimers
- Department of Plastic, Hand and Reconstructive Surgery, Hannover Medical School, D-30659 Hannover, Germany
| | - Peter M Vogt
- Department of Plastic, Hand and Reconstructive Surgery, Hannover Medical School, D-30659 Hannover, Germany
| | - Vesna Bucan
- Department of Plastic, Hand and Reconstructive Surgery, Hannover Medical School, D-30659 Hannover, Germany
| |
Collapse
|
27
|
Ma R, Fredriksson I, Karthik GM, Winn G, Darai-Ramqvist E, Bergh J, Hartman J. Superficial scrapings from breast tumors is a source for biobanking and research purposes. J Transl Med 2014; 94:796-805. [PMID: 24776644 DOI: 10.1038/labinvest.2014.65] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Accepted: 04/01/2014] [Indexed: 01/13/2023] Open
Abstract
Breast cancer is a unique tumor disease in terms of the stringent requirement of predictive biomarker assessments. As recommended by current international guidelines, the established markers consist of estrogen receptor (ER), progesterone receptor, human epidermal growth factor and Ki67, and are primarily analyzed by immunohistochemistry. However, new diagnostic methods based on microarray or next-generation sequencing on DNA and mRNA level are gaining ground. These analyses require fresh-frozen tumor tissue that is generally not available from tumors <10 mm in diameter, comprising almost 25% of all resected breast cancer at our department. We here present a simple and standardized method to generate material from small tumors without risking the histopathological examination. Furthermore, we show that the quality of this material is sufficient for subsequent analysis on mRNA, DNA, and epigenetic level. We were also able to use this method for isolation and expansion of cancer stem cells from the majority of tumors. Consequently, researches can be provided with clinically relevant material for translational studies. In conclusion, this method opens up a new possibility for usage of valuable fresh tumor material for research purposes, biobanking, and next-generation sequencing.
Collapse
Affiliation(s)
- Ran Ma
- Departments of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Irma Fredriksson
- 1] Departments of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden [2] Departments of Breast and Endocrine Surgery, Karolinska Institutet, Stockholm, Sweden
| | | | - Gregory Winn
- Departments of Clinical Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Eva Darai-Ramqvist
- Departments of Clinical Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Bergh
- 1] Departments of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden [2] Radiumhemmet - Karolinska Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Hartman
- 1] Departments of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden [2] Departments of Clinical Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
28
|
In Vitro Efficient Expansion of Tumor Cells Deriving from Different Types of Human Tumor Samples. Med Sci (Basel) 2014. [DOI: 10.3390/medsci2020070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
29
|
Otte A, Bucan V, Reimers K, Hass R. Mesenchymal Stem Cells Maintain Long-Term In Vitro Stemness During Explant Culture. Tissue Eng Part C Methods 2013; 19:937-48. [PMID: 23560527 DOI: 10.1089/ten.tec.2013.0007] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Affiliation(s)
- Anna Otte
- Biochemistry and Tumor Biology Lab, Gynecology Research Unit, Department of Obstetrics and Gynecology, Medical University Hannover, Hannover, Germany
| | - Vesna Bucan
- Department of Plastic, Hand and Reconstructive Surgery, Medical University Hannover, Hannover, Germany
| | - Kerstin Reimers
- Department of Plastic, Hand and Reconstructive Surgery, Medical University Hannover, Hannover, Germany
| | - Ralf Hass
- Biochemistry and Tumor Biology Lab, Gynecology Research Unit, Department of Obstetrics and Gynecology, Medical University Hannover, Hannover, Germany
| |
Collapse
|
30
|
Mandel K, Yang Y, Schambach A, Glage S, Otte A, Hass R. Mesenchymal stem cells directly interact with breast cancer cells and promote tumor cell growth in vitro and in vivo. Stem Cells Dev 2013; 22:3114-27. [PMID: 23895436 DOI: 10.1089/scd.2013.0249] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cellular interactions were investigated between human mesenchymal stem cells (MSC) and human breast cancer cells. Co-culture of the two cell populations was associated with an MSC-mediated growth stimulation of MDA-MB-231 breast cancer cells. A continuous expansion of tumor cell colonies was progressively surrounded by MSC(GFP) displaying elongated cell bodies. Moreover, some MSC(GFP) and MDA-MB-231(cherry) cells spontaneously generated hybrid/chimeric cell populations, demonstrating a dual (green fluorescent protein+cherry) fluorescence. During a co-culture of 5-6 days, MSC also induced expression of the GPI-anchored CD90 molecule in breast cancer cells, which could not be observed in a transwell assay, suggesting the requirement of direct cellular interactions. Indeed, MSC-mediated CD90 induction in the breast cancer cells could be partially blocked by a gap junction inhibitor and by inhibition of the notch signaling pathway, respectively. Similar findings were observed in vivo by which a subcutaneous injection of a co-culture of primary MSC with MDA-MB-231(GFP) cells into NOD/scid mice exhibited an about 10-fold increased tumor size and enhanced metastatic capacity as compared with the MDA-MB-231(GFP) mono-culture. Flow cytometric evaluation of the co-culture tumors revealed more than 90% of breast cancer cells with about 3% of CD90-positive cells, also suggesting an MSC-mediated in vivo induction of CD90 in MDA-MB-231 cells. Furthermore, immunohistochemical analysis demonstrated an elevated neovascularization and viability in the MSC/MDA-MB-231(GFP)-derived tumors. Together, these data suggested an MSC-mediated growth stimulation of breast cancer cells in vitro and in vivo by which the altered MSC morphology and the appearance of hybrid/chimeric cells and breast cancer-expressing CD90(+) cells indicate mutual cellular alterations.
Collapse
Affiliation(s)
- Katharina Mandel
- 1 Biochemistry and Tumor Biology Lab, Gynecology Research Unit , Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | |
Collapse
|
31
|
Mandel K, Seidl D, Rades D, Lehnert H, Gieseler F, Hass R, Ungefroren H. Characterization of spontaneous and TGF-β-induced cell motility of primary human normal and neoplastic mammary cells in vitro using novel real-time technology. PLoS One 2013; 8:e56591. [PMID: 23457587 PMCID: PMC3572945 DOI: 10.1371/journal.pone.0056591] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 01/11/2013] [Indexed: 01/09/2023] Open
Abstract
The clinical complications derived from metastatic disease are responsible for the majority of all breast cancer related deaths. Since cell migration and invasion are a prerequisite for metastasis their assessment in patient cancer cells in vitro may have prognostic value for the tumor's metastatic capacity. We employed real-time cell analysis (RTCA) on the xCELLigence DP system to determine in vitro motility of patient-derived primary human breast cancer epithelial cells (HBCEC). Initially, the RTCA assay was validated using established human breast cancer cell lines with either an invasive (MDA-MB-231, MDA-MB-435s) or a non-invasive phenotype (MCF-7, MDA-MB-468), and primary NSCLC cells (Tu459). Previous standard assays of cell migration/invasion revealed that only MDA-MB-231, −435s, and Tu459 cells exhibited spontaneous and TGF-β1-stimulated migration and invasion through a Matrigel barrier. In the present study, the TGF-β1-stimulated activities could be blocked by SB431542, a potent kinase inhibitor of the TGF-β type I receptor ALK5. Application of the RTCA assay to patient-derived tumor cells showed that 4/4 primary HBCEC and primary NSCLC cells, but not normal human mammary epithelial cells (HMEC), displayed high spontaneous migratory and invasive activity which correlated with higher MMP-2 expression and uPA protein levels in HBCEC compared to HMEC. Upon treatment with TGF-β1, HBCEC exhibited morphologic and gene regulatory alterations indicative of epithelial-to-mesenchymal transition. However, exclusively the invasive but not the migratory activity of HBCEC was further enhanced by TGF-β1. This indicates the requirement for molecular, e.g. integrin interactions with Matrigel components in HBCEC in order to become responsive to pro-invasive TGF-β effects. Together, these results show for the first time that tumorigenic HBCEC but not normal HMEC possess a strong basal migratory as well as a basal and TGF-β1-inducible invasive potential. These findings qualify the RTCA assay as an in vitro migration/invasion testing system for patient-specific primary breast cancer cells.
Collapse
Affiliation(s)
- Katharina Mandel
- Biochemistry and Tumor Biology Laboratory, Gynecology Research Unit, Department of Gynecology and Obstetrics, Medical University, Hannover, Germany
| | - Daniel Seidl
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, Lübeck, Germany
- Department of Radiation Oncology, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, Lübeck, Germany
| | - Dirk Rades
- Department of Radiation Oncology, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, Lübeck, Germany
| | - Hendrik Lehnert
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, Lübeck, Germany
| | - Frank Gieseler
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, Lübeck, Germany
| | - Ralf Hass
- Biochemistry and Tumor Biology Laboratory, Gynecology Research Unit, Department of Gynecology and Obstetrics, Medical University, Hannover, Germany
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, Lübeck, Germany
- * E-mail:
| |
Collapse
|
32
|
Hass R, Otte A. Mesenchymal stem cells as all-round supporters in a normal and neoplastic microenvironment. Cell Commun Signal 2012; 10:26. [PMID: 22943670 PMCID: PMC3444900 DOI: 10.1186/1478-811x-10-26] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 08/28/2012] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSC) represent a heterogeneous population exhibiting stem cell-like properties which are distributed almost ubiquitously among perivascular niches of various human tissues and organs. Organismal requirements such as tissue damage determine interdisciplinary functions of resident MSC including self-renewal, migration and differentiation, whereby MSC support local tissue repair, angiogenesis and concomitant immunomodulation. However, growth of tumor cells and invasion also causes local tissue damage and injury which subsequently activates repair mechanisms and consequently, attracts MSC. Thereby, MSC exhibit a tissue-specific functional biodiversity which is mediated by direct cell-to-cell communication via adhesion molecule signaling and by a tightly regulated exchange of a multifactorial panel of cytokines, exosomes, and micro RNAs. Such interactions determine either tumor-promoting or tumor-inhibitory support by MSC. Moreover, fusion with necrotic/apoptotic tumor cell bodies contributes to re-program MSC into an aberrant phenotype also suggesting that tumor tissue in general represents different types of neoplastic cell populations including tumor-associated stem cell-like cells. The present work summarizes some functional characteristics and biodiversity of MSC and highlights certain controversial interactions with normal and tumorigenic cell populations, including associated modulations within the MSC microenvironment.
Collapse
Affiliation(s)
- Ralf Hass
- Biochemistry and Tumor Biology Lab, Gynecology Research Unit, Department of Obstetrics and Gynecology (OE 6410), Medical University Hannover, Carl-Neuberg-Str, 1, 30625 Hannover, Germany.
| | | |
Collapse
|
33
|
Bucan V, Mandel K, Bertram C, Lazaridis A, Reimers K, Park-Simon TW, Vogt PM, Hass R. LEF-1 regulates proliferation and MMP-7 transcription in breast cancer cells. Genes Cells 2012; 17:559-67. [PMID: 22686279 DOI: 10.1111/j.1365-2443.2012.01613.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 03/26/2012] [Indexed: 11/27/2022]
Abstract
Matrix metalloproteinase-7 (MMP-7) is a small secreted proteolytic enzyme with broad substrate specificity. Its expression is associated with tumor invasion, metastasis, and survival in a variety of cancers including breast cancer. Using bioinformatics analysis, a conserved LEF-1 binding site became obvious that is mapped at the promoter region of the genomic MMP-7 locus. Consequently, electrophoretic mobility shift assay demonstrated in vitro binding of LEF-1 to the predicted MMP-7 promoter binding site. Here, we demonstrate that lymphoid enhancer binding factor-1 (LEF-1) is associated with regulation of the proliferation-associated cyclin D1 and a gene encoding MMP-7 in breast cancer cells. Thus, a decrease of LEF-1 expression using LEF-1 siRNA resulted in down-regulation of cyclin D and MMP-7 expression, respectively. Moreover, cell cycle analysis of LEF-1 siRNA-transfected human breast cancer cells revealed a significant arrest in G2/M phase. Taken together, our results indicate a pivotal role of LEF-1 in the regulation of proliferation and MMP-7 transcription in breast cancer cells.
Collapse
Affiliation(s)
- Vesna Bucan
- Department of Plastic, Hand and Reconstructive Surgery, Medical School Hannover, Podbielskistraße 380, Hannover, D-30659, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Fedrowitz M, Hass R, Bertram C, Löscher W. Salivary α-amylase exhibits antiproliferative effects in primary cell cultures of rat mammary epithelial cells and human breast cancer cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2011; 30:102. [PMID: 22027017 PMCID: PMC3219703 DOI: 10.1186/1756-9966-30-102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 10/25/2011] [Indexed: 01/21/2023]
Abstract
Background Breast cancer is one of the most diagnosed cancers in females, frequently with fatal outcome, so that new strategies for modulating cell proliferation in the mammary tissue are urgently needed. There is some, as yet inconclusive evidence that α-amylase may constitute a novel candidate for affecting cellular growth. Methods The present investigation aimed to examine if salivary α-amylase, an enzyme well known for the metabolism of starch and recently introduced as a stress marker, is able to exert antiproliferative effects on the growth of mammary gland epithelial cells. For this purpose, primary epithelial cultures of breast tissue from two different inbred rat strains, Fischer 344 (F344) and Lewis, as well as breast tumor cells of human origin were used. Treatment with human salivary α-amylase was performed once daily for 2 days followed by cell counting (trypan blue assay) to determine alterations in cell numbers. Cell senescence after α-amylase treatment was assessed by β-galactosidase assay. Endogenous α-amylase was detected in cells from F344 and Lewis by immunofluorescence. Results Salivary α-amylase treatment in vitro significantly decreased the proliferation of primary cells from F344 and Lewis rats in a concentration-dependent manner. Noticeably, the sensitivity towards α-amylase was significantly higher in Lewis cells with stronger impact on cell growth after 5 and 50 U/ml compared to F344 cells. An antiproliferative effect of α-amylase was also determined in mammary tumor cells of human origin, but this effect varied depending on the donor, age, and type of the cells. Conclusions The results presented here indicate for the first time that salivary α-amylase affects cell growth in rat mammary epithelial cells and in breast tumor cells of human origin. Thus, α-amylase may be considered a novel, promising target for balancing cellular growth, which may provide an interesting tool for tumor prophylaxis and treatment.
Collapse
Affiliation(s)
- Maren Fedrowitz
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.
| | | | | | | |
Collapse
|
35
|
Hass R, Kasper C, Böhm S, Jacobs R. Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC. Cell Commun Signal 2011; 9:12. [PMID: 21569606 PMCID: PMC3117820 DOI: 10.1186/1478-811x-9-12] [Citation(s) in RCA: 1184] [Impact Index Per Article: 84.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 05/14/2011] [Indexed: 12/11/2022] Open
Abstract
The mesenchymal stroma harbors an important population of cells that possess stem cell-like characteristics including self renewal and differentiation capacities and can be derived from a variety of different sources. These multipotent mesenchymal stem cells (MSC) can be found in nearly all tissues and are mostly located in perivascular niches. MSC have migratory abilities and can secrete protective factors and act as a primary matrix for tissue regeneration during inflammation, tissue injuries and certain cancers.These functions underlie the important physiological roles of MSC and underscore a significant potential for the clinical use of distinct populations from the various tissues. MSC derived from different adult (adipose tissue, peripheral blood, bone marrow) and neonatal tissues (particular parts of the placenta and umbilical cord) are therefore compared in this mini-review with respect to their cell biological properties, surface marker expression and proliferative capacities. In addition, several MSC functions including in vitro and in vivo differentiation capacities within a variety of lineages and immune-modulatory properties are highlighted. Differences in the extracellular milieu such as the presence of interacting neighbouring cell populations, exposure to proteases or a hypoxic microenvironment contribute to functional developments within MSC populations originating from different tissues, and intracellular conditions such as the expression levels of certain micro RNAs can additionally balance MSC function and fate.
Collapse
Affiliation(s)
- Ralf Hass
- Laboratory of Biochemistry and Tumor Biology, Gynecology Research Unit, Department of Obstetrics and Gynecology, Medical University, Hannover, Carl-Neuberg-Straße 1, 30625 Hannover, Germany.
| | | | | | | |
Collapse
|
36
|
Chaturvedi S, Hass R. Extracellular signals in young and aging breast epithelial cells and possible connections to age-associated breast cancer development. Mech Ageing Dev 2011; 132:213-9. [PMID: 21507328 DOI: 10.1016/j.mad.2011.04.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Revised: 02/07/2011] [Accepted: 04/02/2011] [Indexed: 12/28/2022]
Abstract
Aging of human breast tissue is accompanied by certain structural and functional variations and several studies suggest a possible contribution of these changes to an aging-related breast cancer development. At the cellular level, aging of human mammary epithelial cells is associated with significant morphological and functional alterations such as an increased cell size and a reduced proliferation. Cellular senescence of HMEC cannot be explained by a single mechanism but represents an interaction of numerous extra- and intracellular events and the complexity of such orchestrating pathways is still hardly understood. Besides the contribution of reactive oxygen species and telomere dysfunction to aging, it is the aim of this mini-review, to compare distinct changes to extracellular signals by certain matrix metalloproteinases including MMP-7 and associated growth factor pathways mediated by HB-EGF activation in young and aging HMEC. Such changes can alter hormone receptor levels within aged HMEC, induce tissue fibrosis and promote epithelial-to-mesenchymal transition as a potential prerequisite for breast cancer development. Moreover, an accumulation of aging cells during the normal life span of the breast tissue may also substantially effect and interact with adjacent neighboring populations in the local microenvironment to provide optimized growth conditions which would also support neoplastic cells.
Collapse
Affiliation(s)
- Sukhada Chaturvedi
- Department of Gynecology, Biochemistry and Tumor Biology Lab (OE 6411), Medical School Hannover, Germany.
| | | |
Collapse
|
37
|
Teng PN, Rungruang BJ, Hood BL, Sun M, Flint MS, Bateman NW, Dhir R, Bhargava R, Richard SD, Edwards RP, Conrads TP. Assessment of buffer systems for harvesting proteins from tissue interstitial fluid for proteomic analysis. J Proteome Res 2010; 9:4161-9. [PMID: 20518575 DOI: 10.1021/pr100382v] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tissue interstitial fluid (TIF) bathes cells in tissues, and it is hypothesized that TIF proximal to a developing tumor may contain an enriched population of tumor-specific shed and secreted proteins relative to peripheral blood. Extraction of TIF proteins is typically accomplished through passive incubation of surgically resected tissues in phosphate buffered saline (PBS); however, its influence on cellular activity and viability has not been fully explored. The present investigation sought to characterize whether different buffer systems influence the recovered TIF proteome. Five TIF buffer systems were investigated including PBS, Dulbecco's modified Eagle medium (DMEM), and three organ transplantation preservative solutions: Celsior solution S (CS), histidine-tryptophan-ketoglutarate (HTK), and University of Wisconsin (UW). Kidney tumor, adjacent normal kidney, and ovarian tumor tissues were incubated in each of the buffer systems, and the harvested TIF proteins were analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Although the present results indicate that no significant differences exist in the recovered proteins from these two neoplasms between the five solution groups, additional sample preparative steps are required prior to LC-MS/MS for TIF proteins harvested from DMEM, UW, CS, and HTK. These data support that PBS is a suitable and convenient solution for harvesting TIF proteins for MS-based proteomics.
Collapse
Affiliation(s)
- Pang-ning Teng
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|