1
|
Gao C, Yang B, Li Y, Pei W. Monocarboxylate transporter dependent mechanism is involved in proliferation, migration, and invasion of human glioblastoma cell lines via activation of PI3K/Akt signaling pathway. PLoS One 2024; 19:e0312939. [PMID: 39475905 PMCID: PMC11524508 DOI: 10.1371/journal.pone.0312939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 10/15/2024] [Indexed: 11/02/2024] Open
Abstract
Glioblastoma multiforme is one of the most common primary tumors of the central nervous system, with a very poor prognosis. Cancer cells have been observed to upregulate pH regulators, such as monocarboxylate transporters (MCTs), with an increase in MCT4 expression being observed in several malignancies. MCT4/ recombinant cluster of differentiation 147 (CD147) transporter complex was reported to stimulate vascular endothelial growth factor (VEGF) via the phosphatidylinositol 3 kinase (PI3K) /protein kinase B (Akt) pathway, which has been proven to mediate glioblastoma invasion and migration. The present study aimed to clarify the role of the MCT4/CD147 transporter complex in glioblastoma cell proliferation, migration, and invasion. In this work, lentiviral vectors were used to overexpress MCT4/CD147 and small interfering RNA (siRNA) was used to silence MCT4/CD147 in the human glioma cell lines U87 and U251, respectively. The effects on cell proliferation, migration and invasiveness, as well as the protein expression levels of MCT4 and CD147, extracellular lactate content and Akt activation were assessed by MTT, wound-healing and invasion assays, western blotting and colorimetric method, respectively. The analysis results suggested that cell proliferation, migration, invasion, and Akt activation were decreased by siRNA in all cell lines, but were increased by lentivirus-mediated MCT4 overexpression. These findings suggest that inhibiting the activity and expression of the MCT4/CD147 transporter complex via metabolic-targeting drugs, particularly in cells with a high rate of glycolysis, should be explored as a novel strategy for glioblastoma treatment.
Collapse
Affiliation(s)
- Chen Gao
- Department of General Practice, The 940th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, China
| | - Binni Yang
- Department of General Practice, The 940th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, China
| | - Yurong Li
- Department of General Practice, The 940th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, China
| | - Wenjuan Pei
- Department of General Practice, The 940th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, China
| |
Collapse
|
2
|
Xiong W, Deng Y. BSG Isoform 2 (ENST00000353555) Is a Better Component Than Total BSG Expression in Generating Prognostic Signature for Overall Survival of Liver Cancer. Cureus 2024; 16:e62287. [PMID: 39006665 PMCID: PMC11245721 DOI: 10.7759/cureus.62287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND The basigin (BSG) gene, also known as CD147, has been implicated in the progression and prognosis of various cancers, including liver cancer. This study aimed to comprehensively evaluate the prognostic value of total BSG expression and its specific transcript variants, ENST00000353555 and ENST00000545507, in a large cohort of patients with primary liver cancer. MATERIALS AND METHODS The prognostic values of total BSG, ENST00000353555, and ENST00000545507 expression in overall survival (OS) and progression-free interval (PFI) of patients with primary liver cancer were assessed using The Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC) dataset. Survival analysis, receiver operating characteristic (ROC) analysis, and validation of an extracellular matrix (ECM)-related prognostic signature were performed. RESULTS In univariate and multivariate analyses, total BSG, ENST00000353555, and ENST00000545507 expression were associated with poor OS in liver cancer patients. ENST00000353555 showed the highest hazard ratio among the three prognostic indicators. ROC analysis revealed that ENST00000353555 had better prognostic performance than total BSG expression. Replacing total BSG with ENST00000353555 in an existing ECM-related prognostic signature marginally increased the area under the curve values for one year from 0.79 to 0.80, and five-year OS from 0.72 to 0.73. ENST00000353555 showed isoform-specific positive correlations with EDNRB, IL10, C10orf54, and VEGFA. CONCLUSIONS ENST00000353555 serves as a better prognostic biomarker than total BSG expression in liver cancer, either as an individual marker or as a component of an ECM-related gene signature. Additionally, ENST00000353555 exhibited isoform-specific positive correlations with several immunosuppressive genes, suggesting a potential role in regulating the tumor microenvironment.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Hepatobiliary Surgery, Sichuan Provincial People's Hospital, Chengdu, CHN
| | - Ying Deng
- Cancer Center, Sichuan Provincial People's Hospital, Chengdu, CHN
| |
Collapse
|
3
|
Popa MA, Mihai CM, Șuică VI, Antohe F, Dubey RK, Leeners B, Simionescu M. Dihydrotestosterone Augments the Angiogenic and Migratory Potential of Human Endothelial Progenitor Cells by an Androgen Receptor-Dependent Mechanism. Int J Mol Sci 2024; 25:4862. [PMID: 38732080 PMCID: PMC11084206 DOI: 10.3390/ijms25094862] [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: 03/14/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Endothelial progenitor cells (EPCs) play a critical role in cardiovascular regeneration. Enhancement of their native properties would be highly beneficial to ensuring the proper functioning of the cardiovascular system. As androgens have a positive effect on the cardiovascular system, we hypothesized that dihydrotestosterone (DHT) could also influence EPC-mediated repair processes. To evaluate this hypothesis, we investigated the effects of DHT on cultured human EPCs' proliferation, viability, morphology, migration, angiogenesis, gene and protein expression, and ability to integrate into cardiac tissue. The results showed that DHT at different concentrations had no cytotoxic effect on EPCs, significantly enhanced the cell proliferation and viability and induces fast, androgen-receptor-dependent formation of capillary-like structures. DHT treatment of EPCs regulated gene expression of androgen receptors and the genes and proteins involved in cell migration and angiogenesis. Importantly, DHT stimulation promoted EPC migration and the cells' ability to adhere and integrate into murine cardiac slices, suggesting it has a role in promoting tissue regeneration. Mass spectrometry analysis further highlighted the impact of DHT on EPCs' functioning. In conclusion, DHT increases the proliferation, migration, and androgen-receptor-dependent angiogenesis of EPCs; enhances the cells' secretion of key factors involved in angiogenesis; and significantly potentiates cellular integration into heart tissue. The data offer support for potential therapeutic applications of DHT in cardiovascular regeneration and repair processes.
Collapse
Affiliation(s)
- Mirel Adrian Popa
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (M.A.P.); (C.M.M.); (V.I.Ș.); (F.A.)
| | - Cristina Maria Mihai
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (M.A.P.); (C.M.M.); (V.I.Ș.); (F.A.)
| | - Viorel Iulian Șuică
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (M.A.P.); (C.M.M.); (V.I.Ș.); (F.A.)
| | - Felicia Antohe
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (M.A.P.); (C.M.M.); (V.I.Ș.); (F.A.)
| | - Raghvendra K. Dubey
- Department for Reproductive Endocrinology, University Zurich, 8006 Zürich, Switzerland; (R.K.D.); (B.L.)
| | - Brigitte Leeners
- Department for Reproductive Endocrinology, University Zurich, 8006 Zürich, Switzerland; (R.K.D.); (B.L.)
| | - Maya Simionescu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (M.A.P.); (C.M.M.); (V.I.Ș.); (F.A.)
| |
Collapse
|
4
|
Ghandour F, Kassem S, Simanovich E, Rahat MA. Glucose Promotes EMMPRIN/CD147 and the Secretion of Pro-Angiogenic Factors in a Co-Culture System of Endothelial Cells and Monocytes. Biomedicines 2024; 12:706. [PMID: 38672062 PMCID: PMC11047830 DOI: 10.3390/biomedicines12040706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/17/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Vascular complications in Type 2 diabetes mellitus (T2DM) patients increase morbidity and mortality. In T2DM, angiogenesis is impaired and can be enhanced or reduced in different tissues ("angiogenic paradox"). The present study aimed to delineate differences between macrovascular and microvascular endothelial cells that might explain this paradox. In a monoculture system of human macrovascular (EaHy926) or microvascular (HMEC-1) endothelial cell lines and a monocytic cell line (U937), high glucose concentrations (25 mmole/L) increased the secretion of the pro-angiogenic factors CD147/EMMPRIN, VEGF, and MMP-9 from both endothelial cells, but not from monocytes. Co-cultures of EaHy926/HMEC-1 with U937 enhanced EMMPRIN and MMP-9 secretion, even in low glucose concentrations (5.5 mmole/L), while in high glucose HMEC-1 co-cultures enhanced all three factors. EMMPRIN mediated these effects, as the addition of anti-EMMPRIN antibody decreased VEGF and MMP-9 secretion, and inhibited the angiogenic potential assessed through the wound assay. Thus, the minor differences between the macrovascular and microvascular endothelial cells cannot explain the angiogenic paradox. Metformin, a widely used drug for the treatment of T2DM, inhibited EMMPRIN, VEGF, and MMP-9 secretion in high glucose concentration, and the AMPK inhibitor dorsomorphin enhanced it. Thus, AMPK regulates EMMPRIN, a key factor in diabetic angiogenesis, suggesting that targeting EMMPRIN may help in the treatment of diabetic vascular complications.
Collapse
Affiliation(s)
- Fransis Ghandour
- Department of Internal Medicine A, Carmel Medical Center, Haifa 3436212, Israel
| | - Sameer Kassem
- Department of Internal Medicine A, Carmel Medical Center, Haifa 3436212, Israel
- The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel
| | - Elina Simanovich
- Immunotherapy Laboratory, Carmel Medical Center, Haifa 3436212, Israel
| | - Michal A. Rahat
- The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel
- Immunotherapy Laboratory, Carmel Medical Center, Haifa 3436212, Israel
| |
Collapse
|
5
|
Lee SG, Kim J, Lee YI, Kim J, Choi YS, Ham S, Lee JH. Cutaneous neurogenic inflammation mediated by TRPV1-NGF-TRKA pathway activation in rosacea is exacerbated by the presence of Demodex mites. J Eur Acad Dermatol Venereol 2023; 37:2589-2600. [PMID: 37606610 DOI: 10.1111/jdv.19449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/03/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND Rosacea is a common chronic inflammatory skin condition that is often refractory to treatment, with frequent relapses. Alterations in the skin immunological response and Demodex mite infestation are the primary aetiologic factors targeted for treatment. Transient receptor potential cation channel subfamily V member 1 (TRPV1) is a nociceptive cation channel that plays a role in cutaneous neurogenic pain and can be activated by various rosacea triggers. OBJECTIVES We investigated the effects of TRPV1 modulation in rosacea, focussing on Demodex mite colonization and cutaneous neurogenic inflammation. METHODS We examined mRNA expression levels according to Demodex population counts. An in vitro study using capsazepine as a TRPV1 antagonist was performed to assess the influence of TRPV1 in keratinocytes. A rosacea-like mouse model was generated by the injection of the 37-amino acid C-terminal cathelicidin peptide (LL37), and changes in the skin, dorsal root ganglion (DRG) and ears were examined. RESULTS Increased Demodex mite population counts were associated with increased expression levels of TRPV1, tropomyosin receptor kinase A (TrkA) and nerve growth factor (NGF), and these levels could be reduced by capsazepine treatment in keratinocytes. In an in vivo study, the downstream effects of TRPV1 activation were investigated in the skin, DRG and ears of the rosacea-like mouse model. CONCLUSIONS The findings of this study are instrumental for understanding the underlying causes of rosacea and could potentially lead to the development of new treatments targeting the NGF-TrkA-TRPV1 pathway. The identification of this pathway as a therapeutic target could represent a major breakthrough for rosacea research, potentially resulting in more effective and targeted rosacea treatments. This study contributes to an improved understanding of rosacea pathophysiology, which may lead to the development of more effective treatments in the future.
Collapse
Affiliation(s)
- Sang Gyu Lee
- Department of Dermatology & Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Jihee Kim
- Department of Dermatology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, South Korea
- Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul, South Korea
| | - Young In Lee
- Department of Dermatology & Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, South Korea
- Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul, South Korea
| | - Jemin Kim
- Department of Dermatology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, South Korea
- Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul, South Korea
| | - Ye Seul Choi
- Department of Dermatology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, South Korea
| | - Seoyoon Ham
- Department of Dermatology & Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Ju Hee Lee
- Department of Dermatology & Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, South Korea
- Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul, South Korea
| |
Collapse
|
6
|
Huang D, Rao D, Jin Q, Lai M, Zhang J, Lai Z, Shen H, Zhong T. Role of CD147 in the development and diagnosis of hepatocellular carcinoma. Front Immunol 2023; 14:1149931. [PMID: 37090718 PMCID: PMC10115957 DOI: 10.3389/fimmu.2023.1149931] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer, and the third leading cause of cancer-related deaths worldwide. HCC is characterized by insidious onset, and most patients are diagnosed at an advanced stage with a poor prognosis. Identification of biomarkers for HCC onset and progression is imperative to development of effective diagnostic and therapeutic strategies. CD147 is a glycoprotein that is involved in tumor cell invasion, metastasis and angiogenesis through multiple mechanisms. In this review, we describe the molecular structure of CD147 and its role in regulating HCC invasion, metastasis and angiogenesis. We highlight its potential as a diagnostic and therapeutic target for HCC.
Collapse
Affiliation(s)
- Defa Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Dingyu Rao
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Qing Jin
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Mi Lai
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jiali Zhang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Zhonghong Lai
- Department of traumatology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Haibin Shen
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Haibin Shen, ; Tianyu Zhong,
| | - Tianyu Zhong
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Precision Medicine Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Haibin Shen, ; Tianyu Zhong,
| |
Collapse
|
7
|
Yang S, Bi J, Drnevich J, Li K, Nowak RA. Basigin is necessary for normal decidualization of human uterine stromal cells. Hum Reprod 2022; 37:2885-2898. [PMID: 36303457 PMCID: PMC9712941 DOI: 10.1093/humrep/deac229] [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: 04/21/2022] [Revised: 09/27/2022] [Indexed: 12/14/2022] Open
Abstract
STUDY QUESTION Does basigin (BSG) regulate human endometrial stromal cell (HESC) decidualization in vitro? SUMMARY ANSWER BSG regulates HESCs proliferation and decidualization. WHAT IS KNOWN ALREADY Studies have shown that in the human endometrium, BSG expression is menstrual-cycle dependent and its expression was significantly lower in uterine endometrium during the luteal phase of women experiencing multiple implantation failures after IVF than in women with normal fertility. STUDY DESIGN, SIZE, DURATION We utilized a telomerase-immortalized HESCs in an in vitro cell culture model system to investigate whether BSG regulates decidualization of stromal cells. Further, we used microarray analysis to identify changes in the gene expression profile of HESCs treated with BSG small interfering RNA (siRNA). All experiments were repeated at least three times. PARTICIPANTS/MATERIALS, SETTING, METHODS The effect of BSG knockdown (using siRNA) on HESC proliferation was determined by counting cell number and by tritiated thymidine incorporation assays. The effect of BSG on decidualization of HESCs was determined by RT-qPCR for the decidualization markers insulin-like growth factor-binding protein 1 (IGFBP1) and prolactin (PRL). Immunoblotting was used to determine the effect of BSG siRNA on the expression of MMP-2,3. Microarray analysis was used to identify BSG-regulated genes in HESCs at Day 6 of decidualization. Functional and pathway enrichment analyses were then carried out on the differentially expressed genes (DEGs). The STRING online database was used to analyze protein-protein interaction (PPI) between DEG-encoded proteins, and CytoScape software was used to visualize the interaction. MCODE and CytoHubba were used to construct functional modules and screen hub genes separately. Several BSG-regulated genes identified in the microarray analysis were confirmed by qPCR. MAIN RESULTS AND THE ROLE OF CHANCE Knockdown of BSG expression in cultured stromal cells by siRNA significantly (P < 0.05) inhibited HESC proliferation, disrupted cell decidualization and down-regulated MMP-2 and MMP-3 expression. Microarray analysis identified 721 genes that were down-regulated, and 484 genes up-regulated with P < 0.05 in BSG siRNA treated HESCs. GO term enrichment analysis showed that the DEGs were significantly enriched in cell communication, signaling transduction and regulation, response to stimulus, cell adhesion, anatomical structure morphogenesis, extracellular matrix organization, as well as other functional pathways. KEGG pathway analysis identified upregulated gene enriched in pathways such as the MAPK signaling pathway, colorectal cancer, melanoma and axon guidance. In contrast, downregulated genes were mainly enriched in pathways including ECM-receptor interaction, PI3K-Akt signaling pathway, pathways in cancer, antigen processing, type I diabetes mellitus and focal adhesion. The top 10 hub nodes were identified using 12 methods analyses. The hub genes that showed up in two methods were screened out. Among these genes, upregulated genes included EGFR, HSP90AA1, CCND1, PXN, PRKACB, MGAT4A, EVA1A, LGALS1, STC2, HSPA4; downregulated genes included WNT4/5, FOXO1, CDK1, PIK3R1, IGF1, JAK2, LAMB1, ITGAV, HGF, MXRA8, TMEM132A, UBE2C, QSOX1, ERBB2, GNB4, HSP90B1, LAMB2, LAMC1 and ITGA1. Hub genes and module genes involved in the top three modules of PPI analysis were analyzed through the string database. Analysis showed that hub and module genes were related mainly to the WNT signaling pathway, PI3K-AKT signaling pathway and pathways in cancer. LARGE SCALE DATA The microarray data set generated in this study has been published online at databank.illinois.edu. LIMITATIONS, REASONS FOR CAUTION Most of the findings were obtained using an in vitro cell culture system that may not necessarily reflect in vivo functions. WIDER IMPLICATIONS OF THE FINDINGS Our results demonstrate that BSG plays a vital role in decidualization and that downregulation of BSG in the uterine endometrium may be associated with infertility in women. The identified hub genes and pathways increase our understanding of the genetic etiology and molecular mechanisms underlying the regulation of decidualization by BSG. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the NIH U54 HD40093 (R.A.N.). The authors have no competing interests to declare.
Collapse
Affiliation(s)
- Shuhong Yang
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Jiajia Bi
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| | - Jenny Drnevich
- Roy J. Carver Biotechnology Center, University of Illinois, Urbana, IL, USA
| | - Kailiang Li
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| | - Romana A Nowak
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| |
Collapse
|
8
|
Li C, Yoshimura T, Tian M, Wang Y, Kondo T, Yamamoto KI, Fujisawa M, Ohara T, Sakaguchi M, Matsukawa A. Exosomal Wnt7a from a low metastatic subclone promotes lung metastasis of a highly metastatic subclone in the murine 4t1 breast cancer. BREAST CANCER RESEARCH : BCR 2022; 24:60. [PMID: 36096830 PMCID: PMC9469633 DOI: 10.1186/s13058-022-01557-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/29/2022] [Indexed: 12/03/2022]
Abstract
Background Patients with triple-negative breast cancer (TNBC) often have poorer prognosis than those with other subtypes because of its aggressive behaviors. Cancer cells are heterogeneous, and only a few highly metastatic subclones metastasize. Although the majority of subclones may not metastasize, they could contribute by releasing factors that increase the capacity of highly metastatic cells and/or provide a favorable tumor microenvironment (TME). Here, we analyzed the interclonal communication in TNBC which leads to efficient cancer progression, particularly lung metastasis, using the polyclonal murine 4T1 BC model. Methods We isolated two 4T1 subclones, LM.4T1 and HM.4T1 cells with a low and a high metastatic potential, respectively, and examined the effects of LM.4T1 cells on the behaviors of HM.4T1 cells using the cell scratch assay, sphere-forming assay, sphere invasion assay, RT-qPCR, and western blotting in vitro. We also examined the contribution of LM.4T1 cells to the lung metastasis of HM.4T1 cells and TME in vivo. To identify a critical factor which may be responsible for the effects by LM.4T1 cells, we analyzed the data obtained from the GEO database. Results Co-injection of LM.4T1 cells significantly augmented lung metastases by HM.4T1 cells. LM.4T1-derived exosomes promoted the migration and invasion of HM.4T1 cells in vitro, and blocking the secretion of exosome abrogated their effects on HM.4T1 cells. Analyses of data obtained from the GEO database suggested that Wnt7a might be a critical factor responsible for the enhancing effects. In fact, a higher level of Wnt7a was detected in LM.4T1 cells, especially in exosomes, than in HM.4T1 cells, and deletion of Wnt7a in LM.4T1 cells significantly decreased the lung metastasis of HM.4T1 cells. Further, treatment with Wnt7a increased the spheroid formation by HM.4T1 cells via activation of the PI3K/Akt/mTOR signaling pathway. Finally, infiltration of αSMA-positive fibroblasts and angiogenesis was more prominent in tumors of LM.4T1 cells and deletion of Wnt7a in LM.4T1 cells markedly reduced angiogenesis. Conclusions We demonstrated, for the first time, that a low metastatic subclone can enhance lung metastasis of highly metastatic subclone via exosomal Wnt7a and propose Wnt7a as a molecular target to treat TNBC patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-022-01557-5.
Collapse
Affiliation(s)
- Chunning Li
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan.
| | - Miao Tian
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan.,Department of Cell Biology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Yuze Wang
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan.,Department of Cell Biology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Takamasa Kondo
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan.,Department of Cell Biology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Ken-Ichi Yamamoto
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan.,Department of Cell Biology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Masayoshi Fujisawa
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan.,Department of Cell Biology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Toshiaki Ohara
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan.,Department of Cell Biology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Masakiyo Sakaguchi
- Department of Cell Biology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Akihiro Matsukawa
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan.
| |
Collapse
|
9
|
The Multiple Roles of CD147 in the Development and Progression of Oral Squamous Cell Carcinoma: An Overview. Int J Mol Sci 2022; 23:ijms23158336. [PMID: 35955471 PMCID: PMC9369056 DOI: 10.3390/ijms23158336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 01/27/2023] Open
Abstract
Cluster of differentiation (CD)147, also termed extracellular matrix metalloprotease inducer or basigin, is a glycoprotein ubiquitously expressed throughout the human body, the oral cavity included. CD147 actively participates in physiological tissue development or growth and has important roles in reactive processes such as inflammation, immunity, and tissue repair. It is worth noting that deregulated expression and/or activity of CD147 is observed in chronic inflammatory or degenerative diseases, as well as in neoplasms. Among the latter, oral squamous cell carcinoma (OSCC) is characterized by an upregulation of CD147 in both the neoplastic and normal cells constituting the tumor mass. Most interestingly, the expression and/or activity of CD147 gradually increase as healthy oral mucosa becomes inflamed; hyperplastic/dysplastic lesions are then set on, and, eventually, OSCC develops. Based on these findings, here we summarize published studies which evaluate whether CD147 could be employed as a marker to monitor OSCC development and progression. Moreover, we describe CD147-promoted cellular and molecular events which are relevant to oral carcinogenesis, with the aim to provide useful information for assessing whether CD147 may be the target of novel therapeutic approaches directed against OSCC.
Collapse
|
10
|
Rahat MA. Mini-Review: Can the Metastatic Cascade Be Inhibited by Targeting CD147/EMMPRIN to Prevent Tumor Recurrence? Front Immunol 2022; 13:855978. [PMID: 35418981 PMCID: PMC8995701 DOI: 10.3389/fimmu.2022.855978] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 03/07/2022] [Indexed: 12/05/2022] Open
Abstract
Solid tumors metastasize very early in their development, and once the metastatic cell is lodged in a remote organ, it can proliferate to generate a metastatic lesion or remain dormant for long periods. Dormant cells represent a real risk for future tumor recurrence, but because they are typically undetectable and insensitive to current modalities of treatment, it is difficult to treat them in time. We describe the metastatic cascade, which is the process that allows tumor cells to detach from the primary tumor, migrate in the tissue, intravasate and extravasate the lymphatics or a blood vessel, adhere to a remote tissue and eventually outgrow. We focus on the critical enabling role of the interactions between tumor cells and immune cells, especially macrophages, in driving the metastatic cascade, and on those stages that can potentially be targeted. In order to prevent the metastatic cascade and tumor recurrence, we would need to target a molecule that is involved in all of the steps of the process, and evidence is brought to suggest that CD147/EMMPRIN is such a protein and that targeting it blocks metastasis and prevents tumor recurrence.
Collapse
Affiliation(s)
- Michal A Rahat
- Immunotherapy Laboratory, Carmel Medical Center, Haifa, Israel.,Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
11
|
Solovyeva NI, Timoshenko OS, Kugaevskaya EV, Gureeva TA. Interstitial collagenase MMP-1 and EMMPRIN in cell lines and in clinical specimens of cervical squamous cell carcinoma. Mol Biol Rep 2021; 48:6879-6886. [PMID: 34495460 DOI: 10.1007/s11033-021-06689-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/24/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND The aim of this study was to elucidate the features of the expression of matrix metalloproteinases inducer-EMMPRIN (EMN) and matrix metalloproteinase 1 (MMP-1) in cell lines and in clinical samples of cervical squamous cell carcinoma (SCC). MATERIAL AND METHODS The study was carried out using RT-PCR, densitometry and immunohistochemical studies (IHC) on commercial cell lines Siha, Caski, transformed with HPV16; HeLa, and C33A transformed with HPV18, line C33A without HPV, and in clinical samples of SCC and morphologically normal tissue adjacent to the tumor. RESULTS The data obtained indicate that the expression of mRNA EMN and MMP-1 occurs in all cell lines at different levels. HPV type and number of genes copies had no effect on expression degree both EMN and MMP-1. Gene expression of EMN and MMP-1 has been investigated in tumor and normal tissues. MMP-1 expression in tumor tissue in SCC, as a rule, has been significantly increased (2-6 times) compared to normal tissue. It was found in 90% of tumor samples. It is known, that MMP-1 promotes the development of invasive and metastatic processes. EMN expression was lower in the tumor tissue than in normal tissue in most cases. An increase in EMN expression was noted only in some cases of SCC. CONCLUSION The data obtained indicate that MMP-1 can serve as a marker of the invasive potential of SCC. EMN, apparently, is not a major factor responsible for MMP-1 expression in SCC. Data are important for understanding the process of tumor development and may have prognostic value for the patient.
Collapse
Affiliation(s)
- Nina I Solovyeva
- V.N. Orekhovich Institute of Biomedical Chemistry, 10 Pogodinskaya Str., 119121, Moscow, Russia.
| | - Olga S Timoshenko
- V.N. Orekhovich Institute of Biomedical Chemistry, 10 Pogodinskaya Str., 119121, Moscow, Russia
| | - Elena V Kugaevskaya
- V.N. Orekhovich Institute of Biomedical Chemistry, 10 Pogodinskaya Str., 119121, Moscow, Russia
| | - Tatyana A Gureeva
- V.N. Orekhovich Institute of Biomedical Chemistry, 10 Pogodinskaya Str., 119121, Moscow, Russia
| |
Collapse
|
12
|
Pan S, Su Y, Sun B, Hao R, Gao X, Han B. Knockout of CD147 inhibits the proliferation, invasion, and drug resistance of human oral cancer CAL27 cells in Vitro and in Vivo. Int J Biol Macromol 2021; 181:378-389. [PMID: 33766595 DOI: 10.1016/j.ijbiomac.2021.03.102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/13/2021] [Accepted: 03/17/2021] [Indexed: 11/17/2022]
Abstract
With the development of modern biomedicine, research on the molecular mechanism of tumors has developed gradually. The CD147 gene has been applied to tumor molecular targeted therapy, and significant differences were found in the expression of the CD147 gene in different tumor tissues and normal tissues. Many previous studies have also shown that the expression of the CD147 gene plays a crucial role in the development of tumors. To understand whether CD147 can be used as a therapeutic target for oral cancer, CRISPR/Cas9 gene-editing technology was used to knock out the CD147 gene in cal27 cells to obtain knockout cell lines. Using CCK-8, Transwell, RT-PCR, and Western blotting, the proliferation and invasion abilities of the knockout cell lines were decreased significantly, and the expression of matrix metalloproteinase was also inhibited. Next, a subcutaneously transplanted tumor model in nude mice was constructed to detect the effect of the CD147 gene on tumors. Subcutaneous tumor growth and immunohistochemistry results showed that the proliferation and doxorubicin resistance of knockout cell line were significantly inhibited compared with those in the wild-type group. These results indicated that knocking out CD147 significantly reduced the proliferation and invasion of cal27 cells, and CD147 may be a potential therapeutic target for oral cancer.
Collapse
Affiliation(s)
- Siqi Pan
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Chang Chun, China
| | - Yu Su
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Chang Chun, China
| | - Bixi Sun
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Chang Chun, China
| | - Rubin Hao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Chang Chun, China
| | - Xiaoshu Gao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Chang Chun, China
| | - Bing Han
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Chang Chun, China.
| |
Collapse
|
13
|
Emam M, Keshta AT, Mohamed YM, Attia YA. Insight on Ameliorative Role of Selenium Nanoparticles and Niacin in Wound Healing on Adult Female Albino Mice. ACTA ACUST UNITED AC 2020. [DOI: 10.2174/2212796814666200818111849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Wound healing is a complex process necessary for repairing damaged
tissues and preventing infection. Selenium nanoparticles (Se NPs) were known due to
their antioxidant and antimicrobial effects, also niacin has angiogenesis and antioxidant effects
that are important in wound healing.
Objective:
The present study was conducted to investigate the effect of Se NPs and niacin in
reducing and accelerating the wound healing time in mice.
Methods:
A simple wet chemical method has been modified to synthesize Se NPs in order to
investigate their effect and niacin on reducing the wound healing in 80 adult female albino
mice (250 mm2 full thickness open excision wound) that were divided into eight groups (10
mice/each). After 30-days, the mice were sacrificed, blood and tissue samples were taken for
analysis.
Results:
The results showed that the percentage of wound area had been significantly reduced
in Se NPs and niacin treated groups compared to the positive control. The level of
Vascular Endothelial cell Growth Factor and Collagenase I in Se NPs and niacin groups significantly
exceed those of other groups while Nitric Oxide (NO) was significantly decreased
in treated groups. Liver and kidney functions showed the lower toxicity effect of Se NPs and
niacin. Skin tissue showed the wound healing effect of Se NPs and niacin by regenerating
skin layer compared to the positive group.
Conclusion:
Se NPs and niacin play an important role in accelerating and reducing the time
of wound healing while they were antagonistic to each other.
Collapse
Affiliation(s)
- Marwa Emam
- Chemistry Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Akaber T. Keshta
- Chemistry Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Yasser M.A. Mohamed
- Photochemistry Department, National Research Center, Dokki, Giza 12622, Egypt
| | - Yasser A. Attia
- National Institute of Laser Enhanced Sciences, Cairo University, Giza 12613, Egypt
| |
Collapse
|
14
|
Chen Y, Luan J, Jiang T, Cai D, Sun C, Wang X, Zhao X, Gou X. Knockdown of EMMPRIN (OX47) in MRMT-1 Carcinoma Cells Inhibits Tumor Growth and Decreases Cancer-Induced Bone Destruction and Pain. Cancer Res Treat 2020; 53:576-583. [PMID: 33138345 PMCID: PMC8053874 DOI: 10.4143/crt.2020.801] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/20/2020] [Indexed: 11/29/2022] Open
Abstract
Purpose Bone destruction and pain caused by cancer is one of the most devastating complications of cancer patients with bone metastases, and it seriously affects the quality of patients’ life. Extracellular matrix metalloproteinase inducer (EMMPRIN) is a cell adhesion molecule with increased expression in a variety of tumors. This study focused to clarify the specific function of EMMPRIN in bone metastasis of breast cancer. Materials and Methods Adenovirus with shRNA-EMMPRIN was transfected into MRMT-1 rat breast carcinoma cells, and the MRMT-1 cells with different expression levels of EMMPRIN were implanted into the bone marrow cavity of rat tibia. Next, the effect of down-regulation of EMMPRIN was evaluated as follows: bone damage was detected by X-ray radiological and tartrate-resistant acid phosphatase staining; the tumor burden was evaluated by hematoxylin and eosin staining; the test of pain-related behaviors was assessed used the bilateral paw withdrawal mechanical threshold; and the levels of secretory factors in tumor conditioned medium were determined by using enzyme-linked immunosorbent assay. Results We found that down-regulation of EMMPRIN in tumor cells can simultaneously reduce tumor burden, relieve cancer-induced bone destruction and pain. Conclusion EMMPRIN is expected to be a therapeutic target for relieving bone metastasis of breast cancer and alleviating cancer-induced bone destruction and pain. The method of targeting EMMPRIN may be a promising strategy for the treatment of cancer in the future.
Collapse
Affiliation(s)
- Yanke Chen
- Department of Cell Biology and Genetics and Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Jing Luan
- Shaanxi Key Laboratory of Brain Disorders and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Ting Jiang
- Department of Cell Biology and Genetics and Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Donghui Cai
- Department of Cell Biology and Genetics and Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Chao Sun
- Department of Obstetrics, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xiaofei Wang
- Department of Cell Biology and Genetics and Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoge Zhao
- Department of Cell Biology and Genetics and Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Xingchun Gou
- Shaanxi Key Laboratory of Brain Disorders and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| |
Collapse
|
15
|
Guo WP, Tang D, Pang YY, Li XJ, Chen G, Huang ZG, Tang XZ, Lai QQ, Gan JY, Huang XL, Liu XF, Wei ZX, Ma W. Immunohistochemical basigin expression level in thyroid cancer tissues. World J Surg Oncol 2020; 18:240. [PMID: 32891152 PMCID: PMC7487720 DOI: 10.1186/s12957-020-01975-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 07/28/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Thyroid cancer (TC) is the most common endocrine malignancy; basigin (also known as BSG) plays a crucial role in tumor cell invasion, metastasis, and angiogenesis. This study was designed to identify the change of BSG expression in TC and its possible potential mechanism. METHODS The BSG expression levels in TC were demonstrated using data collected from in-house immunohistochemical (IHC), RNA-sequencing (RNA-seq), microarrays, and literatures. Integrated analysis was performed to determined BSG expression levels in TC comprehensively. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed with the integration of BSG co-expressed genes and differentially expressed genes (DEGs) in TC tissues to explore the potential mechanisms of BSG in TC. RESULTS The protein expression level of BSG was significantly higher in TC cases based on the IHC experiments. In addition, the combined SMD for BSG expression was 0.39 (p < 0.0001), the diagnostic odds ratio was 3.69, and the AUC of the sROC curve was 0.6986 using 1182 TC cases and 437 non-cancerous cases from 17 independent datasets. Furthermore, BSG co-expressed genes tended to be enriched in gene terms of the extracellular matrix (ECM), cell adhesion, and cell-cell interactions. The expression levels of nine hub BSG co-expressed genes were markedly upregulated in TC cases. CONCLUSION BSG expression levels were closely correlated with the progression of TC and may affect the signals of the ECM, cell adhesion, and cell-cell interactions.
Collapse
Affiliation(s)
- Wan-Ping Guo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Deng Tang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Yu-Yan Pang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xiao-Jiao Li
- Department of Nuclear Medicine, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Zhi-Guang Huang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xiao-Zhun Tang
- Department of Head and Neck Tumor Surgery, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Qin-Qiao Lai
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Jin-Yan Gan
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xiao-Li Huang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xiao-Fan Liu
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Zhi-Xiao Wei
- Department of Nuclear Medicine, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
| | - Wei Ma
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
| |
Collapse
|
16
|
Roles of Proteoglycans and Glycosaminoglycans in Cancer Development and Progression. Int J Mol Sci 2020; 21:ijms21175983. [PMID: 32825245 PMCID: PMC7504257 DOI: 10.3390/ijms21175983] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/18/2020] [Accepted: 08/18/2020] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix (ECM) spatiotemporally controls cell fate; however, dysregulation of ECM remodeling can lead to tumorigenesis and cancer development by providing favorable conditions for tumor cells. Proteoglycans (PGs) and glycosaminoglycans (GAGs) are the major macromolecules composing ECM. They influence both cell behavior and matrix properties through direct and indirect interactions with various cytokines, growth factors, cell surface receptors, adhesion molecules, enzymes, and glycoproteins within the ECM. The classical features of PGs/GAGs play well-known roles in cancer angiogenesis, proliferation, invasion, and metastasis. Several lines of evidence suggest that PGs/GAGs critically affect broader aspects in cancer initiation and the progression process, including regulation of cell metabolism, serving as a sensor of ECM's mechanical properties, affecting immune supervision, and participating in therapeutic resistance to various forms of treatment. These functions may be implemented through the characteristics of PGs/GAGs as molecular bridges linking ECM and cells in cell-specific and context-specific manners within the tumor microenvironment (TME). In this review, we intend to present a comprehensive illustration of the ways in which PGs/GAGs participate in and regulate several aspects of tumorigenesis; we put forward a perspective regarding their effects as biomarkers or targets for diagnoses and therapeutic interventions.
Collapse
|
17
|
Landras A, Reger de Moura C, Jouenne F, Lebbe C, Menashi S, Mourah S. CD147 Is a Promising Target of Tumor Progression and a Prognostic Biomarker. Cancers (Basel) 2019; 11:cancers11111803. [PMID: 31744072 PMCID: PMC6896083 DOI: 10.3390/cancers11111803] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/13/2019] [Accepted: 11/13/2019] [Indexed: 12/18/2022] Open
Abstract
Microenvironment plays a crucial role in tumor development and progression. Cancer cells modulate the tumor microenvironment, which also contribute to resistance to therapy. Identifying biomarkers involved in tumorigenesis and cancer progression represents a great challenge for cancer diagnosis and therapeutic strategy development. CD147 is a glycoprotein involved in the regulation of the tumor microenvironment and cancer progression by several mechanisms—in particular, by the control of glycolysis and also by its well-known ability to induce proteinases leading to matrix degradation, tumor cell invasion, metastasis and angiogenesis. Accumulating evidence has demonstrated the role of CD147 expression in tumor progression and prognosis, suggesting it as a relevant tumor biomarker for cancer diagnosis and prognosis, as well as validating its potential as a promising therapeutic target in cancers.
Collapse
Affiliation(s)
- Alexandra Landras
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
| | - Coralie Reger de Moura
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
- Pharmacogenomics Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint Louis Hospital, 75010 Paris, France
| | - Fanelie Jouenne
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
- Pharmacogenomics Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint Louis Hospital, 75010 Paris, France
| | - Celeste Lebbe
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
- Dermatology Department and Centre d’Investigation Clinique (CIC), Assistance Publique-Hôpitaux de Paris (AP-HP), Saint Louis Hospital, 75010 Paris, France
| | - Suzanne Menashi
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
- Pharmacogenomics Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint Louis Hospital, 75010 Paris, France
| | - Samia Mourah
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
- Pharmacogenomics Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint Louis Hospital, 75010 Paris, France
- Correspondence: ; Tel.: +33-1-42-49-48-85
| |
Collapse
|
18
|
Kumar D, Vetrivel U, Parameswaran S, Subramanian KK. Structural insights on druggable hotspots in CD147: A bull's eye view. Life Sci 2019; 224:76-87. [DOI: 10.1016/j.lfs.2019.03.044] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/11/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022]
|
19
|
Hypo-phosphorylated CD147 promotes migration and invasion of hepatocellular carcinoma cells and predicts a poor prognosis. Cell Oncol (Dordr) 2019; 42:537-554. [DOI: 10.1007/s13402-019-00444-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2019] [Indexed: 02/08/2023] Open
|
20
|
Guindolet D, Gabison EE. Role of CD147 (EMMPRIN/Basigin) in Tissue Remodeling. Anat Rec (Hoboken) 2019; 303:1584-1589. [DOI: 10.1002/ar.24089] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/31/2018] [Accepted: 11/07/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Damien Guindolet
- Fondation Ophtalmologique A. de Rothschild 25 rue Manin, 75019, Paris France
| | - Eric E. Gabison
- Fondation Ophtalmologique A. de Rothschild 25 rue Manin, 75019, Paris France
| |
Collapse
|
21
|
Dai DH, Qazi IH, Ran MX, Liang K, Zhang Y, Zhang M, Zhou GB, Angel C, Zeng CJ. Exploration of miRNA and mRNA Profiles in Fresh and Frozen-Thawed Boar Sperm by Transcriptome and Small RNA Sequencing. Int J Mol Sci 2019; 20:ijms20040802. [PMID: 30781801 PMCID: PMC6413023 DOI: 10.3390/ijms20040802] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 01/21/2019] [Accepted: 02/08/2019] [Indexed: 12/17/2022] Open
Abstract
Due to lower farrowing rate and reduced litter size with frozen-thawed semen, over 90% of artificial insemination (AI) is conducted using liquid stored boar semen. Although substantial progress has been made towards optimizing the cryopreservation protocols for boar sperm, the influencing factors and underlying mechanisms related to cryoinjury and freeze tolerance of boar sperm remain largely unknown. In this study, we report the differential expression of mRNAs and miRNAs between fresh and frozen-thawed boar sperm using high-throughput RNA sequencing. Our results showed that 567 mRNAs and 135 miRNAs were differentially expressed (DE) in fresh and frozen-thawed boar sperm. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed that the majority of DE mRNAs were enriched in environmental information processing such as cytokine-cytokine receptor interactions, PI3K-Akt signaling, cell adhesion, MAPK, and calcium signaling pathways. Moreover, the targets of DE miRNAs were enriched in significant GO terms such as cell process, protein binding, and response to stimuli. In conclusion, we speculate that DE mRNAs and miRNAs are heavily involved in boar sperm response to environment stimuli, apoptosis, and metabolic activities. The differences in expression also reflect the various structural and functional changes in sperm during cryopreservation.
Collapse
Affiliation(s)
- Ding-Hui Dai
- College of Animal Sciences and Technology, and Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| | - Izhar Hyder Qazi
- College of Animal Sciences and Technology, and Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
- Department of Veterinary Anatomy & Histology, Shaheed Benazir Bhutto University of Veterinary and Animal Sciences, Sakrand 67210, Pakistan.
| | - Ming-Xia Ran
- College of Animal Sciences and Technology, and Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| | - Kai Liang
- College of Animal Sciences and Technology, and Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| | - Yan Zhang
- College of Animal Sciences and Technology, and Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| | - Ming Zhang
- College of Animal Sciences and Technology, and Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| | - Guang-Bin Zhou
- College of Animal Sciences and Technology, and Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| | - Christiana Angel
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
- Department of Veterinary Parasitology, Faculty of Veterinary Sciences, Shaheed Benazir Bhutto University of Veterinary and Animal Sciences, Sakrand 67210, Pakistan.
| | - Chang-Jun Zeng
- College of Animal Sciences and Technology, and Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
22
|
Simanovich E, Brod V, Rahat MA. Active Vaccination With EMMPRIN-Derived Multiple Antigenic Peptide (161-MAP) Reduces Angiogenesis in a Dextran Sodium Sulfate (DSS)-Induced Colitis Model. Front Immunol 2018; 9:2919. [PMID: 30619283 PMCID: PMC6295553 DOI: 10.3389/fimmu.2018.02919] [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: 08/21/2018] [Accepted: 11/28/2018] [Indexed: 12/26/2022] Open
Abstract
Ulcerative colitis (UC) is an autoimmune disease that affects the colon and shares many clinical and histological features with the dextran sulfate sodium (DSS)-induced colitis model in mice. Angiogenesis is a critical component in many autoimmune diseases, as well as in the DSS-induced colitis model, and is it partially mediated by EMMPRIN, a multifunctional protein that can induce the expression of both the potent pro-angiogenic vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMPs). We asked whether targeting EMMPRIN by active vaccination, using a novel, specific epitope in the protein, synthesized as a multiple antigenic peptide (MAP), could trigger beneficial effects in the DSS-induced colitic C57BL/6J mice. Mice were vaccinated with four boost injections (50 μg each) of either 161-MAP coding for the EMMPRIN epitope or the scrambled control peptide (Scr-MAP) emulsified in Freund's adjuvant. We show that male mice that were vaccinated with 161-MAP lost less weight, demonstrated improved disease activity indices (DAI), had reduced colitis histological score, and their colons were longer in comparison to mice vaccinated with the Scr-MAP. The 161-MAP vaccination also reduced serum and colon levels of EMMPRIN, colon concentrations of VEGF, MMP-9, and TGFβ, and vessel density assessed by CD31 staining. A similar effect was observed in female mice vaccinated with 161-MAP, including weight loss, colitis histological score, colon length, colon levels of EMMPRIN and colon concentrations of VEGF. However, for female mice, the changes in DAI values, EMMPRIN serum levels, and MMP-9 and TGFβ colon concentrations did not reach significance. We conclude that our strategy of alleviating autoimmunity in this model through targeting angiogenesis by actively vaccinating against EMMPRIN was successful and efficient in reducing angiogenesis.
Collapse
Affiliation(s)
| | - Vera Brod
- Immunotherapy Laboratory, Carmel Medical Center, Haifa, Israel
| | - Michal A Rahat
- Immunotherapy Laboratory, Carmel Medical Center, Haifa, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
23
|
Preisner F, Leimer U, Sandmann S, Zoernig I, Germann G, Koellensperger E. Impact of Human Adipose Tissue-Derived Stem Cells on Malignant Melanoma Cells in An In Vitro Co-culture Model. Stem Cell Rev Rep 2018; 14:125-140. [PMID: 29064018 DOI: 10.1007/s12015-017-9772-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
This study focuses on the interactions of human adipose tissue-derived stem cells (ADSCs) and malignant melanoma cells (MMCs) with regard to future cell-based skin therapies. The aim was to identify potential oncological risks as ADSCs could unintentionally be sited within the proximity of the tumor microenvironment of MMCs. An indirect co-culture model was used to analyze interactions between ADSCs and four different established melanoma cell lines (G-361, SK-Mel-5, MeWo and A2058) as well as two low-passage primary melanoma cell cultures (M1 and M2). Doubling time, migration and invasion, angiogenesis, quantitative real-time PCR of 229 tumor-associated genes and multiplex protein assays of 20 chemokines and growth factors and eight matrix metalloproteinases (MMPs) were evaluated. Co-culture with ADSCs significantly increased migration capacity of G-361, SK-Mel-5, A2058, MeWo and M1 and invasion capacity of G-361, SK-Mel-5 and A2058 melanoma cells. Furthermore, conditioned media from all ADSC-MMC-co-cultures induced tube formation in an angiogenesis assay in vitro. Gene expression analysis of ADSCs and MMCs, especially of low-passage melanoma cell cultures, revealed an increased expression of various genes with tumor-promoting activities, such as CXCL12, PTGS2, IL-6, and HGF upon ADSC-MMC-co-culture. In this context, a significant increase (up to 5,145-fold) in the expression of numerous tumor-associated proteins could be observed, e.g. several pro-angiogenic factors, such as VEGF, IL-8, and CCL2, as well as different matrix metalloproteinases, especially MMP-2. In conclusion, the current report clearly demonstrates that a bi-directional crosstalk between ADSCs and melanoma cells can enhance different malignant properties of melanoma cells in vitro.
Collapse
Affiliation(s)
- Fabian Preisner
- ETHIANUM - Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine, Voßstraße 6, 69115, Heidelberg, Germany
| | - Uwe Leimer
- ETHIANUM - Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine, Voßstraße 6, 69115, Heidelberg, Germany
| | - Stefanie Sandmann
- ETHIANUM - Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine, Voßstraße 6, 69115, Heidelberg, Germany
| | - Inka Zoernig
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Im Neuenheimer Feld 460, 60120, Heidelberg, Germany
| | - Guenter Germann
- ETHIANUM - Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine, Voßstraße 6, 69115, Heidelberg, Germany
| | - Eva Koellensperger
- ETHIANUM - Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine, Voßstraße 6, 69115, Heidelberg, Germany.
| |
Collapse
|
24
|
Jeon H, Han SR, Lee S, Park SJ, Kim JH, Yoo SM, Lee MS. Activation of the complement system in an osteosarcoma cell line promotes angiogenesis through enhanced production of growth factors. Sci Rep 2018; 8:5415. [PMID: 29615744 PMCID: PMC5883033 DOI: 10.1038/s41598-018-23851-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/21/2018] [Indexed: 02/07/2023] Open
Abstract
There is increasing evidence that the complement system is activated in various cancer tissues. Besides being involved in innate immunity against pathogens, the complement system also participates in inflammation and the modulation of tumor microenvironment. Recent studies suggest that complement activation promotes tumor progression in various ways. Among some cancer cell lines, we found that human bone osteosarcoma epithelial cells (U2-OS) can activate the alternative pathway of the complement system by pooled normal human serum. Interestingly, U2-OS cells showed less expression of complement regulatory proteins, compared to other cancer cell lines. Furthermore, the activated complement system enhanced the production of growth factors, which promoted angiogenesis of human endothelial cells. Our results demonstrated a direct linkage between the complement system and angiogenesis using the in vitro model, which suggest the complement system and related mechanisms might be potential targets for cancer treatment.
Collapse
Affiliation(s)
- Hyungtaek Jeon
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, 34824, South Korea
| | - Seung Ro Han
- Eulji Biomedical Science Research Institute, Eulji University School of Medicine, Daejeon, 34824, Republic of Korea
| | - Suhyuk Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, 34824, South Korea
| | - Sang June Park
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, 34824, South Korea
| | - Joo Heon Kim
- Department of Pathology, Eulji University School of Medicine, Daejeon, 34824, South Korea
| | - Seung-Min Yoo
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, 34824, South Korea
| | - Myung-Shin Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, 34824, South Korea.
| |
Collapse
|
25
|
Matsumoto T, Nagashio R, Ryuge S, Igawa S, Kobayashi M, Fukuda E, Goshima N, Ichinoe M, Jiang SX, Satoh Y, Masuda N, Murakumo Y, Saegusa M, Sato Y. Basigin expression as a prognostic indicator in stage I pulmonary adenocarcinoma. Pathol Int 2018; 68:232-240. [PMID: 29431238 DOI: 10.1111/pin.12646] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 01/09/2018] [Indexed: 02/06/2023]
Abstract
We established the KU-Lu-8 monoclonal antibody (MoAb) using a lung cancer cell line as an antigen and a random immunization method. The KU-Lu-8 MoAb recognizes basigin (BSG), which is a transmembrane-type glycoprotein that is strongly expressed on the cell membranes of lung cancer cells. This study aimed to clarify the relationships between BSG expression and clinicopathological parameters and determine the prognostic significance of BSG expression in pulmonary adenocarcinoma (AC) patients. To evaluate the significance of BSG expression in lung cancer, we immunohistochemically analyzed 113 surgically resected pulmonary adenocarcinomas, and the associations between BSG expression and various clinicopathological parameters were evaluated. Kaplan-Meier survival analysis and Cox proportional hazards models were used to investigate the effects of BSG expression on survival. Clinicopathologically, BSG expression was significantly associated with tumor differentiation, vascular invasion, lymphatic invasion, and a poor prognosis. In particular, BSG expression was significantly correlated with poorer survival in patients with stage I AC. The high BSG expression group (compared with the low BSG expression group) exhibited adjusted hazard ratios for mortality of 4.694. BSG expression is indicative of a poor prognosis in AC patients, particularly in those with stage I disease.
Collapse
Affiliation(s)
- Toshihide Matsumoto
- Department of Pathology, School of Medicine, Kitasato University, Kanagawa, Japan
| | - Ryo Nagashio
- Department of Applied Tumor Pathology, Graduate School of Medical Sciences, Kitasato University, Kanagawa, Japan
| | - Shinichiro Ryuge
- Department of Respiratory Medicine, School of Medicine, Kitasato University, Kanagawa, Japan
| | - Satoshi Igawa
- Department of Respiratory Medicine, School of Medicine, Kitasato University, Kanagawa, Japan
| | - Makoto Kobayashi
- Department of Applied Tumor Pathology, Graduate School of Medical Sciences, Kitasato University, Kanagawa, Japan
| | - Eriko Fukuda
- Division of Quantitative Proteomics Team, Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Naoki Goshima
- Division of Quantitative Proteomics Team, Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Masaaki Ichinoe
- Department of Pathology, School of Medicine, Kitasato University, Kanagawa, Japan
| | - Shi-Xu Jiang
- Department of Pathology, School of Medicine, Kitasato University, Kanagawa, Japan
| | - Yukitoshi Satoh
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kitasato University, Kanagawa, Japan
| | - Noriyuki Masuda
- Department of Respiratory Medicine, School of Medicine, Kitasato University, Kanagawa, Japan
| | - Yoshiki Murakumo
- Department of Pathology, School of Medicine, Kitasato University, Kanagawa, Japan
| | - Makoto Saegusa
- Department of Pathology, School of Medicine, Kitasato University, Kanagawa, Japan
| | - Yuichi Sato
- Department of Applied Tumor Pathology, Graduate School of Medical Sciences, Kitasato University, Kanagawa, Japan
| |
Collapse
|
26
|
Li X, Yu X, Dai D, Song X, Xu W. The altered glucose metabolism in tumor and a tumor acidic microenvironment associated with extracellular matrix metalloproteinase inducer and monocarboxylate transporters. Oncotarget 2018; 7:23141-55. [PMID: 27009812 PMCID: PMC5029616 DOI: 10.18632/oncotarget.8153] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 01/13/2016] [Indexed: 02/06/2023] Open
Abstract
Extracellular matrix metalloproteinase inducer, also knowns as cluster of differentiation 147 (CD147) or basigin, is a widely distributed cell surface glycoprotein that is involved in numerous physiological and pathological functions, especially in tumor invasion and metastasis. Monocarboxylate transporters (MCTs) catalyze the proton-linked transport of monocarboxylates such as L-lactate across the plasma membrane to preserve the intracellular pH and maintain cell homeostasis. As a chaperone to some MCT isoforms, CD147 overexpression significantly contributes to the metabolic transformation of tumor. This overexpression is characterized by accelerated aerobic glycolysis and lactate efflux, and it eventually provides the tumor cells with a metabolic advantage and an invasive phenotype in the acidic tumor microenvironment. This review highlights the roles of CD147 and MCTs in tumor cell metabolism and the associated molecular mechanisms. The regulation of CD147 and MCTs may prove to be with a therapeutic potential for tumors through the metabolic modification of the tumor microenvironment.
Collapse
Affiliation(s)
- Xiaofeng Li
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiaozhou Yu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Dong Dai
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiuyu Song
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
27
|
Kusunoki M, Sata E, Nishio K, Tanaka T, Nishida T, Sugano N, Sato S, Asano M. Acid-electrolyzed functional water induces extracellular matrix metalloproteinase inducer, a possible novel alarmin, secretion from oral squamous cell carcinoma cell lines. Int J Med Sci 2018; 15:1365-1372. [PMID: 30275765 PMCID: PMC6158666 DOI: 10.7150/ijms.26186] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/30/2018] [Indexed: 11/05/2022] Open
Abstract
Extracellular matrix metalloproteinase inducer (EMMPRIN) secretion was induced in the oral squamous cell carcinoma cell line HSC3 cell by acid-electrolyzed functional water (FW) stimulation. Augmented EMMPRIN secretion was not under transcriptional control; rather, it was derived from the intracellular storages. EMMPRIN secretion was also induced under oxidative stress and accompanied by the release of lactate dehydrogenase (LDH). The molecules released from cells undergoing necrosis are called as alarmins, and the secretion of IL-1α, a typical alarmin, was induced by FW stimulation and oxidative stress. Intracellular localization was examined by cell fractionation. A significant amount of EMMPRIN was localized in the triton X-100 and DNase sensitive fractions; the levels were drastically reduced following FW treatment. The function of the released EMMPRIN was examined using the monocytic cell line THP1. Culture supernatant derived from FW-treated HSC3 cells induced the expression of matrix metalloproteinases (MMPs) 1, 2, 8, 9, 13, and 14, platelet-derived growth factor, and interleukin-8. In contrast, vascular endothelial growth factor expression was reduced. Induction of these factors was abolished following eliminating of EMMPRIN by immunoprecipitation. These results indicate that EMMPRIN might be considered as a type of alarmin that transduces danger signals to the surrounding cells.
Collapse
Affiliation(s)
- Masafumi Kusunoki
- Department of Periodontology, Nihon University School of Dentistry, 1-8-13 Kanda Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Eri Sata
- Department of Orthodontics, Nihon University School of Dentistry, 1-8-13 Kanda Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan.,Oral Structural and Functional Biology, Nihon University Graduate School of Dentistry, 1-8-13 Kanda Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Kensuke Nishio
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry, 1-8-13 Kanda Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Takayoshi Tanaka
- Maxillofacial Prosthetic Clinic, Nihon University School of Dentistry, Tokyo, Japan.,Department of Oral and Maxillofacial Surgery, Nihon University School of Dentistry, 1-8-13 Kanda Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Tetsuya Nishida
- Department of Periodontology, Nihon University School of Dentistry, 1-8-13 Kanda Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Naoyuki Sugano
- Department of Periodontology, Nihon University School of Dentistry, 1-8-13 Kanda Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Shuichi Sato
- Department of Periodontology, Nihon University School of Dentistry, 1-8-13 Kanda Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Masatake Asano
- Department of Pathology, Nihon University School of Dentistry, 1-8-13 Kanda Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan.,Division of Immunology and Pathobiology, Nihon University School of Dentistry, 1-8-13 Kanda Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| |
Collapse
|
28
|
Abu El-Asrar AM, Ahmad A, Alam K, Siddiquei MM, Mohammad G, Hertogh GD, Mousa A, Opdenakker G. Extracellular matrix metalloproteinase inducer (EMMPRIN) is a potential biomarker of angiogenesis in proliferative diabetic retinopathy. Acta Ophthalmol 2017; 95:697-704. [PMID: 27860331 DOI: 10.1111/aos.13284] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 09/13/2016] [Indexed: 12/12/2022]
Abstract
PURPOSE Extracellular matrix metalloproteinase inducer (EMMPRIN) promotes angiogenesis through matrix metalloproteinases (MMPs) and vascular endothelial growth factor (VEGF) production. We investigated the expression levels of EMMPRIN and correlated these levels with VEGF, MMP-1 and MMP-9 in proliferative diabetic retinopathy (PDR). In addition, we examined the expression of EMMPRIN in the retinas of diabetic rats and the effect of EMMPRIN on the induction of angiogenesis regulatory factors in human retinal microvascular endothelial cells (HRMECs). METHODS Vitreous samples from 40 PDR and 19 non-diabetic patients, epiretinal membranes from 12 patients with PDR, retinas of rats and HRMECs were studied by enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, Western blot analysis, zymography analysis and RT-PCR. RESULTS We showed a significant increase in the expression of EMMPRIN, VEGF, MMP-1 and MMP-9 in vitreous samples from PDR patients compared with non-diabetic controls (p < 0.0001; p = 0.001; p = 0.009; p < 0.0001, respectively). Significant positive correlations were found between the levels of EMMPRIN and the levels of VEGF (r = 0.38; p = 0.003), MMP-1 (r = 0.36; p = 0.005) and MMP-9 (r = 0.46; p = 0.003). In epiretinal membranes, EMMPRIN was expressed in vascular endothelial cells and stromal cells. Significant increase of EMMPRIN mRNA was detected in rat retinas after induction of diabetes. EMMPRIN induced hypoxia-inducible factor-1α, VEGF and MMP-1 expression in HRMEC. CONCLUSIONS These results suggest that EMMPRIN/MMPs/VEGF pathway is involved in PDR angiogenesis.
Collapse
Affiliation(s)
- Ahmed M. Abu El-Asrar
- Department of Ophthalmology; College of Medicine; King Saud University; Riyadh Saudi Arabia
- Dr. Nasser Al-Rashid Research Chair in Ophthalmology; College of Medicine; King Saud University; Riyadh Saudi Arabia
| | - Ajmal Ahmad
- Department of Ophthalmology; College of Medicine; King Saud University; Riyadh Saudi Arabia
| | - Kaiser Alam
- Department of Ophthalmology; College of Medicine; King Saud University; Riyadh Saudi Arabia
| | | | - Ghulam Mohammad
- Department of Ophthalmology; College of Medicine; King Saud University; Riyadh Saudi Arabia
| | - Gert De Hertogh
- Laboratory of Histochemistry and Cytochemistry; University of Leuven, KU Leuven; Leuven Belgium
| | - Ahmed Mousa
- Department of Ophthalmology; College of Medicine; King Saud University; Riyadh Saudi Arabia
| | - Ghislain Opdenakker
- Laboratory of Immunobiology; Rega Institute for Medical Research; Department of Microbiology and Immunology; University of Leuven; KU Leuven; Leuven Belgium
| |
Collapse
|
29
|
Dana P, Kariya R, Vaeteewoottacharn K, Sawanyawisuth K, Seubwai W, Matsuda K, Okada S, Wongkham S. Upregulation of CD147 Promotes Metastasis of Cholangiocarcinoma by Modulating the Epithelial-to-Mesenchymal Transitional Process. Oncol Res 2017; 25:1047-1059. [PMID: 28244853 PMCID: PMC7841082 DOI: 10.3727/096504016x14813899000565] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
CD147 is a transmembrane protein that can induce the expression and activity of matrix metalloproteinases (MMPs). Expression of CD147 has been shown to potentiate cell migration, invasion, and metastasis of cancer. In this study, the critical role of CD147 in metastasis was elucidated using CD147-overexpressing cholangiocarcinoma (CCA) cells in vitro and in vivo. The molecular mechanism, demonstrated herein, supported the hypothesis that metastasis increased in CD147-overexpressing cells. Five CD147-overexpressing clones (Ex-CD147) were established from a low CD147-expressing CCA cell line, KKU-055, using lentivirus containing pReceiver-Lenti-CD147. The metastatic capability was determined using the tail vein injection mouse model and an in vitro 3D invasion assay. Liver colonization was assessed using anti-HLA class I immunohistochemistry. Adhesion abilities, cytoskeletal arrangements, MMP activities, the expressions of adhesion molecules, and epithelial-mesenchymal transitional markers were analyzed. All Ex-CD147 clones exhibited a high CD147 expression and high liver colonization in the tail vein-injected mouse model, whereas parental cells lacked this ability. Ex-CD147 clones exhibited metastatic phenotypes (i.e., an increase in F-actin rearrangement) and cell invasion and a decrease in cell adhesion. The molecular mechanisms were shown to be via the induction of MMP-2 activity and enhancement of epithelial-mesenchymal transitions. An increase in mesenchymal markers Slug, vimentin, and N-cadherin, and a decrease in epithelial markers E-cadherin and claudin-1, together with suppression of the adhesion molecule ICAM-1, were observed in the Ex-CD147 clones. Moreover, suppression of CD147 expression using siCD147 in two CCA cell lines with high CD147 expression significantly decreased cell migration and invasion of these CCA cells. These findings emphasize the essential role of CD147 in CCA metastasis and suggest CD147 as a promising target for the effective treatment of CCA.
Collapse
Affiliation(s)
- Paweena Dana
- *Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- †Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- ‡Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Ryusho Kariya
- ‡Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Kulthida Vaeteewoottacharn
- *Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- †Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Kanlayanee Sawanyawisuth
- *Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- †Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Wunchana Seubwai
- †Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- §Department of Forensic Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Kouki Matsuda
- ‡Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Seiji Okada
- ‡Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Sopit Wongkham
- *Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- †Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
30
|
Zhang CY, Yu MS, Li X, Zhang Z, Han CR, Yan B. Overexpression of long non-coding RNA MEG3 suppresses breast cancer cell proliferation, invasion, and angiogenesis through AKT pathway. Tumour Biol 2017. [PMID: 28635399 DOI: 10.1177/1010428317701311] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Long non-coding RNA MEG3 has been identified as a tumor suppressor which plays important roles in tumorigenesis; however, its potential role in breast cancer has not been fully examined. Here, we showed that MEG3 was downregulated in breast cancer tissues and cell lines. Overexpression of MEG3 inhibited breast cancer cell proliferation and invasion, suggesting that MEG3 played an important role in breast cancer progression and metastasis. Moreover, MEG3 upregulation caused marked inhibition of angiogenesis-related factor expression. Conditioned medium derived from MEG3 overexpressed breast cancer cells significantly decreased the capillary tube formation of endothelial cells. Furthermore, elevated expression of MEG3 in breast cancer inhibits in vivo tumorigenesis and angiogenesis in a nude mouse xenograft model. Mechanistically, overexpression of MEG3 results in downregulation of AKT signaling, which is pivotal for breast cancer cell growth, invasion, and tumor angiogenesis. Collectively, these results suggest that MEG3 might suppress the tumor growth and angiogenesis via AKT signaling pathway and MEG3 may serve as a potential novel diagnostic and therapeutic target of breast cancer.
Collapse
Affiliation(s)
- Chen-Yu Zhang
- 1 Department of General Surgery, Shanghai Pudong District People's Hospital, Shanghai, China.,2 Department of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming-Sheng Yu
- 2 Department of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Li
- 2 Department of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhe Zhang
- 2 Department of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ce-Ran Han
- 1 Department of General Surgery, Shanghai Pudong District People's Hospital, Shanghai, China
| | - Bo Yan
- 1 Department of General Surgery, Shanghai Pudong District People's Hospital, Shanghai, China
| |
Collapse
|
31
|
Hahn JN, Kaushik DK, Mishra MK, Wang J, Silva C, Yong VW. Impact of Minocycline on Extracellular Matrix Metalloproteinase Inducer, a Factor Implicated in Multiple Sclerosis Immunopathogenesis. THE JOURNAL OF IMMUNOLOGY 2016; 197:3850-3860. [PMID: 27733550 DOI: 10.4049/jimmunol.1600436] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 09/12/2016] [Indexed: 12/28/2022]
Abstract
Extracellular matrix metalloproteinase inducer (EMMPRIN, CD147) is a transmembrane glycoprotein that is upregulated on leukocytes in active lesions in multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Administration of anti-EMMPRIN Abs reduces the severity of EAE. Minocycline is a tetracycline antibiotic with immune-modulatory properties that decreases the severity of EAE; it was recently found to attenuate the conversion from a first demyelinating event to clinically definite MS in a phase III trial. We investigated whether and how minocycline affects the expression of EMMPRIN on T cells in culture and in mice afflicted with EAE. EMMPRIN expression in cultures of mouse splenocytes or human PBMCs was elevated upon polyclonal T cell activation, and this was reduced by minocycline correspondent with decreased P-Akt levels. An established MS medication, IFN-β, also diminished EMMPRIN levels on human cells whereas this was not readily observed for fingolimod or monomethylfumarate. In EAE-afflicted mice, minocycline treatment significantly reduced EMMPRIN levels on splenic lymphocytes at the presymptomatic (day 7) phase, and prevented the development of disease. Day 7 spleen transcripts from minocycline-treated EAE mice had a significantly lower MMP-9/TIMP-1 ratio, and significantly lower MCT-1 and CD98 levels, factors associated with EMMPRIN function. Day 16 (peak clinical severity) CNS samples from EAE mice had prominent representation of inflammatory perivascular cuffs, inflammatory molecules and EMMPRIN, and these were abrogated by minocycline. Overall, minocycline attenuated the activation-induced elevation of EMMPRIN on T cells in culture and in EAE mice, correspondent with reduced immune function and EAE CNS pathology.
Collapse
Affiliation(s)
- Jennifer N Hahn
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Deepak K Kaushik
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Manoj K Mishra
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Jianxiong Wang
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Claudia Silva
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
32
|
Vrijsen KR, Maring JA, Chamuleau SAJ, Verhage V, Mol EA, Deddens JC, Metz CHG, Lodder K, van Eeuwijk ECM, van Dommelen SM, Doevendans PA, Smits AM, Goumans MJ, Sluijter JPG. Exosomes from Cardiomyocyte Progenitor Cells and Mesenchymal Stem Cells Stimulate Angiogenesis Via EMMPRIN. Adv Healthc Mater 2016; 5:2555-2565. [PMID: 27570124 DOI: 10.1002/adhm.201600308] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 06/21/2016] [Indexed: 11/06/2022]
Abstract
To date, cellular transplantation therapy has not yet fulfilled its high expectations for cardiac repair. A major limiting factor is lack of long-term engraftment of the transplanted cells. Interestingly, transplanted cells can positively affect their environment via secreted paracrine factors, among which are extracellular vesicles, including exosomes: small bi-lipid-layered vesicles containing proteins, mRNAs, and miRNAs. An exosome-based therapy will therefore relay a plethora of effects, without some of the limiting factors of cell therapy. Since cardiomyocyte progenitor cells (CMPC) and mesenchymal stem cells (MSC) induce vessel formation and are frequently investigated for cardiac-related therapies, the pro-angiogenic properties of CMPC and MSC-derived exosome-like vesicles are investigated. Both cell types secrete exosome-like vesicles, which are efficiently taken up by endothelial cells. Endothelial cell migration and vessel formation are stimulated by these exosomes in in vitro models, mediated via ERK/Akt-signaling. Additionally, these exosomes stimulated blood vessel formation into matrigel plugs. Analysis of pro-angiogenic factors revealed high levels of extracellular matrix metalloproteinase inducer (EMMPRIN). Knockdown of EMMPRIN on CMPCs leads to a diminished pro-angiogenic effect, both in vitro and in vivo. Therefore, CMPC and MSC exosomes have powerful pro-angiogenic effects, and this effect is largely mediated via the presence of EMMPRIN on exosomes.
Collapse
Affiliation(s)
- Krijn R. Vrijsen
- Department of Cardiology; Laboratory of Experimental Cardiology; University Medical Center Utrecht; Utrecht 3584CX The Netherlands
| | - Janita A. Maring
- Department of Molecular Cell Biology; Leiden University Medical Center; 2333ZA The Netherlands
| | - Steven A. J. Chamuleau
- Department of Cardiology; Laboratory of Experimental Cardiology; University Medical Center Utrecht; Utrecht 3584CX The Netherlands
- UMC Utrecht Regenerative Medicine Center; University Medical Center; Utrecht 3584CT The Netherlands
| | - Vera Verhage
- Department of Cardiology; Laboratory of Experimental Cardiology; University Medical Center Utrecht; Utrecht 3584CX The Netherlands
| | - Emma A. Mol
- Department of Cardiology; Laboratory of Experimental Cardiology; University Medical Center Utrecht; Utrecht 3584CX The Netherlands
| | - Janine C. Deddens
- Department of Cardiology; Laboratory of Experimental Cardiology; University Medical Center Utrecht; Utrecht 3584CX The Netherlands
| | - Corina H. G. Metz
- Department of Cardiology; Laboratory of Experimental Cardiology; University Medical Center Utrecht; Utrecht 3584CX The Netherlands
- UMC Utrecht Regenerative Medicine Center; University Medical Center; Utrecht 3584CT The Netherlands
| | - Kirsten Lodder
- Department of Molecular Cell Biology; Leiden University Medical Center; 2333ZA The Netherlands
| | - Esther C. M. van Eeuwijk
- Department of Cardiology; Laboratory of Experimental Cardiology; University Medical Center Utrecht; Utrecht 3584CX The Netherlands
| | - Susan M. van Dommelen
- Department of Clinical Chemistry and Haematology; University Medical Center Utrecht; Utrecht 3584CX The Netherlands
| | - Pieter A. Doevendans
- Department of Cardiology; Laboratory of Experimental Cardiology; University Medical Center Utrecht; Utrecht 3584CX The Netherlands
- UMC Utrecht Regenerative Medicine Center; University Medical Center; Utrecht 3584CT The Netherlands
- Netherlands Heart Institute (ICIN); Utrecht 3584CX The Netherlands
| | - Anke M. Smits
- Department of Molecular Cell Biology; Leiden University Medical Center; 2333ZA The Netherlands
| | - Marie-José Goumans
- Department of Molecular Cell Biology; Leiden University Medical Center; 2333ZA The Netherlands
| | - Joost P. G. Sluijter
- Department of Cardiology; Laboratory of Experimental Cardiology; University Medical Center Utrecht; Utrecht 3584CX The Netherlands
- UMC Utrecht Regenerative Medicine Center; University Medical Center; Utrecht 3584CT The Netherlands
- Netherlands Heart Institute (ICIN); Utrecht 3584CX The Netherlands
| |
Collapse
|
33
|
CD147 induces up-regulation of vascular endothelial growth factor in U937-derived foam cells through PI3K/AKT pathway. Arch Biochem Biophys 2016; 609:31-38. [PMID: 27619643 DOI: 10.1016/j.abb.2016.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 08/14/2016] [Accepted: 09/08/2016] [Indexed: 11/23/2022]
Abstract
Intraplaque angiogenesis has been recognized as an important risk factor for the rupture of advanced atherosclerotic plaques in recent years. CD147, also called Extracellular Matrix Metalloproteinase Inducer, has been found the ability to promote angiogenesis in many pathological conditions such as cancer diseases and rheumatoid arthritis via the up-regulation of vascular endothelial growth factor (VEGF), a critical mediator of angiogenesis. We investigated whether CD147 would also induce the up-regulation of VEGF in the foam cells formation process and explored the probable signaling pathway. The results showed the expression of CD147 and VEGF was significantly higher in U937-derived foam cells. After CD147 stealth siRNA transfection treatment, the production of VEGF was reduced depended on the inhibition efficiency of CD147 siRNAs.The special signaling pathway inhibitors LY294002, SP600125, SB203580 and U0126 were added to cultures respectively and the results showed LY294002 dose-dependently inhibited the expression of VEGF. The reduction of phospho-Akt was observed in both LY294002 and siRNA groups, suggested that the phosphatidylinositol 3-kinase/Akt pathway may be the probable signaling pathway underlying CD147 induced up-regulation of VEGF in U937-derived foam cells.
Collapse
|
34
|
Fisel P, Stühler V, Bedke J, Winter S, Rausch S, Hennenlotter J, Nies AT, Stenzl A, Scharpf M, Fend F, Kruck S, Schwab M, Schaeffeler E. MCT4 surpasses the prognostic relevance of the ancillary protein CD147 in clear cell renal cell carcinoma. Oncotarget 2016; 6:30615-27. [PMID: 26384346 PMCID: PMC4741556 DOI: 10.18632/oncotarget.5593] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/22/2015] [Indexed: 12/02/2022] Open
Abstract
Cluster of differentiation 147 (CD147/BSG) is a transmembrane glycoprotein mediating oncogenic processes partly through its role as binding partner for monocarboxylate transporter MCT4/SLC16A3. As demonstrated for MCT4, CD147 is proposed to be associated with progression in clear cell renal cell carcinoma (ccRCC). In this study, we evaluated the prognostic relevance of CD147 in comparison to MCT4/SLC16A3 expression and DNA methylation.
Collapse
Affiliation(s)
- Pascale Fisel
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Viktoria Stühler
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Jens Bedke
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Reseach Center (DKFZ), Heidelberg, Germany
| | - Stefan Winter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Steffen Rausch
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Jörg Hennenlotter
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Anne T Nies
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Reseach Center (DKFZ), Heidelberg, Germany
| | - Marcus Scharpf
- Institute of Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Falko Fend
- Institute of Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Stephan Kruck
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Reseach Center (DKFZ), Heidelberg, Germany.,Department of Clinical Pharmacology, University Hospital Tuebingen, Tuebingen, Germany
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
35
|
Liu Y, Zhang J, Sun X, Li M. EMMPRIN Down-regulating miR-106a/b Modifies Breast Cancer Stem-like Cell Properties via Interaction with Fibroblasts Through STAT3 and HIF-1α. Sci Rep 2016; 6:28329. [PMID: 27325313 PMCID: PMC4914854 DOI: 10.1038/srep28329] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 06/02/2016] [Indexed: 12/17/2022] Open
Abstract
Extracellular matrix metalloproteinase inducer (EMMPRIN) is a heavily glycosylated protein and expresses in cancer cells widely, which plays important roles in tumor progression. However, the role of EMMPRIN in breast cancer stem-like cell properties by interaction with fibroblasts is not known. In the present study, we investigated the effects of fibroblasts on breast cancer stem-like cells. We found that fibroblasts activated by co-cultured breast cancer cells produced higher levels of EMMPRIN, which stimulated the stem-like cell specific, self-renewal and sphere-forming phenotype in breast cancer cells. Increased EMMPRIN expression in activated fibroblasts increased the expression of STAT3 and HIF-1α and showed cancer stem-like cell features in breast cancer cells. We also found that EMMPRIN could down-regulate miR-106a and miR-106b expression in breast cancer cells, which led to activating STAT3 and enhancing HIF-1α expression. Our results illustrated that EMMPRIN has an important role in breast cancer stem-like cells by activation STAT3/HIF-1α through interaction with cancer cells and fibroblasts. The study for the first time indicated that cancer cells and fibroblasts interaction promotes breast cancer cells showing stem-like cells through up-regulation EMMPRIN, and led to inhibiting miR-106a/b expression which targets both STAT3 and HIF-1α expression.
Collapse
Affiliation(s)
- Yonglei Liu
- Research center, Linyi People's Hospital, Shandong, China.,Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jingling Zhang
- Research center, Linyi People's Hospital, Shandong, China
| | - Xiangjun Sun
- Department of Surgery, Linyi People's Hospital, Shandong, China
| | - Meilin Li
- Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
36
|
Zhang X, Huang S, Guo J, Zhou L, You L, Zhang T, Zhao Y. Insights into the distinct roles of MMP-11 in tumor biology and future therapeutics (Review). Int J Oncol 2016; 48:1783-93. [PMID: 26892540 DOI: 10.3892/ijo.2016.3400] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 01/21/2016] [Indexed: 11/06/2022] Open
Abstract
The biological processes of cancer cells such as tumorigenesis, proliferation, angiogenesis, apoptosis and invasion are greatly influenced by the surrounding microenvironment. The ability of solid malignant tumors to alter the microenvironment represents an important characteristic through which tumor cells are able to acquire specific functions necessary for their malignant biological behaviors. Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases with the capacity of remodeling extracellular matrix (ECM) by degrading almost all ECM proteins, which plays essential roles during the invasion and metastasis process of solid malignant tumors, including allowing tumor cells to modify the ECM components and release cytokines, ultimately facilitating protease-dependent tumor progression. MMP-11, also named stromelysin-3, is a member of the stromelysin subgroup belonging to MMPs superfamily, which has been detected in cancer cells, stromal cells and adjacent microenvironment. Differently, MMP-11 exerts a dual effect on tumors. On the one hand MMP-11 promotes cancer development by inhibiting apoptosis as well as enhancing migration and invasion of cancer cells, on the other hand MMP-11 plays a negative role against cancer development via suppressing metastasis in animal models. Overexpression of MMP-11 was discovered in sera of cancer patients compared with normal control group as well as in multiple tumor tissue specimens, such as gastric cancer, breast cancer, and pancreatic cancer. At present, some evidence supports that MMP-11 may work as a significant tumor biomarker for early detection of cancer, tumor staging, prognostic analysis, monitoring recurrence during follow-up and also a potential target for immunotherapy against cancer. In view of the importance of MMP-11 in modifying tumor microenvironment and potent antitumoral effects on solid tumors, there is an urgent need for a deeper understanding of how MMP-11 modulates tumor progression, and exploring its potential clinical application.
Collapse
Affiliation(s)
- Xu Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Shuai Huang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Junchao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
37
|
Liu H, Xu XF, Zhao Y, Tang MC, Zhou YQ, Gao FH. NS-398 promotes pancreatic cancer cell invasion by CD147 and MMP-2 via the activation of P38. Mol Med Rep 2016; 13:2208-14. [PMID: 26782265 DOI: 10.3892/mmr.2016.4783] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 10/29/2015] [Indexed: 11/05/2022] Open
Abstract
The overexpression or abnormal activation of cyclo‑oxygenase‑2 (COX‑2) has been reported in pancreatic cancer cells. NS‑398, a selective inhibitor of COX‑2, is unable to inhibit pancreatic cancer cell proliferation, as determined by a Cell Counting Kit 8 assay. However, it does increase cancer cell invasiveness, and therefore the invasiveness of the PANC‑1 cells was determined, along with the activation of P38, which was assessed by western blotting. In the present study, to evaluate the mechanisms underlying the action of NS‑398 in pancreatic cancer cells, PANC‑1 cells were treated with NS‑398, and the invasion signaling pathways of cluster of differentiation (CD)147‑matrix metalloproteinase (MMP)‑2 and mitogen‑activated protein kinases were evaluated. The results showed that NS‑398‑induced the expression of CD147 and MMP‑2 via the activation of P38, which was involved in antiproliferative activity and induced pancreatic cancer cell invasiveness. The PANC‑1 cells were also co‑treated with CD147 small interfering (si)RNA and NS‑398, and it was found that the NS‑398‑induced activation of P38 was not inhibited by CD147 siRNA, however, the expression of MMP‑2 was inhibited. CD147 siRNA inhibited the invasiveness of the pancreatic cancer cells induced by NS‑398, but also restored NS‑398‑induced antiproliferative activity. These data indicated that P38 in the pancreatic cancer cells was non‑specifically activated by NS‑398. This activation induced the expression of CD147‑MMP‑2, opposed the antiproliferative activity of NS‑398 and increased the invasiveness of the PANC‑1 cells.
Collapse
Affiliation(s)
- Hua Liu
- Department of Gastroenterology, The Tenth Hospital Affiliated to Tongji University, Shanghai 200072, P.R. China
| | - Xuan-Fu Xu
- Department of Gastroenterology, The Tenth Hospital Affiliated to Tongji University, Shanghai 200072, P.R. China
| | - Yan Zhao
- Department of Gastroenterology, The Tenth Hospital Affiliated to Tongji University, Shanghai 200072, P.R. China
| | - Mao-Chun Tang
- Department of Gastroenterology, The Tenth Hospital Affiliated to Tongji University, Shanghai 200072, P.R. China
| | - Ying-Qun Zhou
- Department of Gastroenterology, The Tenth Hospital Affiliated to Tongji University, Shanghai 200072, P.R. China
| | - Feng-Hou Gao
- Institute of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
38
|
Chen Y, Gou X, Kong DK, Wang X, Wang J, Chen Z, Huang C, Zhou J. EMMPRIN regulates tumor growth and metastasis by recruiting bone marrow-derived cells through paracrine signaling of SDF-1 and VEGF. Oncotarget 2015; 6:32575-85. [PMID: 26416452 PMCID: PMC4741713 DOI: 10.18632/oncotarget.5331] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 09/11/2015] [Indexed: 12/23/2022] Open
Abstract
EMMPRIN, a cell adhesion molecule highly expressed in a variety of tumors, is associated with poor prognosis in cancer patients. Mechanistically, EMMPRIN has been characterized to contribute to tumor development and progression by controlling the expression of MMPs and VEGF. In the present study, by using fluorescently labeled bone marrow-derived cells (BMDCs), we found that the down-regulation of EMMPRIN expression in cancer cells reduces tumor growth and metastasis, and is associated with the reduced recruitment of BMDCs. Further protein profiling studies suggest that EMMPRIN controls BMDC recruitment through regulating the secretion of soluble factors, notably, VEGF and SDF-1. We demonstrate that the expression and secretion of SDF-1 in tumor cells are regulated by EMMPRIN. This study reveals a novel mechanism by which EMMPRIN promotes tumor growth and metastasis by recruitment of BMDCs through controlling secretion and paracrine signaling of SDF-1 and VEGF.
Collapse
Affiliation(s)
- Yanke Chen
- Experiment Center of Biomedical Research School of Medicine, Xi'an Jiaotong University, Xi'an 710061, P. R. China
- Department of Neurosurgery, Yale University, New Haven, CT 06511, USA
| | - Xingchun Gou
- Department of Neurosurgery, Yale University, New Haven, CT 06511, USA
- Laboratory of Cell Biology and Translational Medicine, Xi'an Medical University, Xi'an 710021, P. R. China
| | - Derek Kai Kong
- Department of Neurosurgery, Yale University, New Haven, CT 06511, USA
| | - Xiaofei Wang
- Experiment Center of Biomedical Research School of Medicine, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Jianhui Wang
- Department of Pathology, Yale University, New Haven, CT 06511, USA
| | - Zeming Chen
- Department of Neurosurgery, Yale University, New Haven, CT 06511, USA
| | - Chen Huang
- Experiment Center of Biomedical Research School of Medicine, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Jiangbing Zhou
- Department of Neurosurgery, Yale University, New Haven, CT 06511, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
39
|
Li HW, Yang XM, Tang J, Wang SJ, Chen ZN, Jiang JL. Effects of HAb18G/CD147 knockout on hepatocellular carcinoma cells in vitro using a novel zinc-finger nuclease-targeted gene knockout approach. Cell Biochem Biophys 2015; 71:881-90. [PMID: 25308857 DOI: 10.1007/s12013-014-0278-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
HAb18G/CD147 belongs to the immunoglobulin superfamily and predominantly functions as an inducer of matrix metalloproteinase secretion for tumor invasion and metastasis. This study was designed to investigate the effects of HAb18G/CD147 knockout on hepatocellular carcinoma cells using zinc-finger nuclease (ZFNs)-targeted gene knockout approach. The HCC cell line SMMC-7721 was used for ZFNs-targeted cleavage of the HAb18G/CD147 gene. RT-PCR and Western blot assays were used to detect HAb18G/CD147 expression. HAb18G phenotypic changes following HAb18G/CD147 knockout in SMMC-K7721 cells were assessed using tumor cell adhesion, invasion, migration and colony formation and flow cytometric assays. These data demonstrated that tumor cell adhesion, invasion, migration, and colony formation capabilities of SMMC-K7721 were significantly reduced compared to parental cells or SMMC-7721 with re-expression of HAb18G/CD147 protein transfected with HAb18G/CD147 cDNA. Moreover, knockout of HAb18G/CD147 expression also induced SMMC-K7721 cells to undergo apoptosis compared to SMMC-7721 and SMMC-R7721 (P < 0.01). Molecularly, protein expression of p53 was induced in these cells, but re-expression of HAb18G/CD147 reduced p53 levels in SMMC-R7721 cells, possibly through inhibition of the PI3K-Akt-MDM2 signaling pathway. The findings provide a novel insight into the mechanisms underlying HAb18G/CD147-induced progression of HCC cells.
Collapse
Affiliation(s)
- Hong-Wei Li
- Cell Engineering Research Centre and Department of Cell Biology, State Key Discipline of Cell Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | | | | | | | | | | |
Collapse
|
40
|
Knutti N, Kuepper M, Friedrich K. Soluble extracellular matrix metalloproteinase inducer (EMMPRIN, EMN) regulates cancer-related cellular functions by homotypic interactions with surface CD147. FEBS J 2015; 282:4187-200. [PMID: 26277583 DOI: 10.1111/febs.13414] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 07/12/2015] [Accepted: 08/12/2015] [Indexed: 12/27/2022]
Abstract
EMMPRIN (extracellular matrix metalloproteinase inducer) is a widely expressed glycoprotein and a member of the immunoglobulin superfamily which exists in both a membrane-spanning and a soluble form. Homotypic interactions of EMMPRIN underlie its multiple roles in normal development and pathological situations such as viral infections, Alzheimer's disease and cancer. This study employed a recombinant soluble, fully glycosylated EMMPRIN domain (rhsEMN) as a tool to characterize the structural basis of EMMPRIN-EMMPRIN receptor (EMNR) contacts and their functional effects on MCF-7 breast carcinoma cells. rhsEMN did not form dimers in solution but bound to surface EMMPRIN (EMN) on MCF-7 cells with high affinity and was readily internalized. The interaction interface for the homotypic contact was localized to the N-terminal Ig domain. rhsEMN exerted a stimulatory effect on proliferation of MCF-7 cells whereas it reduced cell migration in a dose-dependent manner. These effects were accompanied by an upregulation of endogenous EMMPRIN as well as of matrix metalloproteinase-14 (MMP-14), a membrane-bound protease involved in the extracellular release of soluble EMMPRIN, indicating a regulatory feedback mechanism. The proliferation-promoting activity of rhsEMN was mimicked by a novel functional antibody directed to EMMPRIN, underscoring that crosslinking of cell surface EMMPRIN (EMNR) is crucial for eliciting intracellular signalling. Addressing malignancy-related signal transduction in HEK-293 cells, we could show that rhsEMN triggers the oncogenic Wnt pathway.
Collapse
Affiliation(s)
- Nadine Knutti
- Institute of Biochemistry II, Jena University Hospital, Germany
| | | | | |
Collapse
|
41
|
Walter M, Simanovich E, Brod V, Lahat N, Bitterman H, Rahat MA. An epitope-specific novel anti-EMMPRIN polyclonal antibody inhibits tumor progression. Oncoimmunology 2015; 5:e1078056. [PMID: 27057452 DOI: 10.1080/2162402x.2015.1078056] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/22/2015] [Accepted: 07/22/2015] [Indexed: 01/14/2023] Open
Abstract
Extracellular matrix metalloproteinase inducer (EMMPRIN/CD147) mediates tumor cell-macrophage interactions, and has been shown to induce both matrix metalloproteinases (MMPs) and vascular endothelial growth factor (VEGF). However, the epitope responsible for MMP induction is controversial, and the epitope responsible for VEGF induction is yet unknown. We generated a novel anti-EMMPRIN antibody directed against a specific epitope that successfully inhibited the production of both MMP-9 and VEGF in tumor cell-macrophage in vitro co-culture systems, exhibiting a U-shaped dose response. Furthermore, this antibody efficiently inhibited in vivo tumor progression in both the RENCA renal cell carcinoma and CT26 colon carcinoma subcutaneous tumor models, and reduced tumor size and number of metastatic foci in the 4T1 orthotopic model. This was achieved by inhibiting angiogenesis as assessed by immunohistochemical staining for the endothelial marker CD31, by inhibiting tumor cell proliferation as assessed by the staining for Ki-67, and by enhancing tumor cell apoptosis as assessed in the TUNEL assay. Moreover, administration of the antibody recruited more macrophages into the tumor, and skewed the tumor microenvironment for macrophages from TGFβ-dominated anti-inflammatory microenvironment, to a less immunosuppressive one. The antibody improved the ability of stimulated macrophages to perform antibody-dependent cell cytotoxicity (ADCC) and kill tumor cells. Thus, our new antibody maps the epitope capable of inducing both MMPs and VEGF, and places EMMPRIN as a good target for cancer therapy.
Collapse
Affiliation(s)
- Miriam Walter
- Immunology Research Unit, Carmel Medical Center , Haifa, Israel
| | | | - Vera Brod
- Ischemia-shock Research Laboratory, Carmel Medical Center, The Ruth and Bruce Rappaport Faculty of Medicine , Technion, Haifa, Israel
| | - Nitza Lahat
- Immunology Research Unit, Carmel Medical Center , Haifa, Israel
| | - Haim Bitterman
- Ischemia-shock Research Laboratory, Carmel Medical Center, The Ruth and Bruce Rappaport Faculty of Medicine , Technion, Haifa, Israel
| | - Michal A Rahat
- Immunology Research Unit, Carmel Medical Center , Haifa, Israel
| |
Collapse
|
42
|
Wang SJ, Cui HY, Liu YM, Zhao P, Zhang Y, Fu ZG, Chen ZN, Jiang JL. CD147 promotes Src-dependent activation of Rac1 signaling through STAT3/DOCK8 during the motility of hepatocellular carcinoma cells. Oncotarget 2015; 6:243-57. [PMID: 25428919 PMCID: PMC4381592 DOI: 10.18632/oncotarget.2801] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 11/15/2014] [Indexed: 12/19/2022] Open
Abstract
Metastasis is considered a dynamic process in tumor development that is related to abnormal migration and invasion. Tumor cells can move as individual cells in two interconvertible modes: mesenchymal-type and amoeboid. Previously, we reported that the interaction between CD147 and Annexin II can inhibit the amoeboid movement in hepatocellular carcinoma (HCC) cells. However, the mechanism of CD147 involved in mesenchymal movement is still unclear. Notably, our results show overexpression of CD147 led to mesenchymal-type movement in HCC cells. Evidence indicated that the mesenchymal-type cell movement induced by CD147 was Src dependent, as observed by confocal microscopy and Rac1 activity assay. The phosphorylation of Src (pY416-Src) can be up-regulated by CD147, and this regulation is mediated by focal adhesion kinase (FAK). Next, we identified DOCK8 as a GEF for Rac1, a key molecule driving mesenchymal-type movement. We also found that Src promotes STAT3 phosphorylation and STAT3 facilitates DOCK8 transcription, thus enhancing DOCK8 expression and Rac1 activation. This study provides a novel mechanism of CD147 regulating mesenchymal-type movement in HCC cells.
Collapse
Affiliation(s)
- Shi-Jie Wang
- Cell Engineering Research Center & Department of Cell Biology, State Key Laboratory of Cancer Biology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Hong-Yong Cui
- Cell Engineering Research Center & Department of Cell Biology, State Key Laboratory of Cancer Biology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Yan-Mei Liu
- Cell Engineering Research Center & Department of Cell Biology, State Key Laboratory of Cancer Biology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Pu Zhao
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yang Zhang
- Cell Engineering Research Center & Department of Cell Biology, State Key Laboratory of Cancer Biology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Zhi-Guang Fu
- Cell Engineering Research Center & Department of Cell Biology, State Key Laboratory of Cancer Biology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Zhi-Nan Chen
- Cell Engineering Research Center & Department of Cell Biology, State Key Laboratory of Cancer Biology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Jian-Li Jiang
- Cell Engineering Research Center & Department of Cell Biology, State Key Laboratory of Cancer Biology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
43
|
CD147 promotes liver fibrosis progression via VEGF-A/VEGFR2 signalling-mediated cross-talk between hepatocytes and sinusoidal endothelial cells. Clin Sci (Lond) 2015. [PMID: 26201021 DOI: 10.1042/cs20140823] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although previous evidence indicates close involvement of CD147 in the pathogenesis of liver fibrosis, the underlying molecular mechanisms and its therapeutic value remain largely unknown. In the present study, we investigated the biological roles of CD147 in liver fibrosis and assessed its therapeutic value as a target molecule in the CCl4-induced liver fibrosis mouse model. We found that CD147 was highly expressed in both hepatocytes and SECs (sinusoidal endothelial cells) in fibrotic liver tissues. Additionally, it was significantly associated with the fibrosis stage. TGF-β1 (transforming growth factor β1) was found to be mainly responsible for the up-regulation of CD147. Bioinformatic and experimental data suggest a functional link between CD147 expression and VEGF-A (vascular endothelial growth factor A)/VEGR-2 (VEGF receptor 2) signalling-mediated angiogenesis in fibrotic liver tissues. Furthermore, we observed that the CD147-induced activation of the PI3K (phosphoinositide 3-kinase)/Akt signalling pathway promotes the production of VEGF-A in hepatocytes and expression of VEGFR-2 in SECs, which was found to enhance the angiogenic capability of SECs. Finally, our data indicate that blocking of CD147 using an mAb (monoclonal antibody) attenuated liver fibrosis progression via inhibition of VEGF-A/VEGFR-2 signalling and subsequent amelioration of microvascular abnormality in the CCl4-induced mouse model. Our findings suggest a novel functional mechanism that CD147 may promote liver fibrosis progression via inducing the VEGF-A/VEGFR-2 signalling pathway-mediated cross-talk between hepatocytes and SECs. New strategies based on the intervention of CD147 can be expected for prevention of liver fibrosis.
Collapse
|
44
|
Hahn JN, Kaushik DK, Yong VW. The role of EMMPRIN in T cell biology and immunological diseases. J Leukoc Biol 2015; 98:33-48. [PMID: 25977287 PMCID: PMC7166407 DOI: 10.1189/jlb.3ru0215-045r] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 04/03/2015] [Indexed: 12/30/2022] Open
Abstract
Review on EMMPRIN in numerous immunological/inflammatory disease conditions and its complex roles in T cell biology. EMMPRIN (CD147), originally described as an inducer of the expression of MMPs, has gained attention in its involvement in various immunologic diseases, such that anti‐EMMPRIN antibodies are considered as potential therapeutic medications. Given that MMPs are involved in the pathogenesis of various disease states, it is relevant that targeting an upstream inducer would make for an effective therapeutic strategy. Additionally, EMMPRIN is now appreciated to have multiple roles apart from MMP induction, including in cellular functions, such as migration, adhesion, invasion, energy metabolism, as well as T cell activation and proliferation. Here, we review what is known about EMMPRIN in numerous immunologic/inflammatory disease conditions with a particular focus on its complex roles in T cell biology.
Collapse
Affiliation(s)
| | | | - V Wee Yong
- Hotchkiss Brain Institute, University of Calgary, Alberta, Canada
| |
Collapse
|
45
|
Yuan Y, Shen N, Yang SY, Zhao L, Guan YM. Extracellular matrix metalloproteinase inducer and matrix metalloproteinase-2 overexpression is associated with loss of hormone receptor expression and poor prognosis in endometrial cancer. Oncol Lett 2015; 10:342-348. [PMID: 26171027 DOI: 10.3892/ol.2015.3177] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Accepted: 02/02/2015] [Indexed: 11/06/2022] Open
Abstract
Extracellular matrix metalloproteinase inducer (CD147) and matrix metalloproteinase-2 (MMP-2) have been documented in various malignancies. CD147 is a member of the immunoglobulin superfamily, which promotes the production and release of MMPs in mesenchymal cells and tumor cells. MMP-2 has been extensively studied and is considered to be particularly important in cancer invasion and metastasis. However, studies investigating the expression and prognostic value of CD147 in endometrioid endometrial carcinoma (EEC) are limited. The present study analyzed the expression of CD147 and MMP-2 by immunohistochemistry in endometrial tissue samples from 107 patients with EEC and 30 patients with benign uterus myoma. The association between CD147 and MMP-2 expression and clinicopathological characteristics was evaluated. The results showed that the overexpression of MMP-2 was significantly associated with International Federation of Gynecology and Obstetrics stage (P=0.007), depth of invasion (P=0.037) and reduced expression of progesterone receptor (P=0.005). Kaplan-Meier analyses indicated that CD147 overexpression alone (P<0.05 for disease-specific survival) or in combination with MMP-2 (P<0.001 for disease-specific survival) was correlated with adverse prognosis in EEC patients. Multivariate analysis revealed that the combined overexpression of CD147 and MMP-2 was an independent prognostic factor for disease-specific survival (hazard ratio=5.141, P=0.001) in EEC patients. CD147 and MMP-2 overexpression was positively correlated with aggressive phenotypic features in EEC, however it was negatively correlated with hormone receptor expression. The combination of CD147 and MMP-2 overexpression in EEC further distinguished a subgroup of patients with poor prognosis. Thus, the results of present study indicate that the co-expression of CD147 and MMP-2 may be an independent prognostic factor in EEC patients.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Gynecology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Ning Shen
- Department of Pathophysiology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Shu-Yan Yang
- Department of Gynecology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Ling Zhao
- Department of Gynecology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yong-Mei Guan
- Department of Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
46
|
Abstract
Cancer metastasis is highly inefficient and complex. Common features of metastatic cancer cells have been observed using cancer cell lines and genetically reconstituted mouse and human tumor xenograft models. These include cancer cell interaction with the tumor microenvironment and the ability of cancer cells to sense extracellular stimuli and adapt to adverse growth conditions. This review summarizes the coordinated response of cancer cells to soluble growth factors, such as RANKL, by a unique feed forward mechanism employing coordinated upregulation of RANKL and c-Met with downregulation of androgen receptor. The RANK-mediated signal network was found to drive epithelial to mesenchymal transition in prostate cancer cells, promote osteomimicry and the ability of prostate cancer cells to assume stem cell and neuroendocrine phenotypes, and confer the ability of prostate cancer cells to home to bone. Prostate cancer cells with activated RANK-mediated signal network were observed to recruit and even transform the non-tumorigenic prostate cancer cells to participate in bone and soft tissue colonization. The coordinated regulation of cancer cell invasion and metastasis by the feed forward mechanism involving RANKL, c-Met, transcription factors, and VEGF-neuropilin could offer new therapeutic opportunities to target prostate cancer bone and soft tissue metastases.
Collapse
Affiliation(s)
- Gina Chia-Yi Chu
- Departments of Medicine and Surgery, Samuel Orchin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA,
| | | |
Collapse
|
47
|
Seizer P, Ungern-Sternberg SNIV, Schönberger T, Borst O, Münzer P, Schmidt EM, Mack AF, Heinzmann D, Chatterjee M, Langer H, Malešević M, Lang F, Gawaz M, Fischer G, May AE. Extracellular cyclophilin A activates platelets via EMMPRIN (CD147) and PI3K/Akt signaling, which promotes platelet adhesion and thrombus formation in vitro and in vivo. Arterioscler Thromb Vasc Biol 2014; 35:655-63. [PMID: 25550208 DOI: 10.1161/atvbaha.114.305112] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Cyclophilin A (CyPA) is secreted under inflammatory conditions by various cell types. Whereas the important role of intracellular CyPA for platelet function has been reported, the effect of extracellular CyPA on platelet function has not been investigated yet. APPROACH AND RESULTS Inhibition of extracellular CyPA through a novel specific inhibitor MM284 reduced thrombus after ferric chloride-induced injury in vivo. In vitro extracellular CyPA enhanced thrombus formation even in CyPA(-/-) platelets. Treatment of isolated platelets with recombinant CyPA resulted in platelet degranulation in a time- and dose-dependent manner. Inhibition of the platelet surface receptor extracellular matrix metalloproteinase inducer (cluster of differentiation 147) by an anticluster of differentiation 147 monoclonal antibody significantly reduced CyPA-dependent platelet degranulation. Pretreatment of platelets with CyPA enhanced their recruitment to mouse carotid arteries after arterial injury, which could be inhibited by an anticluster of differentiation 147 monoclonal antibody (intravital microscopy). The role of extracellular CyPA in adhesion could be confirmed by infusing CyPA(-/-) platelets in CyPA(+/+) mice and by infusing CyPA(+/+) platelets in CyPA(-/-) mice. Stimulation of platelets with CyPA induced phosphorylation of Akt, which could in turn be inhibited in the presence of phosphoinositid-3-kinase inhibitors. Akt-1(-/-) platelets revealed a markedly decreased degranulation on CyPA stimulation. Finally, ADP-induced platelet aggregation was attenuated by MM284, as well as by inhibiting paracrine-secreted CyPA without directly affecting Ca(2+)-signaling. CONCLUSIONS Extracellular CyPA activates platelets via cluster of differentiation 147-mediated phosphoinositid-3-kinase/Akt-signaling, leading to enhanced adhesion and thrombus formation independently of intracellular CyPA. Targeting extracellular CyPA via a specific inhibitor may be a promising strategy for platelet inhibition without affecting critical functions of intracellular CyPA.
Collapse
Affiliation(s)
- Peter Seizer
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., S.N.I.v.U.-S., T.S., O.B., D.H., M.C., H.L., M.G., A.E.M.), Institute of Physiology (P.M., E.-M.S., F.L.), and Institute of Anatomy (A.F.M.), Eberhard Karls-University Tübingen, Tübingen, Germany; Institute of Biochemistry, Abteilung Enzymology, Martin-Luther-University Halle-Wittenberg, Halle, Germany (M.M.); and Max-Planck-Institute für Biophysikalische Chemie Göttingen, BO Halle (Saale), Germany (G.F.).
| | - Saskia N I V Ungern-Sternberg
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., S.N.I.v.U.-S., T.S., O.B., D.H., M.C., H.L., M.G., A.E.M.), Institute of Physiology (P.M., E.-M.S., F.L.), and Institute of Anatomy (A.F.M.), Eberhard Karls-University Tübingen, Tübingen, Germany; Institute of Biochemistry, Abteilung Enzymology, Martin-Luther-University Halle-Wittenberg, Halle, Germany (M.M.); and Max-Planck-Institute für Biophysikalische Chemie Göttingen, BO Halle (Saale), Germany (G.F.)
| | - Tanja Schönberger
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., S.N.I.v.U.-S., T.S., O.B., D.H., M.C., H.L., M.G., A.E.M.), Institute of Physiology (P.M., E.-M.S., F.L.), and Institute of Anatomy (A.F.M.), Eberhard Karls-University Tübingen, Tübingen, Germany; Institute of Biochemistry, Abteilung Enzymology, Martin-Luther-University Halle-Wittenberg, Halle, Germany (M.M.); and Max-Planck-Institute für Biophysikalische Chemie Göttingen, BO Halle (Saale), Germany (G.F.)
| | - Oliver Borst
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., S.N.I.v.U.-S., T.S., O.B., D.H., M.C., H.L., M.G., A.E.M.), Institute of Physiology (P.M., E.-M.S., F.L.), and Institute of Anatomy (A.F.M.), Eberhard Karls-University Tübingen, Tübingen, Germany; Institute of Biochemistry, Abteilung Enzymology, Martin-Luther-University Halle-Wittenberg, Halle, Germany (M.M.); and Max-Planck-Institute für Biophysikalische Chemie Göttingen, BO Halle (Saale), Germany (G.F.)
| | - Patrick Münzer
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., S.N.I.v.U.-S., T.S., O.B., D.H., M.C., H.L., M.G., A.E.M.), Institute of Physiology (P.M., E.-M.S., F.L.), and Institute of Anatomy (A.F.M.), Eberhard Karls-University Tübingen, Tübingen, Germany; Institute of Biochemistry, Abteilung Enzymology, Martin-Luther-University Halle-Wittenberg, Halle, Germany (M.M.); and Max-Planck-Institute für Biophysikalische Chemie Göttingen, BO Halle (Saale), Germany (G.F.)
| | - Eva-Maria Schmidt
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., S.N.I.v.U.-S., T.S., O.B., D.H., M.C., H.L., M.G., A.E.M.), Institute of Physiology (P.M., E.-M.S., F.L.), and Institute of Anatomy (A.F.M.), Eberhard Karls-University Tübingen, Tübingen, Germany; Institute of Biochemistry, Abteilung Enzymology, Martin-Luther-University Halle-Wittenberg, Halle, Germany (M.M.); and Max-Planck-Institute für Biophysikalische Chemie Göttingen, BO Halle (Saale), Germany (G.F.)
| | - Andreas F Mack
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., S.N.I.v.U.-S., T.S., O.B., D.H., M.C., H.L., M.G., A.E.M.), Institute of Physiology (P.M., E.-M.S., F.L.), and Institute of Anatomy (A.F.M.), Eberhard Karls-University Tübingen, Tübingen, Germany; Institute of Biochemistry, Abteilung Enzymology, Martin-Luther-University Halle-Wittenberg, Halle, Germany (M.M.); and Max-Planck-Institute für Biophysikalische Chemie Göttingen, BO Halle (Saale), Germany (G.F.)
| | - David Heinzmann
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., S.N.I.v.U.-S., T.S., O.B., D.H., M.C., H.L., M.G., A.E.M.), Institute of Physiology (P.M., E.-M.S., F.L.), and Institute of Anatomy (A.F.M.), Eberhard Karls-University Tübingen, Tübingen, Germany; Institute of Biochemistry, Abteilung Enzymology, Martin-Luther-University Halle-Wittenberg, Halle, Germany (M.M.); and Max-Planck-Institute für Biophysikalische Chemie Göttingen, BO Halle (Saale), Germany (G.F.)
| | - Madhumita Chatterjee
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., S.N.I.v.U.-S., T.S., O.B., D.H., M.C., H.L., M.G., A.E.M.), Institute of Physiology (P.M., E.-M.S., F.L.), and Institute of Anatomy (A.F.M.), Eberhard Karls-University Tübingen, Tübingen, Germany; Institute of Biochemistry, Abteilung Enzymology, Martin-Luther-University Halle-Wittenberg, Halle, Germany (M.M.); and Max-Planck-Institute für Biophysikalische Chemie Göttingen, BO Halle (Saale), Germany (G.F.)
| | - Harald Langer
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., S.N.I.v.U.-S., T.S., O.B., D.H., M.C., H.L., M.G., A.E.M.), Institute of Physiology (P.M., E.-M.S., F.L.), and Institute of Anatomy (A.F.M.), Eberhard Karls-University Tübingen, Tübingen, Germany; Institute of Biochemistry, Abteilung Enzymology, Martin-Luther-University Halle-Wittenberg, Halle, Germany (M.M.); and Max-Planck-Institute für Biophysikalische Chemie Göttingen, BO Halle (Saale), Germany (G.F.)
| | - Miroslav Malešević
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., S.N.I.v.U.-S., T.S., O.B., D.H., M.C., H.L., M.G., A.E.M.), Institute of Physiology (P.M., E.-M.S., F.L.), and Institute of Anatomy (A.F.M.), Eberhard Karls-University Tübingen, Tübingen, Germany; Institute of Biochemistry, Abteilung Enzymology, Martin-Luther-University Halle-Wittenberg, Halle, Germany (M.M.); and Max-Planck-Institute für Biophysikalische Chemie Göttingen, BO Halle (Saale), Germany (G.F.)
| | - Florian Lang
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., S.N.I.v.U.-S., T.S., O.B., D.H., M.C., H.L., M.G., A.E.M.), Institute of Physiology (P.M., E.-M.S., F.L.), and Institute of Anatomy (A.F.M.), Eberhard Karls-University Tübingen, Tübingen, Germany; Institute of Biochemistry, Abteilung Enzymology, Martin-Luther-University Halle-Wittenberg, Halle, Germany (M.M.); and Max-Planck-Institute für Biophysikalische Chemie Göttingen, BO Halle (Saale), Germany (G.F.)
| | - Meinrad Gawaz
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., S.N.I.v.U.-S., T.S., O.B., D.H., M.C., H.L., M.G., A.E.M.), Institute of Physiology (P.M., E.-M.S., F.L.), and Institute of Anatomy (A.F.M.), Eberhard Karls-University Tübingen, Tübingen, Germany; Institute of Biochemistry, Abteilung Enzymology, Martin-Luther-University Halle-Wittenberg, Halle, Germany (M.M.); and Max-Planck-Institute für Biophysikalische Chemie Göttingen, BO Halle (Saale), Germany (G.F.)
| | - Gunter Fischer
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., S.N.I.v.U.-S., T.S., O.B., D.H., M.C., H.L., M.G., A.E.M.), Institute of Physiology (P.M., E.-M.S., F.L.), and Institute of Anatomy (A.F.M.), Eberhard Karls-University Tübingen, Tübingen, Germany; Institute of Biochemistry, Abteilung Enzymology, Martin-Luther-University Halle-Wittenberg, Halle, Germany (M.M.); and Max-Planck-Institute für Biophysikalische Chemie Göttingen, BO Halle (Saale), Germany (G.F.)
| | - Andreas E May
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., S.N.I.v.U.-S., T.S., O.B., D.H., M.C., H.L., M.G., A.E.M.), Institute of Physiology (P.M., E.-M.S., F.L.), and Institute of Anatomy (A.F.M.), Eberhard Karls-University Tübingen, Tübingen, Germany; Institute of Biochemistry, Abteilung Enzymology, Martin-Luther-University Halle-Wittenberg, Halle, Germany (M.M.); and Max-Planck-Institute für Biophysikalische Chemie Göttingen, BO Halle (Saale), Germany (G.F.)
| |
Collapse
|
48
|
Functional role of EMMPRIN in the formation and mineralisation of dental matrix in mouse molars. J Mol Histol 2014; 46:21-32. [PMID: 25501684 DOI: 10.1007/s10735-014-9603-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 12/08/2014] [Indexed: 12/11/2022]
Abstract
Our previous research has shown that the extracellular matrix metalloproteinase inducer (EMMPRIN) is expressed during and may function in the early development of tooth germs. In the present study, we observed the specific expression of EMMPRIN in ameloblasts and odontoblasts during the middle and late stages of tooth germ development using immunohistochemistry. Furthermore, to extend our understanding of the function of EMMPRIN in odontogenesis, we used an anti-EMMPRIN function-blocking antibody to remove EMMPRIN activity in tooth germ culture in vitro. Both the formation and mineralisation of dental hard tissues were suppressed in the tooth germ culture after the abrogation of EMMPRIN. Meanwhile, significant reductions in VEGF, MMP-9, ALPL, ameloblastin, amelogenin and enamelin expression were observed in antibody-treated tooth germ explants compared to control and normal serum-treated explants. The current results illustrate that EMMPRIN may play a critical role in the processing and maturation of the dental matrix.
Collapse
|
49
|
Huang Q, Li J, Xing J, Li W, Li H, Ke X, Zhang J, Ren T, Shang Y, Yang H, Jiang J, Chen Z. CD147 promotes reprogramming of glucose metabolism and cell proliferation in HCC cells by inhibiting the p53-dependent signaling pathway. J Hepatol 2014; 61:859-66. [PMID: 24801417 DOI: 10.1016/j.jhep.2014.04.035] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 03/18/2014] [Accepted: 04/17/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Cancer cells exhibit the reprogrammed metabolism characterized by high level of glycolysis even in the presence of oxygen. Aerobic glycolysis, known as the Warburg effect, supplies cancer cells with the substrates required for biomass generation. To date, several intracellular signaling mediators have been identified in metabolic regulation of cancer cells. However, it remains largely ambiguous how molecules on the cell surface are involved in regulation of cancer metabolism. METHODS In the current study, we established several HCC cell lines differing in their CD147 (a typical transmembrane glycoprotein) expression status by zinc-finger nuclease and RNAi techniques. Then, we systematically investigated the role of CD147 in the regulation of the Warburg effect in HCC cells and explored the underlying mechanism. RESULTS We found that CD147 significantly contributed to the reprogramming of glucose metabolism in HCC cells through a p53-dependent way. CD147 facilitated the cell surface expression of MCT1 and lactate export, which led to activation of the PI3K/Akt/MDM2 pathway and thus increased p53 degradation. The gain/loss-of-function studies demonstrated that while CD147 promoted glycolysis, mediated by p53-dependent upregulation of GLUT1 and activation of PFKL, it inhibited mitochondrial biogenesis and functions, mediated by p53-dependent downregulation of PGC1α, TFAM, and p53R2. Additionally, proliferation of HCC cells was suppressed by blocking CD147 and/or MCT1, which resulted in down-regulation of glucose metabolism. CONCLUSIONS We demonstrate that CD147 is a crucial regulator of glucose metabolism.
Collapse
Affiliation(s)
- Qichao Huang
- State Key Laboratory of Cancer Biology, Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University, Xi'an 710032, China; Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Jibin Li
- State Key Laboratory of Cancer Biology, Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University, Xi'an 710032, China; Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Jinliang Xing
- State Key Laboratory of Cancer Biology, Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University, Xi'an 710032, China; Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Weiwei Li
- State Key Laboratory of Cancer Biology, Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University, Xi'an 710032, China; Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Hongwei Li
- State Key Laboratory of Cancer Biology, Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Xia Ke
- State Key Laboratory of Cancer Biology, Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Jing Zhang
- Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Tingting Ren
- Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yukui Shang
- State Key Laboratory of Cancer Biology, Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Hushan Yang
- Division of Population Science, Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jianli Jiang
- State Key Laboratory of Cancer Biology, Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University, Xi'an 710032, China.
| | - Zhinan Chen
- State Key Laboratory of Cancer Biology, Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
50
|
CD147 (EMMPRIN/Basigin) in kidney diseases: from an inflammation and immune system viewpoint. Nephrol Dial Transplant 2014; 30:1097-103. [DOI: 10.1093/ndt/gfu302] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 08/18/2014] [Indexed: 11/15/2022] Open
|