1
|
Loggenberg S, Twilley D, Lall N. Evaluating the effects of various ethanolic medicinal plant extracts on metastatic breast cancer proliferation, invasion, and expression of a novel potential drug target; CD82 metastatic suppressor protein, and on in vivo angiogenesis using the ex ovo yolk sac membrane (YSM) assay. J Cancer Res Clin Oncol 2024; 150:257. [PMID: 38753184 PMCID: PMC11098903 DOI: 10.1007/s00432-024-05751-0] [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: 01/18/2024] [Accepted: 04/15/2024] [Indexed: 05/19/2024]
Abstract
PURPOSE Breast cancer metastasis relies on cellular invasion and angiogenesis facilitated by the downregulation of metastatic suppressor proteins like Cluster of Differentiation 82 (CD82). Currently, no medicines target multiple systems to prevent metastatic progression through CD82 upregulation. This study screened for plant extracts displaying effects on cell proliferation, invasion, and CD82 expression in breast cancer cells, and in vivo angiogenesis, and further correlated between the biological activities and effect on CD82 expression. METHODS Seventeen ethanolic plant extracts were screened for their effect on cell proliferation (against MDA-MB-231 and MCF-7 breast cancer and Hek293 kidney cells), cell invasion and effect on CD82 expression in metastatic MDA-MB-231 cells. Selected extracts were further evaluated for in vivo anti-angiogenesis. RESULTS Extracts displayed varying antiproliferative activity against the different cell lines, and those that showed selectivity indexes (SI) > 0.5 against MDA-MB-231 were selected for anti-invasion evaluation. Buddleja saligna Willd. (BS), Combretum apiculatum Sond. (CA), Foeniculum vulgare, Greyia radlkoferi, Gunnera perpensa and Persicaria senegalensis (Meisn.) Soják (PS) displayed 50% inhibitory concentration (IC50) values of 44.46 ± 3.46, 74.00 ± 4.48, 180.43 ± 4.51, 96.97 ± 2.29, 55.29 ± 9.88 and 243.60 ± 2.69 µg/mL, respectively against MDA-MB-231, and compared to Hek293 showed SI of 0.9, 0.7, 1.4, 1.1, 2.2 and 0.5. Significant invasion inhibition was observed at both 20 and 40 µg/mL for BS (94.10 ± 0.74 and 96.73 ± 0.95%) and CA (87.42 ± 6.54 and 98.24 ± 0.63%), whereas GR (14.91 ± 1.62 and 41 ± 1.78%) and PS (36.58 ± 0.54 and 51.51 ± 0.83%), only showed significant inhibition at 40 µg/mL, and FV (< 5% inhibition) and GP (10 ± 1.03 and 22 ± 1.31%) did not show significant inhibition at both concentrations. Due to the significant anti-invasive activity of BS, CA and PS at 40 µg/mL, these extracts were further evaluated for their potential to stimulate CD82. BS showed significant (p < 0.05) reduction in CD82 at 20 and 40 µg/mL (13.2 ± 2.2% and 20.3 ± 1.5% decrease, respectively), whereas both CA and PS at 20 µg/mL increased (p < 0.05) CD82 expression (16.4 ± 0.8% and 5.4 ± 0.6% increase, respectively), and at 40 µg/mL significantly reduced CD82 expression (23.4 ± 3.1% and 11.2 ± 2.9% decrease, respectively). Using the yolk sac membrane assay, BS (59.52 ± 4.12 and 56.72 ± 3.13% newly formed vessels) and CA (83.33 ± 3.17 and 74.00 ± 2.12%) at both 20 and 40 µg/egg showed significant (p < 0.001) angiogenesis inhibition, with BS showing statistical similar activity to the positive control, combretastatin A4 (10 nmol/egg), whereas PS only displayed significant (p < 0.001) angiogenesis stimulation at 40 µg/egg (120.81 ± 3.34% newly formed vessels). CONCLUSION BS exhibits antiproliferative, anti-invasive, and anti-angiogenic activity despite inhibiting CD82, suggesting an alternative mode of action. CA at 20 µg/mL shows moderate anti-invasive and anti-angiogenic potential by stimulating CD82, while at 40 µg/mL it still displays these properties but inhibits CD82, suggesting an additional mode of action. PS, with the least antiproliferative activity, stimulates CD82 and inhibits angiogenesis at 20 µg/mL but inhibits CD82 and increases angiogenesis at 40 µg/mL, indicating CD82 targeting as a major mode of action. Future studies should explore breast cancer xenograft models to assess the extracts' impact on CD82 expression and angiogenesis in the tumor microenvironment, along with isolating bioactive compounds from the extracts.
Collapse
Affiliation(s)
- Samantha Loggenberg
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria, 0002, South Africa
| | - Danielle Twilley
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria, 0002, South Africa
| | - Namrita Lall
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria, 0002, South Africa.
- School of Natural Resources, University of Missouri, Columbia, MO, 65211, USA.
- College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka, 570015, India.
- Bio-Tech Research and Development Institute, University of the West Indies, Kingston, Jamaica.
| |
Collapse
|
2
|
Lee JW, Hur J, Kwon YW, Chae CW, Choi JI, Hwang I, Yun JY, Kang JA, Choi YE, Kim YH, Lee SE, Lee C, Jo DH, Seok H, Cho BS, Baek SH, Kim HS. KAI1(CD82) is a key molecule to control angiogenesis and switch angiogenic milieu to quiescent state. J Hematol Oncol 2021; 14:148. [PMID: 34530889 PMCID: PMC8444549 DOI: 10.1186/s13045-021-01147-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Little is known about endogenous inhibitors of angiogenic growth factors. In this study, we identified a novel endogenous anti-angiogenic factor expressed in pericytes and clarified its underlying mechanism and clinical significance. METHODS Herein, we found Kai1 knockout mice showed significantly enhanced angiogenesis. Then, we investigated the anti-angiogenic roll of Kai1 in vitro and in vivo. RESULTS KAI1 was mainly expressed in pericytes rather than in endothelial cells. It localized at the membrane surface after palmitoylation by zDHHC4 enzyme and induced LIF through the Src/p53 pathway. LIF released from pericytes in turn suppressed angiogenic factors in endothelial cells as well as in pericytes themselves, leading to inhibition of angiogenesis. Interestingly, KAI1 had another mechanism to inhibit angiogenesis: It directly bound to VEGF and PDGF and inhibited activation of their receptors. In the two different in vivo cancer models, KAI1 supplementation significantly inhibited tumor angiogenesis and growth. A peptide derived from the large extracellular loop of KAI1 has been shown to have anti-angiogenic effects to block the progression of breast cancer and retinal neovascularization in vivo. CONCLUSIONS KAI1 from PC is a novel molecular regulator that counterbalances the effect of angiogenic factors.
Collapse
Affiliation(s)
- Jin-Woo Lee
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Center of Cell- and Bio-Therapy (CBT), Seoul National University Hospital, Seoul, Republic of Korea
| | - Jin Hur
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Yoo-Wook Kwon
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Center of Cell- and Bio-Therapy (CBT), Seoul National University Hospital, Seoul, Republic of Korea
| | - Cheong-Whan Chae
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Center of Cell- and Bio-Therapy (CBT), Seoul National University Hospital, Seoul, Republic of Korea
| | - Jae-Il Choi
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Center of Cell- and Bio-Therapy (CBT), Seoul National University Hospital, Seoul, Republic of Korea
| | - Injoo Hwang
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Center of Cell- and Bio-Therapy (CBT), Seoul National University Hospital, Seoul, Republic of Korea
| | - Ji-Yeon Yun
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Center of Cell- and Bio-Therapy (CBT), Seoul National University Hospital, Seoul, Republic of Korea
| | - Jin-A Kang
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Center of Cell- and Bio-Therapy (CBT), Seoul National University Hospital, Seoul, Republic of Korea
| | - Young-Eun Choi
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Center of Cell- and Bio-Therapy (CBT), Seoul National University Hospital, Seoul, Republic of Korea
| | - Young Hyun Kim
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Center of Cell- and Bio-Therapy (CBT), Seoul National University Hospital, Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Sang Eun Lee
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Center of Cell- and Bio-Therapy (CBT), Seoul National University Hospital, Seoul, Republic of Korea
| | - Cheol Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dong Hyun Jo
- Department of Anatomy, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Heeyoung Seok
- Genomics Core Facility, Department of Transdisciplinary Research and Collaboration, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Byong Seung Cho
- ExoCoBio Inc, Gasan digital 1-ro, Geumcheon-gu, Seoul, 08594, Republic of Korea
| | - Sung Hee Baek
- Creative Research Initiative Center for Chromatin Dynamics, School of Biological Sciences, Seoul National University, Seoul, 151-742, Republic of Korea
| | - Hyo-Soo Kim
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea.
- Center of Cell- and Bio-Therapy (CBT), Seoul National University Hospital, Seoul, Republic of Korea.
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea.
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 110-744, Korea.
| |
Collapse
|
3
|
Viera M, Yip GWC, Shen HM, Baeg GH, Bay BH. Targeting CD82/KAI1 for Precision Therapeutics in Surmounting Metastatic Potential in Breast Cancer. Cancers (Basel) 2021; 13:4486. [PMID: 34503296 PMCID: PMC8431267 DOI: 10.3390/cancers13174486] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/27/2021] [Accepted: 09/03/2021] [Indexed: 11/16/2022] Open
Abstract
Metastasis is the main cause of mortality in breast cancer patients. There is an unmet need to develop therapies that can impede metastatic spread. Precision oncology has shown great promise for the treatment of cancers, as the therapeutic approach is tailored to a specific group of patients who are likely to benefit from the treatment, rather than the traditional approach of "one size fits all". CD82, also known as KAI1, a glycoprotein belonging to the tetraspanin family and an established metastasis suppressor, could potentially be exploited to hinder metastases in breast cancer. This review explores the prospect of targeting CD82 as an innovative therapeutic approach in precision medicine for breast cancer patients, with the goal of preventing cancer progression and metastasis. Such an approach would entail the selection of a subset of breast cancer patients with low levels of CD82, and instituting an appropriate treatment scheme tailored towards restoring the levels of CD82 in this group of patients. Proposed precision treatment regimens include current modalities of treating breast cancer, in combination with either clinically approved drugs that could restore the levels of CD82, CD82 peptide mimics or non-coding RNA-based therapeutics.
Collapse
Affiliation(s)
- Maximillian Viera
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (M.V.); (G.W.C.Y.)
| | - George Wai Cheong Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (M.V.); (G.W.C.Y.)
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore;
- Faculty of Health Sciences, University of Macau, Taipa, China
| | - Gyeong Hun Baeg
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (M.V.); (G.W.C.Y.)
- Ciechanover Institute of Precision and Regenerative Medicine, School of Life and Health Sciences, Chinese University of Hong Kong, Shenzhen 518172, China
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (M.V.); (G.W.C.Y.)
| |
Collapse
|
4
|
Ordas L, Costa L, Lozano A, Chevillard C, Calovoulos A, Kantar D, Fernandez L, Chauvin L, Dosset P, Doucet C, Heron-Milhavet L, Odintsova E, Berditchevski F, Milhiet PE, Bénistant C. Mechanical Control of Cell Migration by the Metastasis Suppressor Tetraspanin CD82/KAI1. Cells 2021; 10:cells10061545. [PMID: 34207462 PMCID: PMC8234748 DOI: 10.3390/cells10061545] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 01/16/2023] Open
Abstract
The plasma membrane is a key actor of cell migration. For instance, its tension controls persistent cell migration and cell surface caveolae integrity. Then, caveolae constituents such as caveolin-1 can initiate a mechanotransduction loop that involves actin- and focal adhesion-dependent control of the mechanosensor YAP to finely tune cell migration. Tetraspanin CD82 (also named KAI-1) is an integral membrane protein and a metastasis suppressor. Its expression is lost in many cancers including breast cancer. It is a strong inhibitor of cell migration by a little-known mechanism. We demonstrated here that CD82 controls persistent 2D migration of EGF-induced single cells, stress fibers and focal adhesion sizes and dynamics. Mechanistically, we found that CD82 regulates membrane tension, cell surface caveolae abundance and YAP nuclear translocation in a caveolin-1-dependent manner. Altogether, our data show that CD82 controls 2D cell migration using membrane-driven mechanics involving caveolin and the YAP pathway.
Collapse
Affiliation(s)
- Laura Ordas
- Centre de Biologie Structurale (CBS), CNRS, INSERM, University Montpellier, 34090 Montpellier, France; (L.O.); (L.C.); (A.L.); (C.C.); (A.C.); (L.F.); (P.D.); (C.D.)
| | - Luca Costa
- Centre de Biologie Structurale (CBS), CNRS, INSERM, University Montpellier, 34090 Montpellier, France; (L.O.); (L.C.); (A.L.); (C.C.); (A.C.); (L.F.); (P.D.); (C.D.)
| | - Anthony Lozano
- Centre de Biologie Structurale (CBS), CNRS, INSERM, University Montpellier, 34090 Montpellier, France; (L.O.); (L.C.); (A.L.); (C.C.); (A.C.); (L.F.); (P.D.); (C.D.)
- Institut de Génétique Moléculaire de Montpellier, University Montpellier, CNRS, 34293 Montpellier, France
| | - Christopher Chevillard
- Centre de Biologie Structurale (CBS), CNRS, INSERM, University Montpellier, 34090 Montpellier, France; (L.O.); (L.C.); (A.L.); (C.C.); (A.C.); (L.F.); (P.D.); (C.D.)
| | - Alexia Calovoulos
- Centre de Biologie Structurale (CBS), CNRS, INSERM, University Montpellier, 34090 Montpellier, France; (L.O.); (L.C.); (A.L.); (C.C.); (A.C.); (L.F.); (P.D.); (C.D.)
| | - Diala Kantar
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194—University Montpellier—Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France; (D.K.); (L.H.-M.)
| | - Laurent Fernandez
- Centre de Biologie Structurale (CBS), CNRS, INSERM, University Montpellier, 34090 Montpellier, France; (L.O.); (L.C.); (A.L.); (C.C.); (A.C.); (L.F.); (P.D.); (C.D.)
- European Institute of Chemistry and Biology (IECB), University of Bordeaux, 33607 Pessac, France
| | - Lucie Chauvin
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), CNRS UMR 5237, University Montpellier, 34293 Montpellier, France;
| | - Patrice Dosset
- Centre de Biologie Structurale (CBS), CNRS, INSERM, University Montpellier, 34090 Montpellier, France; (L.O.); (L.C.); (A.L.); (C.C.); (A.C.); (L.F.); (P.D.); (C.D.)
| | - Christine Doucet
- Centre de Biologie Structurale (CBS), CNRS, INSERM, University Montpellier, 34090 Montpellier, France; (L.O.); (L.C.); (A.L.); (C.C.); (A.C.); (L.F.); (P.D.); (C.D.)
| | - Lisa Heron-Milhavet
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194—University Montpellier—Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France; (D.K.); (L.H.-M.)
| | - Elena Odintsova
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (E.O.); (F.B.)
| | - Fedor Berditchevski
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (E.O.); (F.B.)
| | - Pierre-Emmanuel Milhiet
- Centre de Biologie Structurale (CBS), CNRS, INSERM, University Montpellier, 34090 Montpellier, France; (L.O.); (L.C.); (A.L.); (C.C.); (A.C.); (L.F.); (P.D.); (C.D.)
- Correspondence: (P.-E.M.); (C.B.)
| | - Christine Bénistant
- Centre de Biologie Structurale (CBS), CNRS, INSERM, University Montpellier, 34090 Montpellier, France; (L.O.); (L.C.); (A.L.); (C.C.); (A.C.); (L.F.); (P.D.); (C.D.)
- Correspondence: (P.-E.M.); (C.B.)
| |
Collapse
|
5
|
Yuan P, Zhou Q, Hu X. WS 2 Nanosheets at Noncytotoxic Concentrations Enhance the Cytotoxicity of Organic Pollutants by Disturbing the Plasma Membrane and Efflux Pumps. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:1698-1709. [PMID: 31916439 DOI: 10.1021/acs.est.9b05537] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Emerging transition-metal dichalcogenide (TMDC) nanosheets, such as WS2 nanosheets, have shown tremendous potential for use in many fields such as intelligent manufacturing and environmental protection. However, considerable knowledge gaps still exist regarding the impact of TMDCs on environmental risks, especially risks involving organic pollutants. Here, a synergistic toxicity between WS2 nanosheets and organic pollutants (triclosan or tris(1,3-dichloro-2-propyl) phosphate) was found using the median-effect and combination index equations. In particular, the effect of synergy had a higher magnitude at low cytotoxicity levels and a noncytotoxic concentration of WS2 nanosheets clearly enhanced the cytotoxicity and intracellular accumulation of organic pollutants. On the one hand, WS2 nanosheets damaged the plasma membrane and cytoskeleton, resulting in increased membrane permeability and organic pollutant uptake. On the other hand, as shown by fluorescence substrate accumulation experiments and molecular dynamics simulations, WS2 nanosheets affected the secondary structure of the efflux pumps and competitively bound with efflux pumps, blocking xenobiotic removal. This work emphasized that TMDCs, especially at the noncytotoxic level, in combination with organic pollutants caused damage that cannot be ignored, providing insight into comprehensive safety assessment and the specific toxicological mechanisms of TMDCs that accompany organic pollutant exposure.
Collapse
Affiliation(s)
- Peng Yuan
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education)/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering , Nankai University , Tianjin 300350 , China
| | - Qixing Zhou
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education)/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering , Nankai University , Tianjin 300350 , China
| | - Xiangang Hu
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education)/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering , Nankai University , Tianjin 300350 , China
| |
Collapse
|
6
|
Huang C, Hays FA, Tomasek JJ, Benyajati S, Zhang XA. Tetraspanin CD82 interaction with cholesterol promotes extracellular vesicle-mediated release of ezrin to inhibit tumour cell movement. J Extracell Vesicles 2019; 9:1692417. [PMID: 31807237 PMCID: PMC6882436 DOI: 10.1080/20013078.2019.1692417] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/16/2019] [Accepted: 10/26/2019] [Indexed: 01/04/2023] Open
Abstract
Tumour metastasis suppressor KAI1/CD82 inhibits tumour cell movement. As a transmembrane protein, tetraspanin CD82 bridges the interactions between membrane microdomains of lipid rafts and tetraspanin-enriched microdomains (TEMs). In this study, we found that CD82 and other tetraspanins contain cholesterol recognition/interaction amino-acid consensus (CRAC) sequences in their transmembrane domains and revealed that cholesterol binding of CD82 determines its interaction with lipid rafts but not with TEMs. Functionally, CD82 needs cholesterol binding to inhibit solitary migration, collective migration, invasion and infiltrative outgrowth of tumour cells. Importantly, CD82–cholesterol/–lipid raft interaction not only promotes extracellular release of lipid raft components such as cholesterol and gangliosides but also facilitates extracellular vesicle (EV)–mediated release of ezrin–radixin–moesin (ERM) protein Ezrin. Since ERM proteins link actin cytoskeleton to the plasma membrane, we show for the first time that cell movement can be regulated by EV-mediated releases, which disengage the plasma membrane from cytoskeleton and then impair cell movement. Our findings also conceptualize that interactions between membrane domains, in this case converge of lipid rafts and TEMs by CD82, can change cell movement. Moreover, CD82 coalescences with both lipid rafts and TEMs are essential for its inhibition of tumour cell movement and for its enhancement of EV release. Finally, our study underpins that tetraspanins as a superfamily of functionally versatile molecules are cholesterol-binding proteins. Abbreviations:Ab: antibody; CBM: cholesterol-binding motif; CCM: cholesterol consensus motif; CRAC/CARC: cholesterol recognition or interaction amino-acid consensus; CTxB: cholera toxin B subunit; ECM: extracellular matrix; ERM: ezrin, radixin and moesin; EV: extracellular vesicles; FBS: foetal bovine serum; mAb: monoclonal antibody; MST: microscale thermophoresis; pAb: polyclonal antibody; and TEM: tetraspanin-enriched microdomain
Collapse
Affiliation(s)
- Chao Huang
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Franklin A Hays
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - James J Tomasek
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Siribhinya Benyajati
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xin A Zhang
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
7
|
Dai C, Liu Y, Yang R, Zhou L. Clinical significance of MACC1, Twist1, and KAI1 expressions in infiltrating urothelial carcinoma of the bladder. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:3877-3885. [PMID: 31933777 PMCID: PMC6949747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 08/28/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Metastasis-associated in colon cancer 1 (MACC1), a candidate oncogene, promotes tumor cell invasion and metastasis in various cancers. Twist1, a key transcriptional gene of the epithelial-mesenchymal transition (EMT), is involved in EMT and metastasis in many cancers. KAI1, also known as CD82, was originally considered as a suppressor gene of tumor metastasis. In this study, we investigated the expressions and significance of MACC1, Twist1, and KAI1 in infiltrating urothelial carcinoma of bladder (IUCB). METHODS The expressions of MACC1, Twist1, and KAI1 in 195 IUCB specimens and their corresponding control specimens were investigated by immunohistochemistry. The patients' clinical, demographic, and follow-up data were collected. RESULTS The rates of the positive expressions of MACC1 and Twist1 were significantly higher in IUCB tissues than in normal bladder mucosa tissues, and their expressions were positively correlated with tumor stages, grades of differentiation, lymph node metastasis (LNM), and tumor-node-metastasis (TNM) stages. The rate of positive expression of KAI1 was significantly lower in IUCB than in the control tissues, and its expression was inversely associated with tumor stages, grades of differentiation, LNM, and TNM stages. Patients who expressed MACC1 or Twist1 had an unfavorable overall survival (OS) time when compared with patients who did not express these proteins. However, patients who expressed KAI1 had a favorable OS when compared with patients who did not express this protein. A multivariate analysis demonstrated that the expressions of MACC1, Twist1, and KAI1, tumor stages, grades of differentiation, LNM, as well as TNM stages were independent prognostic indicators for IUCB patients. CONCLUSION Therefore, MACC1, Twist1, and KAI1 should be considered potentially promising biomarkers of IUCB prognosis.
Collapse
Affiliation(s)
- Changyuan Dai
- Department of Urology, The First Affiliated Hospital of Bengbu Medical UniversityBengbu, Anhui, China
| | - Yuanqun Liu
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical UniversityBengbu, Anhui, China
| | - Ruixue Yang
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical UniversityBengbu, Anhui, China
| | - Lei Zhou
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical UniversityBengbu, Anhui, China
| |
Collapse
|
8
|
Khan NS, Lukason DP, Feliu M, Ward RA, Lord AK, Reedy JL, Ramirez-Ortiz ZG, Tam JM, Kasperkovitz PV, Negoro PE, Vyas TD, Xu S, Brinkmann MM, Acharaya M, Artavanis-Tsakonas K, Frickel EM, Becker CE, Dagher Z, Kim YM, Latz E, Ploegh HL, Mansour MK, Miranti CK, Levitz SM, Vyas JM. CD82 controls CpG-dependent TLR9 signaling. FASEB J 2019; 33:12500-12514. [PMID: 31408613 DOI: 10.1096/fj.201901547r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The tetraspanin CD82 is a potent suppressor of tumor metastasis and regulates several processes including signal transduction, cell adhesion, motility, and aggregation. However, the mechanisms by which CD82 participates in innate immunity are unknown. We report that CD82 is a key regulator of TLR9 trafficking and signaling. TLR9 recognizes unmethylated cytosine-phosphate-guanine (CpG) motifs present in viral, bacterial, and fungal DNA. We demonstrate that TLR9 and CD82 associate in macrophages, which occurs in the endoplasmic reticulum (ER) and post-ER. Moreover, CD82 is essential for TLR9-dependent myddosome formation in response to CpG stimulation. Finally, CD82 modulates TLR9-dependent NF-κB nuclear translocation, which is critical for inflammatory cytokine production. To our knowledge, this is the first time a tetraspanin has been implicated as a key regulator of TLR signaling. Collectively, our study demonstrates that CD82 is a specific regulator of TLR9 signaling, which may be critical in cancer immunotherapy approaches and coordinating the innate immune response to pathogens.-Khan, N. S., Lukason, D. P., Feliu, M., Ward, R. A., Lord, A. K., Reedy, J. L., Ramirez-Ortiz, Z. G., Tam, J. M., Kasperkovitz, P. V., Negoro, P. E., Vyas, T. D., Xu, S., Brinkmann, M. M., Acharaya, M., Artavanis-Tsakonas, K., Frickel, E.-M., Becker, C. E., Dagher, Z., Kim, Y.-M., Latz, E., Ploegh, H. L., Mansour, M. K., Miranti, C. K., Levitz, S. M., Vyas, J. M. CD82 controls CpG-dependent TLR9 signaling.
Collapse
Affiliation(s)
- Nida S Khan
- Division of Infectious Disease, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Biomedical Engineering and Biotechnology, University of Massachusetts Medical School, Worcester, Massachusetts, USA.,Biomedical Engineering and Biotechnology, University of Massachusetts Lowell, Lowell, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel P Lukason
- Division of Infectious Disease, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marianela Feliu
- Division of Infectious Disease, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Rebecca A Ward
- Division of Infectious Disease, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Allison K Lord
- Division of Infectious Disease, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jennifer L Reedy
- Division of Infectious Disease, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Zaida G Ramirez-Ortiz
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.,Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jenny M Tam
- Division of Infectious Disease, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Paige E Negoro
- Division of Infectious Disease, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Tammy D Vyas
- Division of Infectious Disease, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shuying Xu
- Division of Infectious Disease, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Melanie M Brinkmann
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Mridu Acharaya
- Benaroya Research Institute, Seattle, Washington, USA.,Center for Immunity and Immunotherapy, Seattle Children's Research Institute, Seattle, Washington, USA
| | | | - Eva-Maria Frickel
- Host-Toxoplasma Interaction Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Christine E Becker
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zeina Dagher
- Division of Infectious Disease, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - You-Me Kim
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Eicke Latz
- Department of Medicine, University of Massachusetts Medical School, Boston, Massachusetts, USA.,Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Michael K Mansour
- Division of Infectious Disease, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Cindy K Miranti
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, Michigan, USA.,Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, Tucson, Arizona, USA
| | - Stuart M Levitz
- Department of Medicine, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Jatin M Vyas
- Division of Infectious Disease, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Bergsma A, Ganguly SS, Wiegand ME, Dick D, Williams BO, Miranti CK. Regulation of cytoskeleton and adhesion signaling in osteoclasts by tetraspanin CD82. Bone Rep 2019; 10:100196. [PMID: 30788390 PMCID: PMC6369370 DOI: 10.1016/j.bonr.2019.100196] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 01/18/2019] [Accepted: 01/28/2019] [Indexed: 12/11/2022] Open
Abstract
We used a myeloid-specific Cre to conditionally delete CD82 in mouse osteoclasts and their precursors. In contrast to global loss of CD82 (gKO), conditional loss of CD82 (cKO) in osteoclasts does not affect cortical bone, osteoblasts, or adipocytes. CD82 loss results in greater trabecular volume and trabecular number but reduced trabecular space in 6-month old male mice. Though this trend is present in females it did not reach significance; whereas there was an increase in osteoclast numbers and eroded surface area only in female cKO mice. In vitro, there is an increase in osteoclast fusion and defects in actin assembly in both gKO and cKO mice, irrespective of sex. This is accompanied by altered osteoclast morphology and decreased release of CTX in vitro. Integrin αvβ3 expression is reduced, while integrin β1 is increased. Signaling to Src, Syk, and Vav are also compromised. We further discovered that expression of Clec2 and its ligand, Podoplanin, molecules that also signal to Syk and Vav, are increased in differentiated osteoclasts. Loss of CD82 reduces their expression. Thus, CD82 is required for correct assembly of the cytoskeleton and to limit osteoclast fusion, both needed for normal osteoclast function.
Collapse
Affiliation(s)
- Alexis Bergsma
- Center for Cancer and Cell Biology, Program for Skeletal Disease and Tumor Microenvironment, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Sourik S Ganguly
- Center for Cancer and Cell Biology, Program for Skeletal Disease and Tumor Microenvironment, Van Andel Research Institute, Grand Rapids, MI, USA.,Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Mollie E Wiegand
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Daniel Dick
- Center for Cancer and Cell Biology, Program for Skeletal Disease and Tumor Microenvironment, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Bart O Williams
- Center for Cancer and Cell Biology, Program for Skeletal Disease and Tumor Microenvironment, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Cindy K Miranti
- Center for Cancer and Cell Biology, Program for Skeletal Disease and Tumor Microenvironment, Van Andel Research Institute, Grand Rapids, MI, USA.,Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
10
|
Neumann E, Schwarz MC, Hasseli R, Hülser ML, Classen S, Sauerbier M, Rehart S, Mueller-Ladner U. Tetraspanin CD82 affects migration, attachment and invasion of rheumatoid arthritis synovial fibroblasts. Ann Rheum Dis 2018; 77:1619-1626. [PMID: 29980577 DOI: 10.1136/annrheumdis-2018-212954] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 12/22/2022]
Abstract
Tetraspanins function as membrane adaptors altering cell-cell fusion, antigen presentation, receptor-mediated signal transduction and cell motility via interaction with membrane proteins including other tetraspanins and adhesion molecules such as integrins. CD82 is expressed in several malignant cells and well described as tumour metastasis suppressor. Rheumatoid arthritis (RA) is based on persistent synovial inflammation and joint destruction driven to a large extent by transformed-appearing activated synovial fibroblasts (SF) with an increased migratory potential. OBJECTIVE CD82 is upregulated in RA synovial fibroblasts (RASF) compared with osteoarthritis (OA) SF as well as within RA compared with OA synovial lining layer (LL) and the role of CD82 in RASF was evaluated. METHODS CD82 and integrin immunofluorescence was performed. Lentiviral CD82 overexpression and siRNA-mediated knockdown was confirmed (realtime-PCR, Western blot, immunocytochemistry). RASF migration (Boyden chamber, scrape assay), attachment towards plastic/Matrigel, RASF-binding to endothelial cells (EC) and CD82 expression during long-term invasion in the SCID-mouse-model were evaluated. RESULTS CD82 was induced by proinflammatory stimuli in SF. In RA-synovium, CD82 was expressed in RASF close to blood vessels, LL, sites of cartilage invasion and colocalised with distinct integrins involved in tumour metastasis suppression but also in RA-synovium by RASF. CD82 overexpression led to reduced RASF migration, cell-matrix and RASF-EC adhesion. Reduced CD82 expression (observed in the sublining) increased RASF migration and matrix adhesion whereas RASF-EC-interaction was reduced. In SCID mice, the presence of CD82 on cartilage-invading RASF was confirmed. CONCLUSION CD82 could contribute to RASF migration to sites of inflammation and tissue damage, where CD82 keeps aggressive RASF on site.
Collapse
Affiliation(s)
- Elena Neumann
- Department of Rheumatology and Clinical Immunology, Campus Kerckhoff, Justus-Liebig University Giessen, Bad Nauheim, Germany
| | - Maria C Schwarz
- Department of Rheumatology and Clinical Immunology, Campus Kerckhoff, Justus-Liebig University Giessen, Bad Nauheim, Germany
| | - Rebecca Hasseli
- Department of Rheumatology and Clinical Immunology, Campus Kerckhoff, Justus-Liebig University Giessen, Bad Nauheim, Germany
| | - Marie-Lisa Hülser
- Department of Rheumatology and Clinical Immunology, Campus Kerckhoff, Justus-Liebig University Giessen, Bad Nauheim, Germany
| | - Simon Classen
- Division of Vascular Surgery, Harvey-Vascular-Healthcare Center, Kerckhoff-Klinik GmbH, Bad Nauheim, Germany
| | - Michael Sauerbier
- Department of Plastic, Hand and reconstructive Surgery, BGU Frankfurt, Frankfurt, Germany
| | - Stefan Rehart
- Department of Orthopaedics and Trauma Surgery, Agaplesion Markus Hospital, Frankfurt, Germany
| | - Ulf Mueller-Ladner
- Department of Rheumatology and Clinical Immunology, Campus Kerckhoff, Justus-Liebig University Giessen, Bad Nauheim, Germany
| |
Collapse
|
11
|
Huang C, Fu C, Wren JD, Wang X, Zhang F, Zhang YH, Connel SA, Chen T, Zhang XA. Tetraspanin-enriched microdomains regulate digitation junctions. Cell Mol Life Sci 2018; 75:3423-3439. [PMID: 29589089 PMCID: PMC6615572 DOI: 10.1007/s00018-018-2803-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 02/18/2018] [Accepted: 03/21/2018] [Indexed: 12/22/2022]
Abstract
Tetraspanins co-emerged with multi-cellular organisms during evolution and are typically localized at the cell–cell interface, [corrected] and form tetraspanin-enriched microdomains (TEMs) by associating with each other and other membrane molecules. Tetraspanins affect various biological functions, but how tetraspanins engage in multi-faceted functions at the cellular level is largely unknown. When cells interact, the membrane microextrusions at the cell-cell interfaces form dynamic, digit-like structures between cells, which we term digitation junctions (DJs). We found that (1) tetraspanins CD9, CD81, and CD82 and (2) TEM-associated molecules integrin α3β1, CD44, EWI2/PGRL, and PI-4P are present in DJs of epithelial, endothelial, and cancer cells. Tetraspanins and their associated molecules also regulate the formation and development of DJs. Moreover, (1) actin cytoskeleton, RhoA, and actomyosin activities and (2) growth factor receptor-Src-MAP kinase signaling, but not PI-3 kinase, regulate DJs. Finally, we showed that DJs consist of various forms in different cells. Thus, DJs are common, interactive structures between cells, and likely affect cell adhesion, migration, and communication. TEMs probably modulate various cell functions through DJs. Our findings highlight that DJ morphogenesis reflects the transition between cell-matrix adhesion and cell-cell adhesion and involves both cell-cell and cell-matrix adhesion molecules.
Collapse
Affiliation(s)
- Chao Huang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC Building West Room 1474, 975 N.E. 10th Street, Oklahoma City, OK, 73104, USA
| | - Chenying Fu
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC Building West Room 1474, 975 N.E. 10th Street, Oklahoma City, OK, 73104, USA
| | - Jonathan D Wren
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Xuejun Wang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC Building West Room 1474, 975 N.E. 10th Street, Oklahoma City, OK, 73104, USA
| | - Feng Zhang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC Building West Room 1474, 975 N.E. 10th Street, Oklahoma City, OK, 73104, USA
| | - Yanhui H Zhang
- University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Taosheng Chen
- St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xin A Zhang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC Building West Room 1474, 975 N.E. 10th Street, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
12
|
Saito-Reis CA, Marjon KD, Pascetti EM, Floren M, Gillette JM. The tetraspanin CD82 regulates bone marrow homing and engraftment of hematopoietic stem and progenitor cells. Mol Biol Cell 2018; 29:2946-2958. [PMID: 30133344 PMCID: PMC6329911 DOI: 10.1091/mbc.e18-05-0305] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hematopoietic stem and progenitor cell (HSPC) transplantation represents a treatment option for patients with malignant and nonmalignant hematological diseases. Initial steps in transplantation involve the bone marrow homing and engraftment of peripheral blood–injected HSPCs. In recent work, we identified the tetraspanin CD82 as a potential regulator of HSPC homing to the bone marrow, although its mechanism remains unclear. In the present study, using a CD82 knockout (CD82KO) mouse model, we determined that CD82 modulates HSPC bone marrow maintenance, homing, and engraftment. Bone marrow characterization identified a significant decrease in the number of long-term hematopoietic stem cells in the CD82KO mice, which we linked to cell cycle activation and reduced stem cell quiescence. Additionally, we demonstrate that CD82 deficiency disrupts bone marrow homing and engraftment, with in vitro analysis identifying further defects in migration and cell spreading. Moreover, we find that the CD82KO HSPC homing defect is due at least in part to the hyperactivation of Rac1, as Rac1 inhibition rescues homing capacity. Together, these data provide evidence that CD82 is an important regulator of HSPC bone marrow maintenance, homing, and engraftment and suggest exploiting the CD82 scaffold as a therapeutic target for improved efficacy of stem cell transplants.
Collapse
Affiliation(s)
- Chelsea A Saito-Reis
- Department of Pathology, University of New Mexico Health Science Center, University of New Mexico, Albuquerque, NM 87131
| | - Kristopher D Marjon
- Department of Pathology, University of New Mexico Health Science Center, University of New Mexico, Albuquerque, NM 87131
| | - Erica M Pascetti
- Department of Pathology, University of New Mexico Health Science Center, University of New Mexico, Albuquerque, NM 87131
| | - Muskan Floren
- Department of Pathology, University of New Mexico Health Science Center, University of New Mexico, Albuquerque, NM 87131
| | - Jennifer M Gillette
- Department of Pathology, University of New Mexico Health Science Center, University of New Mexico, Albuquerque, NM 87131
| |
Collapse
|
13
|
Suárez H, Rocha-Perugini V, Álvarez S, Yáñez-Mó M. Tetraspanins, Another Piece in the HIV-1 Replication Puzzle. Front Immunol 2018; 9:1811. [PMID: 30127789 PMCID: PMC6088189 DOI: 10.3389/fimmu.2018.01811] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
Despite the great research effort placed during the last decades in HIV-1 study, still some aspects of its replication cycle remain unknown. All this powerful research has succeeded in developing different drugs for AIDS treatment, but none of them can completely remove the virus from infected patients, who require life-long medication. The classical approach was focused on the study of virus particles as the main target, but increasing evidence highlights the importance of host cell proteins in HIV-1 cycle. In this context, tetraspanins have emerged as critical players in different steps of the viral infection cycle. Through their association with other molecules, including membrane receptors, cytoskeletal proteins, and signaling molecules, tetraspanins organize specialized membrane microdomains called tetraspanin-enriched microdomains (TEMs). Within these microdomains, several tetraspanins have been described to regulate HIV-1 entry, assembly, and transfer between cells. Interestingly, the importance of tetraspanins CD81 and CD63 in the early steps of viral replication has been recently pointed out. Indeed, CD81 can control the turnover of the HIV-1 restriction factor SAMHD1. This deoxynucleoside triphosphate triphosphohydrolase counteracts HIV-1 reverse transcription (RT) in resting cells via its dual function as dNTPase, catalyzing deoxynucleotide triphosphates into deoxynucleosides and inorganic triphosphate, and as exonuclease able to degrade single-stranded RNAs. SAMHD1 has also been related with the detection of viral nucleic acids, regulating the innate immune response and would promote viral latency. New evidences demonstrating the ability of CD81 to control SAMHD1 expression, and as a consequence, HIV-1 RT activity, highlight the importance of TEMs for viral replication. Here, we will briefly review how tetraspanins modulate HIV-1 infection, focusing on the latest findings that link TEMs to viral replication.
Collapse
Affiliation(s)
- Henar Suárez
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Vera Rocha-Perugini
- Servicio de Inmunología, Hospital de la Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain.,Vascular Pathophysiology Research Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Susana Álvarez
- Servicio de Inmunobiología Molecular, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - María Yáñez-Mó
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Biología Molecular Severo Ochoa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| |
Collapse
|
14
|
Sun B, Cheng Z, Sun J. Associations of MACC1, AGR2, and KAI1 expression with the metastasis and prognosis in head and neck squamous cell carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:822-830. [PMID: 31938171 PMCID: PMC6958041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/12/2017] [Indexed: 06/10/2023]
Abstract
BACKGROUND Metastasis-associated in colon cancer-1 (MACC1, was firstly found in colon cancer and associated metastasis and prognosis in various cancers), anterior gradient 2 (AGR2, was considered as a valuable prognostic factor for some cancers), and Kangai 1 (KAI1, was a tumor metastasis suppressor gene) are all related to metastasis and prognosis of many cancers. However, the associations of MACC1, AGR2, and KAI1 in head and neck squamous cell carcinoma (HNSCC) are still unclear. In this study, we analyzed associations among MACC1, AGR2, and KAI1 in HNSCC, and their respective associations with clinicopathological parameters and overall survival (OS) in HNSCC. METHODS Positive expression of MACC1, AGR2, and KAI1 in 106 whole HNSCC tissue samples was detected by immunohistochemical staining. Patient's clinical data and demographics were both collected. RESULTS Positive rates of MACC1 and AGR2 were significantly higher, and positive rate of KAI1 was significantly lower, in HNSCC and than those in control tissues. Positive rates of MACC1 and AGR2 were positively correlated with grades of tumor, TNM stages, and lymph node metastasis (LNM) stages, and negatively with patients OS; positive rate of KAI1 was negatively associated with grades of tumor, TNM stages, and LNM stages, and the positive expression of KAI1 subgroup had significantly longer OS than did the negative KAI1 subgroup. In multivariate analysis, positive expression MACC1, AGR2, and KAI1, and tumor stages, as well as LNM stages were potential to be independent prognostic factors for OS in patients with HNSCC. CONCLUSIONS MACC1, AGR2, and KAI1 may represent potential metastatic and prognostic biomarkers, as well as promising therapeutic targets for HNSCC.
Collapse
Affiliation(s)
- Benlu Sun
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Anhui Provincial Hospital of Anhui Medical UniversityAnhui Province, China
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Bengbu Medical CollegeAnhui Province, China
| | - Zenong Cheng
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical CollegeAnhui Province, China
- Department of Pathology, Bengbu Medical CollegeAnhui Province, China
| | - Jingwu Sun
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Anhui Provincial Hospital of Anhui Medical UniversityAnhui Province, China
| |
Collapse
|
15
|
How tetraspanins shape endothelial and leukocyte nano-architecture during inflammation. Biochem Soc Trans 2017; 45:999-1006. [PMID: 28710286 DOI: 10.1042/bst20170163] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 06/07/2017] [Accepted: 06/09/2017] [Indexed: 01/13/2023]
Abstract
Tetraspanins are ubiquitous membrane proteins that induce local membrane curvature and hence co-ordinate cell-to-cell contacts. This review highlights their role in inflammation, which requires control of the nano-architecture of attachment sites between endothelial cells and leukocytes. The active role of endothelial cells in preparing for transmigration of leukocytes and determining the severity of an inflammation is often underscored. A clear hint to endothelial pre-activation is their ability to protrude clustered adhesion proteins upward prior to leukocyte contact. The elevation of molecular adhesive platforms toward the blood stream is crucially dependent on tetraspanins. In addition, leukocytes require tetraspanins for their activation. The example of the B-cell receptor is referenced in some detail here, since it provides deeper insights into the receptor-coreceptor interplay. To lift the role of tetraspanins from an abstract model of inflammation toward a player of clinical significance, two pathologies are analyzed for the known contributions of tetraspanins. The recent publication of the first crystal structure of a full-length tetraspanin revealed a cholesterol-binding site, which provides a strong link to the pathophysiological condition of atherosclerosis. Dysregulation of the inflammatory cascade in autoimmune diseases by endothelial cells is exemplified by the involvement of tetraspanins in multiple sclerosis.
Collapse
|
16
|
Munkley J, McClurg UL, Livermore KE, Ehrmann I, Knight B, Mccullagh P, Mcgrath J, Crundwell M, Harries LW, Leung HY, Mills IG, Robson CN, Rajan P, Elliott DJ. The cancer-associated cell migration protein TSPAN1 is under control of androgens and its upregulation increases prostate cancer cell migration. Sci Rep 2017; 7:5249. [PMID: 28701765 PMCID: PMC5507901 DOI: 10.1038/s41598-017-05489-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/30/2017] [Indexed: 02/06/2023] Open
Abstract
Cell migration drives cell invasion and metastatic progression in prostate cancer and is a major cause of mortality and morbidity. However the mechanisms driving cell migration in prostate cancer patients are not fully understood. We previously identified the cancer-associated cell migration protein Tetraspanin 1 (TSPAN1) as a clinically relevant androgen regulated target in prostate cancer. Here we find that TSPAN1 is acutely induced by androgens, and is significantly upregulated in prostate cancer relative to both normal prostate tissue and benign prostate hyperplasia (BPH). We also show for the first time, that TSPAN1 expression in prostate cancer cells controls the expression of key proteins involved in cell migration. Stable upregulation of TSPAN1 in both DU145 and PC3 cells significantly increased cell migration and induced the expression of the mesenchymal markers SLUG and ARF6. Our data suggest TSPAN1 is an androgen-driven contributor to cell survival and motility in prostate cancer.
Collapse
Affiliation(s)
- Jennifer Munkley
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, UK.
| | - Urszula L McClurg
- Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Karen E Livermore
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, UK
| | - Ingrid Ehrmann
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, UK
| | - Bridget Knight
- NIHR Exeter Clinical Research Facility, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Paul Mccullagh
- Department of Pathology, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - John Mcgrath
- Exeter Surgical Health Services Research Unit, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Malcolm Crundwell
- Department of Urology, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Lorna W Harries
- Institute of Biomedical and Clinical Sciences, University of Exeter, Devon, UK
| | - Hing Y Leung
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Ian G Mills
- Prostate Cancer Research Group, Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo and Oslo University Hospitals, Forskningsparken, Gaustadalléen 21, N-0349, Oslo, Norway
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital HE - Norwegian Radium Hospital, Montebello, Ian G. Mills, NO-0424, Oslo, Norway
- Movember/Prostate Cancer UK Centre of Excellence for Prostate Cancer Research, Centre for Cancer Research and Cell Biology (CCRCB), Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Craig N Robson
- Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Prabhakar Rajan
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - David J Elliott
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, UK
| |
Collapse
|
17
|
Termini CM, Gillette JM. Tetraspanins Function as Regulators of Cellular Signaling. Front Cell Dev Biol 2017; 5:34. [PMID: 28428953 PMCID: PMC5382171 DOI: 10.3389/fcell.2017.00034] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/22/2017] [Indexed: 01/10/2023] Open
Abstract
Tetraspanins are molecular scaffolds that distribute proteins into highly organized microdomains consisting of adhesion, signaling, and adaptor proteins. Many reports have identified interactions between tetraspanins and signaling molecules, finding unique downstream cellular consequences. In this review, we will explore these interactions as well as the specific cellular responses to signal activation, focusing on tetraspanin regulation of adhesion-mediated (integrins/FAK), receptor-mediated (EGFR, TNF-α, c-Met, c-Kit), and intracellular signaling (PKC, PI4K, β-catenin). Additionally, we will summarize our current understanding for how tetraspanin post-translational modifications (palmitoylation, N-linked glycosylation, and ubiquitination) can regulate signal propagation. Many of the studies outlined in this review suggest that tetraspanins offer a potential therapeutic target to modulate aberrant signal transduction pathways that directly impact a host of cellular behaviors and disease states.
Collapse
Affiliation(s)
- Christina M Termini
- Department of Pathology, University of New Mexico Health Sciences CenterAlbuquerque, NM, USA
| | - Jennifer M Gillette
- Department of Pathology, University of New Mexico Health Sciences CenterAlbuquerque, NM, USA
| |
Collapse
|
18
|
Lu G, Zhou L, Zhang X, Zhu B, Wu S, Song W, Gong X, Wang D, Tao Y. The expression of metastasis-associated in colon cancer-1 and KAI1 in gastric adenocarcinoma and their clinical significance. World J Surg Oncol 2016; 14:276. [PMID: 27793161 PMCID: PMC5084408 DOI: 10.1186/s12957-016-1033-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 10/21/2016] [Indexed: 11/19/2022] Open
Abstract
Background The most common reason for malignant tumor treatment failure is recurrence and metastasis. Metastasis-associated in colon cancer-1 (MACC1) was originally identified as a metastatic and prognostic biomarker for colon cancer and later other solid tumors. Kangai 1 (KAI1), a marker of suppressor of metastasis, is also associated with metastasis and poor prognosis in many tumors. However, the prognostic value of either MACC1 or KAI1 in gastric adenocarcinoma (GAC) is unclear. In this study, we explored the relationship between MACC1 and KAI1 expression, as well as their respective correlation with clinicopathological features, to determine if either could be helpful for improvement of survival prognosis in GAC patients. Methods The expression levels of both MACC1 and KAI1 in 325 whole-tissue sections of GAC were examined by immunohistochemistry. Clinical data was also collected. Results MACC1 was significantly overexpressed in GAC tissues when compared to levels in normal gastric tissues; KAI1 was significantly down-expressed in GAC tissues when compared to levels in normal gastric tissues. Investigation of association between MACC1 and KAI1 protein levels with clinicopathological parameters of GAC indicated association between the expression of each with tumor grade, lymph node metastasis, invasive depth, and TNM stages. The overall survival time of patients with MACC1- or KAI1-positive GAC tumors was significantly shorter or longer than that of those who were negative. Importantly, multivariate analysis suggested that positive expression of either MACC1 or KAI1, as well as TNM stage, could be independent prognostic factors for overall survival in patients with GAC. Conclusions MACC1 and KAI1 may represent promising metastatic and prognostic biomarkers, as well as potential therapeutic targets, for GAC.
Collapse
Affiliation(s)
- Guoyu Lu
- Department of Emergence, The First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Bengbu, China
| | - Lei Zhou
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Bengbu, China.,Department of Pathology, Bengbu Medical College, No.2600, Donghai Street, Anhui Province, China
| | - Xiaohua Zhang
- Department of Emergence, The First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Bengbu, China
| | - Bo Zhu
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Bengbu, China.,Department of Pathology, Bengbu Medical College, No.2600, Donghai Street, Anhui Province, China
| | - Shiwu Wu
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Bengbu, China. .,Department of Pathology, Bengbu Medical College, No.2600, Donghai Street, Anhui Province, China.
| | - Wenqing Song
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Bengbu, China.,Department of Pathology, Bengbu Medical College, No.2600, Donghai Street, Anhui Province, China
| | - Xiaomeng Gong
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Bengbu, China.,Department of Pathology, Bengbu Medical College, No.2600, Donghai Street, Anhui Province, China
| | - Danna Wang
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Bengbu, China.,Department of Pathology, Bengbu Medical College, No.2600, Donghai Street, Anhui Province, China
| | - Yanyan Tao
- Department of Emergence, The First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Bengbu, China
| |
Collapse
|
19
|
Feng J, Huang C, Wren JD, Wang DW, Yan J, Zhang J, Sun Y, Han X, Zhang XA. Tetraspanin CD82: a suppressor of solid tumors and a modulator of membrane heterogeneity. Cancer Metastasis Rev 2016; 34:619-33. [PMID: 26335499 DOI: 10.1007/s10555-015-9585-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Tetraspanin CD82 suppresses the progression and metastasis of a wide range of solid malignant tumors. However, its roles in tumorigenesis and hematopoietic malignancy remain unclear. Ubiquitously expressed CD82 restrains cell migration and cell invasion by modulating both cell-matrix and cell-cell adhesiveness and confining outside-in pro-motility signaling. This restraint at least contributes to, if not determines, the metastasis-suppressive activity and, also likely, the physiological functions of CD82. As a modulator of cell membrane heterogeneity, CD82 alters microdomains, trafficking, and topography of the membrane by changing the membrane molecular landscape. The functional activities of membrane molecules and the cytoskeletal interaction of the cell membrane are subsequently altered, followed by changes in cellular functions. Given its pathological and physiological importance, CD82 is a promising candidate for clinically predicting and blocking tumor progression and metastasis and also an emerging model protein for mechanistically understanding cell membrane organization and heterogeneity.
Collapse
Affiliation(s)
- Jin Feng
- Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Huang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC 1474, 975 NE 10th Street, Oklahoma City, OK, 73104, USA
| | - Jonathan D Wren
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Dao-Wen Wang
- Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhou Yan
- Institute for Marine Biosystem and Neurosciences, Shanghai Ocean University, Shanghai, China
| | - Jiexin Zhang
- Department of Biochemistry, Nanjing Medical University, Nanjing, China
| | - Yujie Sun
- Department of Biochemistry, Nanjing Medical University, Nanjing, China
| | - Xiao Han
- Department of Biochemistry, Nanjing Medical University, Nanjing, China
| | - Xin A Zhang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC 1474, 975 NE 10th Street, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
20
|
Wang DD, Chen X, Yu DD, Yang SJ, Shen HY, Sha HH, Zhong SL, Zhao JH, Tang JH. miR-197: A novel biomarker for cancers. Gene 2016; 591:313-9. [PMID: 27320730 DOI: 10.1016/j.gene.2016.06.035] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 06/06/2016] [Accepted: 06/15/2016] [Indexed: 12/19/2022]
Abstract
microRNAs (miRNAs) are small noncoding RNAs that could regulate post-transcription level through binding to 3' untranslated region (3'UTR) of target messenger RNAs (mRNAs), which were reported to be related with the incidence and development of diverse neoplasms. Among them, miR-197 was confirmed to play a vital role of oncogene or anti-oncogene in different cancers via targeting key tumorigenic or tumor-suppressive genes. Additionally, miR-197 had extensively been studied in carcinogenesis progression of cancers through various mechanisms, including apoptosis, proliferation, angiogenesis, metastasis, drug resistance and tumor suppressor, and also played a role in prognosis of cancers. In this review, we summarized the roles of miR-197 in cancers and considered it as a potentially novel biomarker for different cancers, offering an alternatively secure and effective tool in molecular targeting cancer treatment.
Collapse
Affiliation(s)
- Dan-Dan Wang
- The First Clinical School of Nanjing Medical University, Nanjing 210009, China; The Forth Clinical School of Nanjing Medical University, Nanjing 210009, China
| | - Xiu Chen
- The Forth Clinical School of Nanjing Medical University, Nanjing 210009, China; Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China
| | - Dan-Dan Yu
- The First Clinical School of Nanjing Medical University, Nanjing 210009, China; Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China
| | - Su-Jin Yang
- The Forth Clinical School of Nanjing Medical University, Nanjing 210009, China; Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China
| | - Hong-Yu Shen
- The Forth Clinical School of Nanjing Medical University, Nanjing 210009, China; Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China
| | - Huan-Huan Sha
- The Forth Clinical School of Nanjing Medical University, Nanjing 210009, China; Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China
| | - Shan-Liang Zhong
- Center of Clinical Laboratory, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China
| | - Jian-Hua Zhao
- Center of Clinical Laboratory, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China
| | - Jin-Hai Tang
- Department of General Surgery, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China; Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China.
| |
Collapse
|
21
|
Tsui KH, Lin YH, Chung LC, Chuang ST, Feng TH, Chiang KC, Chang PL, Yeh CJ, Juang HH. Prostate-derived ets factor represses tumorigenesis and modulates epithelial-to-mesenchymal transition in bladder carcinoma cells. Cancer Lett 2016; 375:142-151. [PMID: 26965996 DOI: 10.1016/j.canlet.2016.02.056] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/27/2016] [Accepted: 02/29/2016] [Indexed: 12/27/2022]
Abstract
Prostate-derived Ets (E-twenty six) factor (PDEF), an epithelium-specific member of the Ets family of transcription factors, has been shown to play a role in suppressing the development of many epithelium-derived cancers such as prostate and breast cancer. It is not clear, however, whether PDEF is involved in the development or progression of bladder cancer. In a comparison between normal urothelium and bladder tumor tissue, we identified significant decreases of PDEF in the tumor tissue. Further, the immunohistochemistry assays indicated a significantly higher immunostaining of PDEF in low-grade bladder tumors. Additionally, the highly differentiated transitional-cell bladder carcinoma RT-4 cells expressed significantly more PDEF levels than the bladder carcinoma HT1376 and the T24 cells. Ectopic overexpression of PDEF attenuated proliferation, invasion, and tumorigenesis of bladder carcinoma cells in vitro and in vivo. PDEF enhanced the expression levels of mammary serine protease inhibitor (MASPIN), N-myc downstream regulated gene 1 (NDRG1), KAI1, and B-cell translocation gene 2 (BTG2). PDEF modulated epithelial-mesenchymal-transition (EMT) by upregulating E-cadherin expression and downregulating the expression of N-cadherin, SNAIL, SLUG, and vimentin, leading to lower migration and invasion abilities of bladder carcinoma cells. Filamentous actin (F-actin) polarization and remodeling were observed in PDEF-knockdown RT-4 cells. Our results suggest that PDEF gene expression is associated with the extent of bladder neoplasia and PDEF modulated the expressions of EMT-related genes. The induction of BTG2, NDRG1, MASPIN, and KAI1 gene expressions by PDEF may explain the inhibitory functions of PDEF on the proliferation, invasion, and tumorigenesis in bladder carcinoma cells.
Collapse
Affiliation(s)
- Ke-Hung Tsui
- Department of Urology, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan, Taiwan; Department of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Yu-Hsiang Lin
- Department of Urology, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan, Taiwan; Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Li-Chuan Chung
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Sung-Ting Chuang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Tsui-Hsia Feng
- School of Nursing, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Kun-Chun Chiang
- Zebafish Center, General Surgery Department, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Phei-Lang Chang
- Department of Urology, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan, Taiwan; Department of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Chi-Ju Yeh
- Department of Pathology, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Horng-Heng Juang
- Department of Urology, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan, Taiwan; Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan.
| |
Collapse
|
22
|
Rocha-Perugini V, Sánchez-Madrid F, Martínez Del Hoyo G. Function and Dynamics of Tetraspanins during Antigen Recognition and Immunological Synapse Formation. Front Immunol 2016; 6:653. [PMID: 26793193 PMCID: PMC4707441 DOI: 10.3389/fimmu.2015.00653] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/18/2015] [Indexed: 12/31/2022] Open
Abstract
Tetraspanin-enriched microdomains (TEMs) are specialized membrane platforms driven by protein–protein interactions that integrate membrane receptors and adhesion molecules. Tetraspanins participate in antigen recognition and presentation by antigen-presenting cells (APCs) through the organization of pattern-recognition receptors (PRRs) and their downstream-induced signaling, as well as the regulation of MHC-II–peptide trafficking. T lymphocyte activation is triggered upon specific recognition of antigens present on the APC surface during immunological synapse (IS) formation. This dynamic process is characterized by a defined spatial organization involving the compartmentalization of receptors and adhesion molecules in specialized membrane domains that are connected to the underlying cytoskeleton and signaling molecules. Tetraspanins contribute to the spatial organization and maturation of the IS by controlling receptor clustering and local accumulation of adhesion receptors and integrins, their downstream signaling, and linkage to the actin cytoskeleton. This review offers a perspective on the important role of TEMs in the regulation of antigen recognition and presentation and in the dynamics of IS architectural organization.
Collapse
Affiliation(s)
- Vera Rocha-Perugini
- Servicio de Inmunología, Instituto de Investigación Sanitaria La Princesa, Hospital de la Princesa, Madrid, Spain; Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Servicio de Inmunología, Instituto de Investigación Sanitaria La Princesa, Hospital de la Princesa, Madrid, Spain; Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Gloria Martínez Del Hoyo
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) , Madrid , Spain
| |
Collapse
|
23
|
Jones EL, Wee JL, Demaria MC, Blakeley J, Ho PK, Vega-Ramos J, Villadangos JA, van Spriel AB, Hickey MJ, Hämmerling GJ, Wright MD. Dendritic Cell Migration and Antigen Presentation Are Coordinated by the Opposing Functions of the Tetraspanins CD82 and CD37. THE JOURNAL OF IMMUNOLOGY 2016; 196:978-87. [PMID: 26729805 DOI: 10.4049/jimmunol.1500357] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 12/01/2015] [Indexed: 11/19/2022]
Abstract
This study supports a new concept where the opposing functions of the tetraspanins CD37 and CD82 may coordinate changes in migration and Ag presentation during dendritic cell (DC) activation. We have previously published that CD37 is downregulated upon monocyte-derived DC activation, promotes migration of both skin and bone marrow-derived dendritic cells (BMDCs), and restrains Ag presentation in splenic and BMDCs. In this article, we show that CD82, the closest phylogenetic relative to CD37, appears to have opposing functions. CD82 is upregulated upon activation of BMDCs and monocyte-derived DCs, restrains migration of skin and BMDCs, supports MHC class II maturation, and promotes stable interactions between T cells and splenic DCs or BMDCs. The underlying mechanism involves the rearrangement of the cytoskeleton via a differential activation of small GTPases. Both CD37(-/-) and CD82(-/-) BMDCs lack cellular projections, but where CD37(-/-) BMDCs spread poorly on fibronectin, CD82(-/-) BMDCs are large and spread to a greater extent than wild-type BMDCs. At the molecular level, CD82 is a negative regulator of RhoA, whereas CD37 promotes activation of Rac-1; both tetraspanins negatively regulate Cdc42. Thus, this study identifies a key aspect of DC biology: an unactivated BMDC is CD37(hi)CD82(lo), resulting in a highly motile cell with a limited ability to activate naive T cells. By contrast, a late activated BMDC is CD37(lo)CD82(hi), and thus has modified its migratory, cytoskeletal, and Ag presentation machinery to become a cell superbly adapted to activating naive T cells.
Collapse
Affiliation(s)
- Eleanor L Jones
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3004, Australia
| | - Janet L Wee
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3004, Australia; Department of Medicine, Centre for Inflammatory Diseases, Monash University, Clayton, Victoria 3168, Australia
| | - Maria C Demaria
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3004, Australia
| | - Jessica Blakeley
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3004, Australia
| | - Po Ki Ho
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3004, Australia
| | - Javier Vega-Ramos
- Department of Microbiology and Immunology, University of Melbourne, Melbourne 3010, Australia
| | - Jose A Villadangos
- Department of Microbiology and Immunology, University of Melbourne, Melbourne 3010, Australia; Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne 3010, Australia
| | - Annemiek B van Spriel
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, G525 GA Nijmegen, the Netherlands; and
| | - Michael J Hickey
- Department of Medicine, Centre for Inflammatory Diseases, Monash University, Clayton, Victoria 3168, Australia
| | | | - Mark D Wright
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3004, Australia;
| |
Collapse
|
24
|
Wee JL, Schulze KE, Jones EL, Yeung L, Cheng Q, Pereira CF, Costin A, Ramm G, van Spriel AB, Hickey MJ, Wright MD. Tetraspanin CD37 Regulates β2 Integrin-Mediated Adhesion and Migration in Neutrophils. THE JOURNAL OF IMMUNOLOGY 2015; 195:5770-9. [PMID: 26566675 DOI: 10.4049/jimmunol.1402414] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 10/14/2015] [Indexed: 01/13/2023]
Abstract
Deciphering the molecular basis of leukocyte recruitment is critical to the understanding of inflammation. In this study, we investigated the contribution of the tetraspanin CD37 to this key process. CD37-deficient mice showed impaired neutrophil recruitment in a peritonitis model. Intravital microscopic analysis indicated that the absence of CD37 impaired the capacity of leukocytes to follow a CXCL1 chemotactic gradient accurately in the interstitium. Moreover, analysis of CXCL1-induced leukocyte-endothelial cell interactions in postcapillary venules revealed that CXCL1-induced neutrophil adhesion and transmigration were reduced in the absence of CD37, consistent with a reduced capacity to undergo β2 integrin-dependent adhesion. This result was supported by in vitro flow chamber experiments that demonstrated an impairment in adhesion of CD37-deficient neutrophils to the β2 integrin ligand, ICAM-1, despite the normal display of high-affinity β2 integrins. Superresolution microscopic assessment of localization of CD37 and CD18 in ICAM-1-adherent neutrophils demonstrated that these molecules do not significantly cocluster in the cell membrane, arguing against the possibility that CD37 regulates β2 integrin function via a direct molecular interaction. Moreover, CD37 ablation did not affect β2 integrin clustering. In contrast, the absence of CD37 in neutrophils impaired actin polymerization, cell spreading and polarization, dysregulated Rac-1 activation, and accelerated β2 integrin internalization. Together, these data indicate that CD37 promotes neutrophil adhesion and recruitment via the promotion of cytoskeletal function downstream of integrin-mediated adhesion.
Collapse
Affiliation(s)
- Janet L Wee
- Department of Immunology, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia; Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - Keith E Schulze
- Monash Micro Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Eleanor L Jones
- Department of Immunology, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia
| | - Louisa Yeung
- Department of Immunology, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia; Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - Qiang Cheng
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - Candida F Pereira
- Burnet Institute, Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia; and
| | - Adam Costin
- Monash Micro Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Georg Ramm
- Monash Micro Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Annemiek B van Spriel
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - Mark D Wright
- Department of Immunology, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia;
| |
Collapse
|
25
|
Detchokul S, Williams ED, Parker MW, Frauman AG. Tetraspanins as regulators of the tumour microenvironment: implications for metastasis and therapeutic strategies. Br J Pharmacol 2015; 171:5462-90. [PMID: 23731188 DOI: 10.1111/bph.12260] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/16/2013] [Accepted: 05/16/2013] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED One of the hallmarks of cancer is the ability to activate invasion and metastasis. Cancer morbidity and mortality are largely related to the spread of the primary, localized tumour to adjacent and distant sites. Appropriate management and treatment decisions based on predicting metastatic disease at the time of diagnosis is thus crucial, which supports better understanding of the metastatic process. There are components of metastasis that are common to all primary tumours: dissociation from the primary tumour mass, reorganization/remodelling of extracellular matrix, cell migration, recognition and movement through endothelial cells and the vascular circulation and lodgement and proliferation within ectopic stroma. One of the key and initial events is the increased ability of cancer cells to move, escaping the regulation of normal physiological control. The cellular cytoskeleton plays an important role in cancer cell motility and active cytoskeletal rearrangement can result in metastatic disease. This active change in cytoskeletal dynamics results in manipulation of plasma membrane and cellular balance between cellular adhesion and motility which in turn determines cancer cell movement. Members of the tetraspanin family of proteins play important roles in regulation of cancer cell migration and cancer-endothelial cell interactions, which are critical for cancer invasion and metastasis. Their involvements in active cytoskeletal dynamics, cancer metastasis and potential clinical application will be discussed in this review. In particular, the tetraspanin member, CD151, is highlighted for its major role in cancer invasion and metastasis. LINKED ARTICLES This article is part of a themed section on Cytoskeleton, Extracellular Matrix, Cell Migration, Wound Healing and Related Topics. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-24.
Collapse
Affiliation(s)
- S Detchokul
- Clinical Pharmacology and Therapeutics Unit, Department of Medicine (Austin Health/Northern Health), The University of Melbourne, Heidelberg, Vic., Australia
| | | | | | | |
Collapse
|
26
|
Charming neighborhoods on the cell surface: plasma membrane microdomains regulate receptor tyrosine kinase signaling. Cell Signal 2015; 27:1963-76. [PMID: 26163824 DOI: 10.1016/j.cellsig.2015.07.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/07/2015] [Indexed: 12/14/2022]
Abstract
Receptor tyrosine kinases (RTK) are an important family of growth factor and hormone receptors that regulate many aspects of cellular physiology. Ligand binding by RTKs at the plasma membrane elicits activation of many signaling intermediates. The spatial and temporal regulation of RTK signaling within cells is an important determinant of receptor signaling outcome. In particular, the compartmentalization of the plasma membrane into a number of microdomains allows context-specific control of RTK signaling. Indeed various RTKs are recruited to and enriched within specific plasma membrane microdomains under various conditions, including lipid-ordered domains such as caveolae and lipid rafts, clathrin-coated structures, tetraspanin-enriched microdomains, and actin-dependent protrusive membrane microdomains such as dorsal ruffles and invadosomes. We examine the evidence for control of RTK signaling by each of these plasma membrane microdomains, as well as molecular mechanisms for how this spatial organization controls receptor signaling.
Collapse
|
27
|
Abstract
Tetraspanins are a superfamily of small transmembrane proteins that are expressed in almost all eukaryotic cells. Through interacting with one another and with other membrane and intracellular proteins, tetraspanins regulate a wide range of proteins such as integrins, cell surface receptors, and signaling molecules, and thereby engage in diverse cellular processes ranging from cell adhesion and migration to proliferation and differentiation. In particular, tetraspanins modulate the function of proteins involved in all determining factors of cell migration including cell-cell adhesion, cell-ECM adhesion, cytoskeletal protrusion/contraction, and proteolytic ECM remodeling. We herein provide a brief overview of collective in vitro and in vivo studies of tetraspanins to illustrate their regulatory functions in the migration and trafficking of cancer cells, vascular endothelial cells, skin cells (keratinocytes and fibroblasts), and leukocytes. We also discuss the involvement of tetraspanins in various pathologic and remedial processes that rely on cell migration and their potential value as targets for therapeutic intervention.
Collapse
Affiliation(s)
| | - Jiaping Zhang
- a Institute of Burn Research ; State Key Laboratory of Trauma; Burns and Combined Injury; Southwest Hospital; The Third Military Medical University ; Chongqing , China
| | - Yuesheng Huang
- a Institute of Burn Research ; State Key Laboratory of Trauma; Burns and Combined Injury; Southwest Hospital; The Third Military Medical University ; Chongqing , China
| |
Collapse
|
28
|
Blumenthal A, Giebel J, Warsow G, Li L, Ummanni R, Schordan S, Schordan E, Klemm P, Gretz N, Endlich K, Endlich N. Mechanical stress enhances CD9 expression in cultured podocytes. Am J Physiol Renal Physiol 2014; 308:F602-13. [PMID: 25503725 DOI: 10.1152/ajprenal.00190.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Elevated glomerular pressure represents a high risk for the development of severe kidney diseases and causes an increase in mechanical load to podocytes. In this study, we investigated whether mechanical stress alters gene expression in cultured podocytes using gene arrays. We found that tetraspanin CD9 is significantly upregulated in cultured podocytes after mechanical stress. The differential expression of CD9 was confirmed by RT-PCR and Western blotting under stretched and unstretched conditions. Furthermore, mechanical stress resulted in a relocalization of CD9. To get an insight into the functional role of CD9, podocytes were transfected with pEGFP-CD9. The expression of CD9 induced the formation of substratum-attached thin arborized protrusions. Ca(2+) depletion revealed that podocytes overexpressing CD9 possess altered adhesive properties in contrast to the control transfected cells. Finally, elevated CD9 expression increased migration of podocytes in a wound assay. In summary, our results suggest that upregulation of CD9 may play an important role in podocyte morphology, adhesion, and migration.
Collapse
Affiliation(s)
- A Blumenthal
- Department of Anatomy and Cell Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - J Giebel
- Department of Anatomy and Cell Biology, Universitätsmedizin Greifswald, Greifswald, Germany;
| | - G Warsow
- Department of Anatomy and Cell Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - L Li
- Department of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - R Ummanni
- Center for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India; and
| | - S Schordan
- Department of Anatomy and Cell Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - E Schordan
- Department of Anatomy and Cell Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - P Klemm
- Department of Anatomy and Cell Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - N Gretz
- Medical Faculty Mannheim, Medical Research Center, University of Heidelberg, Mannheim, Germany
| | - K Endlich
- Department of Anatomy and Cell Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - N Endlich
- Department of Anatomy and Cell Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
29
|
Rocha-Perugini V, Gordon-Alonso M, Sánchez-Madrid F. PIP2: choreographer of actin-adaptor proteins in the HIV-1 dance. Trends Microbiol 2014; 22:379-88. [PMID: 24768560 DOI: 10.1016/j.tim.2014.03.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 03/11/2014] [Accepted: 03/25/2014] [Indexed: 02/06/2023]
Abstract
The actin cytoskeleton plays a key role during the replication cycle of human immunodeficiency virus-1 (HIV-1). HIV-1 infection is affected by cellular proteins that influence the clustering of viral receptors or the subcortical actin cytoskeleton. Several of these actin-adaptor proteins are controlled by the second messenger phosphatidylinositol 4,5-biphosphate (PIP2), an important regulator of actin organization. PIP2 production is induced by HIV-1 attachment and facilitates viral infection. However, the importance of PIP2 in regulating cytoskeletal proteins and thus HIV-1 infection has been overlooked. This review examines recent reports describing the roles played by actin-adaptor proteins during HIV-1 infection of CD4+ T cells, highlighting the influence of the signaling lipid PIP2 in this process.
Collapse
Affiliation(s)
- Vera Rocha-Perugini
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria de la Princesa, Madrid, Spain; Vascular Biology and Inflammation Department, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Mónica Gordon-Alonso
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria de la Princesa, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria de la Princesa, Madrid, Spain; Vascular Biology and Inflammation Department, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.
| |
Collapse
|
30
|
Dai W, Wang C, Wang F, Wang Y, Shen M, Chen K, Cheng P, Zhang Y, Yang J, Zhu R, Zhang H, Li J, Zheng Y, Lu J, Zhou Y, Xu L, Guo C. Anti-miR-197 inhibits migration in HCC cells by targeting KAI 1/CD82. Biochem Biophys Res Commun 2014; 446:541-548. [PMID: 24613834 DOI: 10.1016/j.bbrc.2014.03.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 03/01/2014] [Indexed: 11/28/2022]
Abstract
AIM To investigate the metastatic effects and mechanisms of miR-197 in hepatocellular carcinoma (HCC). METHODS AND RESULTS The levels of miR-197 increased in HCC cells and tissues compared with a normal hepatic cell line (LO2) and adjacent nontumorous liver tissues, respectively. miR-197 expression negatively correlated with CD82 mRNA expression in these cell lines and tissues. Dual luciferase reporter assay and Western blot confirmed a direct interaction between miR-197 and CD82 3'UTR sequences. After miR-197 was silenced in HCC cells, CD82 expression increased. In the presence of human hepatocyte growth factor (HGF), cells silenced for anti-miR-197 exhibited elongated cellular tails and diminished lamellipodia due to reductions in both ROCK activity and the levels of Rac 1 protein. Downregulation of miR-197 along with the upregulation of CD82 in HCC cells resulted in the inhibition of HCC migration and invasion in vitro and in vivo. CONCLUSION Taken together, these data suggest that anti-miR-197 suppresses HCC migration and invasion by targeting CD82. The regulation of the miR-197/CD82 axis could be a novel therapeutic target in future HCC effective therapy.
Collapse
Affiliation(s)
- Weiqi Dai
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Chengfen Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Fan Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Yugang Wang
- Department of Gastroenterology, Tong Ren Hospital, Jiaotong University, School of Medicine, Shanghai, China
| | - Miao Shen
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Kan Chen
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Ping Cheng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Yan Zhang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Jing Yang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Rong Zhu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Huawei Zhang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Jingjing Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Yuanyuan Zheng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Jie Lu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Yingqun Zhou
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Ling Xu
- Department of Gastroenterology, Tong Ren Hospital, Jiaotong University, School of Medicine, Shanghai, China.
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China.
| |
Collapse
|
31
|
Khanna P, Chung CY, Neves RI, Robertson GP, Dong C. CD82/KAI expression prevents IL-8-mediated endothelial gap formation in late-stage melanomas. Oncogene 2013; 33:2898-908. [PMID: 23873025 DOI: 10.1038/onc.2013.249] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 03/23/2013] [Accepted: 04/19/2013] [Indexed: 12/27/2022]
Abstract
Melanoma cells facilitate endothelial gap formation, the first step during tumor transendothelial migration, which is mediated by both adhesion and endogenously produced chemokines (in particular, interleukin-8 (IL-8)). Tetraspanins are localized to the cell surface in cancer and participate in various functions including invasion of tissues mediated by secretion of cytokines and matrix metalloproteinases. However, little is known about the role of CD82 tetraspanins in malignant melanomas during cancer cell invasion. In this study, we investigated the functional importance of CD82 expression in melanoma-mediated gap formation by using cDNAs to induce CD82 expression in highly invasive melanoma cell lines. Results showed that CD82 expression inhibited melanoma cell-induced gap formation, melanoma cell extravasation in vitro and subsequent lung metastasis development in vivo. Mechanistic studies showed that inducible expression of CD82 in highly metastatic melanoma cells significantly increased p21 expression upon binding of Duffy antigen receptor group (DARC), inducing tumor cell senescence and interrupting IL-8-mediated vascular endothelial (VE)-cadherin disassembly. Taken together, these studies provide a rationale for using drug therapies that restore CD82 expression and inhibit IL-8 production to inhibit late-stage melanoma cell extravasation and subsequent metastasis development.
Collapse
Affiliation(s)
- P Khanna
- Department of Bioengineering, The Pennsylvania State University, University Park, PA, USA
| | - C-Y Chung
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - R I Neves
- 1] Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA [2] Department of Dermatology, The Pennsylvania State University College of Medicine, Hershey, PA, USA [3] Penn State Melanoma Therapeutic Program, The Pennsylvania State University College of Medicine, Hershey, PA, USA [4] Cutaneous Oncology Program, The Pennsylvania State University College of Medicine, Hershey, PA, USA [5] Department of Surgery, Division of Plastic Surgery, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - G P Robertson
- 1] Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA [2] Department of Dermatology, The Pennsylvania State University College of Medicine, Hershey, PA, USA [3] Penn State Melanoma Therapeutic Program, The Pennsylvania State University College of Medicine, Hershey, PA, USA [4] Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, PA, USA [5] Pennsylvania State Melanoma Center, The Pennsylvania State University College of Medicine, Hershey, PA, USA [6] The Foreman Foundation for Melanoma Research, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - C Dong
- 1] Department of Bioengineering, The Pennsylvania State University, University Park, PA, USA [2] Pennsylvania State Melanoma Center, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|