1
|
McIntyre DC, Nance J. Niche cells regulate primordial germ cell quiescence in response to basement membrane signaling. Development 2023; 150:dev201640. [PMID: 37497562 PMCID: PMC10445801 DOI: 10.1242/dev.201640] [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: 01/25/2023] [Accepted: 07/17/2023] [Indexed: 07/28/2023]
Abstract
Stem cell quiescence, proliferation and differentiation are controlled by interactions with niche cells and a specialized extracellular matrix called basement membrane (BM). Direct interactions with adjacent BM are known to regulate stem cell quiescence; however, it is less clear how niche BM relays signals to stem cells that it does not contact. Here, we examine how niche BM regulates Caenorhabditis elegans primordial germ cells (PGCs). BM regulates PGC quiescence even though PGCs are enwrapped by somatic niche cells and do not contact the BM; this can be demonstrated by depleting laminin, which causes normally quiescent embryonic PGCs to proliferate. We show that following laminin depletion, niche cells relay proliferation-inducing signals from the gonadal BM to PGCs via integrin receptors. Disrupting the BM proteoglycan perlecan blocks PGC proliferation when laminin is depleted, indicating that laminin functions to inhibit a proliferation-inducing signal originating from perlecan. Reducing perlecan levels in fed larvae hampers germline growth, suggesting that BM signals regulate germ cell proliferation under physiological conditions. Our results reveal how BM signals can regulate stem cell quiescence indirectly, by activating niche cell integrin receptors.
Collapse
Affiliation(s)
- Daniel C. McIntyre
- Skirball Institute of Biomolecular Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
- University of Virginia, Department of Biology, 90 Geldard Drive, Physical Life Science Building Room 318, Charlottesville, VA 22904, USA
| | - Jeremy Nance
- Skirball Institute of Biomolecular Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
2
|
Chen Z, Zhao H, Meng L, Yu S, Liu Z, Xue J. Microfibril-Associated Glycoprotein-2 Promoted Fracture Healing via Integrin αvβ3/PTK2/AKT Signaling. J Transl Med 2023; 103:100121. [PMID: 36934797 DOI: 10.1016/j.labinv.2023.100121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/08/2023] [Accepted: 02/16/2023] [Indexed: 03/19/2023] Open
Abstract
Fracture healing is a complex physiological process in which angiogenesis plays an essential role. Microfibril-associated glycoprotein-2 (MAGP2) has been reported to possess a proangiogenic activity via integrin αvβ3, yet its role in bone repair is unexplored. In this study, a critical-sized femoral defect (2 mm) was created in mice, followed by the delivery of an adenovirus-based MAGP2 overexpression vector or its negative control at the fracture site. At days 7, 14, 21, and 28 postfracture, bone fracture healing was evaluated by radiography, micro-computed tomography, and histopathologic analysis. Adenovirus-based MAGP2 overexpression vector-treated mice exhibited increased bone mineral density and bone volume fraction. MAGP2 overexpression contributed to an advanced stage of endochondral ossification and induced cartilage callus into the bony callus. Further analysis indicated that MAGP2 was associated with enhanced angiogenesis, as evidenced by marked MAGP2 and integrin αvβ3 costaining and increased endothelial cell markers such as endomucin and CD31 levls, as well as elevated phosphorylation of protein tyrosine kinase 2 (PTK2) and AKT serine/threonine kinase 1 (AKT) in the callus. In vitro, recombinant human MAGP2 treatment enhanced the viability, migration, and tube formation ability of human microvascular endothelial cells, which was partially reversed by integrin αvβ3 inhibition or MK-2206, a specific AKT inhibitor. Inhibition of integrin αvβ3 abolished MAGP2-induced PTK2 and AKT activation. Taken together, our data provide the first evidence that MAGP2 promotes angiogenesis and bone formation by activating the integrin αvβ3/PTK2/AKT signaling pathway.
Collapse
Affiliation(s)
- Zhiguang Chen
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Haibin Zhao
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lingshuai Meng
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shengwei Yu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhenning Liu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jinqi Xue
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
3
|
Han C, Leonardo TR, Romana-Souza B, Shi J, Keiser S, Yuan H, Altakriti M, Ranzer MJ, Ferri-Borgogno S, Mok SC, Koh TJ, Hong SJ, Chen L, DiPietro LA. Microfibril-associated protein 5 and the regulation of skin scar formation. Sci Rep 2023; 13:8728. [PMID: 37253753 PMCID: PMC10229580 DOI: 10.1038/s41598-023-35558-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/20/2023] [Indexed: 06/01/2023] Open
Abstract
Many factors regulate scar formation, which yields a modified extracellular matrix (ECM). Among ECM components, microfibril-associated proteins have been minimally explored in the context of skin wound repair. Microfibril-associated protein 5 (MFAP5), a small 25 kD serine and threonine rich microfibril-associated protein, influences microfibril function and modulates major extracellular signaling pathways. Though known to be associated with fibrosis and angiogenesis in certain pathologies, MFAP5's role in wound healing is unknown. Using a murine model of skin wound repair, we found that MFAP5 is significantly expressed during the proliferative and remodeling phases of healing. Analysis of existing single-cell RNA-sequencing data from mouse skin wounds identified two fibroblast subpopulations as the main expressors of MFAP5 during wound healing. Furthermore, neutralization of MFAP5 in healing mouse wounds decreased collagen deposition and refined angiogenesis without altering wound closure. In vitro, recombinant MFAP5 significantly enhanced dermal fibroblast migration, collagen contractility, and expression of pro-fibrotic genes. Additionally, TGF-ß1 increased MFAP5 expression and production in dermal fibroblasts. Our findings suggest that MFAP5 regulates fibroblast function and influences scar formation in healing wounds. Our work demonstrates a previously undescribed role for MFAP5 and suggests that microfibril-associated proteins may be significant modulators of wound healing outcomes and scarring.
Collapse
Affiliation(s)
- Chen Han
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA
| | - Trevor R Leonardo
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Bruna Romana-Souza
- Department of Histology and Embryology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Junhe Shi
- NMPA Key Laboratory for Clinical Research and Evaluation of Traditional Chinese Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shalyn Keiser
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA
| | - Heidi Yuan
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA
| | - Mohamad Altakriti
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA
| | - Matthew J Ranzer
- Department of Surgery, University of Illinois Chicago, Chicago, IL, USA
| | - Sammy Ferri-Borgogno
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samuel C Mok
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy J Koh
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL, USA
| | - Seok Jong Hong
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, IL, USA
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA.
| | - Luisa A DiPietro
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
4
|
Dong CX, Malecki C, Robertson E, Hambly B, Jeremy R. Molecular Mechanisms in Genetic Aortopathy-Signaling Pathways and Potential Interventions. Int J Mol Sci 2023; 24:ijms24021795. [PMID: 36675309 PMCID: PMC9865322 DOI: 10.3390/ijms24021795] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Thoracic aortic disease affects people of all ages and the majority of those aged <60 years have an underlying genetic cause. There is presently no effective medical therapy for thoracic aneurysm and surgery remains the principal intervention. Unlike abdominal aortic aneurysm, for which the inflammatory/atherosclerotic pathogenesis is well established, the mechanism of thoracic aneurysm is less understood. This paper examines the key cell signaling systems responsible for the growth and development of the aorta, homeostasis of endothelial and vascular smooth muscle cells and interactions between pathways. The evidence supporting a role for individual signaling pathways in pathogenesis of thoracic aortic aneurysm is examined and potential novel therapeutic approaches are reviewed. Several key signaling pathways, notably TGF-β, WNT, NOTCH, PI3K/AKT and ANGII contribute to growth, proliferation, cell phenotype and survival for both vascular smooth muscle and endothelial cells. There is crosstalk between pathways, and between vascular smooth muscle and endothelial cells, with both synergistic and antagonistic interactions. A common feature of the activation of each is response to injury or abnormal cell stress. Considerable experimental evidence supports a contribution of each of these pathways to aneurysm formation. Although human information is less, there is sufficient data to implicate each pathway in the pathogenesis of human thoracic aneurysm. As some pathways i.e., WNT and NOTCH, play key roles in tissue growth and organogenesis in early life, it is possible that dysregulation of these pathways results in an abnormal aortic architecture even in infancy, thereby setting the stage for aneurysm development in later life. Given the fine tuning of these signaling systems, functional polymorphisms in key signaling elements may set up a future risk of thoracic aneurysm. Multiple novel therapeutic agents have been developed, targeting cell signaling pathways, predominantly in cancer medicine. Future investigations addressing cell specific targeting, reduced toxicity and also less intense treatment effects may hold promise for effective new medical treatments of thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Charlotte Xue Dong
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Cassandra Malecki
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
| | - Elizabeth Robertson
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Brett Hambly
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Richmond Jeremy
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
- Correspondence:
| |
Collapse
|
5
|
Rincón-Ortega L, Valencia-Expósito A, Kabanova A, González-Reyes A, Martin-Bermudo MD. Integrins control epithelial stem cell proliferation in the Drosophila ovary by modulating the Notch pathway. Front Cell Dev Biol 2023; 11:1114458. [PMID: 36926523 PMCID: PMC10011466 DOI: 10.3389/fcell.2023.1114458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/07/2023] [Indexed: 03/08/2023] Open
Abstract
Cell proliferation and differentiation show a remarkable inverse relationship. The temporal coupling between cell cycle withdrawal and differentiation of stem cells (SCs) is crucial for epithelial tissue growth, homeostasis and regeneration. Proliferation vs. differentiation SC decisions are often controlled by the surrounding microenvironment, of which the basement membrane (BM; a specialized form of extracellular matrix surrounding cells and tissues), is one of its main constituents. Years of research have shown that integrin-mediated SC-BM interactions regulate many aspects of SC biology, including the proliferation-to-differentiation switch. However, these studies have also demonstrated that the SC responses to interactions with the BM are extremely diverse and depend on the cell type and state and on the repertoire of BM components and integrins involved. Here, we show that eliminating integrins from the follicle stem cells (FSCs) of the Drosophila ovary and their undifferentiated progeny increases their proliferation capacity. This results in an excess of various differentiated follicle cell types, demonstrating that cell fate determination can occur in the absence of integrins. Because these phenotypes are similar to those found in ovaries with decreased laminin levels, our results point to a role for the integrin-mediated cell-BM interactions in the control of epithelial cell division and subsequent differentiation. Finally, we show that integrins regulate proliferation by restraining the activity of the Notch/Delta pathway during early oogenesis. Our work increases our knowledge of the effects of cell-BM interactions in different SC types and should help improve our understanding of the biology of SCs and exploit their therapeutic potential.
Collapse
Affiliation(s)
- Lourdes Rincón-Ortega
- Centro Andaluz de Biología del Desarrollo, CSIC/Universidad Pablo de Olavide/JA, Sevilla, Spain
| | | | - Anna Kabanova
- Centro Andaluz de Biología del Desarrollo, CSIC/Universidad Pablo de Olavide/JA, Sevilla, Spain
| | - Acaimo González-Reyes
- Centro Andaluz de Biología del Desarrollo, CSIC/Universidad Pablo de Olavide/JA, Sevilla, Spain
| | - Maria D Martin-Bermudo
- Centro Andaluz de Biología del Desarrollo, CSIC/Universidad Pablo de Olavide/JA, Sevilla, Spain
| |
Collapse
|
6
|
Wang Y, Chen P, Zhao M, Cao H, Zhao Y, Ji M, Hou P, Chen M. EGFL7 drives the evolution of resistance to EGFR inhibitors in lung cancer by activating NOTCH signaling. Cell Death Dis 2022; 13:910. [PMID: 36309484 PMCID: PMC9617940 DOI: 10.1038/s41419-022-05354-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 11/07/2022]
Abstract
Accumulating evidence supports evolutionary trait of drug resistance. Like resilience in other systems, most tumor cells experience drug-tolerant state before full resistance acquired. However, the underlying mechanism is still poorly understood. Here, we identify that EGF like domain multiple 7 (EGFL7) is a responsive gene to epidermal growth factor receptor (EGFR) kinase inhibition during a period when tumors are decimated. Moreover, our data reveal that the adaptive increase of EGFL7 during this process is controlled by the depression of nonsense-mediated mRNA decay (NMD) pathway. Upregulation of EGFL7 activates NOTCH signaling in lung cancer cells, which slows down the decrease of c-Myc caused by EGFR inhibition, thereby helping the survival of cancer cells. Our data, taken together, demonstrate that EGFL7 is a driver gene for resistance to EGFR kinase inhibition, and suggest that targeting EGFL7/NOTCH signaling may improve the clinical benefits of EGFR inhibitors in patients with EGFR mutant tumors.
Collapse
Affiliation(s)
- Yubo Wang
- grid.452438.c0000 0004 1760 8119Department of Respiratory Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 Shaanxi PR China ,grid.452438.c0000 0004 1760 8119Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 PR China
| | - Pu Chen
- grid.452438.c0000 0004 1760 8119Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 PR China ,grid.452438.c0000 0004 1760 8119Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 Shaanxi PR China
| | - Man Zhao
- grid.452438.c0000 0004 1760 8119Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 PR China ,grid.452438.c0000 0004 1760 8119Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 Shaanxi PR China
| | - Hongxin Cao
- grid.452438.c0000 0004 1760 8119Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 PR China ,grid.452438.c0000 0004 1760 8119Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 Shaanxi PR China
| | - Yuelei Zhao
- grid.452438.c0000 0004 1760 8119Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 PR China ,grid.452438.c0000 0004 1760 8119Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 Shaanxi PR China
| | - Meiju Ji
- grid.452438.c0000 0004 1760 8119Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 Shaanxi PR China
| | - Peng Hou
- grid.452438.c0000 0004 1760 8119Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 PR China ,grid.452438.c0000 0004 1760 8119Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 Shaanxi PR China
| | - Mingwei Chen
- grid.452438.c0000 0004 1760 8119Department of Respiratory Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 Shaanxi PR China
| |
Collapse
|
7
|
Wu L, Zhou F, Xin W, Li L, Liu L, Yin X, Xu X, Wang Y, Hua Z. MAGP2 induces tumor progression by enhancing uPAR-mediated cell proliferation. Cell Signal 2021; 91:110214. [PMID: 34915136 DOI: 10.1016/j.cellsig.2021.110214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/03/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022]
Abstract
Microfibril-associated glycoprotein 2 (MAGP2) plays an important role in regulating cell signaling and acts as a biomarker to predict the prognostic effect of tumor therapy. However, research on MAGP2 mostly focuses on its extracellular signal transmission features, and its potential intracellular function is rarely reported. Here, we reported that intracellular MAGP2 increased the stability of urokinase-type plasminogen activator receptor (uPAR) in the cell by direct interaction which inhibits the lysosomal-mediated degradation of uPAR. Furthermore, with the detection of protein content changes and proteomics analysis, we found that highly expressed MAGP2 promoted the proliferation of tumor cells through uPAR-mediated p38-NF-ĸB signaling axis activation, enhancement of DNA damage repair and reduction of cell stagnation in the S phase of the cell cycle. In the nude mouse xenograft model of colorectal cancer, the upregulation of MAGP2 in tumor cells significantly promoted tumor progression, while the downregulation of uPAR significantly attenuated tumor progression. These studies elucidate the role of MAGP2 inside the cell and provide a new explanation for why patients with higher MAGP2 expression in tumors are associated with a worse prognosis. In addition, we also determined a mechanism for the stable existence of uPAR in the cell, providing information for the development of tumor drugs targeting uPAR.
Collapse
Affiliation(s)
- Leyang Wu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Feng Zhou
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Wenjie Xin
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Lin Li
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Lina Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Xingpeng Yin
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Xuebo Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Yao Wang
- Division of Critical Care and Surgery, St. George Hospital, University of New South Wales, Sydney, NSW 2217, Australia
| | - Zichun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China; Changzhou High-Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc., Changzhou 213164, Jiangsu, China; School of Biopharmacy, China Pharmaceutical University, Nanjing 210023, Jiangsu, China.
| |
Collapse
|
8
|
Dysregulated Notch Signaling in the Airway Epithelium of Children with Wheeze. J Pers Med 2021; 11:jpm11121323. [PMID: 34945795 PMCID: PMC8707470 DOI: 10.3390/jpm11121323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/16/2021] [Accepted: 11/29/2021] [Indexed: 11/17/2022] Open
Abstract
The airway epithelium of children with wheeze is characterized by defective repair that contributes to disease pathobiology. Dysregulation of developmental processes controlled by Notch has been identified in chronic asthma. However, its role in airway epithelial cells of young children with wheeze, particularly during repair, is yet to be determined. We hypothesized that Notch is dysregulated in primary airway epithelial cells (pAEC) of children with wheeze contributing to defective repair. This study investigated transcriptional and protein expression and function of Notch in pAEC isolated from children with and without wheeze. Primary AEC of children with and without wheeze were found to express all known Notch receptors and ligands, although pAEC from children with wheeze expressed significantly lower NOTCH2 (10-fold, p = 0.004) and higher JAG1 (3.5-fold, p = 0.002) mRNA levels. These dysregulations were maintained in vitro and cultures from children with wheeze displayed altered kinetics of both NOTCH2 and JAG1 expression during repair. Following Notch signaling inhibition, pAEC from children without wheeze failed to repair (wound closure rate of 76.9 ± 3.2%). Overexpression of NOTCH2 in pAEC from children with wheeze failed to rescue epithelial repair following wounding. This study illustrates the involvement of the Notch pathway in airway epithelial wound repair in health and disease, where its dysregulation may contribute to asthma development.
Collapse
|
9
|
Li H, Zhou W, Sun S, Zhang T, Zhang T, Huang H, Wang M. Microfibrillar-associated protein 5 regulates osteogenic differentiation by modulating the Wnt/β-catenin and AMPK signaling pathways. Mol Med 2021; 27:153. [PMID: 34865619 PMCID: PMC8647299 DOI: 10.1186/s10020-021-00413-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/20/2021] [Indexed: 12/18/2022] Open
Abstract
Background Dysfunctional osteogenesis of bone marrow mesenchymal stem cells (BMSCs) plays an important role in osteoporosis occurrence and development. However, the molecular mechanisms of osteogenic differentiation remain unclear. This study explored whether microfibrillar-associated protein 5 (MFAP5) regulated BMSCs osteogenic differentiation. Methods We used shRNA or cDNA to knock down or overexpress MFAP5 in C3H10 and MC3T3-E1 cells. AR-S- and ALP-staining were performed to quantify cellular osteogenic differentiation. The mRNA levels of the classical osteogenic differentiation biomarkers Runx2, Col1α1, and OCN were quantified by qRT-PCR. Finally, we employed Western blotting to measure the levels of Wnt/β-catenin and AMPK signaling proteins. Results At days 0, 3, 7, and 14 after osteogenic induction, AR-S- and ALP-staining was lighter in MFAP5 knockdown compared to control cells, as were the levels of Runx2, Col1α1 and OCN. During osteogenesis, the levels of β-catenin, p-GSK-3β, AMPK, and p-AMPK were upregulated, while that of GSK-3β was downregulated, indicating that Wnt/β-catenin and AMPK signaling were activated. The relevant molecules were expressed at lower levels in the knockdown than control group; the opposite was seen for overexpressing cell lines. Conclusions MFAP5 regulates osteogenesis via Wnt/β‑catenin- and AMPK-signaling; MFAP5 may serve as a therapeutic target in patients with osteoporosis. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-021-00413-0.
Collapse
Affiliation(s)
- Haoran Li
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Wuling Zhou
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Shiwei Sun
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Tianlong Zhang
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Tieqi Zhang
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Haitian Huang
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Minghai Wang
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Sottoriva K, Pajcini KV. Notch Signaling in the Bone Marrow Lymphopoietic Niche. Front Immunol 2021; 12:723055. [PMID: 34394130 PMCID: PMC8355626 DOI: 10.3389/fimmu.2021.723055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Lifelong mammalian hematopoiesis requires continuous generation of mature blood cells that originate from Hematopoietic Stem and Progenitor Cells (HSPCs) situated in the post-natal Bone Marrow (BM). The BM microenvironment is inherently complex and extensive studies have been devoted to identifying the niche that maintains HSPC homeostasis and supports hematopoietic potential. The Notch signaling pathway is required for the emergence of the definitive Hematopoietic Stem Cell (HSC) during embryonic development, but its role in BM HSC homeostasis is convoluted. Recent work has begun to explore novel roles for the Notch signaling pathway in downstream progenitor populations. In this review, we will focus an important role for Notch signaling in the establishment of a T cell primed sub-population of Common Lymphoid Progenitors (CLPs). Given that its activation mechanism relies primarily on cell-to-cell contact, Notch signaling is an ideal means to investigate and define a novel BM lymphopoietic niche. We will discuss how new genetic model systems indicate a pre-thymic, BM-specific role for Notch activation in early T cell development and what this means to the paradigm of lymphoid lineage commitment. Lastly, we will examine how leukemic T-cell acute lymphoblastic leukemia (T-ALL) blasts take advantage of Notch and downstream lymphoid signals in the pathological BM niche.
Collapse
Affiliation(s)
- Kilian Sottoriva
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, United States
| | - Kostandin V Pajcini
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, United States
| |
Collapse
|
11
|
Condorelli AG, El Hachem M, Zambruno G, Nystrom A, Candi E, Castiglia D. Notch-ing up knowledge on molecular mechanisms of skin fibrosis: focus on the multifaceted Notch signalling pathway. J Biomed Sci 2021; 28:36. [PMID: 33966637 PMCID: PMC8106838 DOI: 10.1186/s12929-021-00732-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
Fibrosis can be defined as an excessive and deregulated deposition of extracellular matrix proteins, causing loss of physiological architecture and dysfunction of different tissues and organs. In the skin, fibrosis represents the hallmark of several acquired (e.g. systemic sclerosis and hypertrophic scars) and inherited (i.e. dystrophic epidermolysis bullosa) diseases. A complex series of interactions among a variety of cellular types and a wide range of molecular players drive the fibrogenic process, often in a context-dependent manner. However, the pathogenetic mechanisms leading to skin fibrosis are not completely elucidated. In this scenario, an increasing body of evidence has recently disclosed the involvement of Notch signalling cascade in fibrosis of the skin and other organs. Despite its apparent simplicity, Notch represents one of the most multifaceted, strictly regulated and intricate pathways with still unknown features both in health and disease conditions. Starting from the most recent advances in Notch activation and regulation, this review focuses on the pro-fibrotic function of Notch pathway in fibroproliferative skin disorders describing molecular networks, interplay with other pro-fibrotic molecules and pathways, including the transforming growth factor-β1, and therapeutic strategies under development.
Collapse
Affiliation(s)
- Angelo Giuseppe Condorelli
- Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant' Onofrio 4, 00165, Rome, Italy.
| | - May El Hachem
- Dermatology Unit and Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant' Onofrio 4, 00165, Rome, Italy
| | - Giovanna Zambruno
- Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant' Onofrio 4, 00165, Rome, Italy
| | - Alexander Nystrom
- Department of Dermatology, Medical Faculty, Medical Center, University of Freiburg, Freiburg, Germany
| | - Eleonora Candi
- Department of Experimental Medicine, University of Rome "Tor Vergata", via Montpellier, 1, 00133, Rome, Italy.,IDI-IRCCS, via Monti di Creta 104, 00167, Rome, Italy
| | - Daniele Castiglia
- Laboratory of Molecular and Cell Biology, IDI-IRCCS, via Monti di Creta 104, 00167, Rome, Italy
| |
Collapse
|
12
|
Jung IH, Elenbaas JS, Alisio A, Santana K, Young EP, Kang CJ, Kachroo P, Lavine KJ, Razani B, Mecham RP, Stitziel NO. SVEP1 is a human coronary artery disease locus that promotes atherosclerosis. Sci Transl Med 2021; 13:eabe0357. [PMID: 33762433 PMCID: PMC8109261 DOI: 10.1126/scitranslmed.abe0357] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/09/2020] [Accepted: 02/18/2021] [Indexed: 01/07/2023]
Abstract
A low-frequency variant of sushi, von Willebrand factor type A, EGF, and pentraxin domain-containing protein 1 (SVEP1), an extracellular matrix protein, is associated with risk of coronary disease in humans independent of plasma lipids. Despite a robust statistical association, if and how SVEP1 might contribute to atherosclerosis remained unclear. Here, using Mendelian randomization and complementary mouse models, we provide evidence that SVEP1 promotes atherosclerosis in humans and mice and is expressed by vascular smooth muscle cells (VSMCs) within the atherosclerotic plaque. VSMCs also interact with SVEP1, causing proliferation and dysregulation of key differentiation pathways, including integrin and Notch signaling. Fibroblast growth factor receptor transcription increases in VSMCs interacting with SVEP1 and is further increased by the coronary disease-associated SVEP1 variant p.D2702G. These effects ultimately drive inflammation and promote atherosclerosis. Together, our results suggest that VSMC-derived SVEP1 is a proatherogenic factor and support the concept that pharmacological inhibition of SVEP1 should protect against atherosclerosis in humans.
Collapse
Affiliation(s)
- In-Hyuk Jung
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jared S Elenbaas
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Arturo Alisio
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Katherine Santana
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Erica P Young
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO 63108, USA
| | - Chul Joo Kang
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO 63108, USA
| | - Puja Kachroo
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Babak Razani
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
- John Cochran VA Medical Center, Saint Louis, MO 63106, USA
| | - Robert P Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Nathan O Stitziel
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA.
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO 63108, USA
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, USA
| |
Collapse
|
13
|
Stassen OMJA, Ristori T, Sahlgren CM. Notch in mechanotransduction - from molecular mechanosensitivity to tissue mechanostasis. J Cell Sci 2020; 133:133/24/jcs250738. [PMID: 33443070 DOI: 10.1242/jcs.250738] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tissue development and homeostasis are controlled by mechanical cues. Perturbation of the mechanical equilibrium triggers restoration of mechanostasis through changes in cell behavior, while defects in these restorative mechanisms lead to mechanopathologies, for example, osteoporosis, myopathies, fibrosis or cardiovascular disease. Therefore, sensing mechanical cues and integrating them with the biomolecular cell fate machinery is essential for the maintenance of health. The Notch signaling pathway regulates cell and tissue fate in nearly all tissues. Notch activation is directly and indirectly mechanosensitive, and regulation of Notch signaling, and consequently cell fate, is integral to the cellular response to mechanical cues. Fully understanding the dynamic relationship between molecular signaling, tissue mechanics and tissue remodeling is challenging. To address this challenge, engineered microtissues and computational models play an increasingly large role. In this Review, we propose that Notch takes on the role of a 'mechanostat', maintaining the mechanical equilibrium of tissues. We discuss the reciprocal role of Notch in the regulation of tissue mechanics, with an emphasis on cardiovascular tissues, and the potential of computational and engineering approaches to unravel the complex dynamic relationship between mechanics and signaling in the maintenance of cell and tissue mechanostasis.
Collapse
Affiliation(s)
- Oscar M J A Stassen
- Faculty of Science and Engineering, Biosciences, Åbo Akademi University, 20500 Turku, Finland.,Turku Bioscience Centre, Åbo Akademi University and University of Turku, 20520 Turku, Finland.,Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.,Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Cecilia M Sahlgren
- Faculty of Science and Engineering, Biosciences, Åbo Akademi University, 20500 Turku, Finland .,Turku Bioscience Centre, Åbo Akademi University and University of Turku, 20520 Turku, Finland.,Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
14
|
Zhao L, Xu L, Hemmerich A, Ferguson NL, Guy CD, McCall SJ, Cardona DM, Westerhoff M, Pai RK, Xiao SY, Liu B, Green CL, Hart J, Zhang X. Reduced MFAP5 expression in stroma of gallbladder adenocarcinoma and its potential diagnostic utility. Virchows Arch 2020; 478:427-434. [PMID: 32895766 DOI: 10.1007/s00428-020-02925-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 08/04/2020] [Accepted: 09/02/2020] [Indexed: 11/28/2022]
Abstract
The diagnosis of invasive adenocarcinoma of the gallbladder can sometimes be challenging. The presence of true desmoplastic reaction facilitates the diagnosis of invasion. However, desmoplasia-like changes can be observed in benign gallbladder conditions, and recognition of desmoplasia may be challenging based on morphology. In this study, we tested the expression pattern of microfibril-associated protein 5 (MFAP5), a promising immunohistochemical marker for desmoplasia, in benign gallbladders with desmoplasia-like reaction and gallbladders with invasive adenocarcinoma. We also evaluated the diagnostic utility of MFAP5 in challenging cases with an interobserver agreement study. The results showed that all benign cases retained intact/positive MFAP5 staining pattern in periglandular connective tissue, whereas 79.3% (23 out of 29) of cases of adenocarcinomas demonstrated diffuse and complete loss of MFAP5 staining in the tumor stroma. Interobserver agreement was improved by 2.66 times when images of MFAP5 immunohistochemistry were provided. In conclusion, MFAP5 expression is downregulated in the desmoplastic stroma of gallbladder adenocarcinoma and may provide a useful diagnostic marker in difficult cases.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Liyan Xu
- Department of Pathology, St. Luke's University Health Network, Bethlehem, PA, USA
| | - Amanda Hemmerich
- Department of Pathology, Foundation Medicine, Inc., Morrisville, NC, USA
| | - N Lynn Ferguson
- Department of Pathology, Foundation Medicine, Inc., Morrisville, NC, USA
| | - Cynthia D Guy
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Shannon J McCall
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Diana M Cardona
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Maria Westerhoff
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Rish K Pai
- Department of Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Shu-Yuan Xiao
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Beiyu Liu
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Cynthia L Green
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - John Hart
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Xuefeng Zhang
- Department of Pathology, Duke University Medical Center, Durham, NC, USA. .,Department of Anatomic Pathology, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
15
|
Zhou Z, Cui D, Sun MH, Huang JL, Deng Z, Han BM, Sun XW, Xia SJ, Sun F, Shi F. CAFs-derived MFAP5 promotes bladder cancer malignant behavior through NOTCH2/HEY1 signaling. FASEB J 2020; 34:7970-7988. [PMID: 32293074 DOI: 10.1096/fj.201902659r] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/10/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022]
Abstract
Cancer-associated fibroblasts (CAFs) are an important component of the tumor microenvironment and contribute to tumor cell proliferation and metastasis. Microfibrillar-associated protein 5 (MFAP5), a component of elastic microfibers and an oncogenic protein in several types of tumors, is secreted by CAFs. However, the role of MFAP5 in the bladder cancer remains unclear. Here, we report that MFAP5 is upregulated in bladder cancer and is associated with poor patient survival. Downregulation of MFAP5 in CAFs led to an impairment in proliferation and invasion of bladder cancer cells. Luciferase reporter assays and electrophoretic mobility shift assays (EMSA) showed QKI directly downregulates MFAP5 in CAFs. In addition, CAFs-derived MFAP5 led to an activation of the NOTCH2/HEY1 signaling pathway through direct interaction with the NOTCH2 receptor, thereby stimulating the N2ICD release. RNA-sequencing revealed that MFAP5-mediated PI3K-AKT signaling activated the DLL4/NOTCH2 pathway axis in bladder cancer. Moreover, downregulation of NOTCH2 by short hairpin RNA or the inactivating anti-body NRR2Mab was able to reverse the adverse effects of MFAP5 stimulation in vitro and in vivo. Together, these results demonstrate CAFs-derived MFAP5 promotes the bladder cancer proliferation and metastasis and provides new insight for targeting CAFs as novel diagnostic and therapeutic strategy.
Collapse
Affiliation(s)
- Zheng Zhou
- Department of Urology, Shanghai General Hospital, Nanjing Medical University, Shanghai, China
| | - Di Cui
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Meng-Hao Sun
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Lang Huang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Deng
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bang-Min Han
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Wen Sun
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Shu-Jie Xia
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Sun
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Fei Shi
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
16
|
Feifei W, Hui G, Ruiqiang Z, Qunxiang J, Yu'an X. MAGP2, a Component of Extracellular Matrix, Is Upregulated in Colorectal Cancer and Negatively Modulated by miR-200b-3p. Technol Cancer Res Treat 2020; 18:1533033819870777. [PMID: 31426719 PMCID: PMC6702771 DOI: 10.1177/1533033819870777] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background: Colorectal cancer is one of the leading causes of cancer-related death worldwide, but its mechanism has not been clarified clearly. Microfibrial-associated glycoprotein 2 is mainly located in extracellular matrix, and its role in colorectal cancer is obscure. Methods: Immunohistochemical staining and quantitative real-time polymerase chain reaction were used to compare the expression level of microfibrial-associated glycoprotein 2 in colorectal cancer tissues and adjacent tissues. Western blot was used to detect the expression of microfibrial-associated glycoprotein 2 in colorectal cancer cell lines and normal colonic epithelium cell line. Kaplan-Meier analysis and χ2 test were applied to evaluate the potential of microfibrial-associated glycoprotein 2 to function as cancer biomarker. Lentiviral transduction was used to induce microfibrial-associated glycoprotein 2 overexpression in HCT116 cells and NCM460 cells, followed by detecting cell proliferation, migration, and invasion. Quantitative real-time polymerase chain reaction was used to investigate the changes in downstream genes after microfibrial-associated glycoprotein 2 overexpression. Luciferase assay was conducted to validate whether miR-200b-3p can directly target microfibrial-associated glycoprotein 2. Results: We validated that microfibrial-associated glycoprotein 2 was upregulated in colorectal cancer samples and cells. We also demonstrated its upregulation was associated with several clinicopathologic features such as Dukes stage (P = .048), differentiation status (P = .034), and local lymphatic metastasis (P = .036) of patients with colorectal cancer, and its high expression indicated shorter overall survival of the patients. Microfibrial-associated glycoprotein 2 overexpression remarkably promoted cell proliferation and metastasis via regulating the downstream genes of Notch, including hes family bHLH transcription factor 1 (HES1), Slug, Snail, matrix metalloproteinase 2, matrix metalloproteinase 9, and Kruppel-like factor 4. We also identified miR-200b-3p as a posttranscriptional regulator of microfibrial-associated glycoprotein 2, which partly explain the high expression mechanism of microfibrial-associated glycoprotein 2 in cancer tissues. Conclusion: Microfibrial-associated glycoprotein 2, negatively modulated by miR-200b-3p, is an oncogene of colorectal cancer associated with patients’ prognosis. It may function as a potential biomarker and therapeutic target for colorectal cancer.
Collapse
Affiliation(s)
- Wei Feifei
- 1 Department of Experimental Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Guo Hui
- 1 Department of Experimental Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Zhao Ruiqiang
- 2 Department of Biochemistry and Molecular Biology, Guangxi Medical University, Nanning, China
| | - Jiang Qunxiang
- 1 Department of Experimental Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Xie Yu'an
- 1 Department of Experimental Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
17
|
Xu Q, Chang H, Tian X, Lou C, Ma H, Yang X. Hypoxia-induced MFAP5 Promotes Tumor Migration and Invasion via AKT Pathway in Head and Neck Squamous Cell Carcinoma. J Cancer 2020; 11:1596-1605. [PMID: 32047565 PMCID: PMC6995394 DOI: 10.7150/jca.38217] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 11/20/2019] [Indexed: 02/06/2023] Open
Abstract
Objective: Microfibrillar-associated protein 5 (MFAP5) is highly expressed in many types of cancers. Our previous study has observed that overexpression of MFAP5 was correlated with lymph nodes metastasis and poor prognosis in head and neck squamous cell carcinoma (HNSCC), but the underlying mechanism is poorly understood. Materials and methods: The MFAP5 expression is detected under hypoxia condition. HNSCC cell lines are transfected with MFAP5-expressing lentivirus vector to establish stable overexpression model. Wound-healing, migration and invasion assay are used to determine the effect of MFAP5 on HNSCC and metastasis-related proteins are examined by Western blot. In vivo lung metastasis assays are conducted by the tail vein injection. In addition, immunohistochemistry is applied to analyze the correlation of MFAP5, hypoxia-induced factor-1 α (HIF-1α), and vimentin in 84 HNSCC patients' tissue samples. Results: Firstly, MFAP5 expression can be markedly induced under hypoxia condition in HNSCC cell lines. Cell lines with MFAP5 overexpression has a significant higher ability of migration and invasion. In addition, in vivo assay observes that overexpression of MFAP5 can promote tumor lung metastasis. Furthermore, MFAP5 facilitates this process by activating epithelial-mesenchymal transition (EMT) program via AKT pathway in HNSCC cell lines. The pro-metastatic effect of MFAP5 can be reversed by MK2206, an AKT phosphorylation inhibitor. Lastly, the positive correlation among HIF-1α, MFAP5 and vimentin from tissue samples and TCGA dataset are also observed in HNSCC. Conclusion: Our study demonstrates MFAP5 plays a critical role in hypoxia-induced EMT program via AKT pathway in HNSCC, which would be a very promising therapeutic target.
Collapse
Affiliation(s)
- Qiaoshi Xu
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, No 639, Zhizaoju Rd, Shanghai 200011, China.,National Clinical Research Center for Oral Diseases, Shanghai 200011, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Hanyue Chang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, No 639, Zhizaoju Rd, Shanghai 200011, China.,National Clinical Research Center for Oral Diseases, Shanghai 200011, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China.,Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510140, China
| | - Xuerui Tian
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, No 639, Zhizaoju Rd, Shanghai 200011, China.,National Clinical Research Center for Oral Diseases, Shanghai 200011, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Chao Lou
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, No 639, Zhizaoju Rd, Shanghai 200011, China.,National Clinical Research Center for Oral Diseases, Shanghai 200011, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Hailong Ma
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, No 639, Zhizaoju Rd, Shanghai 200011, China.,National Clinical Research Center for Oral Diseases, Shanghai 200011, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Xi Yang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, No 639, Zhizaoju Rd, Shanghai 200011, China.,National Clinical Research Center for Oral Diseases, Shanghai 200011, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China
| |
Collapse
|
18
|
Immunohistochemical Expression of Microfibrillar-associated Protein 5 (MFAP5) in Invasive Breast Carcinoma of No Special Type. Appl Immunohistochem Mol Morphol 2019; 27:649-657. [DOI: 10.1097/pai.0000000000000686] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
19
|
Zhang Y, Xie ZY, Guo XT, Xiao XH, Xiong LX. Notch and breast cancer metastasis: Current knowledge, new sights and targeted therapy. Oncol Lett 2019; 18:2743-2755. [PMID: 31452752 PMCID: PMC6704289 DOI: 10.3892/ol.2019.10653] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 06/21/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most common type of invasive cancer in females and metastasis is one of the major causes of breast cancer-associated mortality. Following detachment from the primary site, disseminated tumor cells (DTCs) enter the bloodstream and establish secondary colonies during the metastatic process. An increasing amount of studies have elucidated the importance of Notch signaling in breast cancer metastasis; therefore, the present review focuses on the mechanisms by which Notch contributes to the occurrence of breast cancer DTCs, increases their motility, establishes interactions with the tumor microenvironment, protects DTCs from host surveillance and finally facilitates secondary colonization. Identification of the underlying mechanisms of Notch-associated breast cancer metastasis will provide additional insights that may contribute towards the development of novel Notch-targeted therapeutic strategies, which may aid in reducing metastasis, culminating in an improved patient prognosis.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pathophysiology, Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zi-Yan Xie
- Department of Pathophysiology, Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xuan-Tong Guo
- Department of Pathophysiology, Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xing-Hua Xiao
- Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Li-Xia Xiong
- Department of Pathophysiology, Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
20
|
Loss of microfibril-associated protein 5 (MFAP5) expression in colon cancer stroma. Virchows Arch 2019; 476:383-390. [DOI: 10.1007/s00428-019-02649-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/31/2019] [Accepted: 08/13/2019] [Indexed: 11/26/2022]
|
21
|
Chen Z, Yan X, Li K, Ling Y, Kang H. Stromal fibroblast-derived MFAP5 promotes the invasion and migration of breast cancer cells via Notch1/slug signaling. Clin Transl Oncol 2019; 22:522-531. [PMID: 31190277 DOI: 10.1007/s12094-019-02156-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The tumor microenvironment (TME) regulates tumor progression, and cancer-associated fibroblasts (CAFs) are the primary stromal components of the TME, with the potential to drive tumor metastasis via the secretion of paracrine factors, but the specific mechanisms driving this process have not been defined. METHODS Proteins secreted from CAFs and normal fibroblasts (NFs) were analyzed via proteomic analysis (fold change > 2, p < 0.05) to identify tumor-promoting proteins secreted by CAFs. RESULTS Proteomic analysis revealed that microfibrillar-associated protein 5 (MFAP5) is preferentially expressed and secreted by CAFs relative to NFs, which was confirmed by Western blotting and RT-qPCR. Transwell and wound healing assays confirmed that MFAP5 is secreted by CAFs, and drives the invasion and migration of MCF7 breast cancer cells. We further found that in MCF7 cells MFAP5 promoted epithelial-mesenchymal transition, activating Notch1 signaling and consequently upregulating NICD1 and slug. When Notch1 was knocked down in MCF7 cells, the ability of MFAP5 to promote invasion and migration decreased. CONCLUSION CAFs promote cancer cells invasion and migration via MFAP5 secretion and activation of the Notch1/slug signaling. These data highlight this pathway as a therapeutic target to disrupt tumor progression through the interference of CAF-tumor crosstalk.
Collapse
Affiliation(s)
- Z Chen
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - X Yan
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - K Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Y Ling
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - H Kang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
22
|
LaFoya B, Munroe JA, Albig AR. A comparison of resveratrol and other polyphenolic compounds on Notch activation and endothelial cell activity. PLoS One 2019; 14:e0210607. [PMID: 30653610 PMCID: PMC6336259 DOI: 10.1371/journal.pone.0210607] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 12/30/2018] [Indexed: 01/01/2023] Open
Abstract
Resveratrol is a polyphenolic compound produced by plants which makes its way into the human diet through plant-based foods. It has been shown to provide many health benefits, helping to ward of age-related diseases and promoting cardiovascular health. Additionally, resveratrol is a potent activator of the Notch signaling pathway. While resveratrol receives the most attention as a polyphenolic nutraceutical, other compounds with similar structures may be more potent regulators of specific cellular processes. Here, we compare resveratrol, apigenin, chrysin, genistein, luteolin, myricetin, piceatannol, pterostilbene, and quercetin for their ability to regulate Notch signaling. In addition, we compare the ability of these polyphenolic compounds to regulate endothelial cell viability, proliferation, and migration. Out of these compounds we found that resveratrol is the best activator of Notch signaling, however, other similar compounds are also capable of stimulating Notch. We also discovered that several of these polyphenols were able to inhibit endothelial cell proliferation. Finally, we found that many of these polyphenols are potent inhibitors of endothelial migration during wound healing assays. These findings provide the first side-by-side comparison of the regulation of Notch signaling, and endothelial cell proliferation and migration, by nine polyphenolic compounds.
Collapse
Affiliation(s)
- Bryce LaFoya
- Biomolecular Sciences PhD Program, Boise State University, Boise, Idaho, United States of America
| | - Jordan A. Munroe
- Department of Biological Sciences, Boise State University, Boise, Idaho, United States of America
| | - Allan R. Albig
- Biomolecular Sciences PhD Program, Boise State University, Boise, Idaho, United States of America
- Department of Biological Sciences, Boise State University, Boise, Idaho, United States of America
- * E-mail:
| |
Collapse
|
23
|
Villain G, Lelievre E, Broekelmann T, Gayet O, Havet C, Werkmeister E, Mecham R, Dusetti N, Soncin F, Mattot V. MAGP
‐1 and fibronectin control
EGFL
7 functions by driving its deposition into distinct endothelial extracellular matrix locations. FEBS J 2018; 285:4394-4412. [DOI: 10.1111/febs.14680] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/31/2018] [Accepted: 10/16/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Gaëlle Villain
- CNRS Institut Pasteur de Lille UMR 8161 – M3T – Mechanisms of Tumorigenesis and Target Therapies Univ. Lille France
| | - Etienne Lelievre
- CNRS Institut Pasteur de Lille UMR 8161 – M3T – Mechanisms of Tumorigenesis and Target Therapies Univ. Lille France
| | - Tom Broekelmann
- Department of Cell Biology and Physiology Washington University School of Medicine St. Louis MO USA
| | - Odile Gayet
- Centre de Recherche en Cancérologie de Marseille (CRCM) INSERM U1068 CNRS UMR 7258 Aix‐Marseille Université and Institut Paoli‐Calmettes, Parc Scientifique et Technologique de Luminy France
| | - Chantal Havet
- CNRS Institut Pasteur de Lille UMR 8161 – M3T – Mechanisms of Tumorigenesis and Target Therapies Univ. Lille France
| | - Elisabeth Werkmeister
- Cellular Microbiology and Physics of Infection Group – Center for Infection and Immunity of Lille CNRS UMR8204 Inserm U1019 CHU Lille Institut Pasteur de Lille Univ. Lille. France
| | - Robert Mecham
- Department of Cell Biology and Physiology Washington University School of Medicine St. Louis MO USA
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM) INSERM U1068 CNRS UMR 7258 Aix‐Marseille Université and Institut Paoli‐Calmettes, Parc Scientifique et Technologique de Luminy France
| | - Fabrice Soncin
- CNRS Institut Pasteur de Lille UMR 8161 – M3T – Mechanisms of Tumorigenesis and Target Therapies Univ. Lille France
| | - Virginie Mattot
- CNRS Institut Pasteur de Lille UMR 8161 – M3T – Mechanisms of Tumorigenesis and Target Therapies Univ. Lille France
| |
Collapse
|
24
|
Abstract
Notch signaling is a form of intercellular communication which plays pivotal roles at various stages in development and disease. Previous findings have hinted that integrins and extracellular matrix may regulate Notch signaling, although a mechanistic basis for this interaction had not been identified. Here, we reveal that the regulation of Notch by integrins and extracellular matrix is carried out by Src family kinases (SFKs) working downstream of integrins. We identify a physical interaction between the SFK member, c-Src, and the Notch intracellular domain (NICD) that is enhanced by β3 integrin and the integrin binding ECM protein, MAGP2. Our results demonstrate that c-Src directly phosphorylates the NICD at specific tyrosine residues and that mutation of these phosphorylation sites increases Notch responsive transcriptional activity. Furthermore, we also find that phosphorylation of the NICD by SFKs attenuates Notch mediated transcription by decreasing recruitment of MAML to the Notch co-transcriptional complex. Finally, we also find that SFK activity decreases NICD half-life. Collectively, our results provide important mechanistic data that underlie the emerging role of Notch as a general sensor and responder to extracellular signals.
Collapse
|
25
|
Høgdall D, Lewinska M, Andersen JB. Desmoplastic Tumor Microenvironment and Immunotherapy in Cholangiocarcinoma. Trends Cancer 2018; 4:239-255. [PMID: 29506673 DOI: 10.1016/j.trecan.2018.01.007] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/15/2018] [Accepted: 01/25/2018] [Indexed: 02/07/2023]
Abstract
Cholangiocarcinoma (CCA) is a dismal disease which often is diagnosed at a late stage where the tumor is locally advanced, metastatic, and, as a result, is associated with low resectability. The heterogeneity of this cancer type is a major reason why the majority of patients fail to respond to therapy, and surgery remains their only curative option. Among patients who undergo surgical intervention, such tumors typically recur in 50% of cases within 1year. Thus, CCA is among the most aggressive and chemoresistant malignancies. CCA is characterized by marked tumor reactive stroma, a fibrogenic connective tissue which surrounds and infiltrates the tumor epithelium. This desmoplastic environment presents a clinical challenge, limiting drug delivery and supporting the growth of the tumor mass. In this review we attempt to highlight key pathways involved in cell to cell communication between the tumor epithelium and stroma, the immune components, and opportunities for novel strategies to improve patient outcome.
Collapse
Affiliation(s)
- Dan Høgdall
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark; Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark; These authors contributed equally
| | - Monika Lewinska
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark; These authors contributed equally
| | - Jesper B Andersen
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark.
| |
Collapse
|
26
|
Notch signaling pathway networks in cancer metastasis: a new target for cancer therapy. Med Oncol 2017; 34:180. [DOI: 10.1007/s12032-017-1039-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/13/2017] [Indexed: 12/19/2022]
|
27
|
LaFoya B, Munroe JA, Mia MM, Detweiler MA, Crow JJ, Wood T, Roth S, Sharma B, Albig AR. Notch: A multi-functional integrating system of microenvironmental signals. Dev Biol 2016; 418:227-41. [PMID: 27565024 PMCID: PMC5144577 DOI: 10.1016/j.ydbio.2016.08.023] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/15/2016] [Accepted: 08/19/2016] [Indexed: 12/20/2022]
Abstract
The Notch signaling cascade is an evolutionarily ancient system that allows cells to interact with their microenvironmental neighbors through direct cell-cell interactions, thereby directing a variety of developmental processes. Recent research is discovering that Notch signaling is also responsive to a broad variety of stimuli beyond cell-cell interactions, including: ECM composition, crosstalk with other signaling systems, shear stress, hypoxia, and hyperglycemia. Given this emerging understanding of Notch responsiveness to microenvironmental conditions, it appears that the classical view of Notch as a mechanism enabling cell-cell interactions, is only a part of a broader function to integrate microenvironmental cues. In this review, we summarize and discuss published data supporting the idea that the full function of Notch signaling is to serve as an integrator of microenvironmental signals thus allowing cells to sense and respond to a multitude of conditions around them.
Collapse
Affiliation(s)
- Bryce LaFoya
- Biomolecular Sciences PhD Program, Boise State University, Boise, ID 83725, USA
| | - Jordan A Munroe
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | - Masum M Mia
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | - Michael A Detweiler
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | - Jacob J Crow
- Biomolecular Sciences PhD Program, Boise State University, Boise, ID 83725, USA
| | - Travis Wood
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | - Steven Roth
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | - Bikram Sharma
- Department of Biological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Allan R Albig
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA; Biomolecular Sciences PhD Program, Boise State University, Boise, ID 83725, USA.
| |
Collapse
|