1
|
Eissazadeh S, Fikrova P, Rathouska JU, Nemeckova I, Tripska K, Vasinova M, Havelek R, Mohammadi S, Igreja Sa IC, Theuer C, König M, Micuda S, Nachtigal P. Anti-Endoglin monoclonal antibody prevents the progression of liver sinusoidal endothelial inflammation and fibrosis in MASH. Life Sci 2025; 364:123428. [PMID: 39889923 DOI: 10.1016/j.lfs.2025.123428] [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: 11/11/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Liver sinusoidal endothelial inflammation/dysfunction and fibrosis are a crucial part of Metabolic Dysfunction Associated Steatohepatitis (MASH) development. TRC105 and M1043 are anti-endoglin (ENG) monoclonal antibodies that bind ENG. In this study, we hypothesized that treatment with anti-ENG antibodies would prevent the progression of LSECs inflammation and fibrosis in vivo and in vitro. MASH was induced in male C57BL/6 mice fed a choline-deficient L-amino acid-defined high-fat diet (CDAA-HFD) for 4 or 8 weeks. In the rescue study, mice were divided into three groups: a control group (chow diet), a MASH group (CDAA-HFD + IgG), and a rescue group (CDAA-HFD + M1043). Later, two groups received rat IgG1 (10 mg/kg) and M1043 (10 mg/kg). In in vitro experiments, inflammation was induced in human LSECs by ox-LDL (50 μg/mL) and treated with TRC105 (300 μg/mL). Liver sinusoidal endothelial inflammation/dysfunction in MASH animals was characterized by endothelial overexpression of ENG, VCAM-1, and ICAM-1 and reduced VE-cadherin and p-eNOS/eNOS expression. M1043 treatment prevented the overexpression of ENG, VCAM-1, and ICAM-1, the progression of liver fibrosis, and the increase of liver-to-body weight ratio. In vitro experiments with TRC105 confirmed the prevention of LSECs inflammation development by reduced ENG and VCAM-1 expression, as well as decreased THP-1 monocytic cell adhesion in ox-LDL activated LSECs. In conclusion, we demonstrate that anti-ENG antibody treatment can prevent LSECs inflammation and fibrosis progression in a MASH animal model and LSECs inflammation in vitro. Thus, we propose directly targeted ENG may represent a promising pharmacological approach for addressing LSECs inflammation and liver fibrosis.
Collapse
Affiliation(s)
- Samira Eissazadeh
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Petra Fikrova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Jana Urbankova Rathouska
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Ivana Nemeckova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Katarina Tripska
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Martina Vasinova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Radim Havelek
- Department of Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - SeyedehNiloufar Mohammadi
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Ivone Cristina Igreja Sa
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic; Department of Clinical Microbiology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Charles Theuer
- Tracon Pharmaceuticals, Inc., San Diego, CA, United States
| | - Matthias König
- Institute for Theoretical Biology, Institute for Biology, Systems Medicine of the Liver, Humboldt University Berlin, Germany
| | - Stanislav Micuda
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic.
| |
Collapse
|
2
|
Tang W, Gu Z, Guo J, Lin M, Tao H, Jia D, Jia P. Activins and Inhibins in Cardiovascular Pathophysiology. Biomolecules 2024; 14:1462. [PMID: 39595638 PMCID: PMC11592067 DOI: 10.3390/biom14111462] [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: 09/25/2024] [Revised: 10/30/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Activins and inhibins, members of the transforming growth factor β (TGFβ) superfamily, were initially recognized for their opposing effects on the secretion of follicle-stimulating hormone. Subsequent research has demonstrated their broader biological roles across various tissue types. Primarily, activins and inhibins function through the classical TGFβ SMAD signaling pathway, but studies suggest that they also act through other pathways, with their specific signaling being complex and context-dependent. Recent research has identified significant roles for activins and inhibins in the cardiovascular system. Their actions in other systems and their signaling pathways show strong correlations with the development and progression of cardiovascular diseases, indicating potential broader roles in the cardiovascular system. This review summarizes the progress in research on the biological functions and mechanisms of activins and inhibins and their signaling pathways in cardiovascular diseases, offering new insights for the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Dalin Jia
- Department of Cardiology, The First Hospital of China Medical University, Shenyang 110001, China; (W.T.); (Z.G.); (J.G.); (M.L.); (H.T.)
| | - Pengyu Jia
- Department of Cardiology, The First Hospital of China Medical University, Shenyang 110001, China; (W.T.); (Z.G.); (J.G.); (M.L.); (H.T.)
| |
Collapse
|
3
|
Gerrits T, Dijkstra KL, Bruijn JA, Scharpfenecker M, Bijkerk R, Baelde HJ. Antisense oligonucleotide-mediated terminal intron retention of endoglin: A potential strategy to inhibit renal interstitial fibrosis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167186. [PMID: 38642778 DOI: 10.1016/j.bbadis.2024.167186] [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: 01/04/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
TGF-β is considered an important cytokine in the development of interstitial fibrosis in chronic kidney disease. The TGF-β co-receptor endoglin (ENG) tends to be upregulated in kidney fibrosis. ENG has two membrane bound isoforms generated via alternative splicing. Long-ENG was shown to enhance the extent of renal fibrosis in an unilateral ureteral obstruction mouse model, while short-ENG inhibited renal fibrosis. Here we aimed to achieve terminal intron retention of endoglin using antisense-oligo nucleotides (ASOs), thereby shifting the ratio towards short-ENG to inhibit the TGF-β1-mediated pro-fibrotic response. We isolated mRNA from kidney biopsies of patients with chronic allograft disease (CAD) (n = 12) and measured total ENG and short-ENG mRNA levels. ENG mRNA was upregulated 2.3 fold (p < 0.05) in kidneys of CAD patients compared to controls, while the percentage short-ENG of the total ENG mRNA was significantly lower (1.8 fold; p < 0.05). Transfection of ASOs that target splicing regulatory sites of ENG into TK173 fibroblasts led to higher levels of short-ENG (2 fold; p < 0.05). In addition, we stimulated these cells with TGF-β1 and measured a decrease in upregulation of ACTA2, COL1A1 and FN1 mRNA levels, and protein expression of αSMA, collagen type I, and fibronectin. These results show a potential for ENG ASOs as a therapy to reduce interstitial fibrosis in CKD.
Collapse
Affiliation(s)
- Tessa Gerrits
- Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands.
| | - Kyra L Dijkstra
- Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Jan Anthonie Bruijn
- Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Marion Scharpfenecker
- Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Roel Bijkerk
- Department of Nephrology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Hans J Baelde
- Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| |
Collapse
|
4
|
Hong YK, Lin YC, Cheng TL, Lai CH, Chang YH, Huang YL, Hung CY, Wu CH, Hung KS, Ku YC, Ho YT, Tang MJ, Lin SW, Shi GY, McGrath JA, Wu HL, Hsu CK. TEM1/endosialin/CD248 promotes pathologic scarring and TGF-β activity through its receptor stability in dermal fibroblasts. J Biomed Sci 2024; 31:12. [PMID: 38254097 PMCID: PMC10804696 DOI: 10.1186/s12929-024-01001-0] [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: 07/06/2023] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Pathologic scars, including keloids and hypertrophic scars, represent a common form of exaggerated cutaneous scarring that is difficult to prevent or treat effectively. Additionally, the pathobiology of pathologic scars remains poorly understood. We aim at investigating the impact of TEM1 (also known as endosialin or CD248), which is a glycosylated type I transmembrane protein, on development of pathologic scars. METHODS To investigate the expression of TEM1, we utilized immunofluorescence staining, Western blotting, and single-cell RNA-sequencing (scRNA-seq) techniques. We conducted in vitro cell culture experiments and an in vivo stretch-induced scar mouse model to study the involvement of TEM1 in TGF-β-mediated responses in pathologic scars. RESULTS The levels of the protein TEM1 are elevated in both hypertrophic scars and keloids in comparison to normal skin. A re-analysis of scRNA-seq datasets reveals that a major profibrotic subpopulation of keloid and hypertrophic scar fibroblasts greatly expresses TEM1, with expression increasing during fibroblast activation. TEM1 promotes activation, proliferation, and ECM production in human dermal fibroblasts by enhancing TGF-β1 signaling through binding with and stabilizing TGF-β receptors. Global deletion of Tem1 markedly reduces the amount of ECM synthesis and inflammation in a scar in a mouse model of stretch-induced pathologic scarring. The intralesional administration of ontuxizumab, a humanized IgG monoclonal antibody targeting TEM1, significantly decreased both the size and collagen density of keloids. CONCLUSIONS Our data indicate that TEM1 plays a role in pathologic scarring, with its synergistic effect on the TGF-β signaling contributing to dermal fibroblast activation. Targeting TEM1 may represent a novel therapeutic approach in reducing the morbidity of pathologic scars.
Collapse
Affiliation(s)
- Yi-Kai Hong
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Chen Lin
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan
| | - Tsung-Lin Cheng
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- College of Professional Studies, National Pingtung University of Science Technology, Pingtung, Taiwan
| | - Chao-Han Lai
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Han Chang
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Lun Huang
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Yi Hung
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chen-Han Wu
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan
| | - Kuo-Shu Hung
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Chu Ku
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen-Ting Ho
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Ming-Jer Tang
- International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shu-Wha Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University Hospital, Taipei, Taiwan
| | - Guey-Yueh Shi
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - John A McGrath
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, UK
| | - Hua-Lin Wu
- International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan.
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Chao-Kai Hsu
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan.
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
5
|
Sharma S, Ehrlich M, Zhang M, Blobe GC, Henis YI. NRP1 interacts with endoglin and VEGFR2 to modulate VEGF signaling and endothelial cell sprouting. Commun Biol 2024; 7:112. [PMID: 38242992 PMCID: PMC10799020 DOI: 10.1038/s42003-024-05798-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 01/09/2024] [Indexed: 01/21/2024] Open
Abstract
Endothelial cells express neuropilin 1 (NRP1), endoglin (ENG) and vascular endothelial growth factor receptor 2 (VEGFR2), which regulate VEGF-A-mediated vascular development and angiogenesis. However, the link between complex formation among these receptors with VEGF-A-induced signaling and biology is yet unclear. Here, we quantify surface receptor interactions by IgG-mediated immobilization of one receptor, and fluorescence recovery after photobleaching (FRAP) measurements of the mobility of another coexpressed receptor. We observe stable ENG/NRP1, ENG/VEGFR2, and NRP1/VEGFR2 complexes, which are enhanced by VEGF-A. ENG augments NRP1/VEGFR2 interactions, suggesting formation of tripartite complexes bridged by ENG. Effects on signaling are measured in murine embryonic endothelial cells expressing (MEEC+/+) or lacking (MEEC-/-) ENG, along with NRP1 and/or ENG overexpression or knockdown. We find that optimal VEGF-A-mediated phosphorylation of VEGFR2 and Erk1/2 requires ENG and NRP1. ENG or NRP1 increase VEGF-A-induced sprouting, becoming optimal in cells expressing all three receptors, and both processes are inhibited by a MEK1/2 inhibitor. We propose a model where the maximal potency of VEGF-A involves a tripartite complex where ENG bridges VEGFR2 and NRP1, providing an attractive therapeutic target for modulation of VEGF-A signaling and biological responses.
Collapse
Affiliation(s)
- Swati Sharma
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Manqi Zhang
- Department of Medicine, Duke University Medical Center, Durham, NC, 27708, USA
| | - Gerard C Blobe
- Department of Medicine, Duke University Medical Center, Durham, NC, 27708, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Yoav I Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel.
| |
Collapse
|
6
|
Sun YL, Hennessey EE, Heins H, Yang P, Villacorta-Martin C, Kwan J, Gopalan K, James M, Emili A, Cole FS, Wambach JA, Kotton DN. Human pluripotent stem cell modeling of alveolar type 2 cell dysfunction caused by ABCA3 mutations. J Clin Invest 2024; 134:e164274. [PMID: 38226623 PMCID: PMC10786693 DOI: 10.1172/jci164274] [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: 08/08/2022] [Accepted: 11/14/2023] [Indexed: 01/17/2024] Open
Abstract
Mutations in ATP-binding cassette A3 (ABCA3), a phospholipid transporter critical for surfactant homeostasis in pulmonary alveolar type II epithelial cells (AEC2s), are the most common genetic causes of childhood interstitial lung disease (chILD). Treatments for patients with pathological variants of ABCA3 mutations are limited, in part due to a lack of understanding of disease pathogenesis resulting from an inability to access primary AEC2s from affected children. Here, we report the generation of AEC2s from affected patient induced pluripotent stem cells (iPSCs) carrying homozygous versions of multiple ABCA3 mutations. We generated syngeneic CRISPR/Cas9 gene-corrected and uncorrected iPSCs and ABCA3-mutant knockin ABCA3:GFP fusion reporter lines for in vitro disease modeling. We observed an expected decreased capacity for surfactant secretion in ABCA3-mutant iPSC-derived AEC2s (iAEC2s), but we also found an unexpected epithelial-intrinsic aberrant phenotype in mutant iAEC2s, presenting as diminished progenitor potential, increased NFκB signaling, and the production of pro-inflammatory cytokines. The ABCA3:GFP fusion reporter permitted mutant-specific, quantifiable characterization of lamellar body size and ABCA3 protein trafficking, functional features that are perturbed depending on ABCA3 mutation type. Our disease model provides a platform for understanding ABCA3 mutation-mediated mechanisms of alveolar epithelial cell dysfunction that may trigger chILD pathogenesis.
Collapse
Affiliation(s)
- Yuliang L. Sun
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Erin E. Hennessey
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Hillary Heins
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine and St. Louis Children’s Hospital, St. Louis, Missouri, USA
| | - Ping Yang
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine and St. Louis Children’s Hospital, St. Louis, Missouri, USA
| | - Carlos Villacorta-Martin
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
| | - Julian Kwan
- Departments of Biology and Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Krithi Gopalan
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Marianne James
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
| | - Andrew Emili
- Departments of Biology and Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - F. Sessions Cole
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine and St. Louis Children’s Hospital, St. Louis, Missouri, USA
| | - Jennifer A. Wambach
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine and St. Louis Children’s Hospital, St. Louis, Missouri, USA
| | - Darrell N. Kotton
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Leblanc R, Ghossoub R, Goubard A, Castellano R, Fares J, Camoin L, Audebert S, Balzano M, Bou‐Tayeh B, Fauriat C, Vey N, Garciaz S, Borg J, Collette Y, Aurrand‐Lions M, David G, Zimmermann P. Downregulation of stromal syntenin sustains AML development. EMBO Mol Med 2023; 15:e17570. [PMID: 37819151 PMCID: PMC10630886 DOI: 10.15252/emmm.202317570] [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: 02/15/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 10/13/2023] Open
Abstract
The crosstalk between cancer and stromal cells plays a critical role in tumor progression. Syntenin is a small scaffold protein involved in the regulation of intercellular communication that is emerging as a target for cancer therapy. Here, we show that certain aggressive forms of acute myeloid leukemia (AML) reduce the expression of syntenin in bone marrow stromal cells (BMSC). Stromal syntenin deficiency, in turn, generates a pro-tumoral microenvironment. From serial transplantations in mice and co-culture experiments, we conclude that syntenin-deficient BMSC stimulate AML aggressiveness by promoting AML cell survival and protein synthesis. This pro-tumoral activity is supported by increased expression of endoglin, a classical marker of BMSC, which in trans stimulates AML translational activity. In short, our study reveals a vicious signaling loop potentially at the heart of AML-stroma crosstalk and unsuspected tumor-suppressive effects of syntenin that need to be considered during systemic targeting of syntenin in cancer therapy.
Collapse
Affiliation(s)
- Raphael Leblanc
- Team Spatio‐Temporal Regulation of Cell Signaling‐Scaffolds and Phosphoinositides, Equipe Labellisée Ligue 2018, Centre de Recherche en Cancérologie de Marseille (CRCM)Institut Paoli‐Calmettes, Aix‐Marseille Université, Inserm, CNRSMarseilleFrance
| | - Rania Ghossoub
- Team Spatio‐Temporal Regulation of Cell Signaling‐Scaffolds and Phosphoinositides, Equipe Labellisée Ligue 2018, Centre de Recherche en Cancérologie de Marseille (CRCM)Institut Paoli‐Calmettes, Aix‐Marseille Université, Inserm, CNRSMarseilleFrance
| | - Armelle Goubard
- TrGET Preclinical Platform, Centre de Recherche en Cancérologie de Marseille, Inserm, CNRSAix‐Marseille Université, Institut Paoli‐CalmettesMarseilleFrance
| | - Rémy Castellano
- TrGET Preclinical Platform, Centre de Recherche en Cancérologie de Marseille, Inserm, CNRSAix‐Marseille Université, Institut Paoli‐CalmettesMarseilleFrance
| | - Joanna Fares
- Team Spatio‐Temporal Regulation of Cell Signaling‐Scaffolds and Phosphoinositides, Equipe Labellisée Ligue 2018, Centre de Recherche en Cancérologie de Marseille (CRCM)Institut Paoli‐Calmettes, Aix‐Marseille Université, Inserm, CNRSMarseilleFrance
| | - Luc Camoin
- Proteomics and Mass Spectrometry Platform, Centre de Recherche en Cancérologie de MarseilleAix‐Marseille Université, Inserm, CNRS, Institut Paoli CalmettesMarseilleFrance
| | - Stephane Audebert
- Proteomics and Mass Spectrometry Platform, Centre de Recherche en Cancérologie de MarseilleAix‐Marseille Université, Inserm, CNRS, Institut Paoli CalmettesMarseilleFrance
| | - Marielle Balzano
- Team Spatio‐Temporal Regulation of Cell Signaling‐Scaffolds and Phosphoinositides, Equipe Labellisée Ligue 2018, Centre de Recherche en Cancérologie de Marseille (CRCM)Institut Paoli‐Calmettes, Aix‐Marseille Université, Inserm, CNRSMarseilleFrance
| | - Berna Bou‐Tayeh
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de MarseilleAix‐Marseille Université, Inserm, CNRS, Institut Paoli CalmettesMarseilleFrance
| | - Cyril Fauriat
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de MarseilleAix‐Marseille Université, Inserm, CNRS, Institut Paoli CalmettesMarseilleFrance
| | - Norbert Vey
- Aix‐Marseille Univ, Inserm, CNRS, Institut Paoli‐Calmettes, CRCMMarseilleFrance
| | - Sylvain Garciaz
- Aix‐Marseille Univ, Inserm, CNRS, Institut Paoli‐Calmettes, CRCMMarseilleFrance
| | - Jean‐Paul Borg
- Proteomics and Mass Spectrometry Platform, Centre de Recherche en Cancérologie de MarseilleAix‐Marseille Université, Inserm, CNRS, Institut Paoli CalmettesMarseilleFrance
| | - Yves Collette
- TrGET Preclinical Platform, Centre de Recherche en Cancérologie de Marseille, Inserm, CNRSAix‐Marseille Université, Institut Paoli‐CalmettesMarseilleFrance
| | - Michel Aurrand‐Lions
- Team Leuko/Stromal Interactions in Normal and Pathological Hematopoiesis, Centre de Recherche en Cancérologie de Marseille, Aix‐Marseille Université, Inserm, CNRS, Institut Paoli CalmettesMarseilleFrance
| | - Guido David
- Team Spatio‐Temporal Regulation of Cell Signaling‐Scaffolds and Phosphoinositides, Equipe Labellisée Ligue 2018, Centre de Recherche en Cancérologie de Marseille (CRCM)Institut Paoli‐Calmettes, Aix‐Marseille Université, Inserm, CNRSMarseilleFrance
- Department of Human GeneticsK U LeuvenLeuvenBelgium
| | - Pascale Zimmermann
- Team Spatio‐Temporal Regulation of Cell Signaling‐Scaffolds and Phosphoinositides, Equipe Labellisée Ligue 2018, Centre de Recherche en Cancérologie de Marseille (CRCM)Institut Paoli‐Calmettes, Aix‐Marseille Université, Inserm, CNRSMarseilleFrance
- Department of Human GeneticsK U LeuvenLeuvenBelgium
| |
Collapse
|
8
|
Yang J, Wu X, Song Y. Recent advances in novel mutation genes of Parkinson's disease. J Neurol 2023:10.1007/s00415-023-11781-4. [PMID: 37222843 DOI: 10.1007/s00415-023-11781-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/12/2023] [Accepted: 05/13/2023] [Indexed: 05/25/2023]
Abstract
With increasing life expectancy, a growing number of individuals are being affected by Parkinson's Disease (PD), a Neurodegenerative Disease (ND). Approximately, 5-10% of PD is explained by genetic causes linked to known PD genes. With improvements in genetic testing and high-throughput technologies, more PD-associated susceptibility genes have been reported in recent years. However, a comprehensive review of the pathogenic mechanisms and physiological roles of these genes is still lacking. This article reviews novel genes with putative or confirmed pathogenic mutations in PD reported since 2019, summarizes the physiological functions and potential associations with PD. Newly reported PD-related genes include ANK2, DNAH1, STAB1, NOTCH2NLC, UQCRC1, ATP10B, TFG, CHMP1A, GIPC1, KIF21B, KIF24, SLC25A39, SPTBN1 and TOMM22. However, the evidence for pathogenic effects of many of these genes is inconclusive. A variety of novel PD-associated genes have been identified through clinical cases of PD patients and analysis of Genome-Wide Association Studies (GWAS). However, more evidence is needed in confirm the strong association of novel genes with disease.
Collapse
Affiliation(s)
- Jie Yang
- Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun, 130062, China
| | - Xinyu Wu
- Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun, 130062, China
| | - Yuning Song
- Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun, 130062, China.
| |
Collapse
|
9
|
Ahmed T, Flores PC, Pan CC, Ortiz HR, Lee YS, Langlais PR, Mythreye K, Lee NY. EPDR1 is a noncanonical effector of insulin-mediated angiogenesis regulated by an endothelial-specific TGF-β receptor complex. J Biol Chem 2022; 298:102297. [PMID: 35872017 PMCID: PMC9396412 DOI: 10.1016/j.jbc.2022.102297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/05/2022] [Accepted: 07/08/2022] [Indexed: 02/05/2023] Open
Abstract
Insulin signaling in blood vessels primarily functions to stimulate angiogenesis and maintain vascular homeostasis through the canonical PI3K and MAPK signaling pathways. However, angiogenesis is a complex process coordinated by multiple other signaling events. Here, we report a distinct crosstalk between the insulin receptor and endoglin/activin receptor-like kinase 1 (ALK1), an endothelial cell-specific TGF-β receptor complex essential for angiogenesis. While the endoglin-ALK1 complex normally binds to TGF-β or bone morphogenetic protein 9 (BMP9) to promote gene regulation via transcription factors Smad1/5, we show that insulin drives insulin receptor oligomerization with endoglin-ALK1 at the cell surface to trigger rapid Smad1/5 activation. Through quantitative proteomic analysis, we identify ependymin-related protein 1 (EPDR1) as a major Smad1/5 gene target induced by insulin but not by TGF-β or BMP9. We found endothelial EPDR1 expression is minimal at the basal state but is markedly enhanced upon prolonged insulin treatment to promote cell migration and formation of capillary tubules. Conversely, we demonstrate EPDR1 depletion strongly abrogates these angiogenic effects, indicating that EPDR1 is a crucial mediator of insulin-induced angiogenesis. Taken together, these results suggest important therapeutic implications for EPDR1 and the TGF-β pathways in pathologic angiogenesis during hyperinsulinemia and insulin resistance.
Collapse
Affiliation(s)
- Tasmia Ahmed
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, Arizona, USA
| | - Paola Cruz Flores
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, Arizona, USA
| | - Christopher C. Pan
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - Hannah R. Ortiz
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Yeon S. Lee
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Paul R. Langlais
- Department of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Karthikeyan Mythreye
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA,For correspondence: Nam Y. Lee; Karthikeyan Mythreye
| | - Nam Y. Lee
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, Arizona, USA,Department of Pharmacology, University of Arizona, Tucson, Arizona, USA,Comprehensive Cancer Center, University of Arizona, Tucson, Arizona, USA,For correspondence: Nam Y. Lee; Karthikeyan Mythreye
| |
Collapse
|
10
|
Betterton RD, Abdullahi W, Williams EI, Lochhead JJ, Brzica H, Stanton J, Reddell E, Ogbonnaya C, Davis TP, Ronaldson PT. Regulation of Blood-Brain Barrier Transporters by Transforming Growth Factor- β/Activin Receptor-Like Kinase 1 Signaling: Relevance to the Brain Disposition of 3-Hydroxy-3-Methylglutaryl Coenzyme A Reductase Inhibitors (i.e., Statins). Drug Metab Dispos 2022; 50:942-956. [PMID: 35504656 PMCID: PMC11022862 DOI: 10.1124/dmd.121.000781] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/08/2022] [Indexed: 11/22/2022] Open
Abstract
Our laboratory has shown that activation of transforming growth factor- β (TGF- β )/activin receptor-like kinase 1 (ALK1) signaling can increase protein expression and transport activity of organic anion transporting polypeptide 1a4 (Oatp1a4) at the blood-brain barrier (BBB). These results are relevant to treatment of ischemic stroke because Oatp transport substrates such as 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (i.e., statins) improve functional neurologic outcomes in patients. Advancement of our work requires determination if TGF- β /ALK1 signaling alters Oatp1a4 functional expression differently across brain regions and if such disparities affect central nervous system (CNS) statin disposition. Therefore, we studied regulation of Oatp1a4 by the TGF- β /ALK1 pathway, in vivo, in rat brain microvessels isolated from cerebral cortex, hippocampus, and cerebellum using the ALK1 agonist bone morphogenetic protein-9 (BMP-9) and the ALK1 inhibitor 4-[6-[4-(1-piperazinyl)phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]quinoline dihydrochloride 193189. We showed that Oatp1a4 protein expression and brain distribution of three currently marketed statin drugs (i.e., atorvastatin, pravastatin, and rosuvastatin) were increased in cortex relative to hippocampus and cerebellum. Additionally, BMP-9 treatment enhanced Oatp-mediated statin transport in cortical tissue but not in hippocampus or cerebellum. Although brain drug delivery is also dependent upon efflux transporters, such as P-glycoprotein and/or Breast Cancer Resistance Protein, our data showed that administration of BMP-9 did not alter the relative contribution of these transporters to CNS disposition of statins. Overall, this study provides evidence for differential regulation of Oatp1a4 by TGF- β /ALK1 signaling across brain regions, knowledge that is critical for development of therapeutic strategies to target Oatps at the BBB for CNS drug delivery. SIGNIFICANCE STATEMENT: Organic anion transporting polypeptides (Oatps) represent transporter targets for brain drug delivery. We have shown that Oatp1a4 statin uptake is higher in cortex versus hippocampus and cerebellum. Additionally, we report that the transforming growth factor- β /activin receptor-like kinase 1 agonist bone morphogenetic protein-9 increases Oatp1a4 functional expression, but not efflux transporters P-glycoprotein and Breast Cancer Resistance Protein, in cortical brain microvessels. Overall, this study provides critical data that will advance treatment for neurological diseases where drug development has been challenging.
Collapse
Affiliation(s)
- Robert D Betterton
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Wazir Abdullahi
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Erica I Williams
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Jeffrey J Lochhead
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Hrvoje Brzica
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Joshua Stanton
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Elizabeth Reddell
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Chidinma Ogbonnaya
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Thomas P Davis
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Patrick T Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
11
|
Pinkaew D, Martinez-Hackert E, Jia W, King MD, Miao F, Enger NR, Silakit R, Ramana K, Chen SY, Fujise K. Fortilin interacts with TGF-β1 and prevents TGF-β receptor activation. Commun Biol 2022; 5:157. [PMID: 35197550 PMCID: PMC8866402 DOI: 10.1038/s42003-022-03112-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/28/2022] [Indexed: 11/21/2022] Open
Abstract
Fortilin is a 172-amino acid multifunctional protein present in both intra- and extracellular spaces. Although fortilin binds and regulates various cellular proteins, the biological role of extracellular fortilin remains unknown. Here we report that fortilin specifically interacts with TGF-β1 and prevents it from activating the TGF-β1 signaling pathway. In a standard immunoprecipitation-western blot assay, fortilin co-immunoprecipitates TGF-β1 and its isoforms. The modified ELISA assay shows that TGF-β1 remains complexed with fortilin in human serum. Both bio-layer interferometry and surface plasmon resonance (SPR) reveal that fortilin directly bind TGF-β1. The SPR analysis also reveals that fortilin and the TGF-β receptor II (TGFβRII) compete for TGF-β1. Both luciferase and secreted alkaline phosphatase reporter assays show that fortilin prevents TGF-β1 from activating Smad3 binding to Smad-binding element. Fortilin inhibits the phosphorylation of Smad3 in both quantitative western blot assays and ELISA. Finally, fortilin inhibits TGFβ-1-induced differentiation of C3H10T1/2 mesenchymal progenitor cells to smooth muscle cells. A computer-assisted virtual docking reveals that fortilin occupies the pocket of TGF-β1 that is normally occupied by TGFβRII and that TGF-β1 can bind either fortilin or TGFβRII at any given time. These data support the role of extracellular fortilin as a negative regulator of the TGF-β1 signaling pathway. Fortilin prevents the activation of the TGF-β1 receptor by occupying the pocket of TGF-β1 and competing with TGF-βRII to bind with TGF-β1. This inhibits Smad3 phosphorylation and the differentiation of C3H10T1/2 mesenchymal progenitor cells to smooth muscle cells.
Collapse
Affiliation(s)
- Decha Pinkaew
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Erik Martinez-Hackert
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Wei Jia
- Department of Surgery, University of Missouri, Columbia, MO, 65212, USA
| | - Matthew D King
- Department of Chemistry and Biochemistry, Boise State University, Boise, ID, 83725, USA
| | - Fei Miao
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, 98109, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nicole R Enger
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Runglawan Silakit
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Kota Ramana
- Department of Biochemistry, Noorda College of Osteopathic Medicine, Provo, UT, 84606, USA
| | - Shi-You Chen
- Department of Surgery, University of Missouri, Columbia, MO, 65212, USA
| | - Ken Fujise
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, 98109, USA.
| |
Collapse
|
12
|
Neurodegenerative diseases associated with non-coding CGG tandem repeat expansions. Nat Rev Neurol 2022; 18:145-157. [PMID: 35022573 DOI: 10.1038/s41582-021-00612-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2021] [Indexed: 02/07/2023]
Abstract
Non-coding CGG repeat expansions cause multiple neurodegenerative disorders, including fragile X-associated tremor/ataxia syndrome, neuronal intranuclear inclusion disease, oculopharyngeal myopathy with leukodystrophy, and oculopharyngodistal myopathy. The underlying genetic causes of several of these diseases have been identified only in the past 2-3 years. These expansion disorders have substantial overlapping clinical, neuroimaging and histopathological features. The shared features suggest common mechanisms that could have implications for the development of therapies for this group of diseases - similar therapeutic strategies or drugs may be effective for various neurodegenerative disorders induced by non-coding CGG expansions. In this Review, we provide an overview of clinical and pathological features of these CGG repeat expansion diseases and consider the likely pathological mechanisms, including RNA toxicity, CGG repeat-associated non-AUG-initiated translation, protein aggregation and mitochondrial impairment. We then discuss future research needed to improve the identification and diagnosis of CGG repeat expansion diseases, to improve modelling of these diseases and to understand their pathogenesis. We also consider possible therapeutic strategies. Finally, we propose that CGG repeat expansion diseases may represent manifestations of a single underlying neuromyodegenerative syndrome in which different organs are affected to different extents depending on the gene location of the repeat expansion.
Collapse
|
13
|
Liu Y, Wang Y, Yang J, Xu T, Tan C, Zhang P, Liu Q, Chen Y. G-alpha interacting protein interacting protein, C terminus 1 regulates epileptogenesis by increasing the expression of metabotropic glutamate receptor 7. CNS Neurosci Ther 2021; 28:126-138. [PMID: 34676980 PMCID: PMC8673704 DOI: 10.1111/cns.13746] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/31/2022] Open
Abstract
Aims It has been reported that the G‐alpha interacting protein (GAIP) interacting protein, C terminus 1 (GIPC1/GIPC) engages in vesicular trafficking, receptor transport and expression, and endocytosis. However, its role in epilepsy is unclear. Therefore, in this study, we aimed to explore the role of GIPC1 in epilepsy and its possible underlying mechanism. Methods The expression patterns of GIPC1 in patients with temporal lobe epilepsy (TLE) and in mice with kainic acid (KA)‐induced epilepsy were detected. Behavioral video monitoring and hippocampal local field potential (LFP) recordings were carried out to determine the role of GIPC1 in epileptogenesis after overexpression of GIPC1. Coimmunoprecipitation (Co‐IP) assay and high‐resolution immunofluorescence staining were conducted to investigate the relationship between GIPC1 and metabotropic glutamate receptor 7 (mGluR7). In addition, the expression of mGluR7 after overexpression of GIPC1 was measured, and behavioral video monitoring and LFP recordings after antagonism of mGluR7 were performed to explore the possible mechanism mediated by GIPC1. Results GIPC1 was downregulated in the brain tissues of patients with TLE and mice with KA‐induced epilepsy. After overexpression of GIPC1, prolonged latency period, decreased epileptic seizures and reduced seizure severity in behavioral analyses, and fewer and shorter abnormal brain discharges in LFP recordings of KA‐induced epileptic mice were observed. The result of the Co‐IP assay showed the interaction between GIPC1 and mGluR7, and the high‐resolution immunofluorescence staining also showed the colocalization of these two proteins. Additionally, along with GIPC1 overexpression, the total and cell membrane expression levels of mGluR7 were also increased. And after antagonism of mGluR7, increased epileptic seizures and aggravated seizure severity in behavioral analyses and more and longer abnormal brain discharges in LFP recordings were observed. Conclusion GIPC1 regulates epileptogenesis by interacting with mGluR7 and increasing its expression.
Collapse
Affiliation(s)
- Yong Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chonqing, China
| | - You Wang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chonqing, China
| | - Juan Yang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chonqing, China.,Department of Neurology, The Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Tao Xu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chonqing, China
| | - Changhong Tan
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chonqing, China
| | - Peng Zhang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chonqing, China
| | - Qiankun Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chonqing, China
| | - Yangmei Chen
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chonqing, China
| |
Collapse
|
14
|
Mahapatra N, Uma Rao KD, Ranganathan K, Joshua E, Thavarajah R. Study of expression of endoglin (CD105) in oral squamous cell carcinoma. J Oral Maxillofac Pathol 2021; 25:552. [PMID: 35281173 PMCID: PMC8859600 DOI: 10.4103/jomfp.jomfp_13_21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 10/29/2021] [Indexed: 11/26/2022] Open
Abstract
Context: Oral cancer is the 8th most common cancer in the world. An important feature of carcinogenesis is angiogenesis. Endoglin is a powerful marker of neovascularization in solid malignancies. This study was done to ascertain its role as an indicator of metastasis and prognosis. Aim: This study aimed to evaluate and compare the expression of endoglin (CD105) in metastatic primary tumor, lymph node of the metastasized tumor, nonmetastatic primary tumor and in normal buccal mucosa immunohistochemically. Settings and Design: The total sample size comprised 45 formalin-fixed paraffin-embedded tissue blocks, n = 10 metastasized primary tumor, n = 10 lymph nodes of metastasized primary tumor, n = 20 nonmetastasized oral squamous cell carcinoma and n = 5 normal buccal mucosa were studied. Subjects and Methods: Immunohistochemistry for endoglin was performed and microvessel density (MVD) was determined by hot spot method. Microvessel density was compared between the groups. Statistical Analysis: Statistical analysis was used using one-way ANOVA. P < 0.05 was statistically significant. Results: Endoglin expression in metastatic cases (0.68 + 0.10) was higher than nonmetastatic cases (0.45 + 0.20) and the difference was statistically significant (P = 0.002). Conclusion: This study shows that presence of endoglin determines the metastatic potential of the tumor and its prognosis, thus, could be considered as a potential target of therapy.
Collapse
Affiliation(s)
- Niva Mahapatra
- Department of Oral and Maxillofacial Pathology, Kalinga Institute of Dental Sciences, Kalinga Institute of Industrial Technology Deemed to be University, Bhubaneswar, Odisha, India
| | - Krishna Devi Uma Rao
- Department Oral and Maxillofacial Pathology, Ragas Dental College and Hospital, Chennai, Tamil Nadu, India
| | - Kannan Ranganathan
- Department Oral and Maxillofacial Pathology, Ragas Dental College and Hospital, Chennai, Tamil Nadu, India
| | - Elizabeth Joshua
- Department Oral and Maxillofacial Pathology, Ragas Dental College and Hospital, Chennai, Tamil Nadu, India
| | - Rooban Thavarajah
- Department Oral and Maxillofacial Pathology, Ragas Dental College and Hospital, Chennai, Tamil Nadu, India
| |
Collapse
|
15
|
Listik E, Horst B, Choi AS, Lee NY, Győrffy B, Mythreye K. A bioinformatic analysis of the inhibin-betaglycan-endoglin/CD105 network reveals prognostic value in multiple solid tumors. PLoS One 2021; 16:e0249558. [PMID: 33819300 PMCID: PMC8021191 DOI: 10.1371/journal.pone.0249558] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/21/2021] [Indexed: 12/13/2022] Open
Abstract
Inhibins and activins are dimeric ligands belonging to the TGFβ superfamily with emergent roles in cancer. Inhibins contain an α-subunit (INHA) and a β-subunit (either INHBA or INHBB), while activins are mainly homodimers of either βA (INHBA) or βB (INHBB) subunits. Inhibins are biomarkers in a subset of cancers and utilize the coreceptors betaglycan (TGFBR3) and endoglin (ENG) for physiological or pathological outcomes. Given the array of prior reports on inhibin, activin and the coreceptors in cancer, this study aims to provide a comprehensive analysis, assessing their functional prognostic potential in cancer using a bioinformatics approach. We identify cancer cell lines and cancer types most dependent and impacted, which included p53 mutated breast and ovarian cancers and lung adenocarcinomas. Moreover, INHA itself was dependent on TGFBR3 and ENG/CD105 in multiple cancer types. INHA, INHBA, TGFBR3, and ENG also predicted patients' response to anthracycline and taxane therapy in luminal A breast cancers. We also obtained a gene signature model that could accurately classify 96.7% of the cases based on outcomes. Lastly, we cross-compared gene correlations revealing INHA dependency to TGFBR3 or ENG influencing different pathways themselves. These results suggest that inhibins are particularly important in a subset of cancers depending on the coreceptor TGFBR3 and ENG and are of substantial prognostic value, thereby warranting further investigation.
Collapse
Affiliation(s)
- Eduardo Listik
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Ben Horst
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, United States of America
| | - Alex Seok Choi
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Nam. Y. Lee
- Division of Pharmacology, Chemistry and Biochemistry, College of Medicine, University of Arizona, Tucson, Arizona, United States of America
| | - Balázs Győrffy
- TTK Cancer Biomarker Research Group, Institute of Enzymology, and Semmelweis University Department of Bioinformatics and 2nd Department of Pediatrics, Budapest, Hungary
| | - Karthikeyan Mythreye
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
16
|
Endoglin in the Spotlight to Treat Cancer. Int J Mol Sci 2021; 22:ijms22063186. [PMID: 33804796 PMCID: PMC8003971 DOI: 10.3390/ijms22063186] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/06/2021] [Accepted: 03/17/2021] [Indexed: 01/02/2023] Open
Abstract
A spotlight has been shone on endoglin in recent years due to that fact of its potential to serve as both a reliable disease biomarker and a therapeutic target. Indeed, endoglin has now been assigned many roles in both physiological and pathological processes. From a molecular point of view, endoglin mainly acts as a co-receptor in the canonical TGFβ pathway, but also it may be shed and released from the membrane, giving rise to the soluble form, which also plays important roles in cell signaling. In cancer, in particular, endoglin may contribute to either an oncogenic or a non-oncogenic phenotype depending on the cell context. The fact that endoglin is expressed by neoplastic and non-neoplastic cells within the tumor microenvironment suggests new possibilities for targeted therapies. Here, we aimed to review and discuss the many roles played by endoglin in different tumor types, as well as the strong evidence provided by pre-clinical and clinical studies that supports the therapeutic targeting of endoglin as a novel clinical strategy.
Collapse
|
17
|
Tazat K, Pomeraniec-Abudy L, Hector-Greene M, Szilágyi SS, Sharma S, Cai EM, Corona AL, Ehrlich M, Blobe GC, Henis YI. ALK1 regulates the internalization of endoglin and the type III TGF-β receptor. Mol Biol Cell 2021; 32:605-621. [PMID: 33566682 PMCID: PMC8101464 DOI: 10.1091/mbc.e20-03-0199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Complex formation and endocytosis of transforming growth factor-β (TGF-β) receptors play important roles in signaling. However, their interdependence remained unexplored. Here, we demonstrate that ALK1, a TGF-β type I receptor prevalent in endothelial cells, forms stable complexes at the cell surface with endoglin and with type III TGF-β receptors (TβRIII). We show that ALK1 undergoes clathrin-mediated endocytosis (CME) faster than ALK5, type II TGF-β receptor (TβRII), endoglin, or TβRIII. These complexes regulate the endocytosis of the TGF-β receptors, with a major effect mediated by ALK1. Thus, ALK1 enhances the endocytosis of TβRIII and endoglin, while ALK5 and TβRII mildly enhance endoglin, but not TβRIII, internalization. Conversely, the slowly endocytosed endoglin has no effect on the endocytosis of either ALK1, ALK5, or TβRII, while TβRIII has a differential effect, slowing the internalization of ALK5 and TβRII, but not ALK1. Such effects may be relevant to signaling, as BMP9-mediated Smad1/5/8 phosphorylation is inhibited by CME blockade in endothelial cells. We propose a model that links TGF-β receptor oligomerization and endocytosis, based on which endocytosis signals are exposed/functional in specific receptor complexes. This has broad implications for signaling, implying that complex formation among various receptors regulates their surface levels and signaling intensities.
Collapse
Affiliation(s)
- Keren Tazat
- Department of Neurobiology, Tel Aviv University, Tel Aviv 6997801, Israel
| | | | | | | | - Swati Sharma
- Department of Neurobiology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Elise M Cai
- Department of Medicine, Duke University Medical Center, Durham, NC 27708
| | - Armando L Corona
- Department of Medicine, Duke University Medical Center, Durham, NC 27708
| | - Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Gerard C Blobe
- Department of Medicine, Duke University Medical Center, Durham, NC 27708
| | - Yoav I Henis
- Department of Neurobiology, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
18
|
Ahmed T, Mythreye K, Lee NY. Strength and duration of GIPC-dependent signaling networks as determinants in cancer. Neoplasia 2021; 23:181-188. [PMID: 33360508 PMCID: PMC7773760 DOI: 10.1016/j.neo.2020.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 10/25/2022]
Abstract
GIPC is a PDZ-domain containing adaptor protein that regulates the cell surface expression and endocytic trafficking of numerous transmembrane receptors and signaling complexes. Interactions with over 50 proteins have been reported to date including VEGFR, insulin-like growth factor-1 receptor (IGF-1R), GPCRs, and APPL, many of which have essential roles in neuronal and cardiovascular development. In cancer, a major subset of GIPC-binding receptors and cytoplasmic effectors have been shown to promote tumorigenesis or metastatic progression, while other subsets have demonstrated strong tumor-suppressive effects. Given that these diverse pathways are widespread in normal tissues and human malignancies, precisely how these opposing signals are integrated and regulated within the same tumor setting likely depend on the strength and duration of their interactions with GIPC. This review highlights the major pathways and divergent mechanisms of GIPC signaling in various cancers and provide a rationale for emerging GIPC-targeted cancer therapies.
Collapse
Affiliation(s)
- Tasmia Ahmed
- Deparment of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Karthikeyan Mythreye
- Division of Molecular and Cellular Pathology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Nam Y Lee
- Deparment of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA; Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA; The University of Arizona Cancer Center, Tucson, AZ, USA.
| |
Collapse
|
19
|
Hu L, Li K, Lin L, Qian F, Li P, Zhu L, Cai H, You L, Song J, Kok SHL, Lee KKH, Yang X, Cheng X. Reversine suppresses osteosarcoma cell growth through targeting BMP-Smad1/5/8-mediated angiogenesis. Microvasc Res 2021; 135:104136. [PMID: 33450295 DOI: 10.1016/j.mvr.2021.104136] [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: 08/11/2020] [Revised: 12/31/2020] [Accepted: 01/11/2021] [Indexed: 11/29/2022]
Abstract
Reversine, or 2-(4-morpholinoanilino)-6cyclohexylaminopurine, is a 2,6-disubstituted purine derivative. This small molecule exhibits tumor-suppressive activities through different molecular mechanisms. In this study, in vitro and in vivo angiogenic models were used to elucidate the effect of Reversine on angiogenesis in the tumor suppression. Firstly, we grafted osteosarcoma-derived MNNG/HOS cell aggregates onto chick embryonic chorioallantoic membrane (CAM) to examine the vascularization of these grafts following Reversine treatment. Following culture, it was determined that Reversine inhibited MNNG/HOS grafts growth, and decreased the density of blood vessels in the chick CAM. We then used CAM and chick embryonic yolk-sac membrane (YSM) to investigate the effects of Reversine on angiogenesis. The results revealed Reversine inhibited the proliferation of endothelial cells, where cells were mainly arrested at G1/S phase of the cell cycle. Scratch-wound assay with HUVECs revealed that Reversine suppressed cell migration in vitro. Furthermore, endothelial cells tube formation assay and chick aortic arch sprouting assay demonstrated Reversine inhibited the sprouting, migration of endothelial cells. Lastly, qPCR and western blot analyses showed BMP-associated Smad1/5/8 signaling expressions were up-regulated by Reversine treatment. Our results showed that Reversine could suppress tumor growth by inhibiting angiogenesis through BMP signaling, and suggests a potential use of Reversine as an anti-tumor therapy.
Collapse
Affiliation(s)
- Lingzhi Hu
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China
| | - Kanghu Li
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China
| | - Li Lin
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China
| | - Fan Qian
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China
| | - Peizhi Li
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China
| | - Liwei Zhu
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China
| | - Hongmei Cai
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China
| | - Lingsen You
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China
| | - Jinhuan Song
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China
| | - Stanton Hon Lung Kok
- Key Laboratory for Regenerative Medicine of the Ministry of Education, School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kenneth Ka Ho Lee
- Key Laboratory for Regenerative Medicine of the Ministry of Education, School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Xuesong Yang
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China.
| | - Xin Cheng
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
20
|
Endoglin: An 'Accessory' Receptor Regulating Blood Cell Development and Inflammation. Int J Mol Sci 2020; 21:ijms21239247. [PMID: 33287465 PMCID: PMC7729465 DOI: 10.3390/ijms21239247] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
Transforming growth factor-β1 (TGF-β1) is a pleiotropic factor sensed by most cells. It regulates a broad spectrum of cellular responses including hematopoiesis. In order to process TGF-β1-responses in time and space in an appropriate manner, there is a tight regulation of its signaling at diverse steps. The downstream signaling is mediated by type I and type II receptors and modulated by the ‘accessory’ receptor Endoglin also termed cluster of differentiation 105 (CD105). Endoglin was initially identified on pre-B leukemia cells but has received most attention due to its high expression on activated endothelial cells. In turn, Endoglin has been figured out as the causative factor for diseases associated with vascular dysfunction like hereditary hemorrhagic telangiectasia-1 (HHT-1), pre-eclampsia, and intrauterine growth restriction (IUPR). Because HHT patients often show signs of inflammation at vascular lesions, and loss of Endoglin in the myeloid lineage leads to spontaneous inflammation, it is speculated that Endoglin impacts inflammatory processes. In line, Endoglin is expressed on progenitor/precursor cells during hematopoiesis as well as on mature, differentiated cells of the innate and adaptive immune system. However, so far only pro-monocytes and macrophages have been in the focus of research, although Endoglin has been identified in many other immune system cell subsets. These findings imply a functional role of Endoglin in the maturation and function of immune cells. Aside the functional relevance of Endoglin in endothelial cells, CD105 is differentially expressed during hematopoiesis, arguing for a role of this receptor in the development of individual cell lineages. In addition, Endoglin expression is present on mature immune cells of the innate (i.e., macrophages and mast cells) and the adaptive (i.e., T-cells) immune system, further suggesting Endoglin as a factor that shapes immune responses. In this review, we summarize current knowledge on Endoglin expression and function in hematopoietic precursors and mature hematopoietic cells of different lineages.
Collapse
|
21
|
Bigaeva E, Gore E, Mutsaers HAM, Oosterhuis D, Kim YO, Schuppan D, Bank RA, Boersema M, Olinga P. Exploring organ-specific features of fibrogenesis using murine precision-cut tissue slices. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165582. [PMID: 31676376 DOI: 10.1016/j.bbadis.2019.165582] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 10/06/2019] [Accepted: 10/16/2019] [Indexed: 12/14/2022]
Abstract
Fibrosis is the hallmark of pathologic tissue remodelling in most chronic diseases. Despite advances in our understanding of the mechanisms of fibrosis, it remains uncured. Fibrogenic processes share conserved core cellular and molecular pathways across organs. In this study, we aimed to elucidate shared and organ-specific features of fibrosis using murine precision-cut tissue slices (PCTS) prepared from small intestine, liver and kidneys. PCTS displayed substantial differences in their baseline gene expression profiles: 70% of the extracellular matrix (ECM)-related genes were differentially expressed across the organs. Culture for 48 h induced significant changes in ECM regulation and triggered the onset of fibrogenesis in all PCTS in organ-specific manner. TGFβ signalling was activated during 48 h culture in all PCTS. However, the degree of its involvement varied: both canonical and non-canonical TGFβ pathways were activated in liver and kidney slices, while only canonical, Smad-dependent, cascade was involved in intestinal slices. The treatment with galunisertib blocked the TGFβRI/SMAD2 signalling in all PCTS, but attenuated culture-induced dysregulation of ECM homeostasis and mitigated the onset of fibrogenesis with organ-specificity. In conclusion, regardless the many common features in pathophysiology of organ fibrosis, PCTS displayed diversity in culture-induced responses and in response to the treatment with TGFβRI kinase inhibitor galunisertib, even though it targets a core fibrosis pathway. A clear understanding of the common and organ-specific features of fibrosis is the basis for developing novel antifibrotic therapies.
Collapse
Affiliation(s)
- Emilia Bigaeva
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, the Netherlands
| | - Emilia Gore
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, the Netherlands
| | - Henricus A M Mutsaers
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, the Netherlands; Department of Clinical Medicine, Aarhus University, Denmark
| | - Dorenda Oosterhuis
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, the Netherlands
| | - Yong Ook Kim
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, Obere Zahlbacherstraße 63, Mainz 55131, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, Obere Zahlbacherstraße 63, Mainz 55131, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, 330 Brookline Avenue, MA 02215, USA
| | - Ruud A Bank
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Miriam Boersema
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, the Netherlands
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, the Netherlands.
| |
Collapse
|
22
|
Chronic exposure to submicromolar arsenite promotes the migration of human esophageal Het1A cells induced by heparin-binding EGF-like growth factor. Arch Toxicol 2019; 93:3523-3534. [DOI: 10.1007/s00204-019-02592-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/08/2019] [Indexed: 12/24/2022]
|
23
|
Meurer S, Wimmer AE, Leur EVD, Weiskirchen R. Endoglin Trafficking/Exosomal Targeting in Liver Cells Depends on N-Glycosylation. Cells 2019; 8:cells8090997. [PMID: 31466384 PMCID: PMC6769735 DOI: 10.3390/cells8090997] [Citation(s) in RCA: 14] [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: 08/19/2019] [Accepted: 08/26/2019] [Indexed: 02/06/2023] Open
Abstract
Injury of the liver involves a wound healing partial reaction governed by hepatic stellate cells and portal fibroblasts. Individual members of the transforming growth factor-β (TGF-β) superfamily including TGF-β itself and bone morphogenetic proteins (BMP) exert diverse and partially opposing effects on pro-fibrogenic responses. Signaling by these ligands is mediated through binding to membrane integral receptors type I/type II. Binding and the outcome of signaling is critically modulated by Endoglin (Eng), a type III co-receptor. In order to learn more about trafficking of Eng in liver cells, we investigated the membranal subdomain localization of full-length (FL)-Eng. We could show that FL-Eng is enriched in Caveolin-1-containing sucrose gradient fractions. Since lipid rafts contribute to the pool of exosomes, we could consequently demonstrate for the first time that exosomes isolated from cultured primary hepatic stellate cells and its derivatives contain Eng. Moreover, via adenoviral overexpression, we demonstrate that all liver cells have the capacity to direct Eng to exosomes, irrespectively whether they express endogenous Eng or not. Finally, we demonstrate that block of N-glycosylation does not interfere with dimerization of the receptor, but abrogates the secretion of soluble Eng (sol-Eng) and prevents exosomal targeting of FL-Eng.
Collapse
Affiliation(s)
- Steffen Meurer
- RWTH University Hospital Aachen, Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, D-52074 Aachen, Germany.
| | - Almut Elisabeth Wimmer
- RWTH University Hospital Aachen, Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, D-52074 Aachen, Germany
| | - Eddy van de Leur
- RWTH University Hospital Aachen, Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, D-52074 Aachen, Germany
| | - Ralf Weiskirchen
- RWTH University Hospital Aachen, Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, D-52074 Aachen, Germany.
| |
Collapse
|
24
|
Castillo J, Wu E, Lowe C, Srinivasan S, McCord R, Wagle MC, Jayakar S, Edick MG, Eastham-Anderson J, Liu B, Hutchinson KE, Jones W, Stokes MP, Tarighat SS, Holcomb T, Glibicky A, Romero FA, Magnuson S, Huang SMA, Plaks V, Giltnane JM, Lackner MR, Mounir Z. CBP/p300 Drives the Differentiation of Regulatory T Cells through Transcriptional and Non-Transcriptional Mechanisms. Cancer Res 2019; 79:3916-3927. [PMID: 31182547 DOI: 10.1158/0008-5472.can-18-3622] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/21/2019] [Accepted: 06/03/2019] [Indexed: 11/16/2022]
Abstract
Regulatory T cells (Treg) are immunosuppressive and negatively impact response to cancer immunotherapies. CREB-binding protein (CBP) and p300 are closely related acetyltransferases and transcriptional coactivators. Here, we evaluate the mechanisms by which CBP/p300 regulate Treg differentiation and the consequences of CBP/p300 loss-of-function mutations in follicular lymphoma. Transcriptional and epigenetic profiling identified a cascade of transcription factors essential for Treg differentiation. Mass spectrometry analysis showed that CBP/p300 acetylates prostacyclin synthase, which regulates Treg differentiation by altering proinflammatory cytokine secretion by T and B cells. Reduced Treg presence in tissues harboring CBP/p300 loss-of-function mutations was observed in follicular lymphoma. Our findings provide novel insights into the regulation of Treg differentiation by CBP/p300, with potential clinical implications on alteration of the immune landscape. SIGNIFICANCE: This study provides insights into the dynamic role of CBP/p300 in the differentiation of Tregs, with potential clinical implications in the alteration of the immune landscape in follicular lymphoma.
Collapse
Affiliation(s)
- Joseph Castillo
- Department of Oncology Biomarker Development, Development Sciences, Genentech, Inc., South San Francisco, California
| | - Esther Wu
- Department of Oncology Biomarker Development, Development Sciences, Genentech, Inc., South San Francisco, California
| | - Christopher Lowe
- Department of Bioanalytical Sciences, Development Sciences, Genentech, Inc., South San Francisco, California
| | - Shrividhya Srinivasan
- Department of Oncology Biomarker Development, Development Sciences, Genentech, Inc., South San Francisco, California
| | - Ron McCord
- Department of Oncology Biomarker Development, Development Sciences, Genentech, Inc., South San Francisco, California
| | - Marie-Claire Wagle
- Department of Oncology Biomarker Development, Development Sciences, Genentech, Inc., South San Francisco, California
| | - Sangeeta Jayakar
- Department of Research Pathology, Genentech, Inc., South San Francisco, California
| | | | | | - Bonnie Liu
- Department of Oncology Biomarker Development, Development Sciences, Genentech, Inc., South San Francisco, California
| | - Katherine E Hutchinson
- Department of Oncology Biomarker Development, Development Sciences, Genentech, Inc., South San Francisco, California
| | - Wendell Jones
- Q Solutions-EA Genomics, Morrisville, North Carolina
| | | | - Somayeh S Tarighat
- Department of Oncology Biomarker Development, Development Sciences, Genentech, Inc., South San Francisco, California
| | - Thomas Holcomb
- Department of Oncology Biomarker Development, Development Sciences, Genentech, Inc., South San Francisco, California
| | - Andrew Glibicky
- Department of Oncology Biomarker Development, Development Sciences, Genentech, Inc., South San Francisco, California
| | - F Anthony Romero
- Department of Discovery Chemistry, Genentech, Inc., South San Francisco, California
| | - Steven Magnuson
- Department of Discovery Chemistry, Genentech, Inc., South San Francisco, California
| | - Shih-Min A Huang
- Department of Oncology Biomarker Development, Development Sciences, Genentech, Inc., South San Francisco, California
| | - Vicki Plaks
- Department of Bioanalytical Sciences, Development Sciences, Genentech, Inc., South San Francisco, California
| | - Jennifer M Giltnane
- Department of Research Pathology, Genentech, Inc., South San Francisco, California
| | - Mark R Lackner
- Department of Oncology Biomarker Development, Development Sciences, Genentech, Inc., South San Francisco, California
| | - Zineb Mounir
- Department of Oncology Biomarker Development, Development Sciences, Genentech, Inc., South San Francisco, California.
| |
Collapse
|
25
|
Egorova AA, Shtykalova SV, Maretina MA, Sokolov DI, Selkov SA, Baranov VS, Kiselev AV. Synergistic Anti-Angiogenic Effects Using Peptide-Based Combinatorial Delivery of siRNAs Targeting VEGFA, VEGFR1, and Endoglin Genes. Pharmaceutics 2019; 11:E261. [PMID: 31174285 PMCID: PMC6631635 DOI: 10.3390/pharmaceutics11060261] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 12/18/2022] Open
Abstract
Angiogenesis is a process of new blood vessel formation, which plays a significant role in carcinogenesis and the development of diseases associated with pathological neovascularization. An important role in the regulation of angiogenesis belongs to several key pathways such as VEGF-pathways, TGF-β-pathways, and some others. Introduction of small interfering RNA (siRNA) against genes of pro-angogenic factors is a promising strategy for the therapeutic suppression of angiogenesis. These siRNA molecules need to be specifically delivered into endothelial cells, and non-viral carriers modified with cellular receptor ligands can be proposed as perspective delivery systems for anti-angiogenic therapy purposes. Here we used modular peptide carrier L1, containing a ligand for the CXCR4 receptor, for the delivery of siRNAs targeting expression of VEGFA, VEGFR1 and endoglin genes. Transfection properties of siRNA/L1 polyplexes were studied in CXCR4-positive breast cancer cells MDA-MB-231 and endothelial cells EA.Hy926. We have demonstrated the efficient down-regulation of endothelial cells migration and proliferation by anti-VEGFA, anti-VEGFR1, and anti-endoglin siRNA-induced silencing. It was found that the efficiency of anti-angiogenic treatment can be synergistically improved via the combinatorial delivery of anti-VEGFA and anti-VEGFR1 siRNAs. Thus, this approach can be useful for the development of therapeutic angiogenesis inhibition.
Collapse
Affiliation(s)
- Anna A Egorova
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia.
| | - Sofia V Shtykalova
- Department of Genetics and Biotechnology, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia.
| | - Marianna A Maretina
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia.
| | - Dmitry I Sokolov
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia.
| | - Sergei A Selkov
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia.
| | - Vladislav S Baranov
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia.
- Department of Genetics and Biotechnology, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia.
| | - Anton V Kiselev
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia.
| |
Collapse
|
26
|
Westbrook JA, Wood SL, Cairns DA, McMahon K, Gahlaut R, Thygesen H, Shires M, Roberts S, Marshall H, Oliva MR, Dunning MJ, Hanby AM, Selby PJ, Speirs V, Mavria G, Coleman RE, Brown JE. Identification and validation of DOCK4 as a potential biomarker for risk of bone metastasis development in patients with early breast cancer. J Pathol 2019; 247:381-391. [PMID: 30426503 PMCID: PMC6618075 DOI: 10.1002/path.5197] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 10/04/2018] [Accepted: 10/30/2018] [Indexed: 12/12/2022]
Abstract
Skeletal metastasis occurs in around 75% of advanced breast cancers, with the disease incurable once cancer cells disseminate to bone, but there remains an unmet need for biomarkers to identify patients at high risk of bone recurrence. This study aimed to identify such a biomarker and to assess its utility in predicting response to adjuvant zoledronic acid (zoledronate). We used quantitative proteomics (stable isotope labelling by amino acids in cell culture-mass spectrometry; SILAC-MS) to compare protein expression in a bone-homing variant (BM1) of the human breast cancer cell line MDA-MB-231 with parental non-bone-homing cells to identify novel biomarkers for risk of subsequent bone metastasis in early breast cancer. SILAC-MS showed that dedicator of cytokinesis protein 4 (DOCK4) was upregulated in bone-homing BM1 cells, confirmed by western blotting. BM1 cells also had enhanced invasive ability compared with parental cells, which could be reduced by DOCK4-shRNA. In a training tissue microarray (TMA) comprising 345 patients with early breast cancer, immunohistochemistry followed by Cox regression revealed that high DOCK4 expression correlated with histological grade (p = 0.004) but not oestrogen receptor status (p = 0.19) or lymph node involvement (p = 0.15). A clinical validation TMA used tissue samples and the clinical database from the large AZURE adjuvant study (n = 689). Adjusted Cox regression analyses showed that high DOCK4 expression in the control arm (no zoledronate) was significantly prognostic for first recurrence in bone (HR 2.13, 95%CI 1.06-4.30, p = 0.034). No corresponding association was found in patients who received zoledronate (HR 0.812, 95%CI 0.176-3.76, p = 0.790), suggesting that treatment with zoledronate may counteract the higher risk for bone relapse from high DOCK4-expressing tumours. High DOCK4 expression was not associated with metastasis to non-skeletal sites when these were assessed collectively. In conclusion, high DOCK4 in early breast cancer is significantly associated with aggressive disease and with future bone metastasis and is a potentially useful biomarker for subsequent bone metastasis risk. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jules A Westbrook
- Department of Oncology and Metabolism, Academic Unit of Clinical OncologyUniversity of SheffieldSheffieldUK
- Clinical and Biomedical Proteomics GroupLeeds Institute of Cancer and Pathology, University of LeedsLeedsUK
| | - Steven L Wood
- Department of Oncology and Metabolism, Academic Unit of Clinical OncologyUniversity of SheffieldSheffieldUK
- Clinical and Biomedical Proteomics GroupLeeds Institute of Cancer and Pathology, University of LeedsLeedsUK
| | - David A Cairns
- Clinical and Biomedical Proteomics GroupLeeds Institute of Cancer and Pathology, University of LeedsLeedsUK
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials ResearchUniversity of LeedsLeedsUK
| | - Kathryn McMahon
- Leeds Institute of Cancer and PathologyUniversity of LeedsLeedsUK
| | - Renu Gahlaut
- Leeds Institute of Cancer and PathologyUniversity of LeedsLeedsUK
| | - Helene Thygesen
- Leeds Institute of Cancer and PathologyUniversity of LeedsLeedsUK
| | - Mike Shires
- Leeds Institute of Cancer and PathologyUniversity of LeedsLeedsUK
| | - Stephanie Roberts
- Clinical and Biomedical Proteomics GroupLeeds Institute of Cancer and Pathology, University of LeedsLeedsUK
| | - Helen Marshall
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials ResearchUniversity of LeedsLeedsUK
| | - Maria R Oliva
- Department of Oncology and Metabolism, Academic Unit of Clinical OncologyUniversity of SheffieldSheffieldUK
| | - Mark J Dunning
- Sheffield Institute of Translational NeuroscienceUniversity of SheffieldSheffieldUK
| | - Andrew M Hanby
- Leeds Institute of Cancer and PathologyUniversity of LeedsLeedsUK
| | - Peter J Selby
- Clinical and Biomedical Proteomics GroupLeeds Institute of Cancer and Pathology, University of LeedsLeedsUK
- Leeds Institute of Cancer and PathologyUniversity of LeedsLeedsUK
| | - Valerie Speirs
- Leeds Institute of Cancer and PathologyUniversity of LeedsLeedsUK
- Institute of Medical SciencesUniversity of AberdeenAberdeenUK
| | - Georgia Mavria
- Leeds Institute of Cancer and PathologyUniversity of LeedsLeedsUK
| | - Robert E Coleman
- Department of Oncology and Metabolism, Academic Unit of Clinical OncologyUniversity of SheffieldSheffieldUK
| | - Janet E Brown
- Department of Oncology and Metabolism, Academic Unit of Clinical OncologyUniversity of SheffieldSheffieldUK
- Clinical and Biomedical Proteomics GroupLeeds Institute of Cancer and Pathology, University of LeedsLeedsUK
| |
Collapse
|
27
|
Pokrywczynska M, Jundzill A, Rasmus M, Adamowicz J, Balcerczyk D, Buhl M, Warda K, Buchholz L, Gagat M, Grzanka D, Drewa T. Understanding the role of mesenchymal stem cells in urinary bladder regeneration-a preclinical study on a porcine model. Stem Cell Res Ther 2018; 9:328. [PMID: 30486856 PMCID: PMC6260700 DOI: 10.1186/s13287-018-1070-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/20/2018] [Accepted: 11/08/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The tissue engineering of urinary bladder advances rapidly reflecting clinical need for a new kind of therapeutic solution for patients requiring urinary bladder replacement. Majority of the bladder augmentation studies have been performed in small rodent or rabbit models. Insufficient number of studies examining regenerative capacity of tissue-engineered graft in urinary bladder augmentation in a large animal model does not allow for successful translation of this technology to the clinical setting. The aim of this study was to evaluate the role of adipose-derived stem cells (ADSCs) in regeneration of clinically significant urinary bladder wall defect in a large animal model. METHODS ADSCs isolated from a superficial abdominal Camper's fascia were labeled with PKH-26 tracking dye and subsequently seeded into bladder acellular matrix (BAM) grafts. Pigs underwent hemicystectomy followed by augmentation cystoplasty with BAM only (n = 10) or BAM seeded with autologous ADSCs (n = 10). Reconstructed bladders were subjected to macroscopic, histological, immunofluoresence, molecular, and radiological evaluations at 3 months post-augmentation. RESULTS Sixteen animals (n = 8 for each group) survived the 3-month follow-up without serious complications. Tissue-engineered bladder function was normal without any signs of post-voiding urine residual in bladders and in the upper urinary tracts. ADSCs enhanced regeneration of tissue-engineered urinary bladder but the process was incomplete in the central graft region. Only a small percentage of implanted ADSCs survived and differentiated into smooth muscle and endothelial cells. CONCLUSIONS The data demonstrate that ADSCs support regeneration of large defects of the urinary bladder wall but the process is incomplete in the central graft region. Stem cells enhance urinary bladder regeneration indirectly through paracrine effect.
Collapse
Affiliation(s)
- Marta Pokrywczynska
- Department of Regenerative Medicine, Cell and Tissue Bank, Chair of Urology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Marii Sklodowskiej Curie 9 Street, 85-094 Bydgoszcz, Poland
| | - Arkadiusz Jundzill
- Department of Regenerative Medicine, Cell and Tissue Bank, Chair of Urology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Marii Sklodowskiej Curie 9 Street, 85-094 Bydgoszcz, Poland
| | - Marta Rasmus
- Department of Regenerative Medicine, Cell and Tissue Bank, Chair of Urology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Marii Sklodowskiej Curie 9 Street, 85-094 Bydgoszcz, Poland
| | - Jan Adamowicz
- Department of Regenerative Medicine, Cell and Tissue Bank, Chair of Urology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Marii Sklodowskiej Curie 9 Street, 85-094 Bydgoszcz, Poland
| | - Daria Balcerczyk
- Department of Regenerative Medicine, Cell and Tissue Bank, Chair of Urology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Marii Sklodowskiej Curie 9 Street, 85-094 Bydgoszcz, Poland
| | - Monika Buhl
- Department of Regenerative Medicine, Cell and Tissue Bank, Chair of Urology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Marii Sklodowskiej Curie 9 Street, 85-094 Bydgoszcz, Poland
| | - Karolina Warda
- Department of Regenerative Medicine, Cell and Tissue Bank, Chair of Urology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Marii Sklodowskiej Curie 9 Street, 85-094 Bydgoszcz, Poland
| | - Lukasz Buchholz
- Department of Regenerative Medicine, Cell and Tissue Bank, Chair of Urology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Marii Sklodowskiej Curie 9 Street, 85-094 Bydgoszcz, Poland
| | - Maciej Gagat
- Department of Embryology and Histology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, 85-092 Bydgoszcz, Poland
| | - Dariusz Grzanka
- Department of Clinical Pathomorphology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, 85-094 Bydgoszcz, Poland
| | - Tomasz Drewa
- Department of Regenerative Medicine, Cell and Tissue Bank, Chair of Urology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Marii Sklodowskiej Curie 9 Street, 85-094 Bydgoszcz, Poland
| |
Collapse
|
28
|
Jin X, Li B, Zhao Y, Liu X, Li Y, Song L, Cui L, Xie D, Li T, Zhang X, Guo Y. Erbin plays a critical role in human umbilical vein endothelial cell migration and tubular structure formation via the Smad1/5 pathway. J Cell Biochem 2018; 120:4654-4664. [PMID: 30367512 DOI: 10.1002/jcb.27754] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 09/06/2018] [Indexed: 12/13/2022]
Abstract
Angiogenesis is an important process in atherosclerosis. ErbB2 was proved to have an important role in vascular development, but it is still unclear whether Erbin expresses in vessels as well as its location and function in the vessels. In the current study, we investigated the location and function of Erbin in human umbilical veins. The human umbilical veins were prepared, and immunofluorescent analysis was performed to determine the expression of Erbin. Human umbilical vein endothelial cells (HUVECs) were cultured and the lentivirus (LV) containing Erbin RNAi was also prepared. After transfection with the lentivirus, CCK-8 assay and Annexin V-PI assay were used for cell proliferation and apoptosis, respectively. Cell migration was studied using the scratch wound healing assay and the transwell assay. The capillary-like tube formation assay was performed to illustrate the effect of Erbin on HUVEC tube formation. Expression of signaling pathway molecules was assessed with Western blot. The immunofluorescent analysis suggested that Erbin expressed in human umbilical veins and the majority of the Erbin is strongly colocalized in endothelial cells. Although knockdown of Erbin did not affect HUVEC proliferation and apoptosis, it significantly suppressed HUVEC migration and tubular structure formation. Erbin knockdown showed no effect on the ERK1/2 and Smad2/3 signaling pathways but significantly promoted Smad1/5 phosphorylation and nuclear translocation. Ablation of the Smad1/5 pathway decreased the effects of Erbin on endothelial cells. Erbin is mainly localized in endothelial cells in human umbilical veins and plays a critical role in endothelial cell migration and tubular formation via the Smad1/5 pathway.
Collapse
Affiliation(s)
- Xiaodong Jin
- Department of Cardiology, Qilu Hospital, Shandong University, Jinan, Shandong, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University, Jinan, Shandong, China.,Department of Geriatrics, Central Hospital of Zibo, Zibo, China
| | - Bo Li
- Heart Center, Central Hospital of Zibo, Zibo, China
| | - Yunhe Zhao
- Heart Center, Central Hospital of Zibo, Zibo, China
| | - Xiqiang Liu
- Department of Geriatrics, Central Hospital of Zibo, Zibo, China
| | - Yuhua Li
- Department of CT and Magnetic Resonance, Central Hospital of Zibo, Zibo, China
| | - Lina Song
- Department of Hospital Office, Maternal and Child Health Care Hospital of Zibo, Zibo, China
| | - Lifang Cui
- Department of Hyperbaric Oxygen, Central Hospital of Zibo, Zibo, China
| | - Dan Xie
- Department of Rehabilitation Medicine, Sixth People's Hospital of Zibo, Zibo, China
| | - Tao Li
- Center of Translational Medicine, Central Hospital of Zibo, Zibo, China
| | - Xiufang Zhang
- Department of Medical Administration Division, Central Hospital of Zibo, Zibo, China
| | - Yuan Guo
- Department of Cardiology, Qilu Hospital, Shandong University, Jinan, Shandong, China
| |
Collapse
|
29
|
Salem SM, Hamed AR, Fayez AG, Nour Eldeen G. Non-target Genes Regulate miRNAs-Mediated Migration Steering of Colorectal Carcinoma. Pathol Oncol Res 2018; 25:559-566. [PMID: 30361904 DOI: 10.1007/s12253-018-0502-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 10/15/2018] [Indexed: 12/14/2022]
Abstract
MicroRNAs (miRNAs) trigger a two-layer regulatory network directly or through transcription factors and their co-regulators. Unlike miR-375, the role of miR-145 and miR-224 in inhibiting or driving cancer cell migration is controversial. This study is a step towards addressing the potential of miR-375, miR-145 and miR-224 expression modulation to inhibit colorectal carcinoma (CRC) cells migration in vitro through regulation of non-target genes VEGFA, TGFβ1, IGF1, CD105 and CD44. Transwell migration assay results revealed a significant subdue of migration ability of cells transfected with miR-375 and miR-145 mimics and miR-224 inhibitor. Real time PCR data showed that expression of VEGFA, TGFβ1, IGF1, CD105 and CD44 was downregulated as a consequence of exogenous re-expression of miR-375 and inhibition of miR-224. On the other hand, ectopic expression of miR-145 did not affect VEGFA, TGFβ1 and CD44 expression, while it elevated CD105 and suppressed IGF1 expression. MAP4K4, a predicted target of miR-145, was validated as a target that could play a role in miR-145-mediated regulation of migration. At mRNA level, no change was observed in expression of MAP4K4 in cells with restored expression of miR-145, while western blotting analysis revealed a 25% reduction of protein level. By applying luciferase reporter assay, a significant decrease in luciferase activity was observed, supporting that miR-145 directly target 3' UTR of MAP4K4. The study highlighted the involvement of non-target genes VEGFA, TGFβ1, IGF1, CD105 and CD44 in mediating anti- and pro-migratory effect of miR-375 and miR-224, respectively, and validated MAP4K4 as a direct target of anti-migratory miR-145.
Collapse
Affiliation(s)
- Sohair M Salem
- Molecular Genetics and Enzymology Department, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt.
| | - Ahmed R Hamed
- Phytochemistry Department, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt.,Biology Unit - Central Laboratory of Pharmaceutical and Drug Industries Research Division, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt
| | - Alaaeldin G Fayez
- Molecular Genetics and Enzymology Department, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt
| | - Ghada Nour Eldeen
- Molecular Genetics and Enzymology Department, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt.,Stem Cell Research Unit, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt
| |
Collapse
|
30
|
Culjat M, Razak J, Saadeh‐Haddad R, Driggers R, Kamholz K, Timofeev J. Perinatal findings in a patient with a novel large chromosome 19p deletion. Clin Case Rep 2018; 6:1525-1530. [PMID: 30147897 PMCID: PMC6099042 DOI: 10.1002/ccr3.1615] [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: 08/08/2017] [Revised: 03/16/2018] [Accepted: 05/08/2018] [Indexed: 12/02/2022] Open
Abstract
We describe the prenatal and postnatal course of an infant with a large 19p deletion. Cases such as ours will improve the knowledge of specific gene functions for every medical specialist. The goal is to allow for a more rapid diagnosis, accurate prognosis and to decrease the likelihood of complications.
Collapse
Affiliation(s)
- Marko Culjat
- MedStar Georgetown University HospitalNeonatal‐Perinatal MedicineWashingtonDCUSA
| | - Jennifer Razak
- Division of Maternal Fetal MedicineSibley Memorial HospitalWashingtonDCUSA
| | - Reem Saadeh‐Haddad
- Department of PediatricsMedStar Georgetown University HospitalWashingtonDCUSA
| | - Rita Driggers
- Division of Maternal Fetal MedicineSibley Memorial HospitalWashingtonDCUSA
- Division of Maternal Fetal MedicineJohns Hopkins School of MedicineBaltimoreMDUSA
| | - Karen Kamholz
- MedStar Georgetown University HospitalNeonatal‐Perinatal MedicineWashingtonDCUSA
| | - Julia Timofeev
- Division of Maternal Fetal MedicineSibley Memorial HospitalWashingtonDCUSA
- Division of Maternal Fetal MedicineJohns Hopkins School of MedicineBaltimoreMDUSA
| |
Collapse
|
31
|
Paauwe M, Schoonderwoerd MJA, Helderman RFCP, Harryvan TJ, Groenewoud A, van Pelt GW, Bor R, Hemmer DM, Versteeg HH, Snaar-Jagalska BE, Theuer CP, Hardwick JCH, Sier CFM, Ten Dijke P, Hawinkels LJAC. Endoglin Expression on Cancer-Associated Fibroblasts Regulates Invasion and Stimulates Colorectal Cancer Metastasis. Clin Cancer Res 2018; 24:6331-6344. [PMID: 29945992 DOI: 10.1158/1078-0432.ccr-18-0329] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/23/2018] [Accepted: 06/18/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE Cancer-associated fibroblasts (CAF) are a major component of the colorectal cancer tumor microenvironment. CAFs play an important role in tumor progression and metastasis, partly through TGF-β signaling pathway. We investigated whether the TGF-β family coreceptor endoglin is involved in CAF-mediated invasion and metastasis. EXPERIMENTAL DESIGN CAF-specific endoglin expression was studied in colorectal cancer resection specimens using IHC and related to metastases-free survival. Endoglin-mediated invasion was assessed in vitro by transwell invasion, using primary colorectal cancer-derived CAFs. Effects of CAF-specific endoglin expression on tumor cell invasion were investigated in a colorectal cancer zebrafish model, whereas liver metastases were assessed in a mouse model. RESULTS CAFs specifically at invasive borders of colorectal cancer express endoglin and increased expression intensity correlated with increased disease stage. Endoglin-expressing CAFs were also detected in lymph node and liver metastases, suggesting a role in colorectal cancer metastasis formation. In stage II colorectal cancer, CAF-specific endoglin expression at invasive borders correlated with poor metastasis-free survival. In vitro experiments revealed that endoglin is indispensable for bone morphogenetic protein (BMP)-9-induced signaling and CAF survival. Targeting endoglin using the neutralizing antibody TRC105 inhibited CAF invasion in vitro. In zebrafish, endoglin-expressing fibroblasts enhanced colorectal tumor cell infiltration into the liver and decreased survival. Finally, CAF-specific endoglin targeting with TRC105 decreased metastatic spread of colorectal cancer cells to the mouse liver. CONCLUSIONS Endoglin-expressing CAFs contribute to colorectal cancer progression and metastasis. TRC105 treatment inhibits CAF invasion and tumor metastasis, indicating an additional target beyond the angiogenic endothelium, possibly contributing to beneficial effects reported during clinical evaluations.See related commentary by Becker and LeBleu, p. 6110.
Collapse
Affiliation(s)
- Madelon Paauwe
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands.,Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, the Netherlands.,Department of Thrombosis & Hemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | - Mark J A Schoonderwoerd
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Roxan F C P Helderman
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tom J Harryvan
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Arwin Groenewoud
- Institute of Biology, Leiden University, Leiden, the Netherlands
| | - Gabi W van Pelt
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Rosalie Bor
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands.,Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Danielle M Hemmer
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Henri H Versteeg
- Department of Thrombosis & Hemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - James C H Hardwick
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Cornelis F M Sier
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands.,Oncode Institute, the Netherlands
| | - Lukas J A C Hawinkels
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands. .,Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
32
|
Kawase J, Aoki JY, Hamada K, Ozaki A, Araki K. Identification of Sex-associated SNPs of Greater Amberjack ( Seriola dumerili). J Genomics 2018; 6:53-62. [PMID: 29861788 PMCID: PMC5970132 DOI: 10.7150/jgen.24788] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 04/25/2018] [Indexed: 11/22/2022] Open
Abstract
The sex determination systems of fish are highly diverse compared with those of mammals. Thus, performing investigations using nonmodel fish species helps to understand the highly diverse sex determination systems of fish. Because greater amberjack (Seriola dumerili) is one of the most important edible fish globally and knowledge of its sex determination system is economically important in the field of aquaculture, we are interested in the mechanisms of sex determination of Seriola species. In this study, we identified sex-associated SNPs of greater amberjack using SNP information of 10 males and 10 females by an association test. We determined that the sex-associated SNPs were on chromosome 12 and mainly covered with two scaffolds (about 7.1 Mbp). Genotypes of sex-associated SNPs indicated that females are the heterogametic sex (ZZ/ZW). Furthermore, we compared the genomic structure of greater amberjack with those of Japanese amberjack (Seriola quinqueradiata), California yellowtail (Seriola dorsalis), and medaka (Oryzias latipes). Whole-genome alignments and synteny analysis indicated that the sex determination system of greater amberjack is markedly different from that of medaka and implied that the sex determination system is conserved in the Seriola species.
Collapse
Affiliation(s)
- Junya Kawase
- Department of Life Sciences, Graduate School of Bioresources, Mie University, 1577 Kurimamachiya-cho, Tsu City, Mie 514-8507, Japan
| | - Jun-Ya Aoki
- Research Center for Aquatic Breeding, National Research Institute of Aquaculture, Fisheries Research Agency, 224 Hiruda, Tamaki-cho, Watarai, Mie 519-0423, Japan
| | - Kazuhisa Hamada
- Marine Farm Laboratory Limited Company, 309 Takahiro, Tachibaura, Otsuki-cho, Hata-gun, Kochi 788-0352, Japan
| | - Akiyuki Ozaki
- Research Center for Aquatic Breeding, National Research Institute of Aquaculture, Fisheries Research Agency, 224 Hiruda, Tamaki-cho, Watarai, Mie 519-0423, Japan
| | - Kazuo Araki
- Research Center for Aquatic Breeding, National Research Institute of Aquaculture, Fisheries Research Agency, 224 Hiruda, Tamaki-cho, Watarai, Mie 519-0423, Japan
| |
Collapse
|
33
|
Antagonizing CD105 enhances radiation sensitivity in prostate cancer. Oncogene 2018; 37:4385-4397. [PMID: 29717261 PMCID: PMC6085281 DOI: 10.1038/s41388-018-0278-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 02/16/2018] [Accepted: 02/20/2018] [Indexed: 12/31/2022]
Abstract
Radiation therapy is the primary intervention for nearly half of the patients with localized advanced prostate cancer and standard of care for recurrent disease following surgery. The development of radiation-resistant disease is an obstacle for nearly 30–50% of patients undergoing radiotherapy. A better understanding of mechanisms that lead to radiation resistance could aid in the development of sensitizing agents to improve outcome. Here we identified a radiation-resistance pathway mediated by CD105, downstream of BMP and TGF-β signaling. Antagonizing CD105-dependent BMP signaling with a partially humanized monoclonal antibody, TRC105, resulted in a significant reduction in clonogenicity when combined with irradiation. In trying to better understand the mechanism for the radio-sensitization, we found that radiation-induced CD105/BMP signaling was sufficient and necessary for the upregulation of sirtuin 1 (SIRT1) in contributing to p53 stabilization and PGC-1α activation. Combining TRC105 with irradiation delayed DNA damage repair compared to irradiation alone. However, in the absence of p53 function, combining TRC105 and radiation resulted in no reduction in clonogenicity compared to radiation alone, despite similar reduction of DNA damage repair observed in p53-intact cells. This suggested DNA damage repair was not the sole determinant of CD105 radio-resistance. As cancer cells undergo an energy deficit following irradiation, due to the demands of DNA and organelle repair, we examined SIRT1’s role on p53 and PGC-1α with respect to glycolysis and mitochondrial biogenesis, respectively. Consequently, blocking the CD105-SIRT1 axis was found to deplete the ATP stores of irradiated cells and cause G2 cell cycle arrest. Xenograft models supported these findings that combining TRC105 with irradiation significantly reduces tumor size over irradiation alone (p value = 10−9). We identified a novel synthetic lethality strategy of combining radiation and CD105 targeting to address the DNA repair and metabolic addiction induced by irradiation in p53-functional prostate cancers.
Collapse
|
34
|
Duran D, Karschnia P, Gaillard JR, Karimy JK, Youngblood MW, DiLuna ML, Matouk CC, Aagaard-Kienitz B, Smith ER, Orbach DB, Rodesch G, Berenstein A, Gunel M, Kahle KT. Human genetics and molecular mechanisms of vein of Galen malformation. J Neurosurg Pediatr 2018; 21:367-374. [PMID: 29350590 DOI: 10.3171/2017.9.peds17365] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Vein of Galen malformations (VOGMs) are rare developmental cerebrovascular lesions characterized by fistulas between the choroidal circulation and the median prosencephalic vein. Although the treatment of VOGMs has greatly benefited from advances in endovascular therapy, including technical innovation in interventional neuroradiology, many patients are recalcitrant to procedural intervention or lack accessibility to specialized care centers, highlighting the need for improved screening, diagnostics, and therapeutics. A fundamental obstacle to identifying novel targets is the limited understanding of VOGM molecular pathophysiology, including its human genetics, and the lack of an adequate VOGM animal model. Herein, the known human mutations associated with VOGMs are reviewed to provide a framework for future gene discovery. Gene mutations have been identified in 2 Mendelian syndromes of which VOGM is an infrequent but associated phenotype: capillary malformation-arteriovenous malformation syndrome ( RASA1) and hereditary hemorrhagic telangiectasia ( ENG and ACVRL1). However, these mutations probably represent only a small fraction of all VOGM cases. Traditional genetic approaches have been limited in their ability to identify additional causative genes for VOGM because kindreds are rare, limited in patient number, and/or seem to have sporadic inheritance patterns, attributable in part to incomplete penetrance and phenotypic variability. The authors hypothesize that the apparent sporadic occurrence of VOGM may frequently be attributable to de novo mutation or incomplete penetrance of rare transmitted variants. Collaboration among treating physicians, patients' families, and investigators using next-generation sequencing could lead to the discovery of novel genes for VOGM. This could improve the understanding of normal vascular biology, elucidate the pathogenesis of VOGM and possibly other more common arteriovenous malformation subtypes, and pave the way for advances in the diagnosis and treatment of patients with VOGM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Beverly Aagaard-Kienitz
- 2Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin; Departments of
| | | | - Darren B Orbach
- 4Neurointerventional Radiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Georges Rodesch
- 5Service de Neuroradiologie Diagnostique et Thérapeutique, Hôpital Foch, Suresnes, France; and
| | - Alejandro Berenstein
- 6Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Murat Gunel
- 1Department of Neurosurgery.,7Department of Genetics.,8Centers for Mendelian Genomics and Yale Program on Neurogenetics, and
| | - Kristopher T Kahle
- 1Department of Neurosurgery.,8Centers for Mendelian Genomics and Yale Program on Neurogenetics, and.,9Department of Pediatrics and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
35
|
Goumans MJ, Zwijsen A, Ten Dijke P, Bailly S. Bone Morphogenetic Proteins in Vascular Homeostasis and Disease. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a031989. [PMID: 28348038 DOI: 10.1101/cshperspect.a031989] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is well established that control of vascular morphogenesis and homeostasis is regulated by vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), Delta-like 4 (Dll4), angiopoietin, and ephrin signaling. It has become clear that signaling by bone morphogenetic proteins (BMPs), which have a long history of studies in bone and early heart development, are also essential for regulating vascular function. Indeed, mutations that cause deregulated BMP signaling are linked to two human vascular diseases, hereditary hemorrhagic telangiectasia and pulmonary arterial hypertension. These observations are corroborated by data obtained with vascular cells in cell culture and in mouse models. BMPs are required for normal endothelial cell differentiation and for venous/arterial and lymphatic specification. In adult life, BMP signaling orchestrates neo-angiogenesis as well as vascular inflammation, remodeling, and calcification responses to shear and oxidative stress. This review emphasizes the pivotal role of BMPs in the vascular system, based on studies of mouse models and human vascular disorders.
Collapse
Affiliation(s)
- Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - An Zwijsen
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium.,KU Leuven Department of Human Genetics, 3000 Leuven, Belgium
| | - Peter Ten Dijke
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.,Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Sabine Bailly
- Institut National de la Santé et de la Recherche Mécale (INSERM), U1036, 38000 Grenoble, France.,Laboratoire Biologie du Cancer et de l'Infection, Commissariat à l'Énergie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France.,University of Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
36
|
Hepatitis C Virus Core Protein Modulates Endoglin (CD105) Signaling Pathway for Liver Pathogenesis. J Virol 2017; 91:JVI.01235-17. [PMID: 28794048 DOI: 10.1128/jvi.01235-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 08/06/2017] [Indexed: 12/11/2022] Open
Abstract
Endoglin is part of the TGF-β receptor complex and has a crucial role in fibrogenesis and angiogenesis. It is also an important protein for tumor growth, survival, and cancer cell metastasis. In a previous study, we have shown that hepatitis C virus (HCV) infection induces epithelial-mesenchymal transition (EMT) state and cancer stem-like cell (CSC) properties in human hepatocytes. Our array data suggested that endoglin (CD105) mRNA is significantly upregulated in HCV-associated CSCs. In this study, we have observed increased endoglin expression on the cell surface of an HCV core-expressing hepatocellular carcinoma (HepG2) cell line or immortalized human hepatocytes (IHH) and activation of its downstream signaling molecules. The status of phospho-SMAD1/5 and the expression of inhibitor of DNA binding protein 1 (ID1) were upregulated in HCV-infected cells or viral core gene-transfected cells. Additionally, we observed upregulation of endoglin/ID1 mRNA expression in chronic HCV patient liver biopsy samples. CSC generation by HCV core protein was dependent on the endoglin signaling pathway using activin receptor-like kinase 1 (ALK1) Fc blocking peptide and endoglin small interfering RNA (siRNA). Further, follow-up from in vitro analysis suggested that the antiapoptosis Bcl2 protein, proliferation-related cyclin D1 protein, and CSC-associated Hes1, Notch1, Nanog, and Sox2 proteins are enhanced during infection or ectopic expression of HCV core protein.IMPORTANCE Endoglin plays a crucial role in fibrogenesis and angiogenesis and is an important protein for tumor growth, survival, and cancer cell metastasis. Endoglin enhances ALK1-SMAD1/5 signaling in different cell types, leading to increased proliferation and migration responses. We have observed endoglin expression on the HCV core-expressing cell surface of human hepatocyte origin and activation of phospho-SMAD1/5 and ID1 downstream signaling molecules. ID1 protein plays a role in CSC properties, and we found that this pathway is important for antiapoptotic and cell proliferation signaling. Blocking of endoglin-ALK1-SMAD1/5 might be a good candidate for therapy for liver cancer stem cells together with liver cirrhosis.
Collapse
|
37
|
Tian H, Ketova T, Hardy D, Xu X, Gao X, Zijlstra A, Blobe GC. Endoglin Mediates Vascular Maturation by Promoting Vascular Smooth Muscle Cell Migration and Spreading. Arterioscler Thromb Vasc Biol 2017; 37:1115-1126. [PMID: 28450296 PMCID: PMC5444426 DOI: 10.1161/atvbaha.116.308859] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 04/19/2017] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Endoglin, a transforming growth factor-β superfamily coreceptor, is predominantly expressed in endothelial cells and has essential roles in vascular development. However, whether endoglin is also expressed in vascular smooth muscle cells (VSMCs), especially in vivo, remains controversial. Furthermore, the roles of endoglin in VSMC biology remain largely unknown. Our objective was to examine the expression and determine the function of endoglin in VSMCs during angiogenesis. Approach and Results— Here, we determine that endoglin is robustly expressed in VSMCs. Using CRISPR/CAS9 knockout and short hairpin RNA knockdown in the VSMC/endothelial coculture model system, we determine that endoglin in VSMCs, but not in endothelial cells, promotes VSMCs recruitment by the endothelial cells both in vitro and in vivo. Using an unbiased bioinformatics analysis of RNA sequencing data and further study, we determine that, mechanistically, endoglin mediates VSMC recruitment by promoting VSMC migration and spreading on endothelial cells via increasing integrin/FAK pathway signaling, whereas endoglin has minimal effects on VSMC adhesion to endothelial cells. In addition, we further determine that loss of endoglin in VSMCs inhibits VSMC recruitment in vivo. Conclusions— These studies demonstrate that endoglin has an important role in VSMC recruitment and blood vessel maturation during angiogenesis and also provide novel insights into how discordant endoglin function in endothelial and VSMCs may regulate vascular maturation and angiogenesis.
Collapse
Affiliation(s)
- Hongyu Tian
- From the Division of Medical Oncology, Department of Medicine (H.T., D.H., G.C.B.) and Department of Pharmacology and Cancer Biology (G.C.B.), Duke University Medical Center, Durham, NC; Department of Pathology, Microbiology, and Immunology (T.K., A.Z.) and Department of Cancer Biology (A.Z.), Vanderbilt University, Nashville, TN; Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC (X.X.); Department of Cell Biology, Duke University School of Medicine, Durham, NC (X.G.); and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN (A.Z.);
| | - Tatiana Ketova
- From the Division of Medical Oncology, Department of Medicine (H.T., D.H., G.C.B.) and Department of Pharmacology and Cancer Biology (G.C.B.), Duke University Medical Center, Durham, NC; Department of Pathology, Microbiology, and Immunology (T.K., A.Z.) and Department of Cancer Biology (A.Z.), Vanderbilt University, Nashville, TN; Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC (X.X.); Department of Cell Biology, Duke University School of Medicine, Durham, NC (X.G.); and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN (A.Z.)
| | - Duriel Hardy
- From the Division of Medical Oncology, Department of Medicine (H.T., D.H., G.C.B.) and Department of Pharmacology and Cancer Biology (G.C.B.), Duke University Medical Center, Durham, NC; Department of Pathology, Microbiology, and Immunology (T.K., A.Z.) and Department of Cancer Biology (A.Z.), Vanderbilt University, Nashville, TN; Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC (X.X.); Department of Cell Biology, Duke University School of Medicine, Durham, NC (X.G.); and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN (A.Z.)
| | - Xiaojiang Xu
- From the Division of Medical Oncology, Department of Medicine (H.T., D.H., G.C.B.) and Department of Pharmacology and Cancer Biology (G.C.B.), Duke University Medical Center, Durham, NC; Department of Pathology, Microbiology, and Immunology (T.K., A.Z.) and Department of Cancer Biology (A.Z.), Vanderbilt University, Nashville, TN; Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC (X.X.); Department of Cell Biology, Duke University School of Medicine, Durham, NC (X.G.); and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN (A.Z.)
| | - Xia Gao
- From the Division of Medical Oncology, Department of Medicine (H.T., D.H., G.C.B.) and Department of Pharmacology and Cancer Biology (G.C.B.), Duke University Medical Center, Durham, NC; Department of Pathology, Microbiology, and Immunology (T.K., A.Z.) and Department of Cancer Biology (A.Z.), Vanderbilt University, Nashville, TN; Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC (X.X.); Department of Cell Biology, Duke University School of Medicine, Durham, NC (X.G.); and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN (A.Z.)
| | - Andries Zijlstra
- From the Division of Medical Oncology, Department of Medicine (H.T., D.H., G.C.B.) and Department of Pharmacology and Cancer Biology (G.C.B.), Duke University Medical Center, Durham, NC; Department of Pathology, Microbiology, and Immunology (T.K., A.Z.) and Department of Cancer Biology (A.Z.), Vanderbilt University, Nashville, TN; Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC (X.X.); Department of Cell Biology, Duke University School of Medicine, Durham, NC (X.G.); and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN (A.Z.)
| | - Gerard C Blobe
- From the Division of Medical Oncology, Department of Medicine (H.T., D.H., G.C.B.) and Department of Pharmacology and Cancer Biology (G.C.B.), Duke University Medical Center, Durham, NC; Department of Pathology, Microbiology, and Immunology (T.K., A.Z.) and Department of Cancer Biology (A.Z.), Vanderbilt University, Nashville, TN; Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC (X.X.); Department of Cell Biology, Duke University School of Medicine, Durham, NC (X.G.); and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN (A.Z.);
| |
Collapse
|
38
|
Vorstenbosch J, Nguyen CM, Zhou S, Seo YJ, Siblini A, Finnson KW, Bizet AA, Tran SD, Philip A. Overexpression of CD109 in the Epidermis Differentially Regulates ALK1 Versus ALK5 Signaling and Modulates Extracellular Matrix Synthesis in the Skin. J Invest Dermatol 2016; 137:641-649. [PMID: 27866969 DOI: 10.1016/j.jid.2016.09.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 09/25/2016] [Indexed: 12/16/2022]
Abstract
Transforming growth factor-β (TGF-β) is a multifunctional growth factor involved in many physiological processes including wound healing and inflammation. Excessive TGF-β signaling in the skin has been implicated in fibrotic skin disorders such as keloids and scleroderma. We previously identified CD109 as a TGF-β co-receptor and inhibitor of TGF-β signaling and have shown that transgenic mice overexpressing CD109 in the epidermis display decreased scarring. In certain cell types, in addition to the canonical type I receptor, ALK5, which activates Smad2/3, TGF-β can signal through another type I receptor, ALK1, which activates Smad1/5. Here we demonstrate that ALK1 is expressed and co-localizes with CD109 in mouse keratinocytes and that mice overexpressing CD109 in the epidermis display enhanced ALK1-Smad1/5 signaling but decreased ALK5-Smad2/3 signaling, TGF-β expression, and extracellular matrix production in the skin when compared with wild-type littermates. Furthermore, treatment with conditioned media from isolated keratinocytes or epidermal explants from CD109 transgenic mouse skin leads to a decrease in extracellular matrix production in mouse skin fibroblasts. Taken together, our findings suggest that CD109 differentially regulates TGF-β-induced ALK1-Smad1/5 versus ALK5-Smad2/3 pathways, leading to decreased extracellular matrix production in the skin and that epidermal CD109 expression regulates dermal function through a paracrine mechanism.
Collapse
Affiliation(s)
- Joshua Vorstenbosch
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec, Canada
| | - Christopher M Nguyen
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec, Canada
| | - Shufeng Zhou
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec, Canada
| | - You Jung Seo
- Faculty of Dentistry, McGill University, McGill University, Montreal, Canada
| | - Aya Siblini
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec, Canada
| | - Kenneth W Finnson
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec, Canada
| | - Albane A Bizet
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec, Canada
| | - Simon D Tran
- Faculty of Dentistry, McGill University, McGill University, Montreal, Canada
| | - Anie Philip
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
39
|
Abstract
Transforming growth factor β (TGF-β) family members signal via heterotetrameric complexes of type I and type II dual specificity kinase receptors. The activation and stability of the receptors are controlled by posttranslational modifications, such as phosphorylation, ubiquitylation, sumoylation, and neddylation, as well as by interaction with other proteins at the cell surface and in the cytoplasm. Activation of TGF-β receptors induces signaling via formation of Smad complexes that are translocated to the nucleus where they act as transcription factors, as well as via non-Smad pathways, including the Erk1/2, JNK and p38 MAP kinase pathways, and the Src tyrosine kinase, phosphatidylinositol 3'-kinase, and Rho GTPases.
Collapse
Affiliation(s)
- Carl-Henrik Heldin
- Ludwig Institute for Cancer Research Ltd., Science for Life Laboratory, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Aristidis Moustakas
- Ludwig Institute for Cancer Research Ltd., Science for Life Laboratory, Uppsala University, SE-751 24 Uppsala, Sweden Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23 Uppsala, Sweden
| |
Collapse
|
40
|
Asai S, Otsuru S, Candela ME, Cantley L, Uchibe K, Hofmann TJ, Zhang K, Wapner KL, Soslowsky LJ, Horwitz EM, Enomoto-Iwamoto M. Tendon progenitor cells in injured tendons have strong chondrogenic potential: the CD105-negative subpopulation induces chondrogenic degeneration. Stem Cells 2015; 32:3266-77. [PMID: 25220576 DOI: 10.1002/stem.1847] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 05/15/2014] [Indexed: 12/29/2022]
Abstract
To study the cellular mechanism of the tendon repair process, we used a mouse Achilles tendon injury model to focus on the cells recruited to the injured site. The cells isolated from injured tendon 1 week after the surgery and uninjured tendons contained the connective tissue progenitor populations as determined by colony-forming capacity, cell surface markers, and multipotency. When the injured tendon-derived progenitor cells (inTPCs) were transplanted into injured Achilles tendons, they were not only integrated in the regenerating area expressing tenogenic phenotype but also trans-differentiated into chondrogenic cells in the degenerative lesion that underwent ectopic endochondral ossification. Surprisingly, the micromass culture of the inTPCs rapidly underwent chondrogenic differentiation even in the absence of exogenous bone morphogenetic proteins or TGFβs. The cells isolated from human ruptured tendon tissues also showed connective tissue progenitor properties and exhibited stronger chondrogenic ability than bone marrow stromal cells. The mouse inTPCs contained two subpopulations one positive and one negative for CD105, a coreceptor of the TGFβ superfamily. The CD105-negative cells showed superior chondrogenic potential in vitro and induced larger chondroid degenerative lesions in mice as compared to the CD105-positive cells. These findings indicate that tendon progenitor cells are recruited to the injured site of tendons and have a strong chondrogenic potential and that the CD105-negative population of these cells would be the cause for chondroid degeneration in injured tendons. The newly identified cells recruited to the injured tendon may provide novel targets to develop therapeutic strategies to facilitate tendon repair.
Collapse
Affiliation(s)
- Shuji Asai
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Koyama T, Ozaki A, Yoshida K, Suzuki J, Fuji K, Aoki JY, Kai W, Kawabata Y, Tsuzaki T, Araki K, Sakamoto T. Identification of Sex-Linked SNPs and Sex-Determining Regions in the Yellowtail Genome. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2015; 17:502-510. [PMID: 25975833 DOI: 10.1007/s10126-015-9636-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 04/15/2015] [Indexed: 06/04/2023]
Abstract
Unlike the conservation of sex-determining (SD) modes seen in most mammals and birds, teleost fishes exhibit a wide variety of SD systems and genes. Hence, the study of SD genes and sex chromosome turnover in fish is one of the most interesting topics in evolutionary biology. To increase resolution of the SD gene evolutionary trajectory in fish, identification of the SD gene in more fish species is necessary. In this study, we focused on the yellowtail, a species widely cultivated in Japan. It is a member of family Carangidae in which no heteromorphic sex chromosome has been observed, and no SD gene has been identified to date. By performing linkage analysis and BAC walking, we identified a genomic region and SNPs with complete linkage to yellowtail sex. Comparative genome analysis revealed the yellowtail SD region ancestral chromosome structure as medaka-fugu. Two inversions occurred in the yellowtail linage after it diverged from the yellowtail-medaka ancestor. An association study using wild yellowtails and the SNPs developed from BAC ends identified two SNPs that can reasonably distinguish the sexes. Therefore, these will be useful genetic markers for yellowtail breeding. Based on a comparative study, it was suggested that a PDZ domain containing the GIPC protein might be involved in yellowtail sex determination. The homomorphic sex chromosomes widely observed in the Carangidae suggest that this family could be a suitable marine fish model to investigate the early stages of sex chromosome evolution, for which our results provide a good starting point.
Collapse
Affiliation(s)
- Takashi Koyama
- Faculty of Marine Science, Tokyo University of Marine Science and Technology, 4-5-7, Konan, Minato-ku, Tokyo, 108-8477, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mężyk-Kopeć R, Wyroba B, Stalińska K, Próchnicki T, Wiatrowska K, Kilarski WW, Swartz MA, Bereta J. ADAM17 Promotes Motility, Invasion, and Sprouting of Lymphatic Endothelial Cells. PLoS One 2015; 10:e0132661. [PMID: 26176220 PMCID: PMC4503755 DOI: 10.1371/journal.pone.0132661] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 06/18/2015] [Indexed: 02/04/2023] Open
Abstract
Tumor-associated lymphatic vessels actively participate in tumor progression and dissemination. ADAM17, a sheddase for numerous growth factors, cytokines, receptors, and cell adhesion molecules, is believed to promote tumor development, facilitating both tumor cell proliferation and migration, as well as tumor angiogenesis. In this work we addressed the issue of whether ADAM17 may also promote tumor lymphangiogenesis. First, we found that ADAM17 is important for the migratory potential of immortalized human dermal lymphatic endothelial cells (LEC). When ADAM17 was stably silenced in LEC, their proliferation was not affected, but: (i) single-cell motility, (ii) cell migration through a 3D Matrigel/collagen type I matrix, and (iii) their ability to form sprouts in a 3D matrix were significantly diminished. The differences in the cell motility between ADAM17-proficient and ADAM17-silenced cells were eliminated by inhibitors of EGFR and HER2, indicating that ADAM17-mediated shedding of growth factors accounts for LEC migratory potential. Interestingly, ADAM17 depletion affected the integrin surface expression/functionality in LEC. ADAM17-silenced cells adhered to plastic, type I collagen, and fibronectin faster than their ADAM17-proficient counterparts. The difference in adhesion to fibronectin was abolished by a cyclic RGD peptide, emphasizing the involvement of integrins in the process. Using a soluble receptor array, we identified BIG-H3 among several candidate proteins involved in the phenotypic and behavioral changes of LEC upon ADAM17 silencing. In additional assays, we confirmed the increased expression of BIG-H3, as well as TGFβ2 in ADAM17-silenced LEC. The antilymphangiogenic effects of ADAM17 silencing in lymphatic endothelial cells suggest further relevance of ADAM17 as a potential target in cancer therapy.
Collapse
Affiliation(s)
- Renata Mężyk-Kopeć
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
- Institute of Bioengineering and Swiss Institute for Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute for Molecular Engineering, University of Chicago, Chicago, Illinois, United States of America
| | - Barbara Wyroba
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Krystyna Stalińska
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Tomasz Próchnicki
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Karolina Wiatrowska
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Witold W. Kilarski
- Institute of Bioengineering and Swiss Institute for Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute for Molecular Engineering, University of Chicago, Chicago, Illinois, United States of America
| | - Melody A. Swartz
- Institute of Bioengineering and Swiss Institute for Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute for Molecular Engineering, University of Chicago, Chicago, Illinois, United States of America
| | - Joanna Bereta
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
- * E-mail:
| |
Collapse
|
43
|
VEGF, Notch and TGFβ/BMPs in regulation of sprouting angiogenesis and vascular patterning. Biochem Soc Trans 2015; 42:1576-83. [PMID: 25399573 DOI: 10.1042/bst20140231] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The blood vasculature is constantly adapting to meet the demand from tissue. In so doing, branches may form, reorganize or regress. These complex processes employ integration of multiple signalling cascades, some of them being restricted to endothelial and mural cells and, hence, suitable for targeting of the vasculature. Both genetic and drug targeting experiments have demonstrated the requirement for the vascular endothelial growth factor (VEGF) system, the Delta-like-Notch system and the transforming growth factor β (TGFβ)/bone morphogenetic protein (BMP) cascades in vascular development. Although several of these signalling cascades in part converge into common downstream components, they differ in temporal and spatial regulation and expression. For example, the pro-angiogenic VEGFA is secreted by cells in need of oxygen, presented to the basal side of the endothelium, whereas BMP9 and BMP10 are supplied via the bloodstream in constant interaction with the apical side to suppress angiogenesis. Delta-like 4 (DLL4), on the other hand, is provided as an endothelial membrane bound ligand. In the present article, we discuss recent data on the integration of these pathways in the process of sprouting angiogenesis and vascular patterning and malformation.
Collapse
|
44
|
Pomeraniec L, Hector-Greene M, Ehrlich M, Blobe GC, Henis YI. Regulation of TGF-β receptor hetero-oligomerization and signaling by endoglin. Mol Biol Cell 2015; 26:3117-27. [PMID: 26157163 PMCID: PMC4551323 DOI: 10.1091/mbc.e15-02-0069] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 06/30/2015] [Indexed: 11/23/2022] Open
Abstract
Endoglin is a modulator of TGF-β signaling in endothelial cells. We show that it forms stable homodimers serving as a scaffold for binding TβRII, ALK5, and ALK1. ALK1 and ALK5 bind endoglin differentially, with TβRII recruiting ALK5. Signaling data indicate a role for this receptor complex in balancing TGF-β signaling between Smad1/5/8 and Smad2/3. Complex formation among transforming growth factor-β (TGF-β) receptors and its modulation by coreceptors represent an important level of regulation for TGF-β signaling. Oligomerization of ALK5 and the type II TGF-β receptor (TβRII) has been thoroughly investigated, both in vitro and in intact cells. However, such studies, especially in live cells, are missing for the endothelial cell coreceptor endoglin and for the ALK1 type I receptor, which enables endothelial cells to respond to TGF-β by activation of both Smad2/3 and Smad1/5/8. Here we combined immunoglobulin G–mediated immobilization of one cell-surface receptor with lateral mobility studies of a coexpressed receptor by fluorescence recovery after photobleaching (FRAP) to demonstrate that endoglin forms stable homodimers that function as a scaffold for binding TβRII, ALK5, and ALK1. ALK1 and ALK5 bind to endoglin with differential dependence on TβRII, which plays a major role in recruiting ALK5 to the complex. Signaling data indicate a role for the quaternary receptor complex in regulating the balance between TGF-β signaling to Smad1/5/8 and to Smad2/3.
Collapse
Affiliation(s)
- Leslie Pomeraniec
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | | | - Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Gerard C Blobe
- Department of Medicine, Duke University Medical Center, Durham, NC 27708
| | - Yoav I Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
45
|
Jerkic M, Letarte M. Increased endothelial cell permeability in endoglin-deficient cells. FASEB J 2015; 29:3678-88. [PMID: 25972355 DOI: 10.1096/fj.14-269258] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 05/04/2015] [Indexed: 01/12/2023]
Abstract
Endoglin (ENG) is a TGF-β superfamily coreceptor essential for vascular endothelium integrity. ENG mutations lead to a vascular dysplasia associated with frequent hemorrhages in multiple organs, whereas ENG null mouse embryos die at midgestation with impaired heart development and leaky vasculature. ENG interacts with several proteins involved in cell adhesion, and we postulated that it regulates vascular permeability. The current study assessed the permeability of ENG homozygous null (Eng(-/-)), heterozygous (Eng(+/-)), and normal (Eng(+/+)) mouse embryonic endothelial cell (EC) lines. Permeability, measured by passage of fluorescent dextran through EC monolayers, was increased 2.9- and 1.7-fold for Eng(-/-) and Eng(+/-) ECs, respectively, compared to control ECs and was not increased by TGF-β1 or VEGF. Prolonged starvation increased Eng(-/-) EC permeability by 3.7-fold with no effect on control ECs; neutrophils transmigrated faster through Eng(-/-) than Eng(+/+) monolayers. Using a pull-down assay, we demonstrate that Ras homolog gene family (Rho) A is constitutively active in Eng(-/-) and Eng(+/-) ECs. We show that the endothelial barrier destabilizing factor thrombospondin-1 and its receptor-like protein tyrosine phosphatase are increased, whereas stabilizing factors VEGF receptor 2, vascular endothelial-cadherin, p21-activated kinase, and Ras-related C3 botulinum toxin substrate 2 are decreased in Eng(-/-) cells. Our findings indicate that ENG deficiency leads to EC hyperpermeability through constitutive activation of RhoA and destabilization of endothelial barrier function.
Collapse
Affiliation(s)
- Mirjana Jerkic
- *Molecular Structure and Function Program, The Hospital for Sick Children, Toronto, Ontario, Canada; and Department of Immunology and Keenan Research Centre for Biomedical Science, Anesthesia Research, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Michelle Letarte
- *Molecular Structure and Function Program, The Hospital for Sick Children, Toronto, Ontario, Canada; and Department of Immunology and Keenan Research Centre for Biomedical Science, Anesthesia Research, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
46
|
Gatza CE, Elderbroom JL, Oh SY, Starr MD, Nixon AB, Blobe GC. The balance of cell surface and soluble type III TGF-β receptor regulates BMP signaling in normal and cancerous mammary epithelial cells. Neoplasia 2015; 16:489-500. [PMID: 25077702 PMCID: PMC4198744 DOI: 10.1016/j.neo.2014.05.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 05/12/2014] [Accepted: 05/19/2014] [Indexed: 12/20/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are members of the TGF-β superfamily that are over-expressed in breast cancer, with context dependent effects on breast cancer pathogenesis. The type III TGF-β receptor (TβRIII) mediates BMP signaling. While TβRIII expression is lost during breast cancer progression, the role of TβRIII in regulating BMP signaling in normal mammary epithelium and breast cancer cells has not been examined. Restoring TβRIII expression in a 4T1 murine syngeneic model of breast cancer suppressed Smad1/5/8 phosphorylation and inhibited the expression of the BMP transcriptional targets, Id1 and Smad6, in vivo. Similarly, restoring TβRIII expression in human breast cancer cell lines or treatment with sTβRIII inhibited BMP-induced Smad1/5/8 phosphorylation and BMP-stimulated migration and invasion. In normal mammary epithelial cells, shRNA-mediated silencing of TβRIII, TβRIII over-expression, or treatment with sTβRIII inhibited BMP-mediated phosphorylation of Smad1/5/8 and BMP induced migration. Inhibition of TβRIII shedding through treatment with TAPI-2 or expression of a non-shedding TβRIII mutant rescued TβRIII mediated inhibition of BMP induced Smad1/5/8 phosphorylation and BMP induced migration and/or invasion in both in normal mammary epithelial cells and breast cancer cells. Conversely, expression of a TβRIII mutant, which exhibited increased shedding, significantly reduced BMP-mediated Smad1/5/8 phosphorylation, migration, and invasion. These data demonstrate that TβRIII regulates BMP-mediated signaling and biological effects, primarily through the ligand sequestration effects of sTβRIII in normal and cancerous mammary epithelial cells and suggest that the ratio of membrane bound versus sTβRIII plays an important role in mediating these effects.
Collapse
Affiliation(s)
| | - Jennifer L Elderbroom
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC
| | - Sun Young Oh
- Department of Medicine, Duke University Medical Center, Durham, NC
| | - Mark D Starr
- Department of Medicine, Duke University Medical Center, Durham, NC
| | - Andrew B Nixon
- Department of Medicine, Duke University Medical Center, Durham, NC
| | - Gerard C Blobe
- Department of Medicine, Duke University Medical Center, Durham, NC; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC.
| |
Collapse
|
47
|
Abstract
Tubulointerstitial fibrosis and glomerulosclerosis, are a major feature of end stage chronic kidney disease (CKD), characterised by an excessive accumulation of extracellular matrix (ECM) proteins. Transforming growth factor beta-1 (TGF-β1) is a cytokine with an important role in many steps of renal fibrosis such as myofibroblast activation and proliferation, ECM protein synthesis and inflammatory cell infiltration. Endoglin is a TGF-β co-receptor that modulates TGF-β responses in different cell types. In numerous cells types, such as mesangial cells or myoblasts, endoglin regulates negatively TGF-β-induced ECM protein expression. However, recently it has been demonstrated that 'in vivo' endoglin promotes fibrotic responses. Furthermore, several studies have demonstrated an increase of endoglin expression in experimental models of renal fibrosis in the kidney and other tissues. Nevertheless, the role of endoglin in renal fibrosis development is unclear and a question arises: Does endoglin protect against renal fibrosis or promotes its development? The purpose of this review is to critically analyse the recent knowledge relating to endoglin and renal fibrosis. Knowledge of endoglin role in this pathology is necessary to consider endoglin as a possible therapeutic target against renal fibrosis.
Collapse
|
48
|
Pal K, Pletnev AA, Dutta SK, Wang E, Zhao R, Baral A, Yadav VK, Aggarwal S, Krishnaswamy S, Alkharfy KM, Chowdhury S, Spaller MR, Mukhopadhyay D. Inhibition of endoglin-GIPC interaction inhibits pancreatic cancer cell growth. Mol Cancer Ther 2014; 13:2264-75. [PMID: 25125675 PMCID: PMC4229952 DOI: 10.1158/1535-7163.mct-14-0291] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Endoglin, a 180-kDa disulfide-linked homodimeric transmembrane receptor protein mostly expressed in tumor-associated endothelial cells, is an endogenous binding partner of GAIP-interacting protein, C terminus (GIPC). Endoglin functions as a coreceptor of TβRII that binds TGFβ and is important for vascular development, and consequently has become a compelling target for antiangiogenic therapies. A few recent studies in gastrointestinal stromal tumor (GIST), breast cancer, and ovarian cancer, however, suggest that endoglin is upregulated in tumor cells and is associated with poor prognosis. These findings indicate a broader role of endoglin in tumor biology, beyond angiogenic effects. The goal of our current study is to evaluate the effects of targeting endoglin in pancreatic cancer both in vitro and in vivo. We analyzed the antiproliferative effect of both RNAi-based and peptide ligand-based inhibition of endoglin in pancreatic cancer cell lines, the latter yielding a GIPC PDZ domain-targeting lipopeptide with notable antiproliferative activity. We further demonstrated that endoglin inhibition induced a differentiation phenotype in the pancreatic cancer cells and sensitized them against conventional chemotherapeutic drug gemcitabine. Most importantly, we have demonstrated the antitumor effect of both RNAi-based and competitive inhibitor-based blocking of endoglin in pancreatic cancer xenograft models in vivo. To our knowledge, this is the first report exploring the effect of targeting endoglin in pancreatic cancer cells.
Collapse
Affiliation(s)
- Krishnendu Pal
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Alexandre A Pletnev
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth and Norris Cotton Cancer Center, Lebanon, New Hampshire
| | - Shamit K Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Ruizhi Zhao
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth and Norris Cotton Cancer Center, Lebanon, New Hampshire
| | - Aradhita Baral
- Proteomics and Structural Biology Unit, Institute of Genomics and Integrative Biology, Council for Scientific and Industrial Research, New Delhi, India
| | - Vinod Kumar Yadav
- G.N.R. Knowledge Center for Genome Informatics, Institute of Genomics and Integrative Biology, Council for Scientific and Industrial Research, New Delhi, India
| | - Suruchi Aggarwal
- G.N.R. Knowledge Center for Genome Informatics, Institute of Genomics and Integrative Biology, Council for Scientific and Industrial Research, New Delhi, India
| | | | - Khalid M Alkharfy
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota. Department of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Shantanu Chowdhury
- Proteomics and Structural Biology Unit, Institute of Genomics and Integrative Biology, Council for Scientific and Industrial Research, New Delhi, India. G.N.R. Knowledge Center for Genome Informatics, Institute of Genomics and Integrative Biology, Council for Scientific and Industrial Research, New Delhi, India
| | - Mark R Spaller
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth and Norris Cotton Cancer Center, Lebanon, New Hampshire
| | | |
Collapse
|
49
|
Solomon O, Bazak L, Levanon EY, Amariglio N, Unger R, Rechavi G, Eyal E. Characterizing of functional human coding RNA editing from evolutionary, structural, and dynamic perspectives. Proteins 2014; 82:3117-31. [DOI: 10.1002/prot.24672] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/28/2014] [Accepted: 08/11/2014] [Indexed: 12/29/2022]
Affiliation(s)
- Oz Solomon
- Cancer Research Center; Chaim Sheba Medical Center; Tel Hashomer 52621 Ramat Gan Israel
- The Everard & Mina Goodman Faculty of Life Sciences; Bar-Ilan University; Ramat Gan 52900 Israel
| | - Lily Bazak
- The Everard & Mina Goodman Faculty of Life Sciences; Bar-Ilan University; Ramat Gan 52900 Israel
| | - Erez Y. Levanon
- The Everard & Mina Goodman Faculty of Life Sciences; Bar-Ilan University; Ramat Gan 52900 Israel
| | - Ninette Amariglio
- Cancer Research Center; Chaim Sheba Medical Center; Tel Hashomer 52621 Ramat Gan Israel
| | - Ron Unger
- The Everard & Mina Goodman Faculty of Life Sciences; Bar-Ilan University; Ramat Gan 52900 Israel
| | - Gideon Rechavi
- Cancer Research Center; Chaim Sheba Medical Center; Tel Hashomer 52621 Ramat Gan Israel
- Sackler School of Medicine; Tel Aviv University; Tel Aviv 69978 Israel
| | - Eran Eyal
- Cancer Research Center; Chaim Sheba Medical Center; Tel Hashomer 52621 Ramat Gan Israel
| |
Collapse
|
50
|
Investigation of endoglin wild-type and missense mutant protein heterodimerisation using fluorescence microscopy based IF, BiFC and FRET analyses. PLoS One 2014; 9:e102998. [PMID: 25080347 PMCID: PMC4117486 DOI: 10.1371/journal.pone.0102998] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 06/25/2014] [Indexed: 11/30/2022] Open
Abstract
The homodimeric transmembrane receptor endoglin (CD105) plays an important role in angiogenesis. This is highlighted by mutations in its gene, causing the vascular disorder HHT1. The main role of endoglin function has been assigned to the modulation of transforming growth factor β and bone morphogenetic protein signalling in endothelial cells. Nevertheless, other functions of endoglin have been revealed to be involved in different cellular functions and in other cell types than endothelial cells. Compared to the exploration of its natural function, little experimental data have been gathered about the mode of action of endoglin HHT mutations at the cellular level, especially missense mutations, and to what degree these might interfere with normal endoglin function. In this paper, we have used fluorescence-based microscopic techniques, such as bimolecular fluorescence complementation (BiFC), immunofluorescence staining with the endoglin specific monoclonal antibody SN6, and protein interaction studies by Förster Resonance Energy Transfer (FRET) to investigate the formation and cellular localisation of possible homo- and heterodimers composed of endoglin wild-type and endoglin missense mutant proteins. The results show that all of the investigated missense mutants dimerise with themselves, as well as with wild-type endoglin, and localise, depending on the position of the affected amino acid, either in the rough endoplasmic reticulum (rER) or in the plasma membrane of the cells. We show that the rER retained mutants reduce the amount of endogenous wild-type endoglin on the plasma membrane through interception in the rER when transiently or stably expressed in HMEC-1 endothelial cells. As a result of this, endoglin modulated TGF-β1 signal transduction is also abrogated, which is not due to TGF-β receptor ER trafficking interference. Protein interaction analyses by FRET show that rER located endoglin missense mutants do not perturb protein processing of other membrane receptors, such as TβRII, ALK5 or ALK1.
Collapse
|