1
|
Fan W, Bradford TM, Török NJ. Metabolic dysfunction-associated liver disease and diabetes: Matrix remodeling, fibrosis, and therapeutic implications. Ann N Y Acad Sci 2024; 1538:21-33. [PMID: 38996214 DOI: 10.1111/nyas.15184] [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] [Indexed: 07/14/2024]
Abstract
Metabolic dysfunction-associated liver disease (MASLD) and steatohepatitis (MASH) are becoming the most common causes of chronic liver disease in the United States and worldwide due to the obesity and diabetes epidemics. It is estimated that by 2030 close to 100 million people might be affected and patients with type 2 diabetes are especially at high risk. Twenty to 30% of patients with MASLD can progress to MASH, which is characterized by steatosis, necroinflammation, hepatocyte ballooning, and in advanced cases, fibrosis progressing to cirrhosis. Clinically, it is recognized that disease progression in diabetic patients is accelerated and the role of various genetic and epigenetic factors, as well as cell-matrix interactions in fibrosis and stromal remodeling, have recently been recognized. While there has been great progress in drug development and clinical trials for MASLD/MASH, the complexity of these pathways highlights the need to improve diagnosis/early detection and develop more successful antifibrotic therapies that not only prevent but reverse fibrosis.
Collapse
Affiliation(s)
- Weiguo Fan
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California, USA
- Palo Alto VA Medical Center, Palo Alto, California, USA
| | - Toby M Bradford
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California, USA
| | - Natalie J Török
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California, USA
- Palo Alto VA Medical Center, Palo Alto, California, USA
| |
Collapse
|
2
|
Lee DH, Imran M, Choi JH, Park YJ, Kim YH, Min S, Park TJ, Choi YW. CDK4/6 inhibitors induce breast cancer senescence with enhanced anti-tumor immunogenic properties compared with DNA-damaging agents. Mol Oncol 2024; 18:216-232. [PMID: 37854019 PMCID: PMC10766199 DOI: 10.1002/1878-0261.13541] [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: 05/22/2023] [Revised: 10/05/2023] [Accepted: 10/17/2023] [Indexed: 10/20/2023] Open
Abstract
Since therapy-induced senescence (TIS) can either support or inhibit cancer progression, identifying which types of chemotherapeutic agents can produce the strongest anti-tumor TIS is an important issue. Here, cyclin-dependent kinase4/6 inhibitors (CDK4/6i)-induced senescence was compared to the TIS induced by conventional DNA-damaging agents. Despite both types of agents eliciting a similar degree of senescence, we observed increased expression of the senescence-associated secretory phenotype (SASP) and ligands related to pro-tumor immunity (IL6, CXCL8, TGFβ, CD274, and CEACAM1) and angiogenesis (VEGFA) mainly in TIS induced by DNA-damaging agents rather than by CDK4/6i. Additionally, although all agents increased the expression of anti-tumor immunomodulatory proteins related to antigen presentation (MHC-I, B2M) and T cell chemokines (CXCL9, 10, 11), CDK4/6i-induced senescent cells still maintained this expression at a similar or even higher intensity than cells treated with DNA-damaging agents, despite the absence of nuclear factor-kappa-B (NF-κB) and p53 activation. These data suggest that in contrast with DNA-damaging agents, which augment the pro-tumorigenic microenvironment via pro-inflammatory SASP, CDK4/6i can generate TIS only with antitumor immunomodulatory proteins.
Collapse
Affiliation(s)
- Dong Hyun Lee
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwonKorea
- Department of Biomedical SciencesAjou University Graduate School of MedicineSuwonKorea
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
| | - Muhammad Imran
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwonKorea
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
| | - Jae Ho Choi
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
- Department of Hematology‐OncologyAjou University School of MedicineSuwonKorea
| | - Yoo Jung Park
- Department of Hematology‐OncologyAjou University School of MedicineSuwonKorea
| | - Young Hwa Kim
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
| | - Sunwoo Min
- Department of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)DaejeonKorea
| | - Tae Jun Park
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwonKorea
- Department of Biomedical SciencesAjou University Graduate School of MedicineSuwonKorea
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
| | - Yong Won Choi
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
- Department of Hematology‐OncologyAjou University School of MedicineSuwonKorea
| |
Collapse
|
3
|
Gençoğlu Ş. Circulating Podocalyxin, Tumstatin/Col-IVα3 and Chitinase 1: New Culprits in Vitiligo Occurrence. Dermatol Pract Concept 2024; 14:dpc.1401a95. [PMID: 38236996 PMCID: PMC10868821 DOI: 10.5826/dpc.1401a95] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2023] [Indexed: 02/16/2024] Open
Abstract
INTRODUCTION The roles of anti-adhesive podocalyxin (PODXL), anti-angiogenetic tumstatin/ Col-IVα3 and neuro-inflammation and innate immunity modulator Chitinase 1 (CHIT-1) in the etiology of vitiligo have not been studied yet. OBJECTIVES This study was planned to detect changes in serum PODXL, tumstatin/Col-IVα3 and CHIT1 levels in vitiligo patients. METHODS This case-controlled study was performed on a total of 50 patients, 25 with vitiligo and 25 healthy controls. Participants in the vitiligo and control groups were matched in pairs for age and sex. At least 8-10 hours of overnight fasting, venous blood samples were taken from the participants in both groups and serum levels of podocalyxin, tumstatin/Col-IVα3 and CHIT 1 levels were measured by sandwich enzyme immunoassay. RESULTS In the classification made according to the vitiligo European Task Force evaluation criteria, 18 of 25 vitiligo patients were in the slowly progressive phase and 7 patients were in the active progressive phase. Serum podocalyxin levels increased significantly in the vitiligo group compared to the controls (7.03±2.09 ng/ml vs. 4.99±1.20 ng/ml, p<0.02). However, serum tumstatin levels in vitiligo patients showed a significantly lower course compared to controls (4.88±1.76 ng/ml vs. 6.05±2.19 ng/nl, p<0.03). Serum CHIT-1 levels of vitiligo patients (42.4±7.22 ng/ml) were found to be significantly higher than the serum levels of the control group (34.5±5.33 ng/ml) (p<0.01). CONCLUSION High podocalyxin and CHIT1versus low tumstatin levels are new biomarkers that support the role of anti-adhesive, anti-angiogenic and neuroinflammatory pathways in the formation of vitiligo.
Collapse
Affiliation(s)
- Şule Gençoğlu
- Department of Dermatology, Gozde Academy Hospitals, Malatya, Turkey
| |
Collapse
|
4
|
Eckersley A, Yamamura T, Lennon R. Matrikines in kidney ageing and age-related disease. Curr Opin Nephrol Hypertens 2023; 32:551-558. [PMID: 37584348 PMCID: PMC10552846 DOI: 10.1097/mnh.0000000000000916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
PURPOSE OF REVIEW Matrikines are cell-signalling extracellular matrix fragments and they have attracted recent attention from basic and translational scientists, due to their diverse roles in age-related disease and their potential as therapeutic agents. In kidney, the matrix undergoes remodelling by proteolytic fragmentation, so matrikines are likely to play a substantial, yet understudied, role in ageing and pathogenesis of age-related diseases. RECENT FINDINGS This review presents an up-to-date description of known matrikines with either a confirmed or highly anticipated role in kidney ageing and disease, including their point of origin, mechanism of cleavage, a summary of known biological actions and the current knowledge which links them to kidney health. We also highlight areas of interest, such as the prospect of matrikine cross-tissue communication, and gaps in knowledge, such as the unexplored signalling potential of many kidney disease-specific matrix fragments. SUMMARY We anticipate that knowledge of specific matrikines, and their roles in controlling processes of kidney pathology, could be leveraged for the development of exciting new future therapies through inhibition or even with their supplementation.
Collapse
Affiliation(s)
- Alexander Eckersley
- Division of Musculoskeletal & Dermatological Sciences, School of Biological Science
| | - Tomohiko Yamamura
- Wellcome Centre for Cell-Matrix Research, School of Biological Science, Faculty of Biology, Medicine and Health, The University of Manchester
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, School of Biological Science, Faculty of Biology, Medicine and Health, The University of Manchester
- Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| |
Collapse
|
5
|
Fließer E, Lins T, Berg JL, Kolb M, Kwapiszewska G. The endothelium in lung fibrosis: a core signaling hub in disease pathogenesis? Am J Physiol Cell Physiol 2023; 325:C2-C16. [PMID: 37184232 DOI: 10.1152/ajpcell.00097.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/05/2023] [Accepted: 05/05/2023] [Indexed: 05/16/2023]
Abstract
Pulmonary fibrosis (PF) is a progressive chronic lung disease characterized by excessive deposition of extracellular matrix (ECM) and structural destruction, associated with a severe 5-year mortality rate. The onset of the disease is thought to be triggered by chronic damage to the alveolar epithelium. Since the pulmonary endothelium is an important component of the alveolar-capillary niche, it is also affected by the initial injury. In addition to ensuring proper gas exchange, the endothelium has critical functional properties, including regulation of vascular tone, inflammatory responses, coagulation, and maintenance of vascular homeostasis and integrity. Recent single-cell analyses have shown that shifts in endothelial cell (EC) subtypes occur in PF. Furthermore, the increased vascular remodeling associated with PF leads to deteriorated outcomes for patients, underscoring the importance of the vascular bed in PF. To date, the causes and consequences of endothelial and vascular involvement in lung fibrosis are poorly understood. Therefore, it is of great importance to investigate the involvement of EC and the vascular system in the pathogenesis of the disease. In this review, we will outline the current knowledge on the role of the pulmonary vasculature in PF, in terms of abnormal cellular interactions, hyperinflammation, vascular barrier disorders, and an altered basement membrane composition. Finally, we will summarize recent advances in extensive therapeutic research and discuss the significant value of novel therapies targeting the endothelium.
Collapse
Affiliation(s)
- Elisabeth Fließer
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Thomas Lins
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Johannes Lorenz Berg
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Martin Kolb
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Member of the German Lung Center (DZL), Cardiopulmonary Institute (CPI), Giessen, Germany
| |
Collapse
|
6
|
LeBleu VS, Dai J, Tsutakawa S, MacDonald BA, Alge JL, Sund M, Xie L, Sugimoto H, Tainer J, Zon LI, Kalluri R. Identification of unique α4 chain structure and conserved antiangiogenic activity of α3NC1 type IV collagen in zebrafish. Dev Dyn 2023; 252:1046-1060. [PMID: 37002899 PMCID: PMC10524752 DOI: 10.1002/dvdy.590] [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: 08/14/2022] [Revised: 01/17/2023] [Accepted: 02/28/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND Type IV collagen is an abundant component of basement membranes in all multicellular species and is essential for the extracellular scaffold supporting tissue architecture and function. Lower organisms typically have two type IV collagen genes, encoding α1 and α2 chains, in contrast with the six genes in humans, encoding α1-α6 chains. The α chains assemble into trimeric protomers, the building blocks of the type IV collagen network. The detailed evolutionary conservation of type IV collagen network remains to be studied. RESULTS We report on the molecular evolution of type IV collagen genes. The zebrafish α4 non-collagenous (NC1) domain, in contrast with its human ortholog, contains an additional cysteine residue and lacks the M93 and K211 residues involved in sulfilimine bond formation between adjacent protomers. This may alter α4 chain interactions with other α chains, as supported by temporal and anatomic expression patterns of collagen IV chains during the zebrafish development. Despite the divergence between zebrafish and human α3 NC1 domain (endogenous angiogenesis inhibitor, Tumstatin), the zebrafish α3 NC1 domain exhibits conserved antiangiogenic activity in human endothelial cells. CONCLUSIONS Our work supports type IV collagen is largely conserved between zebrafish and humans, with a possible difference involving the α4 chain.
Collapse
Affiliation(s)
- Valerie S LeBleu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Feinberg School of Medicine and Kellogg School of Management, Northwestern University, Chicago, Illinois, USA
- Division of Matrix Biology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Jianli Dai
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Susan Tsutakawa
- Lawrence Berkeley National Laboratory, University of California, Berkeley, California, USA
| | - Brian A MacDonald
- Division of Matrix Biology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Joseph L Alge
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Malin Sund
- Division of Matrix Biology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Liang Xie
- Division of Matrix Biology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Hikaru Sugimoto
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Matrix Biology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - John Tainer
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Leonard I Zon
- Department of Hematology/Oncology, Children's Hospital, Boston, Massachusetts, USA
| | - Raghu Kalluri
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Matrix Biology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Bioengineering, Rice University, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
7
|
Das S, Dash BS, Premji TP, Chen JP. Immunotherapeutic Approaches for the Treatment of Glioblastoma Multiforme: Mechanism and Clinical Applications. Int J Mol Sci 2023; 24:10546. [PMID: 37445721 DOI: 10.3390/ijms241310546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Glioma is one of the most aggressive types of primary brain tumor with a high-grade glioma known as glioblastoma multiforme (GBM). Patients diagnosed with GBM usually have an overall survival rate of less than 18 months after conventional therapy. This bleak prognosis underlines the need to consider new therapeutic interventions for GBM treatment to overcome current treatment limitations. By highlighting different immunotherapeutic approaches currently in preclinical and clinical trials, including immune checkpoint inhibitors, chimeric antigen receptors T cells, natural killer cells, vaccines, and combination therapy, this review aims to discuss the mechanisms, benefits, and limitations of immunotherapy in treating GBM patients.
Collapse
Affiliation(s)
- Suprava Das
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Banendu Sunder Dash
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Thejas P Premji
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan
- Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-San, Taoyuan 33305, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan
| |
Collapse
|
8
|
De Bakshi D, Chen YC, Wuerzberger-Davis SM, Ma M, Waters BJ, Li L, Suzuki A, Miyamoto S. Ectopic CH60 mediates HAPLN1-induced cell survival signaling in multiple myeloma. Life Sci Alliance 2023; 6:e202201636. [PMID: 36625202 PMCID: PMC9748848 DOI: 10.26508/lsa.202201636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematological malignancy, is generally considered incurable because of the development of drug resistance. We previously reported that hyaluronan and proteoglycan link protein 1 (HAPLN1) produced by stromal cells induces activation of NF-κB, a tumor-supportive transcription factor, and promotes drug resistance in MM cells. However, the identity of the cell surface receptor that detects HAPLN1 and thereby engenders pro-tumorigenic signaling in MM cells remains unknown. Here, we performed an unbiased cell surface biotinylation assay and identified chaperonin 60 (CH60) as the direct binding partner of HAPLN1 on MM cells. Cell surface CH60 specifically interacted with TLR4 to evoke HAPLN1-induced NF-κB signaling, transcription of anti-apoptotic genes, and drug resistance in MM cells. Collectively, our findings identify a cell surface CH60-TLR4 complex as a HAPLN1 receptor and a potential molecular target to overcome drug resistance in MM cells.
Collapse
Affiliation(s)
- Debayan De Bakshi
- Cellular and Molecular Biology Graduate Program, University of Wisconsin, Madison, WI, USA
- McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, WI, USA
- Department of Oncology, University of Wisconsin, Madison, WI, USA
| | - Yu-Chia Chen
- McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, WI, USA
- Department of Oncology, University of Wisconsin, Madison, WI, USA
| | - Shelly M Wuerzberger-Davis
- McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, WI, USA
- Department of Oncology, University of Wisconsin, Madison, WI, USA
| | - Min Ma
- School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Bayley J Waters
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, USA
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin, Madison, WI, USA
- Department of Chemistry, University of Wisconsin, Madison, WI, USA
| | - Aussie Suzuki
- McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, WI, USA
- Department of Oncology, University of Wisconsin, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Shigeki Miyamoto
- McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, WI, USA
- Department of Oncology, University of Wisconsin, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
9
|
Opposing MMP-9 Expression in Mesenchymal Stromal Cells and Head and Neck Tumor Cells after Direct 2D and 3D Co-Culture. Int J Mol Sci 2023; 24:ijms24021293. [PMID: 36674806 PMCID: PMC9861345 DOI: 10.3390/ijms24021293] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/23/2022] [Accepted: 01/07/2023] [Indexed: 01/11/2023] Open
Abstract
Bone marrow-derived mesenchymal stromal cells (BMSCs) respond to a variety of tumor cell-derived signals, such as inflammatory cytokines and growth factors. As a result, the inflammatory tumor microenvironment may lead to the recruitment of BMSCs. Whether BMSCs in the tumor environment are more likely to promote tumor growth or tumor suppression is still controversial. In our experiments, direct 3D co-culture of BMSCs with tumor cells from the head and neck region (HNSCC) results in strong expression and secretion of MMP-9. The observed MMP-9 secretion mainly originates from BMSCs, leading to increased invasiveness. In addition to our in vitro data, we show in vivo data based on the chorioallantoic membrane (CAM) model. Our results demonstrate that MMP-9 induces hemorrhage and increased perfusion in BMSC/HNSCC co-culture. While we had previously outlined that MMP-9 expression and secretion originate from BMSCs, our data showed a strong downregulation of MMP-9 promoter activity in HNSCC cells upon direct contact with BMSCs using the luciferase activity assay. Interestingly, the 2D and 3D models of direct co-culture suggest different drivers for the downregulation of MMP-9 promoter activity. Whereas the 3D model depicts a BMSC-dependent downregulation, the 2D model shows cell density-dependent downregulation. In summary, our data suggest that the direct interaction of HNSCC cells and BMSCs promotes tumor progression by significantly facilitating angiogenesis via MMP-9 expression. On the other hand, data from 3D and 2D co-culture models indicate opposing regulation of the MMP-9 promoter in tumor cells once stromal cells are involved.
Collapse
|
10
|
Bao F, Liu M, Gai W, Hua Y, Li J, Han C, Zai Z, Li J, Hua Z. Bacteria-mediated tumor-targeted delivery of tumstatin (54-132) significantly suppresses tumor growth in mouse model by inhibiting angiogenesis and promoting apoptosis. Front Med 2022; 16:873-882. [PMID: 36152127 DOI: 10.1007/s11684-022-0925-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 01/30/2022] [Indexed: 01/19/2023]
Abstract
Tumor growth is an angiogenesis-dependent process and accompanied by the formation of hypoxic areas. Tumstatin is a tumor-specific angiogenesis inhibitor that suppresses the proliferation and induces the apoptosis of tumorous vascular endothelial cells. VNP20009, an attenuated Salmonella typhimurium strain, preferentially accumulates in the hypoxic areas of solid tumors. In this study, a novel Salmonella-mediated targeted expression system of tumstatin (VNP-Tum5) was developed under the control of the hypoxia-induced J23100 promoter to obtain anti-tumor efficacy in mice. Treatment with VNP-Tum5 effectively suppressed tumor growth and prolonged survival in the mouse model of B16F10 melanoma. VNP-Tum5 exhibited a higher efficacy in inhibiting the proliferation and inducing the necrosis and apoptosis of B16F10 cells in vitro and in vivo compared with VNP (control). VNP-Tum5 significantly inhibited the proliferation and migration of mouse umbilical vascular endothelial cells to impede angiogenesis. VNP-Tum5 downregulated the expression of anti-vascular endothelial growth factor A, platelet endothelial cell adhesion molecule-1, phosphorylated phosphoinositide 3 kinase, and phosphorylated protein kinase B and upregulated the expression of cleaved-caspase 3 in tumor tissues. This study is the first to use tumstatin-transformed VNP20009 as a tumor-targeted system for treatment of melanoma by combining anti-tumor and anti-angiogenic effects.
Collapse
Affiliation(s)
- Feifei Bao
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Mengjie Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Wenhua Gai
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Yuwei Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Jing Li
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Chao Han
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Ziyu Zai
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Jiahuang Li
- Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou, 213164, China
| | - Zichun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China.
- Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou, 213164, China.
| |
Collapse
|
11
|
Liu C, Wang M, Zhang H, Li C, Zhang T, Liu H, Zhu S, Chen J. Tumor microenvironment and immunotherapy of oral cancer. Eur J Med Res 2022; 27:198. [PMID: 36209263 PMCID: PMC9547678 DOI: 10.1186/s40001-022-00835-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/22/2022] [Indexed: 11/10/2022] Open
Abstract
Oral cancer is one of the most common malignant tumors of the head and neck, not only affects the appearance, but also affects eating and even endangers life. The clinical treatments of oral cancer mainly include surgery, radiotherapy, and chemotherapy. However, unsatisfactory therapeutic effect and toxic side effects are still the main problems in clinical treatment. Tumor microenvironment (TME) is not only closely related to the occurrence, growth, and metastasis of tumor but also works in the diagnosis, prevention, and treatment of tumor and prognosis. Future studies should continue to investigate the relationship of TME and oral cancer therapy. This purpose of this review was to analyze the characteristics of oral cancer microenvironment, summarize the traditional oral cancer therapy and immunotherapy strategies, and finally prospect the development prospects of oral cancer immunotherapy. Immunotherapy targeting tumor microenvironment is expected to provide a new strategy for clinical treatment of oral cancer.
Collapse
Affiliation(s)
- Chang Liu
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Min Wang
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Haiyang Zhang
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Chunyan Li
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Tianshou Zhang
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Hong Liu
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Song Zhu
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China.
| | - Jie Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People's Republic of China.
| |
Collapse
|
12
|
Li H, Zhang S, Zhu R, Zhou Z, Xia L, Lin H, Chen S. Early assessment of chemotherapeutic response in hepatocellular carcinoma based on serum surface-enhanced Raman spectroscopy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 278:121314. [PMID: 35525180 DOI: 10.1016/j.saa.2022.121314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 04/18/2022] [Accepted: 04/24/2022] [Indexed: 06/14/2023]
Abstract
In clinical practice, the transcatheter arterial chemoembolization (TACE) has been widely accepted as the first option for non-surgical hepatocellular carcinoma (HCC) treatment. However, patients with HCC often suffer from poor response to TACE therapy. This can be prevented if the chemotherapeutic response can be early and accurately assessed, which is essential to guide timely and rational management. In this study, the serum SERS technique was for the first time investigated as a potential prognostic tool for early assessment of HCC chemotherapeutic response. According to the SERS spectral analysis results, it is newly found that not only the absolute circulating nucleic acids and collagen levels in pre-therapeutic serum but also the changes in circulating nucleic acids and amino acids between pre-therapeutic and post-therapeutic serum are expected to be potential serum markers for HCC prognosis. By further applying chemometrics methods to establish prognostic models, excellent prognostic accuracies were achieved within only 3 days after TACE therapy. Thus, the proposed method is expected to provide guidance on timely and rational management of HCC to improve its survival rate.
Collapse
Affiliation(s)
- Haiwei Li
- Department of Interventional Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China.
| | - Songqi Zhang
- School of Optical and Electronic Information, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ruochen Zhu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110167, China
| | - Zheng Zhou
- School of Innovation and Entrepreneurship, Liaoning Institute of Science and Technology, Benxi 117004, China
| | - Lu Xia
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110167, China
| | - Hao Lin
- School of Optical and Electronic Information, Huazhong University of Science and Technology, Wuhan 430074, China; College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110167, China; Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shuo Chen
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110167, China; Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, China.
| |
Collapse
|
13
|
Mohindra R, Mohindra R, Agrawal DK, Thankam FG. Bioactive extracellular matrix fragments in tendon repair. Cell Tissue Res 2022; 390:131-140. [PMID: 36074173 DOI: 10.1007/s00441-022-03684-z] [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: 01/06/2022] [Accepted: 08/30/2022] [Indexed: 11/02/2022]
Abstract
Tendinopathy is a common tendon disorder that causes pain, loss of strength and function, and local inflammation mainly characterized by hypoxia, collagen degradation, and extracellular matrix (ECM) disorganization. Generally, ECM degradation and remodeling is tightly regulated; however, hyperactivation of matrix metalloproteases (MMPs) contributes to excessive collagenolysis under pathologic conditions resulting in tendon ECM degradation. This review article focuses on the production, function, and signaling of matrikines for tendon regeneration following injury with insights into the expression, tissue compliance, and cell proliferation exhibited by various matrikines. Furthermore, the regenerative properties suggest translational significance of matrikines to improve the outcomes post-injury by assisting with tendon healing.
Collapse
Affiliation(s)
- Ritika Mohindra
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Rohit Mohindra
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA.
| |
Collapse
|
14
|
Itoh Y. Proteolytic modulation of tumor microenvironment signals during cancer progression. Front Oncol 2022; 12:935231. [PMID: 36132127 PMCID: PMC9483212 DOI: 10.3389/fonc.2022.935231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Under normal conditions, the cellular microenvironment is optimized for the proper functioning of the tissues and organs. Cells recognize and communicate with the surrounding cells and extracellular matrix to maintain homeostasis. When cancer arises, the cellular microenvironment is modified to optimize its malignant growth, evading the host immune system and finding ways to invade and metastasize to other organs. One means is a proteolytic modification of the microenvironment and the signaling molecules. It is now well accepted that cancer progression relies on not only the performance of cancer cells but also the surrounding microenvironment. This mini-review discusses the current understanding of the proteolytic modification of the microenvironment signals during cancer progression.
Collapse
|
15
|
Arikoglu H, Dursunoglu D, Kaya DE, Avci E. The effects of Juglone-Selenium combination on invasion and metastasis in pancreatic cancer cell lines. Afr Health Sci 2022; 22:334-342. [PMID: 36407358 PMCID: PMC9652687 DOI: 10.4314/ahs.v22i2.37] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Pancreatic cancer does not show any symptoms in the early period and metastatic process is already passed when the diagnosis is done. Therefore, in the battle with pancreatic cancer, novel treatment strategies, particularly antiinvasive and antimetastatic strategies, are needed. The cytotoxic and anticancer effects of juglone and sodium selenite (NaSe) have been showed in various cancer cells. OBJECTIVES In this study, it is aimed to investigate the synergistic effects of juglone and selenium on PANC-1 and BxPC-3 pancreatic cancer cells. METHODS Antimetastatic effects of juglone-NaSe were carried out by adhesion and invasion assays and the genes and protein expressions. Expression analysis of the CDH1, ITGB3 and COL4A3 genes and their proteins E-cadherin, β3 integrin and tumstatin which play role in metastasis and angiogenesis processes, were done by qPCR and immunohistochemical analysis, respectively. RESULTS Study findings have provided evidences that the juglone-selenium has a cytotoxic and dose dependent suppressive effect on invasion and metastasis in PANC-1 and BxPC-3 cells. CONCLUSION The juglone-NaSe has the potential to be a promising agent especially to inhibit invasion and metastasis in pancreatic cancer treatment. However, more in depth studies are needed to more clearly demonstrate the effects of juglone-selenium.
Collapse
Affiliation(s)
| | | | | | - Ebru Avci
- Necmettin Erbakan University, Medical Biology
| |
Collapse
|
16
|
Tao D, Wang Y, Zhang X, Wang C, Yang D, Chen J, Long Y, Jiang Y, Zhou X, Zhang N. Identification of Angiogenesis-Related Prognostic Biomarkers Associated With Immune Cell Infiltration in Breast Cancer. Front Cell Dev Biol 2022; 10:853324. [PMID: 35602610 PMCID: PMC9121305 DOI: 10.3389/fcell.2022.853324] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/30/2022] [Indexed: 12/01/2022] Open
Abstract
Background: This study aimed to explore the prognostic value of angiogenesis-related genes (ARGs) and their association with immune cell infiltration (ICI) in breast cancer (BC). Methods: Transcriptome data of BC were obtained from the TCGA and GEO databases. Differentially expressed ARGs were identified by the limma package. The identification of key genes and construction of the risk score model were performed by univariate and multivariate Cox regression algorithms. The prognostic value of the risk score was assessed by ROC curves and nomogram. GO, KEGG pathway, and GSEA were used to investigate the biological functions of differentially expressed genes (DEGs), and CIBERSORT, ssGSEA, and xCell algorithms were performed to estimate the ICI in high-risk and low-risk groups. The correlations between prognostic biomarkers and differentially distributed immune cells were assessed. Moreover, a ceRNA regulatory network based on prognostic biomarkers was constructed and visualized by Cytoscape software. Results: A total of 18 differentially expressed ARGs were identified between tumor and adjacent normal tissue samples. TNFSF12, SCG2, COL4A3, and TNNI3 were identified as key prognostic genes by univariate and multivariate Cox regression analyses. The risk score model was further constructed based on the four-gene signature and validated in GSE7390 and GSE88770 datasets. ROC curves and nomogram indicated that the risk score had good accuracy for determining BC patient survival. Biological function analysis showed that DEGs in high- and low-risk groups had a high enrichment in immune-related biological processes and signaling pathways. Moreover, significantly different ICIs were found between high- and low-risk groups, such as memory B cells, CD8+ T cells, resting memory CD4+ T cells, follicular helper T cells, regulatory T cells, monocytes, M2 macrophages, and neutrophils, and each prognostic biomarker was significantly correlated with one or more immune cell types. Conclusion: The current study identified novel prognostic ARGs and developed a prognostic model for predicting survival in patients with BC. Furthermore, this study indicated that ICI may act as a bond between angiogenesis and BC. These findings enhance our understanding of angiogenesis in BC and provide novel guidance on developing therapeutic targets for BC patients.
Collapse
Affiliation(s)
- Dan Tao
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Ying Wang
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Xin Zhang
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Can Wang
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Dingyi Yang
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Jing Chen
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yanyan Long
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yong Jiang
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Xian Zhou
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Ningning Zhang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- *Correspondence: Ningning Zhang,
| |
Collapse
|
17
|
Augoff K, Hryniewicz-Jankowska A, Tabola R, Stach K. MMP9: A Tough Target for Targeted Therapy for Cancer. Cancers (Basel) 2022; 14:cancers14071847. [PMID: 35406619 PMCID: PMC8998077 DOI: 10.3390/cancers14071847] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/27/2022] [Accepted: 03/31/2022] [Indexed: 02/01/2023] Open
Abstract
Having the capability to proteolyze diverse structural and signaling proteins, matrix metalloproteinase 9 (MMP9), one of the best-studied secretory endopeptidases, has been identified as a crucial mediator of processes closely associated with tumorigenesis, such as the extracellular matrix reorganization, epithelial to mesenchymal transition, cell migration, new blood vessel formation, and immune response. In this review, we present the current state of knowledge on MMP9 and its role in cancer growth in the context of cell adhesion/migration, cancer-related inflammation, and tumor microenvironment formation. We also summarize recent achievements in the development of selective MMP9 inhibitors and the limitations of using them as anticancer drugs.
Collapse
Affiliation(s)
- Katarzyna Augoff
- Department of Surgical Education, Wroclaw Medical University, 50-367 Wroclaw, Poland
- Department of Chemistry and Immunochemistry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
- Correspondence:
| | | | - Renata Tabola
- Department of Thoracic Surgery, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Kamilla Stach
- Department of Chemistry and Immunochemistry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| |
Collapse
|
18
|
Wroński P, Wroński S, Kurant M, Malinowski B, Wiciński M. Curcumin May Prevent Basement Membrane Disassembly by Matrix Metalloproteinases and Progression of the Bladder Cancer. Nutrients 2021; 14:32. [PMID: 35010907 PMCID: PMC8746354 DOI: 10.3390/nu14010032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/18/2021] [Accepted: 12/19/2021] [Indexed: 12/25/2022] Open
Abstract
Authors present a review of crucial mechanisms contributing to the invasion of the basement membrane (BM) of the urothelium by cancer cells and to the progression of bladder cancer (BC). The breeching of the urothelial BM, facilitated by an aberrant activation of matrix metalloproteinases (MMP) is particularly perilous. Inhibition of activation of these proteinases constitutes a logic opportunity to restrain progression. Because of limited efficacy of current therapeutic methods, the search for the development of alternative approaches constitutes "the hot spot" of modern oncology. Recent studies revealed significant anticancer potential of natural phytochemicals. Especially, curcumin has emerged as a one of the most promising phytochemicals and showed its efficacy in several human malignancies. Therefore, this article addresses experimental and clinical data indicating multi-directional inhibitory effect of curcumin on the growth of bladder cancer. We particularly concentrate on the mechanisms, by which curcumin inhibits the MMP's activities, thereby securing BM integrity and alleviating the eventual cancer invasion into the bladder muscles. Authors review the recently accumulating data, that curcumin constitutes a potent factor contributing to the more effective treatment of the bladder cancer.
Collapse
Affiliation(s)
- Paweł Wroński
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland; (P.W.); (B.M.)
- Department of Oncological Urology, The Franciszek Lukaszczyk Oncology Center, Romanowskiej 2, 85-796 Bydgoszcz, Poland
| | - Stanisław Wroński
- Department of Urology, Jan Biziel Memorial University Hospital, Ujejskiego 75, 85-168 Bydgoszcz, Poland;
| | - Marcin Kurant
- Department of Urology, District Hospital, 10 Lesna Street, 89-600 Chojnice, Poland;
| | - Bartosz Malinowski
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland; (P.W.); (B.M.)
| | - Michał Wiciński
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland; (P.W.); (B.M.)
| |
Collapse
|
19
|
Dekkers BG, Saad SI, van Spelde LJ, Burgess JK. Basement membranes in obstructive pulmonary diseases. Matrix Biol Plus 2021; 12:100092. [PMID: 34877523 PMCID: PMC8632995 DOI: 10.1016/j.mbplus.2021.100092] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 11/04/2021] [Accepted: 11/07/2021] [Indexed: 12/24/2022] Open
Abstract
Basement membrane composition is changed in the airways of patients with obstructive airway diseases. Basement membrane changes are linked to disease characteristics in patients. Mechanisms behind the altered BM composition remain to be elucidated. Laminin and collagen IV affect key pathological processes in obstructive airway diseases.
Increased and changed deposition of extracellular matrix proteins is a key feature of airway wall remodeling in obstructive pulmonary diseases, including asthma and chronic obstructive pulmonary disease. Studies have highlighted that the deposition of various basement membrane proteins in the lung tissue is altered and that these changes reflect tissue compartment specificity. Inflammatory responses in both diseases may result in the deregulation of production and degradation of these proteins. In addition to their role in tissue development and integrity, emerging evidence indicates that basement membrane proteins also actively modulate cellular processes in obstructive airway diseases, contributing to disease development, progression and maintenance. In this review, we summarize the changes in basement membrane composition in airway remodeling in obstructive airway diseases and explore their potential application as innovative targets for treatment development.
Collapse
Key Words
- ADAM9, a metalloproteinase domain 9
- ASM, airway smooth muscle
- Airway inflammation
- Airway remodeling
- Asthma
- BM, basement membrane
- COPD, chronic obstructive pulmonary disease
- Chronic obstructive pulmonary disease
- Col IV, collagen IV
- Collagen IV
- ECM, extracellular matrix
- LN, laminin
- Laminin
- MMP, matrix metalloproteinase
- TIMP, tissue inhibitors of metalloproteinase
- Th2, T helper 2
- VSM, vascular smooth muscle
Collapse
Affiliation(s)
- Bart G.J. Dekkers
- University of Groningen, University Medical Center Groningen, Department of Clinical Pharmacy and Pharmacology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Corresponding author at: Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| | - Shehab I. Saad
- University of Groningen, University Medical Centre Groningen, Department of Pathology & Medical Biology, Experimental Pulmonology and Inflammation Research, Groningen, The Netherlands
| | - Leah J. van Spelde
- University of Groningen, University Medical Centre Groningen, Department of Pathology & Medical Biology, Experimental Pulmonology and Inflammation Research, Groningen, The Netherlands
| | - Janette K. Burgess
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- University of Groningen, University Medical Centre Groningen, Department of Pathology & Medical Biology, Experimental Pulmonology and Inflammation Research, Groningen, The Netherlands
| |
Collapse
|
20
|
Pach E, Kümper M, Fromme JE, Zamek J, Metzen F, Koch M, Mauch C, Zigrino P. Extracellular Matrix Remodeling by Fibroblast-MMP14 Regulates Melanoma Growth. Int J Mol Sci 2021; 22:12276. [PMID: 34830157 PMCID: PMC8625044 DOI: 10.3390/ijms222212276] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/04/2021] [Accepted: 11/11/2021] [Indexed: 12/20/2022] Open
Abstract
Maintaining a balanced state in remodeling the extracellular matrix is crucial for tissue homeostasis, and this process is altered during skin cancer progression. In melanoma, several proteolytic enzymes are expressed in a time and compartmentalized manner to support tumor progression by generating a permissive environment. One of these proteases is the matrix metalloproteinase 14 (MMP14). We could previously show that deletion of MMP14 in dermal fibroblasts results in the generation of a fibrotic-like skin in which melanoma growth is impaired. That was primarily due to collagen I accumulation due to lack of the collagenolytic activity of MMP14. However, as well as collagen I processing, MMP14 can also process several extracellular matrices. We investigated extracellular matrix alterations occurring in the MMP14-deleted fibroblasts that can contribute to the modulation of melanoma growth. The matrix deposited by cultured MMP14-deleted fibroblast displayed an antiproliferative and anti-migratory effect on melanoma cells in vitro. Analysis of the secreted and deposited-decellularized fibroblast's matrix identified a few altered proteins, among which the most significantly changed was collagen XIV. This collagen was increased because of post-translational events, while de novo synthesis was unchanged. Collagen XIV as a substrate was not pro-proliferative, pro-migratory, or adhesive, suggesting a negative regulatory role on melanoma cells. Consistent with that, increasing collagen XIV concentration in wild-type fibroblast-matrix led to reduced melanoma proliferation, migration, and adhesion. In support of its anti-tumor activity, enhanced accumulation of collagen XIV was detected in peritumoral areas of melanoma grown in mice with the fibroblast's deletion of MMP14. In advanced human melanoma samples, we detected reduced expression of collagen XIV compared to benign nevi, which showed a robust expression of this molecule around melanocytic nests. This study shows that loss of fibroblast-MMP14 affects melanoma growth through altering the peritumoral extracellular matrix (ECM) composition, with collagen XIV being a modulator of melanoma progression and a new proteolytic substrate to MMP14.
Collapse
Affiliation(s)
- Elke Pach
- Department of Dermatology and Venereology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (E.P.); (M.K.); (J.E.F.); (J.Z.); (C.M.)
| | - Maike Kümper
- Department of Dermatology and Venereology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (E.P.); (M.K.); (J.E.F.); (J.Z.); (C.M.)
| | - Julia E. Fromme
- Department of Dermatology and Venereology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (E.P.); (M.K.); (J.E.F.); (J.Z.); (C.M.)
- Mildred Scheel School of Oncology Aachen Bonn Cologne Düsseldorf (MSSO ABCD), 50937 Cologne, Germany
| | - Jan Zamek
- Department of Dermatology and Venereology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (E.P.); (M.K.); (J.E.F.); (J.Z.); (C.M.)
| | - Fabian Metzen
- Faculty of Medicine and University Hospital, Institute for Dental Research and Oral Musculoskeletal Biology and Center for Biochemistry, University of Cologne, 50937 Cologne, Germany; (F.M.); (M.K.)
| | - Manuel Koch
- Faculty of Medicine and University Hospital, Institute for Dental Research and Oral Musculoskeletal Biology and Center for Biochemistry, University of Cologne, 50937 Cologne, Germany; (F.M.); (M.K.)
| | - Cornelia Mauch
- Department of Dermatology and Venereology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (E.P.); (M.K.); (J.E.F.); (J.Z.); (C.M.)
| | - Paola Zigrino
- Department of Dermatology and Venereology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (E.P.); (M.K.); (J.E.F.); (J.Z.); (C.M.)
| |
Collapse
|
21
|
Karsdal MA, Genovese F, Rasmussen DGK, Bay-Jensen AC, Mortensen JH, Holm Nielsen S, Willumsen N, Jensen C, Manon-Jensen T, Jennings L, Reese-Petersen AL, Henriksen K, Sand JM, Bager C, Leeming DJ. Considerations for understanding protein measurements: Identification of formation, degradation and more pathological relevant epitopes. Clin Biochem 2021; 97:11-24. [PMID: 34453894 DOI: 10.1016/j.clinbiochem.2021.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/06/2021] [Accepted: 08/23/2021] [Indexed: 01/01/2023]
Abstract
OBJECTIVES There is a need for precision medicine and an unspoken promise of an optimal approach for identification of the right patients for value-based medicine based on big data. However, there may be a misconception that measurement of proteins is more valuable than measurement of fewer selected biomarkers. In population-based research, variation may be somewhat eliminated by quantity. However, this fascination of numbers may limit the attention to and understanding of the single. This review highlights that protein measurements (with collagens as examples) may mean different things depending on the targeted epitope - formation or degradation of tissues, and even signaling potential of proteins. DESIGN AND METHODS PubMed was searched for collagen, neo-epitope, biomarkers. RESULTS Ample examples of assays with specific epitopes, either pathological such as HbA1c, or domain specific such as pro-peptides, which total protein arrays would not have identified were evident. CONCLUSIONS We suggest that big data may be considered as the funnel of data points, in which most important parameters will be selected. If the technical precision is low or the biological accuracy is limited, and we include suboptimal quality of biomarkers, disguised as big data, we may not be able to fulfill the promise of helping patients searching for the optimal treatment. Alternatively, if the technical precision of the total protein quantification is high, but we miss the functional domains with the most considerable biological meaning, we miss the most important and valuable information of a given protein. This review highlights that measurements of the same protein in different ways may provide completely different meanings. We need to understand the pathological importance of each epitope quantified to maximize protein measurements.
Collapse
Affiliation(s)
- M A Karsdal
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark.
| | - F Genovese
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - D G K Rasmussen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - A C Bay-Jensen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - J H Mortensen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - S Holm Nielsen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - N Willumsen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - C Jensen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - T Manon-Jensen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | | | | | - K Henriksen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - J M Sand
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - C Bager
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - D J Leeming
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| |
Collapse
|
22
|
Weckmann M, Bahmer T, Bülow Sand JM, Rank Rønnow S, Pech M, Vermeulen C, Faiz A, Leeming DJ, Karsdal MA, Lunding L, Oliver BGG, Wegmann M, Ulrich-Merzenich G, Juergens UR, Duhn J, Laumonnier Y, Danov O, Sewald K, Zissler U, Jonker M, König I, Hansen G, von Mutius E, Fuchs O, Dittrich AM, Schaub B, Happle C, Rabe KF, van de Berge M, Burgess JK, Kopp MV. COL4A3 is degraded in allergic asthma and degradation predicts response to anti-IgE therapy. Eur Respir J 2021; 58:13993003.03969-2020. [PMID: 34326188 DOI: 10.1183/13993003.03969-2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 04/28/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND Asthma is a heterogeneous syndrome substantiating the urgent requirement for endotype-specific biomarkers. Dysbalance of fibrosis and fibrolysis in asthmatic lung tissue leads to reduced levels of the inflammation-protective collagen 4 (COL4A3). OBJECTIVE To delineate the degradation of COL4A3 in allergic airway inflammation and evaluate the resultant product as a biomarker for anti-IgE therapy response. METHODS The serological COL4A3 degradation marker C4Ma3 (Nordic Bioscience, Denmark) and serum cytokines were measured in the ALLIANCE cohort (pediatric cases/controls: 134/35; adult cases/controls: 149/31). Exacerbation of allergic airway disease in mice was induced by sensitising to OVA, challenge with OVA aerosol and instillation of poly(cytidylic-inosinic). Fulacimstat (chymase inhibitor, Bayer) was used to determine the role of mast cell chymase in COL4A3 degradation. Patients with cystic fibrosis (CF, n=14) and CF with allergic broncho-pulmonary aspergillosis (ABPA, n=9) as well as severe allergic, uncontrolled asthmatics (n=19) were tested for COL4A3 degradation. Omalizumab (anti-IgE) treatment was assessed by the Asthma Control Test. RESULTS Serum levels of C4Ma3 were increased in asthma in adults and children alike and linked to a more severe, exacerbating allergic asthma phenotype. In an experimental asthma mouse model, C4Ma3 was dependent on mast cell chymase. Serum C4Ma3 was significantly elevated in CF plus ABPA and at baseline predicted the success of the anti-IgE therapy in allergic, uncontrolled asthmatics (diagnostic odds ratio 31.5). CONCLUSION C4Ma3 level depend on lung mast cell chymase and are increased in a severe, exacerbating allergic asthma phenotype. C4Ma3 may serve as a novel biomarker to predict anti-IgE therapy response.
Collapse
Affiliation(s)
- Markus Weckmann
- Division of Pediatric Pneumology & Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany
| | - Thomas Bahmer
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany.,Department of Pneumology, LungenClinic Grosshansdorf, Grosshansdorf, Germany
| | | | - Sarah Rank Rønnow
- Nordic Bioscience A/S, Herlev, Denmark.,The Faculty of Health Science, University of Southern Denmark, Odense, Denmark
| | - Martin Pech
- Division of Pediatric Pneumology & Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany
| | - Cornelis Vermeulen
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, , GRIAC (Groningen Research Institute for Asthma and COPD), Groningen, The Netherlands
| | - Alen Faiz
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, , GRIAC (Groningen Research Institute for Asthma and COPD), Groningen, The Netherlands.,Department of Pathology & Medical Biology, University of Groningen, University Medical Center Groningen, , GRIAC (Groningen Research Institute for Asthma and COPD), Groningen, The Netherlands.,Woolcock Institute of Medical Research, The University of Sydney, Glebe, NSW, Australia.,School of Medical and Molecular Biosciences, University of Technology, Sydney, NSW, Australia
| | | | | | - Lars Lunding
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany.,Division of Asthma-Exacerbation & -Regulation; Program Area Asthma & Allergy, Leibniz-Center for Medicine and Biosciences Borstel
| | - Brian George G Oliver
- Woolcock Institute of Medical Research, The University of Sydney, Glebe, NSW, Australia.,School of Medical and Molecular Biosciences, University of Technology, Sydney, NSW, Australia
| | - Michael Wegmann
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany.,Division of Asthma-Exacerbation & -Regulation; Program Area Asthma & Allergy, Leibniz-Center for Medicine and Biosciences Borstel
| | | | - Uwe R Juergens
- Department of Pneumonology, Medical Clinic II, University Hospital Bonn
| | - Jannis Duhn
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Olga Danov
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Katherina Sewald
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Ulrich Zissler
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health (CPC-M), Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Marnix Jonker
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, , GRIAC (Groningen Research Institute for Asthma and COPD), Groningen, The Netherlands.,Department of Pathology & Medical Biology, University of Groningen, University Medical Center Groningen, , GRIAC (Groningen Research Institute for Asthma and COPD), Groningen, The Netherlands
| | - Inke König
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany.,Institute for Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | - Gesine Hansen
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center of Lung Research (DZL), Germany
| | - Erika von Mutius
- University Children's Hospital, Ludwig Maximilian's University, Munich, Germany.,German Research Center for Environmental Health (CPC-M), Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Oliver Fuchs
- Division of Pediatric Pneumology & Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany.,Department of Paediatric Respiratory Medicine, Inselspital, University Children's Hospital of Bern, University of Bern, Bern, Switzerland
| | - Anna-Maria Dittrich
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center of Lung Research (DZL), Germany
| | - Bianca Schaub
- University Children's Hospital, Ludwig Maximilian's University, Munich, Germany.,German Research Center for Environmental Health (CPC-M), Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Christine Happle
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center of Lung Research (DZL), Germany
| | - Klaus F Rabe
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany.,Department of Pneumology, LungenClinic Grosshansdorf, Grosshansdorf, Germany
| | - Maarten van de Berge
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, , GRIAC (Groningen Research Institute for Asthma and COPD), Groningen, The Netherlands
| | - Janette Kay Burgess
- Department of Pathology & Medical Biology, University of Groningen, University Medical Center Groningen, , GRIAC (Groningen Research Institute for Asthma and COPD), Groningen, The Netherlands.,Woolcock Institute of Medical Research, The University of Sydney, Glebe, NSW, Australia.,Discipline of Pharmacology, Faculty of Medicine, The University of Sydney, NSW, Australia
| | - Matthias Volkmar Kopp
- Division of Pediatric Pneumology & Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany.,Department of Paediatric Respiratory Medicine, Inselspital, University Children's Hospital of Bern, University of Bern, Bern, Switzerland
| | | |
Collapse
|
23
|
Philip R, Dumont A, Martin Silva N, de Boysson H, Aouba A, Deshayes S. ANCA and anti-glomerular basement membrane double-positive patients: A systematic review of the literature. Autoimmun Rev 2021; 20:102885. [PMID: 34242834 DOI: 10.1016/j.autrev.2021.102885] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Double-positive patients (DPP) exhibiting anti-glomerular basement membrane (GBM) and anti-neutrophil cytoplasmic antibodies (ANCAs) belong to an entity that is newly and poorly described, mainly in short series. We aimed to better characterize the epidemiological features, clinical presentation and therapeutic outcomes of these patients through a systematic review. METHODS We performed a systematic review of English-, German-, Spanish- and French-written publications from February 1987 to March 2020 reporting cases of DPP using the following databases: PubMed, Scielo, ScienceDirect, Google Scholar, The Cochrane Library, Open Grey, The Grey Literature Report, Clinicaltrials.gov and International Clinical Trial Registry Platform of the World Health Organization. RESULTS In total, 538 DPP were identified from 90 articles. Their clinical presentations were often severe, and the majority exhibited acute kidney failure (91.8%) with a median initial serum creatinine level of 873 μmol/L; 50.7% had alveolar haemorrhage. Other manifestations were present in 30.3% of DPP, mainly ear, nose, throat and articular manifestations. ANCAs were predominantly directed against MPO (n = 377/523; 72.1%) compared to PR3 (n = 107/523; 20.5%), with rare cases of triple positivity (n = 15/538; 2.9%). Although most patients received initial immunosuppressive therapy (n = 285/317; 89.9%), the one-year overall, renal and relapse-free survival rates were 64.8%, 38.7% and 71.1%, respectively. CONCLUSION DPP are associated with the characteristics of two eponymous vasculitis types, responsible for a poor overall and renal prognosis. Thus, simultaneous testing of both antibodies and systematic renal biopsy should be recommended in every patient with rapidly progressive glomerulonephritis to recognize this difficult-to-treat and rare disease.
Collapse
Affiliation(s)
- Rémi Philip
- Department of Clinical Immunology and Internal Medicine, CHU of Caen Normandie, 14000 Caen, France
| | - Anael Dumont
- Department of Clinical Immunology and Internal Medicine, CHU of Caen Normandie, 14000 Caen, France; Normandie Univ, UNICAEN, CHU de Caen Normandie, 14000 Caen, France
| | - Nicolas Martin Silva
- Department of Clinical Immunology and Internal Medicine, CHU of Caen Normandie, 14000 Caen, France
| | - Hubert de Boysson
- Department of Clinical Immunology and Internal Medicine, CHU of Caen Normandie, 14000 Caen, France; Normandie Univ, UNICAEN, CHU de Caen Normandie, 14000 Caen, France
| | - Achille Aouba
- Department of Clinical Immunology and Internal Medicine, CHU of Caen Normandie, 14000 Caen, France; Normandie Univ, UNICAEN, CHU de Caen Normandie, 14000 Caen, France.
| | - Samuel Deshayes
- Department of Clinical Immunology and Internal Medicine, CHU of Caen Normandie, 14000 Caen, France; Normandie Univ, UNICAEN, CHU de Caen Normandie, 14000 Caen, France
| |
Collapse
|
24
|
Salamone M, Rigogliuso S, Nicosia A, Campora S, Bruno CM, Ghersi G. 3D Collagen Hydrogel Promotes In Vitro Langerhans Islets Vascularization through ad-MVFs Angiogenic Activity. Biomedicines 2021; 9:biomedicines9070739. [PMID: 34199087 PMCID: PMC8301445 DOI: 10.3390/biomedicines9070739] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023] Open
Abstract
Adipose derived microvascular fragments (ad-MVFs) consist of effective vascularization units able to reassemble into efficient microvascular networks. Because of their content in stem cells and related angiogenic activity, ad-MVFs represent an interesting tool for applications in regenerative medicine. Here we show that gentle dissociation of rat adipose tissue provides a mixture of ad-MVFs with a length distribution ranging from 33–955 μm that are able to maintain their original morphology. The isolated units of ad-MVFs that resulted were able to activate transcriptional switching toward angiogenesis, forming tubes, branches, and entire capillary networks when cultured in 3D collagen type-I hydrogel. The proper involvement of metalloproteases (MMP2/MMP9) and serine proteases in basal lamina and extracellular matrix ECM degradation during the angiogenesis were concurrently assessed by the evaluation of alpha-smooth muscle actin (αSMA) expression. These results suggest that collagen type-I hydrogel provides an adequate 3D environment supporting the activation of the vascularization process. As a proof of concept, we exploited 3D collagen hydrogel for the setting of ad-MVF–islet of Langerhans coculture to improve the islets vascularization. Our results suggest potential employment of the proposed in vitro system for regenerative medicine applications, such as the improving of the islet of Langerhans engraftment before transplantation.
Collapse
Affiliation(s)
- Monica Salamone
- Abiel s.r.l., c/o Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF) University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (M.S.); (S.R.); (C.M.B.)
| | - Salvatrice Rigogliuso
- Abiel s.r.l., c/o Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF) University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (M.S.); (S.R.); (C.M.B.)
| | - Aldo Nicosia
- Institute for Biomedical Research and Innovation-National Research Council (IRIB-CNR), Via Ugo La Malfa 153, 90146 Palermo, Italy;
| | - Simona Campora
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF) University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy;
| | - Carmelo Marco Bruno
- Abiel s.r.l., c/o Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF) University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (M.S.); (S.R.); (C.M.B.)
| | - Giulio Ghersi
- Abiel s.r.l., c/o Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF) University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (M.S.); (S.R.); (C.M.B.)
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF) University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy;
- Correspondence:
| |
Collapse
|
25
|
Liu B, Tian Y, Chen M, Shen H, Xia J, Nan J, Yan T, Wang Y, Shi L, Shen B, Yu H, Cai X. CircUBAP2 Promotes MMP9-Mediated Oncogenic Effect via Sponging miR-194-3p in Hepatocellular Carcinoma. Front Cell Dev Biol 2021; 9:675043. [PMID: 34239873 PMCID: PMC8258265 DOI: 10.3389/fcell.2021.675043] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/31/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The physiological regulatory functions of circRNAs have become a topic of intensive research in recent years. Increasing evidence supports a significant role of circRNAs during cancer initiation and progression, including hepatocellular carcinoma (HCC). MATERIALS AND METHODS A bioinformatics analysis from three independent Gene Expression Omnibus (GEO) databases was performed to profile and screen the dysregulated circRNAs in HCC. RT-qPCR was used to examine the expression level of circUBAP2 in HCC and adjacent non-tumor tissues. Then, proliferation assays (CCK8 and colony formation) and migration assays (transwell and wound healing) were performed to examine effect of circUBAP2 in vitro. Immunoprecipitation, RNA pulldown, FISH, and dual-luciferase reporter assay was conducted to explore the circUBAP2-related mechanism for regulating HCC progression. Moreover, a mouse xenograft model and a mouse lung metastasis model confirmed the effect of circUBAP2 in vivo. RESULTS In this study, we found a novel circRNA: circUBAP2, which was identified by bioinformatics analysis. Among 91 HCC patients, circUBAP2 was significantly upregulated in HCC tissues, and negatively correlated with aggressive clinical characteristics and prognosis. Functional assays demonstrated that circUBAP2 promoted cell proliferation, colony formation, migration, and invasion in vitro. Moreover, circUBAP2 enhanced tumor growth and pulmonary metastasis in vivo. Mechanistically, circUBAP2 acts as a competing endogenous RNA (ceRNA) for miR-194-3p, a tumor suppressor in HCC. We confirmed that MMP9 was direct target for miR-194-3p, which was regulated by circUBAP2. CONCLUSION CircUBAP2 plays a significant role in promoting HCC via the miR-194-3p/MMP9 pathway and could serve as a promising prognostic biomarker and novel therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Boqiang Liu
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuanshi Tian
- Department of Diagnostic Ultrasound and Echocardiography, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Mingyu Chen
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hao Shen
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiafeng Xia
- State Key Laboratory for Diagnosis, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Junjie Nan
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tingting Yan
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yifan Wang
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liang Shi
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bo Shen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hong Yu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiujun Cai
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang University, Hangzhou, China
- Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Zhejiang University, Hangzhou, China
- Zhejiang University Cancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
26
|
Yang J, Zhong J, Zhou M, Zhou Y, Xiu P, Liu F, Wang F, Li Z, Tang Y, Chen Y, Yao S, Huang T, Liu T, Dong X. Targeting of the COX-2/PGE2 axis enhances the antitumor activity of T7 peptide in vitro and in vivo. Drug Deliv 2021; 28:844-855. [PMID: 33928829 PMCID: PMC8812588 DOI: 10.1080/10717544.2021.1914776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
T7 peptide is considered as an antiangiogenic polypeptide. The presents study aimed to further detect the antiangiogenic mechanisms of T7 peptide and determine whether combining T7 peptide and meloxicam (COX-2/PGE2 specific inhibitor) could offer a better therapy to combat hepatocellular carcinoma (HCC). T7 peptide suppressed the proliferation, migration, tube formation, and promoted the apoptosis of endothelial cells under both normoxic and hypoxic conditions via integrin α3β1 and αvβ3 pathways. Cell proliferation, migration, apoptosis, or tube formation ability were detected, and the expression of integrin-associated regulatory proteins was detected. The anti-tumor activity of T7 peptide, meloxicam, and their combination were evaluated in HCC tumor models established in mice. T7 peptide suppressed the proliferation, migration, tube formation, and promoted the apoptosis of endothelial cells under both normoxic and hypoxic conditions via integrin α3β1 and αvβ3 pathways. Meloxicam enhanced the activity of T7 peptide under hypoxic condition. T7 peptide partly inhibited COX-2 expression via integrin α3β1 not αvβ3-dependent pathways under hypoxic condition. T7 peptide regulated apoptosis associated protein through MAPK-dependent and -independent pathways under hypoxic condition. The MAPK pathway was activated by the COX-2/PGE2 axis under hypoxic condition. The combination of T7 and meloxicam showed a stronger anti-tumor effect against HCC tumors in mice. The data highlight that meloxicam enhanced the antiangiogenic activity of T7 peptide in vitro and in vivo.
Collapse
Affiliation(s)
- Jianrong Yang
- Department of Hepatobiliary, Pancreas and Spleen Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jingtao Zhong
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Mi Zhou
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yinghong Zhou
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, China
| | - Peng Xiu
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Feng Liu
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Fuhai Wang
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Zelun Li
- Department of Hepatobiliary, Pancreas and Spleen Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yuntian Tang
- Department of Hepatobiliary, Pancreas and Spleen Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yuanyuan Chen
- Department of Hepatobiliary, Pancreas and Spleen Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Siyang Yao
- Department of Hepatobiliary, Pancreas and Spleen Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Tao Huang
- Department of Hepatobiliary, Pancreas and Spleen Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Tianqi Liu
- Department of Hepatobiliary, Pancreas and Spleen Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xiaofeng Dong
- Department of Hepatobiliary, Pancreas and Spleen Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
27
|
Huang J, Zhang L, Wan D, Zhou L, Zheng S, Lin S, Qiao Y. Extracellular matrix and its therapeutic potential for cancer treatment. Signal Transduct Target Ther 2021; 6:153. [PMID: 33888679 PMCID: PMC8062524 DOI: 10.1038/s41392-021-00544-0] [Citation(s) in RCA: 367] [Impact Index Per Article: 91.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 02/17/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) is one of the major components of tumors that plays multiple crucial roles, including mechanical support, modulation of the microenvironment, and a source of signaling molecules. The quantity and cross-linking status of ECM components are major factors determining tissue stiffness. During tumorigenesis, the interplay between cancer cells and the tumor microenvironment (TME) often results in the stiffness of the ECM, leading to aberrant mechanotransduction and further malignant transformation. Therefore, a comprehensive understanding of ECM dysregulation in the TME would contribute to the discovery of promising therapeutic targets for cancer treatment. Herein, we summarized the knowledge concerning the following: (1) major ECM constituents and their functions in both normal and malignant conditions; (2) the interplay between cancer cells and the ECM in the TME; (3) key receptors for mechanotransduction and their alteration during carcinogenesis; and (4) the current therapeutic strategies targeting aberrant ECM for cancer treatment.
Collapse
Affiliation(s)
- Jiacheng Huang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Lele Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Dalong Wan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Shengzhang Lin
- School of Medicine, Zhejiang University, Hangzhou, 310003, China.
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310000, China.
| | - Yiting Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China.
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China.
| |
Collapse
|
28
|
Infarct in the Heart: What's MMP-9 Got to Do with It? Biomolecules 2021; 11:biom11040491. [PMID: 33805901 PMCID: PMC8064345 DOI: 10.3390/biom11040491] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 12/12/2022] Open
Abstract
Over the past three decades, numerous studies have shown a strong connection between matrix metalloproteinase 9 (MMP-9) levels and myocardial infarction (MI) mortality and left ventricle remodeling and dysfunction. Despite this fact, clinical trials using MMP-9 inhibitors have been disappointing. This review focuses on the roles of MMP-9 in MI wound healing. Infiltrating leukocytes, cardiomyocytes, fibroblasts, and endothelial cells secrete MMP-9 during all phases of cardiac repair. MMP-9 both exacerbates the inflammatory response and aids in inflammation resolution by stimulating the pro-inflammatory to reparative cell transition. In addition, MMP-9 has a dual effect on neovascularization and prevents an overly stiff scar. Here, we review the complex role of MMP-9 in cardiac wound healing, and highlight the importance of targeting MMP-9 only for its detrimental actions. Therefore, delineating signaling pathways downstream of MMP-9 is critical.
Collapse
|
29
|
Han X, Caron JM, Lary CW, Sathyanarayana P, Vary C, Brooks PC. An RGDKGE-Containing Cryptic Collagen Fragment Regulates Phosphorylation of Large Tumor Suppressor Kinase-1 and Controls Ovarian Tumor Growth by a Yes-Associated Protein-Dependent Mechanism. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:527-544. [PMID: 33307038 PMCID: PMC7927278 DOI: 10.1016/j.ajpath.2020.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/28/2020] [Accepted: 11/17/2020] [Indexed: 10/22/2022]
Abstract
The growth and spread of malignant tumors, such as ovarian carcinomas, are governed in part by complex interconnected signaling cascades occurring between stromal and tumor cells. These reciprocal cross-talk signaling networks operating within the local tissue microenvironment may enhance malignant tumor progression. Understanding how novel bioactive molecules generated within the tumor microenvironment regulate signaling pathways in distinct cellular compartments is critical for the development of more effective treatment paradigms. Herein, we provide evidence that blocking cellular interactions with an RGDKGE-containing collagen peptide that selectively binds integrin β3 on ovarian tumor cells enhances the phosphorylation of the hippo effector kinase large tumor suppressor kinase-1 and reduces nuclear accumulation of yes-associated protein and its target gene c-Myc. Selectively targeting this RGDKGE-containing collagen fragment inhibited ovarian tumor growth and the development of ascites fluid in vivo. These findings suggest that this bioactive collagen fragment may represent a previously unknown regulator of the hippo effector kinase large tumor suppressor kinase-1 and regulate ovarian tumor growth by a yes-associated protein-dependent mechanism. Taken together, these data not only provide new mechanistic insight into how a unique collagen fragment may regulate ovarian cancer, but in addition may help provide a useful new alternative strategy to control ovarian tumor progression based on selectively disrupting a previously unappreciated signaling cascade.
Collapse
Affiliation(s)
- XiangHua Han
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Jennifer M Caron
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Christine W Lary
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Pradeep Sathyanarayana
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Calvin Vary
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Peter C Brooks
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine.
| |
Collapse
|
30
|
Yang X, Wu Q, Wu F, Zhong Y. Differential expression of COL4A3 and collagen in upward and downward progressing types of nasopharyngeal carcinoma. Oncol Lett 2021; 21:223. [PMID: 33613712 PMCID: PMC7859474 DOI: 10.3892/ol.2021.12484] [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: 05/04/2020] [Accepted: 12/21/2020] [Indexed: 12/18/2022] Open
Abstract
Upward (local growth and invasion of the base of skull), downward (distant metastasis) and mixed progressing types of nasopharyngeal carcinoma (NPC) have been identified and are distinctly different with respect to clinical symptoms, therapeutic strategies and prognosis. The present study aimed to identify the genetic difference and collagen expression levels in the upward and downward progressing types of NPC. Whole exon sequencing (WES) was used to detect genes differentially mutated between the upward and downward progressing types of NPC. Collagen deposition in the upward and downward progressing types of NPC was determined using Masson trichromatic staining, while the protein expression level of COL4A3 was detected using immunohistochemistry. Survival analysis was also performed using the Kaplan-Meier Plotter database to examine the role of COL4A3 expression level in the prognosis of head and neck squamous cell carcinoma. Knockdown of COL4A3 was performed using short interfering (si)RNA-COL4A3 in a 5-8F NPC cell line. Reverse transcription-quantitative PCR and western blot analyses were utilized to analyze the mRNA and protein expression levels of COL4A3, respectively. The roles of COL4A3 in the migration and invasion of the 5-8F cell line were examined using wound-healing Transwell and Matrigel assays, respectively. A total of 21 genes were differentially mutated between the upward and downward progressing types of NPC. The COL4A3 was investigated further, as it was found to be associated with extracellular matrix deposition and cancer metastasis. The COL4A3 gene was markedly downregulated in the downward progressing type compared with that in the upward progressing type (2.161±1.306 vs. 5.077±3.619; P<0.05). In addition, the deposition of collagen in the downward progressing type was also significantly decreased compared with that in the upward progressing type (5.63±6.83 vs. 10.94±9.60; P<0.05). Kaplan-Meier analysis indicated that high expression level of COL4A3 was positively associated with a favorable prognosis of head and neck squamous cell carcinoma (HR, 0.69; 95% CI, 0.49- 0.97; P=0.031). To confirm the role of COL4A3, the expression level of COL4A3 was knocked down using siRNA in the 5-8F cell line and the results showed that the invasion and migration was significantly increased when the expression of COL4A3 was inhibited (P<0.0001). In conclusion, the gene mutation patterns were significantly different between the upward and downward progressing types of NPC. In addition, the expression level of the COL4A3 gene was decreased in the downward progressing type, which might promote NPC metastasis through the downregulation of extracellular collagen expression.
Collapse
Affiliation(s)
- Xiting Yang
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Fengyang Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yahua Zhong
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
- Correspondence to: Professor Yahua Zhong, Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang, Wuhan, Hubei 430071, P.R. China, E-mail:
| |
Collapse
|
31
|
Relapse of pathological angiogenesis: functional role of the basement membrane and potential treatment strategies. Exp Mol Med 2021; 53:189-201. [PMID: 33589713 PMCID: PMC8080572 DOI: 10.1038/s12276-021-00566-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/14/2020] [Accepted: 12/17/2020] [Indexed: 01/31/2023] Open
Abstract
Blinding eye diseases such as corneal neovascularization, proliferative diabetic retinopathy, and age-related macular degeneration are driven by pathological angiogenesis. In cancer, angiogenesis is key for tumor growth and metastasis. Current antiangiogenic treatments applied clinically interfere with the VEGF signaling pathway-the main angiogenic pathway-to inhibit angiogenesis. These treatments are, however, only partially effective in regressing new pathologic vessels, and the disease relapses following cessation of treatment. Moreover, the relapse of pathological angiogenesis can be rapid, aggressive and more difficult to treat than angiogenesis in the initial phase. The manner in which relapse occurs is poorly understood; however, recent studies have begun to shed light on the mechanisms underlying the revascularization process. Hypotheses have been generated to explain the rapid angiogenic relapse and increased resistance of relapsed disease to treatment. In this context, the present review summarizes knowledge of the various mechanisms of disease relapse gained from different experimental models of pathological angiogenesis. In addition, the basement membrane-a remnant of regressed vessels-is examined in detail to discuss its potential role in disease relapse. Finally, approaches for gaining a better understanding of the relapse process are discussed, including prospects for the management of relapse in the context of disease.
Collapse
|
32
|
Tao QR, Chu YM, Wei L, Tu C, Han YY. Antiangiogenic therapy in diabetic nephropathy: A double‑edged sword (Review). Mol Med Rep 2021; 23:260. [PMID: 33655322 PMCID: PMC7893700 DOI: 10.3892/mmr.2021.11899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 01/21/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetes and the associated complications are becoming a serious global threat and an increasing burden to human health and the healthcare systems. Diabetic nephropathy (DN) is the primary cause of end-stage kidney disease. Abnormal angiogenesis is well established to be implicated in the morphology and pathophysiology of DN. Factors that promote or inhibit angiogenesis serve an important role in DN. In the present review, the current issues associated with the vascular disease in DN are highlighted, and the challenges in the development of treatments are discussed.
Collapse
Affiliation(s)
- Qian-Ru Tao
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Ying-Ming Chu
- Department of Integrated Traditional Chinese Medicine, Peking University First Hospital, Beijing 100034, P.R. China
| | - Lan Wei
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Chao Tu
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Yuan-Yuan Han
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, P.R. China
| |
Collapse
|
33
|
Sugiyama A, Okada M, Otani K, Yamawaki H. [Development of basic research toward clinical application of cleaved fragment of type IV collagen]. Nihon Yakurigaku Zasshi 2021; 156:282-287. [PMID: 34470932 DOI: 10.1254/fpj.21016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Basement membrane is a dense sheet-like extracellular matrix (ECM), which separates cells from surrounding interstitium. Type IV collagen is a major component of basement membrane and three of six α chains (namely α1-α6 chains) form a triple-helix structure. Recently, endogenous bioactive factors called "matricryptins" or "matrikines", which are produced by degrading and cleaving C-terminal domain of type IV collagen, attract attentions as a novel therapeutic target or a candidate for biomarkers. In all type IV collagens, matricryptins called arresten (α1 chain), canstatin (α2), tumstatin (α3), tetrastatin (α4), pentastatin (α5), and hexastatin (α6), have been identified. The type IV collagen-derived matricryptins have been previously studied as new therapeutic targets for neoplastic diseases since they exert anti-angiogenic and/or anti-tumor effects. On the other hand, we have recently demonstrated the cardioprotective effects of matricryptins in addition to the altered expression levels in cardiac diseases. In this review, we introduce the results of fundamental studies for the type IV collagen-derived matricryptins in various diseases, such as neoplastic diseases and cardiac diseases, and discuss the potential clinical application as novel therapeutic agents and biomarkers.
Collapse
Affiliation(s)
- Akira Sugiyama
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University
| | - Muneyoshi Okada
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University
| | - Kosuke Otani
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University
| |
Collapse
|
34
|
Extracellular Matrix Remodeling in Chronic Liver Disease. CURRENT TISSUE MICROENVIRONMENT REPORTS 2021; 2:41-52. [PMID: 34337431 PMCID: PMC8300084 DOI: 10.1007/s43152-021-00030-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/09/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE OF THE REVIEW This review aims to summarize the current knowledge of the extracellular matrix remodeling during hepatic fibrosis. We discuss the diverse interactions of the extracellular matrix with hepatic cells and the surrounding matrix in liver fibrosis, with the focus on the molecular pathways and the mechanisms that regulate extracellular matrix remodeling. RECENT FINDINGS The extracellular matrix not only provides structure and support for the cells, but also controls cell behavior by providing adhesion signals and by acting as a reservoir of growth factors and cytokines. SUMMARY Hepatic fibrosis is characterized by an excessive accumulation of extracellular matrix. During fibrogenesis, the natural remodeling process of the extracellular matrix varies, resulting in the excessive accumulation of its components, mainly collagens. Signals released by the extracellular matrix induce the activation of hepatic stellate cells, which are the major source of extracellular matrix and most abundant myofibroblasts in the liver. GRAPHICAL ABSTRACT
Collapse
|
35
|
Frangogiannis NG, Kovacic JC. Extracellular Matrix in Ischemic Heart Disease, Part 4/4: JACC Focus Seminar. J Am Coll Cardiol 2020; 75:2219-2235. [PMID: 32354387 DOI: 10.1016/j.jacc.2020.03.020] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 02/06/2023]
Abstract
Myocardial ischemia and infarction, both in the acute and chronic phases, are associated with cardiomyocyte loss and dramatic changes in the cardiac extracellular matrix (ECM). It has long been appreciated that these changes in the cardiac ECM result in altered mechanical properties of ischemic or infarcted myocardial segments. However, a growing body of evidence now clearly demonstrates that these alterations of the ECM not only affect the structural properties of the ischemic and post-infarct heart, but they also play a crucial and sometimes direct role in mediating a range of biological pathways, including the orchestration of inflammatory and reparative processes, as well as the pathogenesis of adverse remodeling. This final part of a 4-part JACC Focus Seminar reviews the evidence on the role of the ECM in relation to the ischemic and infarcted heart, as well as its contribution to cardiac dysfunction and adverse clinical outcomes.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York.
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Victor Chang Cardiac Research Institute and St. Vincent's Clinical School, University of New South Wales, Darlinghurst, New South Wales, Australia.
| |
Collapse
|
36
|
Odiatis C, Savva I, Pieri M, Ioannou P, Petrou P, Papagregoriou G, Antoniadou K, Makrides N, Stefanou C, Ljubanović DG, Nikolaou G, Borza DB, Stylianou K, Gross O, Deltas C. A glycine substitution in the collagenous domain of Col4a3 in mice recapitulates late onset Alport syndrome. Matrix Biol Plus 2020; 9:100053. [PMID: 33718859 PMCID: PMC7930875 DOI: 10.1016/j.mbplus.2020.100053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022] Open
Abstract
Alport syndrome (AS) is a severe inherited glomerulopathy caused by mutations in the genes encoding the α-chains of type-IV collagen, the most abundant component of the extracellular glomerular basement membrane (GBM). Currently most AS mouse models are knockout models for one of the collagen-IV genes. In contrast, about half of AS patients have missense mutations, with single aminoacid substitutions of glycine being the most common. The only mouse model for AS with a homozygous knockin missense mutation, Col4a3-p.Gly1332Glu, was partly described before by our group. Here, a detailed in-depth description of the same mouse is presented, along with another compound heterozygous mouse that carries the glycine substitution in trans with a knockout allele. Both mice recapitulate essential features of AS, including shorten lifespan by 30–35%, increased proteinuria, increased serum urea and creatinine, pathognomonic alternate GBM thinning and thickening, and podocyte foot process effacement. Notably, glomeruli and tubuli respond differently to mutant collagen-IV protomers, with reduced expression in tubules but apparently normal in glomeruli. However, equally important is the fact that in the glomeruli the mutant α3-chain as well as the normal α4/α5 chains seem to undergo a cleavage at, or near the point of the mutation, possibly by the metalloproteinase MMP-9, producing a 35 kDa C-terminal fragment. These mouse models represent a good tool for better understanding the spectrum of molecular mechanisms governing collagen-IV nephropathies and could be used for pre-clinical studies aimed at better treatments for AS. Two mouse models were generated that recapitulate essential features of AS patients. Glomeruli and tubuli respond differently to mutant collagen IV protomers. The mutant colIV protomers in glomeruli probably undergo a cleavage process by MMP9. The two AS mouse models represent a good tool for studying collagen-IV nephropathies. These models could be used for pre-clinical studies aimed at better treatments.
Collapse
Key Words
- ARAS, autosomal recessive alport syndrome
- AS, alport syndrome
- Alport syndrome
- BSA, bovine serum albumin
- Collagen-IV
- EM, electron microscopy
- ESRD, end stage renal disease
- GBM, glomerular basement membrane
- Glomerular basement membrane
- Glycine missense mutation
- Kidney disease
- Mouse model
- PAS, periodic acid schiff
- TBM, tubular basement membrane
- TGF-b1, transforming growth factor beta1
- UPR, unfolded protein response
Collapse
Affiliation(s)
- Christoforos Odiatis
- Center of Excellence in Biobanking and Biomedical Research, Molecular Medicine Research Center, University of Cyprus Medical School, Cyprus
| | - Isavella Savva
- Center of Excellence in Biobanking and Biomedical Research, Molecular Medicine Research Center, University of Cyprus Medical School, Cyprus
| | - Myrtani Pieri
- Department of Life and Health Sciences, School of Sciences and Engineering, University of Nicosia, Cyprus
| | - Pavlos Ioannou
- Center of Excellence in Biobanking and Biomedical Research, Molecular Medicine Research Center, University of Cyprus Medical School, Cyprus
| | - Petros Petrou
- Department of Biochemistry, The Cyprus Institute of Neurology and Genetics, Cyprus
| | - Gregory Papagregoriou
- Center of Excellence in Biobanking and Biomedical Research, Molecular Medicine Research Center, University of Cyprus Medical School, Cyprus
| | - Kyriaki Antoniadou
- Center of Excellence in Biobanking and Biomedical Research, Molecular Medicine Research Center, University of Cyprus Medical School, Cyprus
| | - Neoklis Makrides
- Department of Developmental Functional Genetics, The Cyprus Institute of Neurology and Genetics, Cyprus
| | - Charalambos Stefanou
- Center of Excellence in Biobanking and Biomedical Research, Molecular Medicine Research Center, University of Cyprus Medical School, Cyprus
| | | | - Georgios Nikolaou
- Veterinary diagnostic laboratory, Vet ex Machina LTD, Nicosia, Cyprus
| | - Dorin-Bogdan Borza
- Dept. of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN, United States of America
| | - Kostas Stylianou
- Department of Nephrology, University of Crete Medical School, Greece
| | - Oliver Gross
- Clinic for Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| | - Constantinos Deltas
- Center of Excellence in Biobanking and Biomedical Research, Molecular Medicine Research Center, University of Cyprus Medical School, Cyprus
| |
Collapse
|
37
|
Niland S, Eble JA. Hold on or Cut? Integrin- and MMP-Mediated Cell-Matrix Interactions in the Tumor Microenvironment. Int J Mol Sci 2020; 22:ijms22010238. [PMID: 33379400 PMCID: PMC7794804 DOI: 10.3390/ijms22010238] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
The tumor microenvironment (TME) has become the focus of interest in cancer research and treatment. It includes the extracellular matrix (ECM) and ECM-modifying enzymes that are secreted by cancer and neighboring cells. The ECM serves both to anchor the tumor cells embedded in it and as a means of communication between the various cellular and non-cellular components of the TME. The cells of the TME modify their surrounding cancer-characteristic ECM. This in turn provides feedback to them via cellular receptors, thereby regulating, together with cytokines and exosomes, differentiation processes as well as tumor progression and spread. Matrix remodeling is accomplished by altering the repertoire of ECM components and by biophysical changes in stiffness and tension caused by ECM-crosslinking and ECM-degrading enzymes, in particular matrix metalloproteinases (MMPs). These can degrade ECM barriers or, by partial proteolysis, release soluble ECM fragments called matrikines, which influence cells inside and outside the TME. This review examines the changes in the ECM of the TME and the interaction between cells and the ECM, with a particular focus on MMPs.
Collapse
|
38
|
Song R, Zhang L. Cardiac ECM: Its Epigenetic Regulation and Role in Heart Development and Repair. Int J Mol Sci 2020; 21:ijms21228610. [PMID: 33203135 PMCID: PMC7698074 DOI: 10.3390/ijms21228610] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/07/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
The extracellular matrix (ECM) is the non-cellular component in the cardiac microenvironment, and serves essential structural and regulatory roles in establishing and maintaining tissue architecture and cellular function. The patterns of molecular and biochemical ECM alterations in developing and adult hearts depend on the underlying injury type. In addition to exploring how the ECM regulates heart structure and function in heart development and repair, this review conducts an inclusive discussion of recent developments in the role, function, and epigenetic guidelines of the ECM. Moreover, it contributes to the development of new therapeutics for cardiovascular disease.
Collapse
Affiliation(s)
- Rui Song
- Correspondence: (R.S.); (L.Z.); Tel.: +1-909-558-4325 (R.S. & L.Z.)
| | - Lubo Zhang
- Correspondence: (R.S.); (L.Z.); Tel.: +1-909-558-4325 (R.S. & L.Z.)
| |
Collapse
|
39
|
Karsdal MA, Kraus VB, Shevell D, Bay-Jensen AC, Schattenberg J, Rambabu Surabattula R, Schuppan D. Profiling and targeting connective tissue remodeling in autoimmunity - A novel paradigm for diagnosing and treating chronic diseases. Autoimmun Rev 2020; 20:102706. [PMID: 33188918 DOI: 10.1016/j.autrev.2020.102706] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 08/16/2020] [Indexed: 12/14/2022]
Abstract
Connective tissue (ConT) remodeling is an essential process in tissue regeneration, where a balanced replacement of old tissue by new tissue occurs. This balance is disturbed in chronic diseases, often autoimmune diseases, usually resulting in the buld up of fibrosis and a gradual loss of organ function. During progression of liver, lung, skin, heart, joint, skeletal and kidney diseasesboth ConT formation and degradation are elevated, which is tightly linked to immune cell activation and a loss of specific cell types and extracellular matrix (ECM) structures that are required for normal organ function. Here, we address the balance of key general and organ specific components of the ECM during homeostasis and in disease, with a focus on collagens, which are emerging as both structural and signaling molecules harbouring neoepitopes and autoantigens that are released during ConT remodeling. Specific collagen molecular signatures of ConT remodeling are linked to disease activity and stage, and to prognosis across different organs. These signatures accompany and further drive disease progression, and often become detectable before clinical disease manifestation (illness). Recent advances allow to quantify and define the nature of ConT remodeling via blood-based assays that measure the levels of well-defined collagen fragments, reflecting different facets of ConT formation and degradation, and associated immunological processes. These novel serum assays are becoming important tools of precision medicine, to detect various chronic and autoimmune diseases before their clinical manifestation, and to non-invasively monitor the efficacy of a broad range of pharmacological interventions.
Collapse
Affiliation(s)
- Morten Asser Karsdal
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Metabolic Liver Research Program, Denmark
| | - Virginia Byers Kraus
- Duke Molecular Physiology Institute and Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Diane Shevell
- Clinical Biomarkers and Immunology, Bristol-Myers Squibb, Westfield, NJ, USA
| | | | | | - R Rambabu Surabattula
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
40
|
Damjanović A, Kolundžija B, Matić IZ, Krivokuća A, Zdunić G, Šavikin K, Janković R, Stanković JA, Stanojković TP. Mahonia aquifolium Extracts Promote Doxorubicin Effects against Lung Adenocarcinoma Cells In Vitro. Molecules 2020; 25:E5233. [PMID: 33182665 PMCID: PMC7697947 DOI: 10.3390/molecules25225233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/26/2020] [Accepted: 10/26/2020] [Indexed: 11/16/2022] Open
Abstract
Mahonia aquifolium and its secondary metabolites have been shown to have anticancer potential. We performed MTT, scratch, and colony formation assays; analyzed cell cycle phase distribution and doxorubicin uptake and retention with flow cytometry; and detected alterations in the expression of genes involved in the formation of cell-cell interactions and migration using quantitative real-time PCR following treatment of lung adenocarcinoma cells with doxorubicin, M. aquifolium extracts, or their combination. MTT assay results suggested strong synergistic effects of the combined treatments, and their application led to an increase in cell numbers in the subG1 phase of the cell cycle. Both extracts were shown to prolong doxorubicin retention time in cancer cells, while the application of doxorubicin/extract combination led to a decrease in MMP9 expression. Furthermore, cells treated with doxorubicin/extract combinations were shown to have lower migratory and colony formation potentials than untreated cells or cells treated with doxorubicin alone. The obtained results suggest that nontoxic M. aquifolium extracts can enhance the activity of doxorubicin, thus potentially allowing the application of lower doxorubicin doses in vivo, which may decrease its toxic effects in normal tissues.
Collapse
Affiliation(s)
- Ana Damjanović
- Department for Experimental Oncology, Institute of Oncology and Radiology of Serbia, 11 000 Belgrade, Serbia; (A.D.); (B.K.); (I.Z.M.); (A.K.); (R.J.); (T.P.S.)
| | - Branka Kolundžija
- Department for Experimental Oncology, Institute of Oncology and Radiology of Serbia, 11 000 Belgrade, Serbia; (A.D.); (B.K.); (I.Z.M.); (A.K.); (R.J.); (T.P.S.)
| | - Ivana Z. Matić
- Department for Experimental Oncology, Institute of Oncology and Radiology of Serbia, 11 000 Belgrade, Serbia; (A.D.); (B.K.); (I.Z.M.); (A.K.); (R.J.); (T.P.S.)
| | - Ana Krivokuća
- Department for Experimental Oncology, Institute of Oncology and Radiology of Serbia, 11 000 Belgrade, Serbia; (A.D.); (B.K.); (I.Z.M.); (A.K.); (R.J.); (T.P.S.)
| | - Gordana Zdunić
- Department for Pharmaceutical Investigations and Development, Institute for Medicinal Plant Research, Dr. Josif Pančić, 11 070 Belgrade, Serbia; (G.Z.); (K.Š.)
| | - Katarina Šavikin
- Department for Pharmaceutical Investigations and Development, Institute for Medicinal Plant Research, Dr. Josif Pančić, 11 070 Belgrade, Serbia; (G.Z.); (K.Š.)
| | - Radmila Janković
- Department for Experimental Oncology, Institute of Oncology and Radiology of Serbia, 11 000 Belgrade, Serbia; (A.D.); (B.K.); (I.Z.M.); (A.K.); (R.J.); (T.P.S.)
| | - Jelena Antić Stanković
- Department for Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 11 221 Belgrade, Serbia
| | - Tatjana P. Stanojković
- Department for Experimental Oncology, Institute of Oncology and Radiology of Serbia, 11 000 Belgrade, Serbia; (A.D.); (B.K.); (I.Z.M.); (A.K.); (R.J.); (T.P.S.)
| |
Collapse
|
41
|
Quesnel A, Karagiannis GS, Filippou PS. Extracellular proteolysis in glioblastoma progression and therapeutics. Biochim Biophys Acta Rev Cancer 2020; 1874:188428. [PMID: 32956761 DOI: 10.1016/j.bbcan.2020.188428] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/20/2022]
Abstract
Gliomas encompass highly invasive primary central nervous system (CNS) tumours of glial cell origin with an often-poor clinical prognosis. Of all gliomas, glioblastoma is the most aggressive form of primary brain cancer. Current treatments in glioblastoma are insufficient due to the invasive nature of brain tumour cells, which typically results in local tumour recurrence following treatment. The latter represents the most important cause of mortality in glioblastoma and underscores the necessity for an in-depth understanding of the underlying mechanisms. Interestingly, increased synthesis and secretion of several proteolytic enzymes within the tumour microenvironment, such as matrix metalloproteinases, lysosomal proteases, cathepsins and kallikreins for extracellular-matrix component degradation may play a major role in the aforementioned glioblastoma invasion mechanisms. These proteolytic networks are key players in establishing and maintaining a tumour microenvironment that promotes tumour cell survival, proliferation, and migration. Indeed, the targeted inhibition of these proteolytic enzymes has been a promisingly useful therapeutic strategy for glioblastoma management in both preclinical and clinical development. We hereby summarize current advances on the biology of the glioblastoma tumour microenvironment, with a particular emphasis on the role of proteolytic enzyme families in glioblastoma invasion and progression, as well as on their subsequent prognostic value as biomarkers and their therapeutic targeting in the era of precision medicine.
Collapse
Affiliation(s)
- Agathe Quesnel
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BX, United Kingdom; National Horizons Centre, Teesside University, 38 John Dixon Ln, Darlington, DL1 1HG, United Kingdom
| | - George S Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, USA; Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, New York, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Panagiota S Filippou
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BX, United Kingdom; National Horizons Centre, Teesside University, 38 John Dixon Ln, Darlington, DL1 1HG, United Kingdom.
| |
Collapse
|
42
|
Park KC, Dharmasivam M, Richardson DR. The Role of Extracellular Proteases in Tumor Progression and the Development of Innovative Metal Ion Chelators that Inhibit their Activity. Int J Mol Sci 2020; 21:E6805. [PMID: 32948029 PMCID: PMC7555822 DOI: 10.3390/ijms21186805] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/21/2022] Open
Abstract
The crucial role of extracellular proteases in cancer progression is well-known, especially in relation to the promotion of cell invasion through extracellular matrix remodeling. This also occurs by the ability of extracellular proteases to induce the shedding of transmembrane proteins at the plasma membrane surface or within extracellular vesicles. This process results in the regulation of key signaling pathways by the modulation of kinases, e.g., the epidermal growth factor receptor (EGFR). Considering their regulatory roles in cancer, therapeutics targeting various extracellular proteases have been discovered. These include the metal-binding agents di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT) and di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC), which increase c-MET degradation by multiple mechanisms. Both the direct and indirect inhibition of protease expression and activity can be achieved through metal ion depletion. Considering direct mechanisms, chelators can bind zinc(II) that plays a catalytic role in enzyme activity. In terms of indirect mechanisms, Dp44mT and DpC potently suppress the expression of the kallikrein-related peptidase-a prostate-specific antigen-in prostate cancer cells. The mechanism of this activity involves promotion of the degradation of the androgen receptor. Additional suppressive mechanisms of Dp44mT and DpC on matrix metalloproteases (MMPs) relate to their ability to up-regulate the metastasis suppressors N-myc downstream regulated gene-1 (NDRG1) and NDRG2, which down-regulate MMPs that are crucial for cancer cell invasion.
Collapse
Affiliation(s)
- Kyung Chan Park
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building, University of Sydney, Sydney 2006, Australia; (K.C.P.); (M.D.)
| | - Mahendiran Dharmasivam
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building, University of Sydney, Sydney 2006, Australia; (K.C.P.); (M.D.)
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute of Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Des R. Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building, University of Sydney, Sydney 2006, Australia; (K.C.P.); (M.D.)
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute of Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
43
|
Mutgan AC, Jandl K, Kwapiszewska G. Endothelial Basement Membrane Components and Their Products, Matrikines: Active Drivers of Pulmonary Hypertension? Cells 2020; 9:cells9092029. [PMID: 32899187 PMCID: PMC7563239 DOI: 10.3390/cells9092029] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/27/2020] [Accepted: 08/29/2020] [Indexed: 12/19/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a vascular disease that is characterized by elevated pulmonary arterial pressure (PAP) due to progressive vascular remodeling. Extracellular matrix (ECM) deposition in pulmonary arteries (PA) is one of the key features of vascular remodeling. Emerging evidence indicates that the basement membrane (BM), a specialized cluster of ECM proteins underlying the endothelium, may be actively involved in the progression of vascular remodeling. The BM and its steady turnover are pivotal for maintaining appropriate vascular functions. However, the pathologically elevated turnover of BM components leads to an increased release of biologically active short fragments, which are called matrikines. Both BM components and their matrikines can interfere with pivotal biological processes, such as survival, proliferation, adhesion, and migration and thus may actively contribute to endothelial dysfunction. Therefore, in this review, we summarize the emerging role of the BM and its matrikines on the vascular endothelium and further discuss its implications on lung vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Ayse Ceren Mutgan
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, 8010 Graz, Austria;
| | - Katharina Jandl
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria;
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - Grazyna Kwapiszewska
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, 8010 Graz, Austria;
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria;
- Correspondence:
| |
Collapse
|
44
|
Abstract
Chronic liver injury due to viral hepatitis, alcohol abuse, and metabolic disorders is a worldwide health concern. Insufficient treatment of chronic liver injury leads to fibrosis, causing liver dysfunction and carcinogenesis. Most cases of hepatocellular carcinoma (HCC) develop in the fibrotic liver. Pathological features of liver fibrosis include extracellular matrix (ECM) accumulation, mesenchymal cell activation, immune deregulation, and angiogenesis, all of which contribute to the precancerous environment, supporting tumor development. Among liver cells, hepatic stellate cells (HSCs) and macrophages play critical roles in fibrosis and HCC. These two cell types interplay and remodel the ECM and immune microenvironment in the fibrotic liver. Once HCC develops, HCC-derived factors influence HSCs and macrophages to switch to protumorigenic cell populations, cancer-associated fibroblasts and tumor-associated macrophages, respectively. This review aims to summarize currently available data on the roles of HSCs and macrophages in liver fibrosis and HCC, with a focus on their interaction.
Collapse
Affiliation(s)
- Michitaka Matsuda
- Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Ekihiro Seki
- Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
45
|
Amruta N, Rahman AA, Pinteaux E, Bix G. Neuroinflammation and fibrosis in stroke: The good, the bad and the ugly. J Neuroimmunol 2020; 346:577318. [PMID: 32682140 PMCID: PMC7794086 DOI: 10.1016/j.jneuroim.2020.577318] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/01/2020] [Accepted: 07/03/2020] [Indexed: 02/08/2023]
Abstract
Stroke is the leading cause of death and the main cause of disability in surviving patients. The detrimental interaction between immune cells, glial cells, and matrix components in stroke pathology results in persistent inflammation that progresses to fibrosis. A substantial effort is being directed toward understanding the exact neuroinflammatory events that take place as a result of stroke. The initiation of a potent cytokine response, along with immune cell activation and infiltration in the ischemic core, has massive acute deleterious effects, generally exacerbated by comorbid inflammatory conditions. There is secondary neuroinflammation that promotes further injury, resulting in cell death, but conversely plays a beneficial role, by promoting recovery. This highlights the need for a better understanding of the neuroinflammatory and fibrotic processes, as well as the need to identify new mechanisms and potential modulators. In this review, we summarize several aspects of stroke-induced inflammation, fibrosis, and include a discussion of cytokine inhibitors/inducers, immune cells, and fibro-inflammation signaling inhibitors in order to identify new pharmacological means of intervention.
Collapse
Affiliation(s)
- Narayanappa Amruta
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Abir A Rahman
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom.
| | - Gregory Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA; Faculty of Biology, Medicine and Health, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA.
| |
Collapse
|
46
|
Matsuda M, Seki E. The liver fibrosis niche: Novel insights into the interplay between fibrosis-composing mesenchymal cells, immune cells, endothelial cells, and extracellular matrix. Food Chem Toxicol 2020; 143:111556. [PMID: 32640349 DOI: 10.1016/j.fct.2020.111556] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 12/11/2022]
Abstract
Liver fibrosis is a hepatic wound-healing response caused by chronic liver diseases that include viral hepatitis, alcoholic liver disease, non-alcoholic steatohepatitis, and cholestatic liver disease. Liver fibrosis eventually progresses to cirrhosis that is histologically characterized by an abnormal liver architecture that includes distortion of liver parenchyma, formation of regenerative nodules, and a massive accumulation of extracellular matrix (ECM). Despite intensive investigations into the underlying mechanisms of liver fibrosis, developments of anti-fibrotic therapies for liver fibrosis are still unsatisfactory. Recent novel experimental approaches, such as single-cell RNA sequencing and proteomics, have revealed the heterogeneity of ECM-producing cells (mesenchymal cells) and ECM-regulating cells (immune cells and endothelial cells). These approaches have accelerated the identification of fibrosis-specific subpopulations among these cell types. The ECM also consists of heterogenous components. Their production, degradation, deposition, and remodeling are dynamically regulated in liver fibrosis, further affecting the functions of cells responsible for fibrosis. These cellular and ECM elements cooperatively form a unique microenvironment: a fibrotic niche. Understanding the complex interplay between these elements could lead to a better understanding of underlying fibrosis mechanisms and to the development of effective therapies.
Collapse
Affiliation(s)
- Michitaka Matsuda
- Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Ekihiro Seki
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
47
|
van Huizen NA, Ijzermans JNM, Burgers PC, Luider TM. Collagen analysis with mass spectrometry. MASS SPECTROMETRY REVIEWS 2020; 39:309-335. [PMID: 31498911 DOI: 10.1002/mas.21600] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 07/17/2019] [Accepted: 07/17/2019] [Indexed: 06/10/2023]
Abstract
Mass spectrometry-based techniques can be applied to investigate collagen with respect to identification, quantification, supramolecular organization, and various post-translational modifications. The continuous interest in collagen research has led to a shift from techniques to analyze the physical characteristics of collagen to methods to study collagen abundance and modifications. In this review, we illustrate the potential of mass spectrometry for in-depth analyses of collagen.
Collapse
Affiliation(s)
- Nick A van Huizen
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Surgery, Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Jan N M Ijzermans
- Department of Surgery, Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Peter C Burgers
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Theo M Luider
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
48
|
Villesen IF, Daniels SJ, Leeming DJ, Karsdal MA, Nielsen MJ. Review article: the signalling and functional role of the extracellular matrix in the development of liver fibrosis. Aliment Pharmacol Ther 2020; 52:85-97. [PMID: 32419162 DOI: 10.1111/apt.15773] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/17/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Patients with liver fibrosis show a large heterogeneity, and for that reason effective treatments are still lacking. Emerging data suggest that there is more to fibrosis than previously understood. Opposed to earlier belief of being a passive scaffold for cells to reside in, the extracellular matrix (ECM) is now known to hold both signalling and functional properties important for the development of fibrosis. The interaction between the ECM and the collagen-producing cells determines the course of the disease but is still poorly understood. Exploring the dynamics of this interplay will aid in the development of effective treatments. AIM To summarise and discuss the latest advances in the pathogenesis of liver fibrosis as well as key mediators of early disease progression. METHODS Through literature search using databases including PubMed and Google Scholar, manuscripts published between 1961 and 2019 were included to assess both well-established and recent theories of fibrosis development. Both pre-clinical and clinical studies were included. RESULTS Fibrosis alters the structure of the ECM releasing signalling fragments with the potential to escalate disease severity. In a diseased liver, hepatic stellate cells and other fibroblasts, together with hepatocytes and sinusoidal cells, produce an excessive amount of collagens. The cell-to-collagen interactions are unique in the different liver aetiologies, generating ECM profiles with considerable patient-monitoring potential. CONCLUSIONS The local milieu in the injured area affects the course of fibrosis development in a site-specific manner. Future research should focus on the dissimilarities in the ECM profile between different aetiologies of liver fibrosis.
Collapse
Affiliation(s)
- Ida Falk Villesen
- Nordic Bioscience A/S, Herlev, Denmark.,University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
49
|
Barillari G. The Impact of Matrix Metalloproteinase-9 on the Sequential Steps of the Metastatic Process. Int J Mol Sci 2020; 21:ijms21124526. [PMID: 32630531 PMCID: PMC7350258 DOI: 10.3390/ijms21124526] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023] Open
Abstract
In industrialized countries, cancer is the second leading cause of death after cardiovascular disease. Most cancer patients die because of metastases, which consist of the self-transplantation of malignant cells in anatomical sites other than the one from where the tumor arose. Disseminated cancer cells retain the phenotypic features of the primary tumor, and display very poor differentiation indices and functional regulation. Upon arrival at the target organ, they replace preexisting, normal cells, thereby permanently compromising the patient's health; the metastasis can, in turn, metastasize. The spread of cancer cells implies the degradation of the extracellular matrix by a variety of enzymes, among which the matrix metalloproteinase (MMP)-9 is particularly effective. This article reviews the available published literature concerning the important role that MMP-9 has in the metastatic process. Additionally, information is provided on therapeutic approaches aimed at counteracting, or even preventing, the development of metastasis via the use of MMP-9 antagonists.
Collapse
Affiliation(s)
- Giovanni Barillari
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 1 via Montpellier, 00133 Rome, Italy
| |
Collapse
|
50
|
Andreuzzi E, Capuano A, Poletto E, Pivetta E, Fejza A, Favero A, Doliana R, Cannizzaro R, Spessotto P, Mongiat M. Role of Extracellular Matrix in Gastrointestinal Cancer-Associated Angiogenesis. Int J Mol Sci 2020; 21:E3686. [PMID: 32456248 PMCID: PMC7279269 DOI: 10.3390/ijms21103686] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal tumors are responsible for more cancer-related fatalities than any other type of tumors, and colorectal and gastric malignancies account for a large part of these diseases. Thus, there is an urgent need to develop new therapeutic approaches to improve the patients' outcome and the tumor microenvironment is a promising arena for the development of such treatments. In fact, the nature of the microenvironment in the different gastrointestinal tracts may significantly influence not only tumor development but also the therapy response. In particular, an important microenvironmental component and a potential therapeutic target is the vasculature. In this context, the extracellular matrix is a key component exerting an active effect in all the hallmarks of cancer, including angiogenesis. Here, we summarized the current knowledge on the role of extracellular matrix in affecting endothelial cell function and intratumoral vascularization in the context of colorectal and gastric cancer. The extracellular matrix acts both directly on endothelial cells and indirectly through its remodeling and the consequent release of growth factors. We envision that a deeper understanding of the role of extracellular matrix and of its remodeling during cancer progression is of chief importance for the development of new, more efficacious, targeted therapies.
Collapse
Affiliation(s)
- Eva Andreuzzi
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Alessandra Capuano
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Evelina Poletto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Eliana Pivetta
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Albina Fejza
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Andrea Favero
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Roberto Doliana
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Renato Cannizzaro
- Department of Clinical Oncology, Experimental Gastrointestinal Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
| | - Paola Spessotto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Maurizio Mongiat
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| |
Collapse
|