1
|
Rambhia KJ, Sun H, Feng K, Kannan R, Doleyres Y, Holzwarth JM, Doepker M, Franceschi RT, Ma PX. Nanofibrous 3D scaffolds capable of individually controlled BMP and FGF release for the regulation of bone regeneration. Acta Biomater 2024:S1742-7061(24)00636-6. [PMID: 39486780 DOI: 10.1016/j.actbio.2024.10.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
The current clinical applications of bone morphogenetic proteins (BMPs) are limited to only a few specific indications. Locally controlled delivery of combinations of growth factors can be a promising strategy to improve BMP-based bone repair. However, the success of this approach requires the development of an effective release system and the correct choice of growth factors capable of enhancing BMP activity. Basic fibroblast growth factor (bFGF, also known as FGF-2) has shown promise in promoting bone repair, although conflicting results have been reported. Considering the complex biological activities of FGF-2, we hypothesized that FGF-2 can promote BMP-induced bone regeneration only if the dosage and kinetic parameters of the two factors are individually tailored. In this study, we conducted systematic in vitro studies on cell proliferation, differentiation, and mineralization in response to factor dose, delivery mode (sequential or simultaneous), and release rate. Subsequently, we designed individually controlled BMP-7 and FGF-2 release poly(lactide-co-glycolide) (PLGA) nanospheres attached to the poly(l-lactic acid) (PLLA) nanofibrous scaffolds. The data showed that BMP-7-induced bone formation was accelerated by a relatively higher FGF-2 dose (100 ng/scaffold) delivered at a faster release rate, or by a relatively lower FGF-2 dose (10 ng/scaffold) at a slower release rate in an in vivo bone regeneration model. In contrast, a very high dose of FGF-2 (1000 ng/scaffold) inhibited bone regeneration under all conditions. In vitro and in vivo data suggest that FGF-2 improved BMP-7-induced bone regeneration by coordinating FGF-2 dosage and release kinetics to enhance stem cell migration, proliferation, and angiogenesis. STATEMENT OF SIGNIFICANCE: Bone morphogenetic proteins (BMPs) are the most potent growth/differentiation factors in bone development and regeneration. However, the clinical applications of BMPs have been limited to only a few specific indications due to the required supraphysiological dosages with the current BMP products and their side effects. Locally controlled delivery of BMPs and additional growth factors that can enhance their osteogenic potency are highly desired. However, different growth factors act with different mechanisms. Here we report a nanofibrous scaffold that mimics collagen in size and geometry and is immobilized with biodegradable nanospheres to achieve local and distinct release profiles of BMP7 and FGF2. Systematic studies demonstrated low dose BMP7 and FGF2 with different temporal release profiles can optimally enhance bone regeneration.
Collapse
Affiliation(s)
- Kunal J Rambhia
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hongli Sun
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kai Feng
- Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rahasudha Kannan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yasmine Doleyres
- Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeremy M Holzwarth
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mikayla Doepker
- Department of Biology, Kalamazoo College, Kalamazoo, MI 49006, USA
| | - Renny T Franceschi
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter X Ma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, MI 48109, USA; Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
2
|
Shintani T, Higaki M, Rosli SNZ, Okamoto T. Potential treatment of squamous cell carcinoma by targeting heparin-binding protein 17/fibroblast growth factor-binding protein 1 with vitamin D 3 or eldecalcitol. In Vitro Cell Dev Biol Anim 2024; 60:583-589. [PMID: 38713345 PMCID: PMC11286729 DOI: 10.1007/s11626-024-00913-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 04/16/2024] [Indexed: 05/08/2024]
Abstract
Heparin-binding protein 17 (HBp17), first purified in 1991 from the conditioned medium of the human A431 squamous cell carcinoma (SCC) cell line, was later renamed fibroblast growth factor-binding protein 1 (FGFBP-1). HBp17/FGFBP-1 is specifically expressed and secreted by epithelial cells, and it reversibly binds to fibroblast growth factor (FGF)-1 and FGF-2, as well as FGFs-7, -10, and -22, indicating a crucial involvement in the transportation and function of these FGFs. Our laboratory has investigated and reported several studies to elucidate the function of HBp17/FGFBP-1 in SCC cells and its potential as a molecular therapeutic target. HBp17/FGFBP-1 transgene exoression in A431-4 cells, a clonal subline of A431 that lacks tumorigenicity and does not express HBp17/FGFBP-1, demonstrated a significantly enhanced proliferation in vitro compared with A431-4 cells, and it acquired tumorigenicity in the subcutis of nude mice. Knockout (KO) of the HBp17/FGFBP-1 by genome editing significantly suppressed tumor growth, cell motility, and tumorigenicity compared with control cells. A comprehensive analysis of expressed molecules in both cell types revealed that molecules that promote epithelial cell differentiation were highly expressed in HBp17/FGFBP-1 KO cells. Additionally, we reported that 1α,25(OH)2D3 or eldecalcitol (ED-71), which is an analog of 1α,25(OH)2D3, suppresses HBp17/FGFBP-1 expression and tumor growth in vitro and in vivo by inhibiting the nuclear factor kappa-light-chain-enhancer of activated B cells signaling pathway. Here, we discuss the prospects of molecular targeted therapy targeting HBp17/FGFBP-1 with 1α,25(OH)2D3 or ED71 in SCC and oral SCC.
Collapse
Affiliation(s)
- Tomoaki Shintani
- Center of Oral Clinical Examination, Hiroshima University Hospital, Hiroshima, 734-8551, Japan.
| | - Mirai Higaki
- Department of Molecular Oral Medicine and Maxilofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Siti Nur Zawani Rosli
- Department of Molecular Oral Medicine and Maxilofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
- Infectious Disease Research Center, Institute for Medical Research, Bacteriology Unit, National Institutes of Health, Ministry of Health Malaysia, 40170, Setia Alam, Malaysia
| | - Tetsuji Okamoto
- Department of Molecular Oral Medicine and Maxilofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
- School of Medical Sciences, University of East Asia, Shimonoseki, 751-8503, Japan
| |
Collapse
|
3
|
Bhargava K, Raghavendra SS, Mulay S, Hindlekar A, Kharat A, Kheur S. Evaluating the ability of cultivated odontoblasts to form dentin-like tissue in vitro using fibroblast growth factor and insulin-like growth factor. JOURNAL OF CONSERVATIVE DENTISTRY AND ENDODONTICS 2024; 27:598-602. [PMID: 38989496 PMCID: PMC11232758 DOI: 10.4103/jcde.jcde_174_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 07/12/2024]
Abstract
Aim The aim of the study was to evaluate the ability of cultivated odontoblast to form dentin-like tissue using fibroblast growth factor (FGF) and insulin-like growth factor (IGF). Materials and Methods Dental pulp stem cells (DPSCs) were extracted from 10 human teeth. They were isolated and cultivated in vitro with the use of stem cell markers. The human DPSCs were characterized for trilineage differentiation. They were then differentiated into odontoblasts. The ability of cultivated odontoblasts to form dentin-like tissue was evaluated using FGF and IGF. Results IGF showed superior ability to form dentin-like tissue as compared to FGF. The addition of FGF showed no significant difference in the formation of dentin-like tissue. A combination of FGF and IGF in odontoblast showed an enhanced ability to form dentin-like tissue. Conclusion The use of growth factors IGF and FGF with dental stem cells showed a greater potential to form dentin-like tissue. This can profoundly alter the paradigms of conservative vital pulp therapy, which may eventually make it possible to treat dental diseases by regeneration of lost dentine.
Collapse
Affiliation(s)
- Karan Bhargava
- Department of Conservative Dentistry and Endodontics, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Srinidhi Surya Raghavendra
- Department of Conservative Dentistry and Endodontics, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Sanjyot Mulay
- Department of Conservative Dentistry and Endodontics, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Ajit Hindlekar
- Department of Conservative Dentistry and Endodontics, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Avinash Kharat
- Department of Regenerative Medicine, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Supriya Kheur
- Department of Oral Pathology, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
| |
Collapse
|
4
|
Motta I, Soccio M, Guidotti G, Lotti N, Pasquinelli G. Hydrogels for Cardio and Vascular Tissue Repair and Regeneration. Gels 2024; 10:196. [PMID: 38534614 DOI: 10.3390/gels10030196] [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: 01/17/2024] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Cardiovascular disease (CVD), the leading cause of death globally, affects the heart and arteries with a variety of clinical manifestations, the most dramatic of which are myocardial infarction (MI), abdominal aortic aneurysm (AAA), and intracranial aneurysm (IA) rupture. In MI, necrosis of the myocardium, scar formation, and loss of cardiomyocytes result from insufficient blood supply due to coronary artery occlusion. Beyond stenosis, the arteries that are structurally and functionally connected to the cardiac tissue can undergo pathological dilation, i.e., aneurysmal dilation, with high risk of rupture. Aneurysms of the intracranial arteries (IAs) are more commonly seen in young adults, whereas those of the abdominal aorta (AAA) are predominantly seen in the elderly. IAs, unpredictably, can undergo rupture and cause life-threatening hemorrhage, while AAAs can result in rupture, internal bleeding and high mortality rate. In this clinical context, hydrogels, three-dimensional networks of water-seizing polymers, have emerged as promising biomaterials for cardiovascular tissue repair or protection due to their biocompatibility, tunable properties, and ability to encapsulate and release bioactive molecules. This review provides an overview of the current state of research on the use of hydrogels as an innovative platform to promote cardiovascular-specific tissue repair in MI and functional recovery or protection in aneurysmal dilation.
Collapse
Affiliation(s)
- Ilenia Motta
- Alma Mater Institute on Healthy Planet, University of Bologna, Via Massarenti 11, 40138 Bologna, Italy
| | - Michelina Soccio
- Civil, Chemical, Environmental and Materials Engineering Department, University of Bologna, Via Terracini 28, 40131 Bologna, Italy
| | - Giulia Guidotti
- Civil, Chemical, Environmental and Materials Engineering Department, University of Bologna, Via Terracini 28, 40131 Bologna, Italy
| | - Nadia Lotti
- Civil, Chemical, Environmental and Materials Engineering Department, University of Bologna, Via Terracini 28, 40131 Bologna, Italy
| | - Gianandrea Pasquinelli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
5
|
Jimenez SA, Piera-Velazquez S. Cellular Transdifferentiation: A Crucial Mechanism of Fibrosis in Systemic Sclerosis. Curr Rheumatol Rev 2024; 20:388-404. [PMID: 37921216 DOI: 10.2174/0115733971261932231025045400] [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: 05/12/2023] [Revised: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 11/04/2023]
Abstract
Systemic Sclerosis (SSc) is a systemic autoimmune disease of unknown etiology with a highly complex pathogenesis that despite extensive investigation is not completely understood. The clinical and pathologic manifestations of the disease result from three distinct processes: 1) Severe and frequently progressive tissue fibrosis causing exaggerated and deleterious accumulation of interstitial collagens and other extracellular matrix molecules in the skin and various internal organs; 2) extensive fibroproliferative vascular lesions affecting small arteries and arterioles causing tissue ischemic alterations; and 3) cellular and humoral immunity abnormalities with the production of numerous autoantibodies, some with very high specificity for SSc. The fibrotic process in SSc is one of the main causes of disability and high mortality of the disease. Owing to its essentially universal presence and the severity of its clinical effects, the mechanisms involved in the development and progression of tissue fibrosis have been extensively investigated, however, despite intensive investigation, the precise molecular mechanisms have not been fully elucidated. Several recent studies have suggested that cellular transdifferentiation resulting in the phenotypic conversion of various cell types into activated myofibroblasts may be one important mechanism. Here, we review the potential role that cellular transdifferentiation may play in the development of severe and often progressive tissue fibrosis in SSc.
Collapse
Affiliation(s)
- Sergio A Jimenez
- Department of Dermatology and Cutaneous Biology, Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia 19107, USA
| | - Sonsoles Piera-Velazquez
- Department of Dermatology and Cutaneous Biology, Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia 19107, USA
| |
Collapse
|
6
|
Cannon-Albright LA, Teerlink CC, Stevens J, Facelli JC, Carr SR, Allen-Brady K, Puri S, Bailey-Wilson JE, Musolf AM, Akerley W. A rare FGF5 candidate variant (rs112475347) for predisposition to nonsquamous, nonsmall-cell lung cancer. Int J Cancer 2023; 153:364-372. [PMID: 36916144 PMCID: PMC10182245 DOI: 10.1002/ijc.34510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/16/2023]
Abstract
A unique approach with rare resources was used to identify candidate variants predisposing to familial nonsquamous nonsmall-cell lung cancers (NSNSCLC). We analyzed sequence data from NSNSCLC-affected cousin pairs belonging to high-risk lung cancer pedigrees identified in a genealogy of Utah linked to statewide cancer records to identify rare, shared candidate predisposition variants. Variants were tested for association with lung cancer risk in UK Biobank. Evidence for linkage with lung cancer was also reviewed in families from the Genetic Epidemiology of Lung Cancer Consortium. Protein prediction modeling compared the mutation with reference. We sequenced NSNSCLC-affected cousin pairs from eight high-risk lung cancer pedigrees and identified 66 rare candidate variants shared in the cousin pairs. One variant in the FGF5 gene also showed significant association with lung cancer in UKBiobank. This variant was observed in 3/163 additional sampled Utah lung cancer cases, 2 of whom were related in another independent pedigree. Modeling of the predicted protein predicted a second binding site for SO4 that may indicate binding differences. This unique study identified multiple candidate predisposition variants for NSNSCLC, including a rare variant in FGF5 that was significantly associated with lung cancer risk and that segregated with lung cancer in the two pedigrees in which it was observed. FGF5 is an oncogenic factor in several human cancers, and the mutation found here (W81C) changes the binding ability of heparan sulfate to FGF5, which might lead to its deregulation. These results support FGF5 as a potential NSNSCLC predisposition gene and present additional candidate predisposition variants.
Collapse
Affiliation(s)
- Lisa A Cannon-Albright
- Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Craig C Teerlink
- Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Jeff Stevens
- Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Julio C Facelli
- Department of BioMedical Informatics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Clinical and Translational Science Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Shamus R Carr
- Thoracic Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kristina Allen-Brady
- Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Sonam Puri
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
- Medical Oncology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Joan E Bailey-Wilson
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, Maryland, USA
| | - Anthony M Musolf
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, Maryland, USA
| | - Wallace Akerley
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
- Medical Oncology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
7
|
Dong Y, Gu P, Yi Q, Hu H, Cheng X, Zhang Z, Zhang L, Bai Y. Development of polymeric microparticles for controlled release of bioactive drugs using modified solution enhanced dispersion by supercritical CO2. J Supercrit Fluids 2022. [DOI: 10.1016/j.supflu.2022.105723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
8
|
Saker Z, Rizk M, Bahmad HF, Nabha SM. Targeting Angiogenic Factors for the Treatment of Medulloblastoma. Curr Treat Options Oncol 2022; 23:864-886. [PMID: 35412196 DOI: 10.1007/s11864-022-00981-1] [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: 03/20/2022] [Indexed: 11/24/2022]
Abstract
OPINION STATEMENT Medulloblastoma (MB) is the most frequent pediatric brain tumor. Despite conventional therapy, MB patients have high mortality and morbidity rates mainly due to the incomplete understanding of the molecular and cellular processes involved in development of this cancer. Similar to other solid tumors, MB demonstrated high endothelial cell proliferation and angiogenic activity, wherein new blood vessels arise from the pre-existing vasculature, a process named angiogenesis. MB angiogenesis is considered a hallmark for MB development, progression, and metastasis emphasizing its potential target for antitumor therapy. However, angiogenesis is tightly regulated by a set of angiogenic factors making it a complex process to be targeted. Although agents targeting these factors and their receptors are early in development, the potential for their targeting may translate into improvement in the clinical care for MB patients. In this review, we focus on the most potent angiogenic factors and their corresponding receptors, highlighting their basic properties and expression in MB. We describe their contribution to MB tumorigenesis and angiogenesis and the potential therapeutic targeting of these factors.
Collapse
Affiliation(s)
- Zahraa Saker
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Mahdi Rizk
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hisham F Bahmad
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, 4300 Alton Rd, Miami Beach, FL, 33140, USA.
| | - Sanaa M Nabha
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
9
|
Heparin-Binding Protein 17/Fibroblast Growth Factor-Binding Protein-1 Knockout Inhibits Proliferation and Induces Differentiation of Squamous Cell Carcinoma Cells. Cancers (Basel) 2021; 13:cancers13112684. [PMID: 34072393 PMCID: PMC8199440 DOI: 10.3390/cancers13112684] [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: 04/23/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 01/17/2023] Open
Abstract
Simple Summary Fibroblast growth factor (FGF) plays an important role in tumor growth by inducing angiogenesis in addition to promoting the proliferation of squamous cell carcinoma (SCC) and oral squamous cell carcinoma (OSCC) cells. Heparin-binding protein 17/fibroblast growth factor-binding protein-1 (HBp17/FGFBP-1) purified from A431 cell-conditioned media based on its capacity to bind to FGF-1 and FGF-2 is recognized as a pro-angiogenic molecule as a consequence of its interaction with FGF-2. In this study, we have examined the functional role of HBp17/FGFBP-1 in A431 and HO-1-N-1 cells using the CRISPR/Cas9 technology. Our results showed that HBp17/FGFBP-1 knockout inhibited cell proliferation, colony formation, and cell motility compared to control. The amount of FGF-2 was decreased in culture medium conditioned by HBp17/FGFBP-1 knockout cells compared to control. We performed cDNA/protein expression analysis followed by Gene Ontology and protein–protein interaction analysis. The results demonstrate that both gene and protein expression related to epidermal development, cornification, and keratinization were upregulated in HBp17/FGFBP-1-knockout A431 and HO-1-N-1 cells. Abstract Heparin-binding protein 17/fibroblast growth factor-binding protein-1 (HBp17/FGFBP-1) has been observed to induce the tumorigenic potential of epithelial cells and is highly expressed in oral cancer cell lines and tissues. It is also recognized as a pro-angiogenic molecule because of its interaction with fibroblast growth factor (FGF)-2. In this study, we examined the functional role of HBp17/FGFBP-1 in A431 and HO-1-N-1 cells. Originally, HBp17/FGFBP-1 was purified from A431 cell-conditioned media based on its capacity to bind to FGF-1 and FGF-2. We isolated and established HBp17/FGFBP-1-knockout (KO)-A431 and KO-HO-1-N-1 cell lines using the clusters of regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (Cas9) gene editing technology. The amount of FGF-2 secreted into conditioned medium decreased for A431-HBp17-KO and HO-1-N-1-HBp17-KO cells compared to their WT counterparts. Functional assessment showed that HBp17/FGFBP-1 KO inhibited cell proliferation, colony formation, and cell motility in vitro. It also inhibited tumor growth in vivo compared to controls, which confirmed the significant difference in growth in vitro between HBp17-KO cells and wild-type (WT) cells, indicating that HBp17/FGFBP-1 is a potent therapeutic target in squamous cell carcinomas (SCC) and oral squamous cell carcinomas (OSCC). In addition, complementary DNA/protein expression analysis followed by Gene Ontology and protein–protein interaction (PPI) analysis using the Database for Visualization and Integrated Discovery and Search Tool for the Retrieval of Interacting Genes/Proteins showed that both gene and protein expression related to epidermal development, cornification, and keratinization were upregulated in A431-HBp17-KO and HO-1-N-1-KO cells. This is the first discovery of a novel role of HBp17/FGFBP-1 that regulates SCC and OSCC cell differentiation.
Collapse
|
10
|
Mendoza FA, Piera-Velazquez S, Jimenez SA. Tyrosine kinases in the pathogenesis of tissue fibrosis in systemic sclerosis and potential therapeutic role of their inhibition. Transl Res 2021; 231:139-158. [PMID: 33422651 DOI: 10.1016/j.trsl.2021.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/09/2020] [Accepted: 01/04/2021] [Indexed: 12/30/2022]
Abstract
Systemic sclerosis (SSc) is an idiopathic autoimmune disease with a heterogeneous clinical phenotype ranging from limited cutaneous involvement to rapidly progressive diffuse SSc. The most severe SSc clinical and pathologic manifestations result from an uncontrolled fibrotic process involving the skin and various internal organs. The molecular mechanisms responsible for the initiation and progression of the SSc fibrotic process have not been fully elucidated. Recently it has been suggested that tyrosine protein kinases play a role. The implicated kinases include receptor-activated tyrosine kinases and nonreceptor tyrosine kinases. The receptor kinases are activated following specific binding of growth factors (platelet-derived growth factor, fibroblast growth factor, or vascular endothelial growth factor). Other receptor kinases are the discoidin domain receptors activated by binding of various collagens, the ephrin receptors that are activated by ephrins and the angiopoetin-Tie-2s receptors. The nonreceptor tyrosine kinases c-Abl, Src, Janus, and STATs have also been shown to participate in SSc-associated tissue fibrosis. Currently, there are no effective disease-modifying therapies for SSc-associated tissue fibrosis. Therefore, extensive investigation has been conducted to examine whether tyrosine kinase inhibitors (TKIs) may exert antifibrotic effects. Here, we review the role of receptor and nonreceptor tyrosine kinases in the pathogenesis of the frequently progressive cutaneous and systemic fibrotic alterations in SSc, and the potential of TKIs as SSc disease-modifying antifibrotic therapeutic agents.
Collapse
Affiliation(s)
- Fabian A Mendoza
- Rheumatology Division, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania; Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sonsoles Piera-Velazquez
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sergio A Jimenez
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
11
|
Cavalli G, Colafrancesco S, Emmi G, Imazio M, Lopalco G, Maggio MC, Sota J, Dinarello CA. Interleukin 1α: a comprehensive review on the role of IL-1α in the pathogenesis and treatment of autoimmune and inflammatory diseases. Autoimmun Rev 2021; 20:102763. [PMID: 33482337 DOI: 10.1016/j.autrev.2021.102763] [Citation(s) in RCA: 166] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022]
Abstract
The interleukin (IL)-1 family member IL-1α is a ubiquitous and pivotal pro-inflammatory cytokine. The IL-1α precursor is constitutively present in nearly all cell types in health, but is released upon necrotic cell death as a bioactive mediator. IL-1α is also expressed by infiltrating myeloid cells within injured tissues. The cytokine binds the IL-1 receptor 1 (IL-1R1), as does IL-1β, and induces the same pro-inflammatory effects. Being a bioactive precursor released upon tissue damage and necrotic cell death, IL-1α is central to the pathogenesis of numerous conditions characterized by organ or tissue inflammation. These include conditions affecting the lung and respiratory tract, dermatoses and inflammatory skin disorders, systemic sclerosis, myocarditis, pericarditis, myocardial infarction, coronary artery disease, inflammatory thrombosis, as well as complex multifactorial conditions such as COVID-19, vasculitis and Kawasaki disease, Behcet's syndrome, Sjogren Syndrome, and cancer. This review illustrates the clinical relevance of IL-1α to the pathogenesis of inflammatory diseases, as well as the rationale for the targeted inhibition of this cytokine for treatment of these conditions. Three biologics are available to reduce the activities of IL-1α; the monoclonal antibody bermekimab, the IL-1 soluble receptor rilonacept, and the IL-1 receptor antagonist anakinra. These advances in mechanistic understanding and therapeutic management make it incumbent on physicians to be aware of IL-1α and of the opportunity for therapeutic inhibition of this cytokine in a broad spectrum of diseases.
Collapse
Affiliation(s)
- Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy, and Rare Diseases, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University, Milan, Italy.
| | - Serena Colafrancesco
- Dipartimento of Clinical Sciences (Internal Medicine, Anesthesia and Resuscitation, and Cardiology), Rheumatology Unit, Sapienza University of Rome, Rome, Italy
| | - Giacomo Emmi
- Department of Experimental and Clinical Medicine, Careggi University Hospital, Firenze, Italy
| | - Massimo Imazio
- University Division of Cardiology, Cardiovascular and Throracic Department, AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - Giuseppe Lopalco
- Department of Emergency and Organ Transplantation, Rheumatology Unit, University of Bari, Bari, Italy
| | - Maria Cristina Maggio
- Department of Health Promotion, Maternal and Infantile Care, Department of Internal Medicine and Medical Specialties "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Jurgen Sota
- Research Center of Systemic Autoinflammatory Diseases and Behçet's Disease Clinic, Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA.
| |
Collapse
|
12
|
Research Progress of Hair Cell Protection Mechanism. Neural Plast 2020; 2020:8850447. [PMID: 33133179 PMCID: PMC7568815 DOI: 10.1155/2020/8850447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/16/2020] [Accepted: 09/30/2020] [Indexed: 11/17/2022] Open
Abstract
How to prevent and treat hearing-related diseases through the protection of hair cells (HCs) is the focus in the field of hearing in recent years. Hearing loss caused by dysfunction or loss of HCs is the main cause of hearing diseases. Therefore, clarifying the related mechanisms of HC development, apoptosis, protection, and regeneration is the main goal of current hearing research. This review introduces the latest research on mechanism of HC protection and regeneration.
Collapse
|
13
|
Zhang C, Wang J, Xie Y, Wang L, Yang L, Yu J, Miyamoto A, Sun F. Development of FGF-2-loaded electrospun waterborne polyurethane fibrous membranes for bone regeneration. Regen Biomater 2020; 8:rbaa046. [PMID: 33732492 PMCID: PMC7947599 DOI: 10.1093/rb/rbaa046] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/06/2020] [Accepted: 09/11/2020] [Indexed: 12/24/2022] Open
Abstract
Guided bone regeneration (GBR) membrane has been used to improve functional outcomes for periodontal regeneration. However, few studies have focused on the biomimetic membrane mimicking the vascularization of the periodontal membrane. This study aimed to fabricate waterborne polyurethane (WPU) fibrous membranes loaded fibroblast growth factor-2 (FGF-2) via emulsion electrospinning, which can promote regeneration of periodontal tissue via the vascularization of the biomimetic GBR membrane. A biodegradable WPU was synthesized by using lysine and dimethylpropionic acid as chain extenders according to the rule of green chemical synthesis technology. The WPU fibers with FGF-2 was fabricated via emulsion electrospinning. The results confirmed that controlled properties of the fibrous membrane had been achieved with controlled degradation, suitable mechanical properties and sustained release of the factor. The immunohistochemical expression of angiogenic-related factors was positive, meaning that FGF-2 loaded in fibers can significantly promote cell vascularization. The fiber scaffold loaded FGF-2 has the potential to be used as a functional GBR membrane to promote the formation of extraosseous blood vessels during periodontal repairing.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China
| | - Jianxiong Wang
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China
| | - Yujie Xie
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China
| | - Li Wang
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China
| | - Lishi Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China
| | - Jihua Yu
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China
| | - Akira Miyamoto
- Faculty of Rehabilitation, Department of Physical Therapy, Kobe International University, Kobe, Japan
| | - Fuhua Sun
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China
- Correspondence address. Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Taiping Street 25, Luzhou 646000, P.R. China. Tel.: +81-18428397607; E-mail:
| |
Collapse
|
14
|
Tsutsui TW. Dental Pulp Stem Cells: Advances to Applications. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2020; 13:33-42. [PMID: 32104005 PMCID: PMC7025818 DOI: 10.2147/sccaa.s166759] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 12/07/2019] [Indexed: 12/18/2022]
Abstract
Dental pulp stem cells (DPSCs) have a high capacity for differentiation and the ability to regenerate a dentin/pulp-like complex. Numerous studies have provided evidence of DPSCs’ differentiation capacity, such as in neurogenesis, adipogenesis, osteogenesis, chondrogenesis, angiogenesis, and dentinogenesis. The molecular mechanisms and functions of DPSCs’ differentiation process are affected by growth factors and scaffolds. For example, growth factors such as basic fibroblast growth factor (bFGF), transforming growth factor-β (TGF-β), nerve growth factor (NGF), platelet-derived growth factor (PDGF), and bone morphogenic proteins (BMPs) influence DPSC fate, including in differentiation, cell proliferation, and wound healing. In addition, several types of scaffolds, such as collagen, hydrogel, decellularized bioscaffold, and nanofibrous spongy microspheres, have been used to characterize DPSC cellular attachment, migration, proliferation, differentiation, and functions. An appropriate combination of growth factors and scaffolds can enhance the differentiation capacity of DPSCs, in terms of optimizing not only dental-related expression but also dental pulp morphology. For a cell-based clinical approach, focus has been placed on the tissue engineering triad [cells/bioactive molecules (growth factors)/scaffolds] to characterize DPSCs. It is clear that a deep understanding of the mechanisms of stem cells, including their aging, self-renewal, microenvironmental homeostasis, and differentiation correlated with cell activity, the energy for which is provided from mitochondria, should provide new approaches for DPSC research and therapeutics. Mitochondrial functions and dynamics are related to the direction of stem cell differentiation, including glycolysis, oxidative phosphorylation, mitochondrial metabolism, mitochondrial transcription factor A (TFAM), mitochondrial elongation, and mitochondrial fusion and fission proteins. This review summarizes the effects of major growth factors and scaffolds for regenerating dentin/pulp-like complexes, as well as elucidating mitochondrial properties of DPSCs for the development of advanced applications research.
Collapse
Affiliation(s)
- Takeo W Tsutsui
- Department of Pharmacology, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan
| |
Collapse
|
15
|
Cavalli G, Cenci S. Autophagy and Protein Secretion. J Mol Biol 2020; 432:2525-2545. [PMID: 31972172 DOI: 10.1016/j.jmb.2020.01.015] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/08/2020] [Accepted: 01/12/2020] [Indexed: 12/13/2022]
Abstract
Autophagy - conventional for macroautophagy - is a major recycling strategy that ensures cellular homeostasis through the selective engulfment of cytoplasmic supramolecular cargos in double membrane vesicles and their rapid dispatch to the lysosome for digestion. As autophagy operates in the cytoplasm, its interference with secretory proteins, that is, proteins destined to the plasma membrane or the extracellular space, generally synthesized and routed within the endoplasmic reticulum (ER), has been relatively overlooked in the past. However, mounting evidence reveals that autophagy in fact heavily regulates protein secretion through diverse mechanisms. First, autophagy is closely involved in the unconventional secretion of leaderless proteins, a pool of proteins destined extracellularly, but lacking an ER-targeted leader sequence, and thus manufactured in the cytosol. Autophagy-related (ATG) genes now appear instrumental to the underlying pathways, hence the recently coined concept of secretory autophagy, or better ATG gene-dependent secretion. Indeed, ATG genes regulate unconventional protein secretion at multiple levels, ranging from intracellular inflammatory signaling, for example, through the control of mitochondrial health and inflammasome activity, to trafficking of leaderless proteins. Moreover, perhaps less expectedly, autophagy also participates in the control of conventional secretion, intersecting the secretory apparatus at multiple points, though with surprising differences among professional secretory cell types that disclose remarkable and unpredicted specificity. This review synopsizes the multiple mechanisms whereby autophagy interfaces with conventional and unconventional protein secretory pathways and discusses the relative teleology. Altogether, the diverse controls exerted on protein secretion broaden and deepen the homeostatic significance of autophagy within the cell.
Collapse
Affiliation(s)
- Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| | - Simone Cenci
- Vita-Salute San Raffaele University, Milano, Italy; Unit of Age Related Diseases, Division of Genetics and Cell Biology, Ospedale San Raffaele, Milano, Italy.
| |
Collapse
|
16
|
Sun M, Jin L, Bai Y, Wang L, Zhao S, Ma C, Ma D. Fibroblast growth factor 21 protects against pathological cardiac remodeling by modulating galectin-3 expression. J Cell Biochem 2019; 120:19529-19540. [PMID: 31286550 DOI: 10.1002/jcb.29260] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/11/2019] [Indexed: 11/09/2022]
Abstract
BACKGROUND/AIMS Fibroblast growth factor 21 (FGF21) plays a protective role in ischemia/reperfusion induced cardiac injury. However, the exact molecular mechanism of FGF21 action remains unclear. This study was designed the protective effect of FGF21 on the heart and its mechanism. METHOD Adenovirus vector expressing FGF21 or control β-galactosidase was injected into the myocardium of mice. Myocardial injury was observed by tissue staining and immunohistochemical staining. The expression level of caspases-3 and galectin-3 in myocardial cells were observed by immunoblotting. Then, hypoxia-induced cell model was established. Small interfering RNA (SiRNA) and plasmid were transfected into H9c2 using Lipofectamine 2000 reagent (Invitrogen). The expression levels of galectin-3, ECM and cystatin-3 in cells were observed by immunoblotting, and the relationship between fibroblast growth factor 21 and galectin-3 was analyzed. RESULT Cell test in vitro showed that FGF21 could inhibit apoptosis and decrease the expression of ECM (ColIaI, fibronectin, and alpha-SMA) under hypoxia. Western blot data showed that hypoxia-induced cell damage increased galectin-3 levels, while FGF21 decreased galactose lectin-3 levels. In addition, inhibition of galactose agglutinin-3 expression by siRNA enhanced the cardioprotective effect of FGF21, while overexpression of galectin-3 reduced the cardioprotective effect of fibroblast growth factor 21. CONCLUSION FGF21 may be a novel therapy for hypoxia-induced cardiac injury by regulating the expression of galectin-3.
Collapse
Affiliation(s)
- Mengyao Sun
- Department of Cardiac Surgery, first Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Liying Jin
- Department of Cardiac Surgery, first Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Yang Bai
- Department of Cardiac Surgery, first Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Lei Wang
- Department of Cardiac Surgery, first Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Song Zhao
- Department of Spine Surgery, first Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Chunye Ma
- Department of Cardiac Surgery, first Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Dashi Ma
- Department of Cardiac Surgery, first Hospital of Jilin University, Changchun, Jilin, P. R. China
| |
Collapse
|
17
|
Le BT, Raguraman P, Kosbar TR, Fletcher S, Wilton SD, Veedu RN. Antisense Oligonucleotides Targeting Angiogenic Factors as Potential Cancer Therapeutics. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 14:142-157. [PMID: 30594893 PMCID: PMC6307321 DOI: 10.1016/j.omtn.2018.11.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
Abstract
Cancer is one of the leading causes of death worldwide, and conventional cancer therapies such as surgery, chemotherapy, and radiotherapy do not address the underlying molecular pathologies, leading to inadequate treatment and tumor recurrence. Angiogenic factors, such as EGF, PDGF, bFGF, TGF-β, TGF-α, VEGF, endoglin, and angiopoietins, play important roles in regulating tumor development and metastasis, and they serve as potential targets for developing cancer therapeutics. Nucleic acid-based therapeutic strategies have received significant attention in the last two decades, and antisense oligonucleotide-mediated intervention is a prominent therapeutic approach for targeted manipulation of gene expression. Clinical benefits of antisense oligonucleotides have been recognized by the U.S. Food and Drug Administration, with full or conditional approval of Vitravene, Kynamro, Exondys51, and Spinraza. Herein we review the scope of antisense oligonucleotides that target angiogenic factors toward tackling solid cancers.
Collapse
Affiliation(s)
- Bao T Le
- Centre for Comparative Genomics, Murdoch University, Murdoch, WA 6150, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Prithi Raguraman
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Tamer R Kosbar
- Centre for Comparative Genomics, Murdoch University, Murdoch, WA 6150, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Susan Fletcher
- Centre for Comparative Genomics, Murdoch University, Murdoch, WA 6150, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Steve D Wilton
- Centre for Comparative Genomics, Murdoch University, Murdoch, WA 6150, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Rakesh N Veedu
- Centre for Comparative Genomics, Murdoch University, Murdoch, WA 6150, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia.
| |
Collapse
|
18
|
Thiagarajan H, Thiyagamoorthy U, Shanmugham I, Dharmalingam Nandagopal G, Kaliyaperumal A. Angiogenic growth factors in myocardial infarction: a critical appraisal. Heart Fail Rev 2018. [PMID: 28639006 DOI: 10.1007/s10741-017-9630-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In the recent past, substantial advances have been made in the treatment of myocardial infarction (MI). Despite the impact of these positive developments, MI remains to be a leading cause of morbidity as well as mortality. An interesting hypothesis is that the development of new blood vessels (angiogenesis) or the remodeling of preexisting collaterals may form natural bypasses that could compensate for the occlusion of an epicardial coronary artery. A number of angiogenic factors are proven to be elicited during MI. Exogenous supplementation of these growth factors either in the form of recombinant protein or gene would enhance the collateral vessel formation and thereby improve the outcome after MI. The aim of this review is to describe the nature and potentials of different angiogenic factors, their expression, their efficacy in animal studies, and clinical trials pertaining to MI.
Collapse
Affiliation(s)
- Hemalatha Thiagarajan
- Department of Biological Materials, CSIR - Central Leather Research Institute, Adyar, Chennai, 600020, India.
| | - UmaMaheswari Thiyagamoorthy
- Department of Food Science and Nutrition, Home Science College and Research Institute, Tamil Nadu Agricultural University, Madurai, 625 014, India
| | - Iswariya Shanmugham
- Department of Biological Materials, CSIR - Central Leather Research Institute, Adyar, Chennai, 600020, India
| | | | | |
Collapse
|
19
|
Coffin JD, Homer-Bouthiette C, Hurley MM. Fibroblast Growth Factor 2 and Its Receptors in Bone Biology and Disease. J Endocr Soc 2018; 2:657-671. [PMID: 29942929 PMCID: PMC6009610 DOI: 10.1210/js.2018-00105] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 05/23/2018] [Indexed: 01/24/2023] Open
Abstract
The fibroblast growth factor (FGF) regulatory axis is phylogenetically ancient, evolving into a large mammalian/human gene family of 22 ligands that bind to four receptor tyrosine kinases for a complex physiologic system controlling cell growth, differentiation, and metabolism. The tissue targets for the primary FGF function are mainly in cartilage and in bone for morphogenesis, mineralization, and metabolism. A multitude of complexities in the FGF ligand-receptor signaling pathways have made translation into therapies for FGF-related bone disorders such as osteomalacia, osteoarthritis, and osteoporosis difficult but not impossible.
Collapse
Affiliation(s)
| | | | - Marja Marie Hurley
- Department of Medicine, University of Connecticut School of Medicine, UCONN Health, Farmington, Connecticut
| |
Collapse
|
20
|
Sitia R, Rubartelli A. The unconventional secretion of IL-1β: Handling a dangerous weapon to optimize inflammatory responses. Semin Cell Dev Biol 2018; 83:12-21. [PMID: 29571971 DOI: 10.1016/j.semcdb.2018.03.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/13/2018] [Accepted: 03/19/2018] [Indexed: 01/08/2023]
Abstract
Interleukin 1β (IL-1β) is a major mediator of inflammation, with a causative role in many diseases. Unlike most other cytokines, however, it lacks a secretory signal sequence, raising intriguing mechanistic, functional and evolutionary questions. Despite decades of strenuous efforts in many laboratories, how IL-1β is secreted is still a matter of intense debate. Here, we summarize the different mechanisms and pathways that have been proposed for IL-1β secretion. At least two of them, namely the endolysosomal vesicle-based and gasdermin D-dependent pathways (types III and I in the recent Rabouille's classification of unconventional protein secretion), can be triggered in monocytes, the main source of IL-1β in humans, according to the type and strength of the pro-inflammatory stimuli. As during the escalation of human conflicts, monocytes deploy secretory mechanisms of increasing efficiency and dangerousness, shifting from the specific and controlled type III pathway to the much faster release of type I. Thus, the different mechanisms are activated depending on the severity of the conditions, from the self-limiting type III pathways in response of low pathogen load or small trauma, to the uncontrolled responses that underlie autoinflammatory disorders and sepsis.
Collapse
Affiliation(s)
- Roberto Sitia
- Protein Transport and Secretion Unit, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Anna Rubartelli
- Cell Biology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy.
| |
Collapse
|
21
|
Abstract
A key challenge in chemical biology is to identify small molecule regulators for every single protein. However, protein surfaces are notoriously difficult to recognise with synthetic molecules, often having large flat surfaces that are poorly matched to traditional small molecules. In the surface mimetic approach, a supramolecular scaffold is used to project recognition groups in such a manner as to make multivalent non-covalent contacts over a large area of protein surface. Metal based supramolecular scaffolds offer unique advantages over conventional organic molecules for protein binding, including greater stereochemical and geometrical diversity conferred through the metal centre and the potential for direct assessment of binding properties and even visualisation in cells without recourse to further functionalisation. This feature article will highlight the current state of the art in protein surface recognition using metal complexes as surface mimetics.
Collapse
Affiliation(s)
- Sarah H Hewitt
- School of Chemistry, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK. and Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK
| | - Andrew J Wilson
- School of Chemistry, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK. and Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK
| |
Collapse
|
22
|
Bai Y, Bai L, Zhou J, Chen H, Zhang L. Sequential delivery of VEGF, FGF-2 and PDGF from the polymeric system enhance HUVECs angiogenesis in vitro and CAM angiogenesis. Cell Immunol 2017; 323:19-32. [PMID: 29111157 DOI: 10.1016/j.cellimm.2017.10.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 10/17/2017] [Accepted: 10/21/2017] [Indexed: 12/17/2022]
Abstract
Angiogenesis is an organized series of events, beginning with vessel destabilization, followed by endothelial cell re-organization, and ending with vessel maturation. The formation of a mature vascular network requires precise spatial and temporal regulation of a large number of angiogenic factors, including vascular endothelial growth factor (VEGF), basic fibroblast growth factor-2 (FGF-2) and platelet-derived growth factor (PDGF). VEGF aids in vascular permeability and endothelial cell recruitment, FGF-2 activates endothelial cell proliferation and migration while PDGF stimulates vascular stability. Accordingly, VEGF may inhibit vessel stabilization while PDGF may inhibit endothelial cell recruitment. Therefore, a new polymeric system was prepared by the supercritical carbon dioxide foaming technology, which realized sequential delivery of two or more growth factors with the controlled dose and rate. Increased release of VEGF (71.10%) and FGF-2 (69.76%) compared to PDGF (43.17%) was observed for the first 7 days. Thereafter, up till 21 days, an increased rate of release of BMP-2 compared to VEGF 165 was observed. The effects of PDGF-PLAms/VEGF-FGF-2-PLGA scaffolds on angiogenesis were investigated by human umbilical vein endothelial cells (HUVECs) angiogenic differentiation in vitro and chorioallantoic membrane (CAM) angiogenesis in vivo. Sequential delivery of VEGF, FGF-2 and PDGF from structural polymer scaffolds with distinct kinetics resulted in significant angiogenic differentiation of HUVECs and rapid formation of mature vascular networks in chorioallantoic membrane. This study reported a composite scaffold with distinct release kinetics, and these results clearly indicated the importance of sequential delivery of multiple growth factors in tissue regeneration and engineering.
Collapse
Affiliation(s)
- Yan Bai
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China; Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, PR China.
| | - Lijuan Bai
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Jing Zhou
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Huali Chen
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Liangke Zhang
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
23
|
Preparation and in vitro evaluation of FGF-2 incorporated carboxymethyl chitosan nanoparticles. Carbohydr Polym 2017; 173:114-120. [DOI: 10.1016/j.carbpol.2017.05.080] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 05/01/2017] [Accepted: 05/24/2017] [Indexed: 01/09/2023]
|
24
|
Todorović-Raković N, Radulovic M, Vujasinović T, Rabi ZA, Milovanović J, Nikolić-Vukosavljević D. bFGF in tumor tissue independently prognosticates disease outcome of a natural course of invasive breast cancer. Cancer Biomark 2017; 20:151-158. [DOI: 10.3233/cbm-170022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
25
|
Clump formation in mouse pituitary-derived non-endocrine cell line Tpit/F1 promotes differentiation into growth-hormone-producing cells. Cell Tissue Res 2017; 369:353-368. [DOI: 10.1007/s00441-017-2603-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 02/24/2017] [Indexed: 01/08/2023]
|
26
|
Adipose stem cell-derived nanovesicles inhibit emphysema primarily via an FGF2-dependent pathway. Exp Mol Med 2017; 49:e284. [PMID: 28082743 PMCID: PMC5291836 DOI: 10.1038/emm.2016.127] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 08/10/2016] [Accepted: 08/18/2016] [Indexed: 12/23/2022] Open
Abstract
Cell therapy using stem cells has produced therapeutic benefits in animal models of COPD. Secretory mediators are proposed as one mechanism for stem cell effects because very few stem cells engraft after injection into recipient animals. Recently, nanovesicles that overcome the disadvantages of natural exosomes have been generated artificially from cells. We generated artificial nanovesicles from adipose-derived stem cells (ASCs) using sequential penetration through polycarbonate membranes. ASC-derived artificial nanovesicles displayed a 100 nm-sized spherical shape similar to ASC-derived natural exosomes and expressed both exosomal and stem cell markers. The proliferation rate of lung epithelial cells was increased in cells treated with ASC-derived artificial nanovesicles compared with cells treated with ASC-derived natural exosomes. The lower dose of ASC-derived artificial nanovesicles had similar regenerative capacity compared with a higher dose of ASCs and ASC-derived natural exosomes. In addition, FGF2 levels in the lungs of mice treated with ASC-derived artificial nanovesicles were increased. The uptake of ASC-derived artificial nanovesicles was inhibited by heparin, which is a competitive inhibitor of heparan sulfate proteoglycan that is associated with FGF2 signaling. Taken together, the data indicate that lower doses of ASC-derived artificial nanovesicles may have beneficial effects similar to higher doses of ASCs or ASC-derived natural exosomes in an animal model with emphysema, suggesting that artificial nanovesicles may have economic advantages that warrant future clinical studies.
Collapse
|
27
|
Ho YT, Poinard B, Kah JCY. Nanoparticle drug delivery systems and their use in cardiac tissue therapy. Nanomedicine (Lond) 2016; 11:693-714. [DOI: 10.2217/nnm.16.6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cardiovascular diseases make up one of the main causes of death today, with myocardial infarction and ischemic heart disease contributing a large share of the deaths reported. With mainstream clinical therapy focusing on palliative medicine following myocardial infarction, the structural changes that occur in the diseased heart will eventually lead to end-stage heart failure. Heart transplantation remains the only gold standard of cure but a shortage in donor organs pose a major problem that led to clinicians and researchers looking into alternative strategies for cardiac repair. This review will examine some alternative methods of treatment using chemokines and drugs carried by nanoparticles as drug delivering agents for the purposes of treating myocardial infarction through the promotion of revascularization. We will also provide an overview of existing studies involving such nanoparticulate drug delivery systems, their reported efficacy and the challenges facing their translation into ubiquitous clinical use.
Collapse
Affiliation(s)
- Yan Teck Ho
- Department of Biomedical Engineering, National University of Singapore, 9 Engineering Drive 1, Block EA #07–25, Singapore 117575
- NUS Graduate School of Integrative Sciences & Engineering, National University of Singapore, 28 Medical Drive, Singapore 117456
| | - Barbara Poinard
- Department of Biomedical Engineering, National University of Singapore, 9 Engineering Drive 1, Block EA #07–25, Singapore 117575
- NUS Graduate School of Integrative Sciences & Engineering, National University of Singapore, 28 Medical Drive, Singapore 117456
| | - James Chen Yong Kah
- Department of Biomedical Engineering, National University of Singapore, 9 Engineering Drive 1, Block EA #07–25, Singapore 117575
- NUS Graduate School of Integrative Sciences & Engineering, National University of Singapore, 28 Medical Drive, Singapore 117456
| |
Collapse
|
28
|
Choi D, Son B, Park TH, Hong J. Controlled surface functionality of magnetic nanoparticles by layer-by-layer assembled nano-films. NANOSCALE 2015; 7:6703-6711. [PMID: 25798789 DOI: 10.1039/c4nr07373h] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Over the past several years, the preparation of functionalized nanoparticles has been aggressively pursued in order to develop desired structures, compositions, and structural order. Among the various nanoparticles, iron oxide magnetic nanoparticles (MNPs) have shown great promise because the material generated using these MNPs can be used in a variety of biomedical applications and possible bioactive functionalities. In this study, we report the development of various functionalized MNPs (F-MNPs) generated using the layer-by-layer (LbL) self-assembly method. To provide broad functional opportunities, we fabricated F-MNP bio-toolbox by using three different materials: synthetic polymers, natural polymers, and carbon materials. Each of these F-MNPs displays distinct properties, such as enhanced thickness or unique morphologies. In an effort to explore their biomedical applications, we generated basic fibroblast growth factor (bFGF)-loaded F-MNPs. The bFGF-loaded F-MNPs exhibited different release mechanisms and loading amounts, depending on the film material and composition order. Moreover, bFGF-loaded F-MNPs displayed higher biocompatibility and possessed superior proliferation properties than the bare MNPs and pure bFGF, respectively. We conclude that by simply optimizing the building materials and the nanoparticle's film composition, MNPs exhibiting various bioactive properties can be generated.
Collapse
Affiliation(s)
- Daheui Choi
- School of Chemical Engineering & Material Science, Chung-Ang University, 47 Heukseok-ro, Dongjak-gu, Seoul 156-756, Republic of Korea.
| | | | | | | |
Collapse
|
29
|
Lankford L, Selby T, Becker J, Ryzhuk V, Long C, Farmer D, Wang A. Early gestation chorionic villi-derived stromal cells for fetal tissue engineering. World J Stem Cells 2015; 7:195-207. [PMID: 25621120 PMCID: PMC4300931 DOI: 10.4252/wjsc.v7.i1.195] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 11/04/2014] [Accepted: 11/10/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the potential for early gestation placenta-derived mesenchymal stromal cells (PMSCs) for fetal tissue engineering.
METHODS: PMSCs were isolated from early gestation chorionic villus tissue by explant culture. Chorionic villus sampling (CVS)-size tissue samples (mean = 35.93 mg) were used to test the feasibility of obtaining large cell numbers from CVS within a clinically relevant timeframe. We characterized PMSCs isolated from 6 donor placentas by flow cytometry immunophenotyping, multipotency assays, and through immunofluorescent staining. Protein secretion from PMSCs was examined using two cytokine array assays capable of probing for over 70 factors in total. Delivery vehicle compatibility of PMSCs was determined using three common scaffold systems: fibrin glue, collagen hydrogel, and biodegradable nanofibrous scaffolds made from a combination of polylactic acid (PLA) and poly(lactic-co-glycolic acid) (PLGA). Viral transduction of PMSCs was performed using a Luciferase-GFP-containing lentiviral vector and efficiency of transduction was tested by fluorescent microscopy and flow cytometry analysis.
RESULTS: We determined that an average of 2.09 × 106 (SD ± 8.59 × 105) PMSCs could be obtained from CVS-size tissue samples within 30 d (mean = 27 d, SD ± 2.28), indicating that therapeutic numbers of cells can be rapidly expanded from very limited masses of tissue. Immunophenotyping by flow cytometry demonstrated that PMSCs were positive for MSC markers CD105, CD90, CD73, CD44, and CD29, and were negative for hematopoietic and endothelial markers CD45, CD34, and CD31. PMSCs displayed trilineage differentiation capability, and were found to express developmental transcription factors Sox10 and Sox17 as well as neural-related structural proteins NFM, Nestin, and S100β. Cytokine arrays revealed a robust and extensive profile of PMSC-secreted cytokines and growth factors, and detected 34 factors with spot density values exceeding 103. Detected factors had widely diverse functions that include modulation of angiogenesis and immune response, cell chemotaxis, cell proliferation, blood vessel maturation and homeostasis, modulation of insulin-like growth factor activity, neuroprotection, extracellular matrix degradation and even blood coagulation. Importantly, PMSCs were also determined to be compatible with both biological and synthetic material-based delivery vehicles such as collagen and fibrin hydrogels, and biodegradable nanofiber scaffolds made from a combination of PLA and PLGA. Finally, we demonstrated that PMSCs can be efficiently transduced (> 95%) with a Luciferase-GFP-containing lentiviral vector for future in vivo cell tracking after transplantation.
CONCLUSION: Our findings indicate that PMSCs represent a unique source of cells that can be effectively utilized for in utero cell therapy and tissue engineering.
Collapse
|
30
|
Liu Y, Lu J, Li H, Wei J, Li X. Engineering blood vessels through micropatterned co-culture of vascular endothelial and smooth muscle cells on bilayered electrospun fibrous mats with pDNA inoculation. Acta Biomater 2015; 11:114-25. [PMID: 25305234 DOI: 10.1016/j.actbio.2014.10.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 09/25/2014] [Accepted: 10/02/2014] [Indexed: 01/08/2023]
Abstract
Although engineered blood vessels have seen important advances during recent years, proper mechanical strength and vasoactivity remain unsolved problems. In the current study, micropatterned fibrous mats were created to load smooth muscle cells (SMC), and a co-culture with endothelial cells (EC) was established through overlaying on an EC-loaded flat fibrous mat to mimic the layered structure of a blood vessel. A preferential distribution of SMC was determined in the patterned regions throughout the fibrous scaffolds, and aligned fibers in the patterned regions provided topological cues to guide the orientation of SMC with intense actin filaments and extracellular matrix (ECM) production in a circumferential direction. Plasmid DNA encoding basic fibroblast growth factors and vascular endothelial growth factor were integrated into electrospun fibers as biological cues to promote SMC infiltration into fibrous mats, and the viability and ECM production of both EC and SMC. The layered fibrous mats with loaded EC and SMC were wrapped into a cylinder, and engineered vessels were obtained with compact EC and SMC layers after co-culture for 3 months. Randomly oriented ECM productions of EC formed a continuous endothelium covering the entire lumenal surface, and a high alignment of ECM was shown in the circumferential direction of SMC layers. The tensile strength, strain at failure and suture retention strength were higher than those of the human femoral artery, and the burst pressure and radial compliance were in the same range as the human saphenous vein, indicating potential as blood vessel substitutes for transplantation in vivo. Thus, the establishment of topographical cues and biochemical signals in fibrous scaffolds demonstrates advantages in modulating cellular behavior and organization found in complex multicellular tissues.
Collapse
|
31
|
Abstract
Angiogenesis is a vital component of bone healing. The formation of the new blood vessels at the fracture site restores the hypoxia and nutrient deprivation found at the early stages after fracture whilst at a later stage facilitates osteogenesis by the activity of the osteoprogenitor cells. Emerging evidence suggests that there are certain molecules and gene therapies that could promote new blood vessel formation and as a consequence enhance the local bone healing response. This article summarizes the current in vivo evidence on therapeutic approaches aiming at the augmentation of the angiogenic signalling during bone repair.
Collapse
|
32
|
Therapeutic foam scaffolds incorporating biopolymer-shelled mesoporous nanospheres with growth factors. Acta Biomater 2014; 10:2612-21. [PMID: 24530558 DOI: 10.1016/j.actbio.2014.02.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 01/29/2014] [Accepted: 02/03/2014] [Indexed: 12/12/2022]
Abstract
A novel therapeutic scaffolding system of engineered nanocarriers within a foam matrix for the long-term and sequential delivery of growth factors is reported. Mesoporous silica nanospheres were first functionalized to have an enlarged mesopore size (12.2nm) and aminated surface, which was then shelled by a biopolymer, poly(lactic acid) (PLA) or poly(ethylene glycol) (PEG), via electrospraying. The hybrid nanocarrier was subsequently combined with collagen to produce foam scaffolds. Bovine serum albumin (BSA), used as a model protein, was effectively loaded within the enlarged nanospheres. The biopolymer shell substantially prolonged the release period of BSA (2-3weeks from shelled nanospheres vs. within 1week from bare nanospheres), and the release rate was highly dependent on the shell composition (PEG>PLA). Collagen foam scaffolding of the shelled nanocarrier further slowed down the protein release, while enabling the incorporation of a rapidly releasing protein, which is effective for sequential protein delivery. Acidic fibroblast growth factor (aFGF), loaded onto the shelled-nanocarrier scaffolds, was released over a month at a highly sustainable rate, profiling a release pattern similar to that of BSA. The biological activity of the aFGF was evidenced by the significant proliferation of osteoblastic precursor cells in the aFGF-releasing scaffolds. Furthermore, the aFGF-delivering scaffolds implanted in rat subcutaneous tissue for 2weeks showed a substantially enhanced invasion of fibroblasts with a homogeneous population. Taken together, it is concluded that the biopolymer encapsulation of mesoporous nanospheres effectively prolongs the release of growth factors over weeks to a month, providing a nanocarrier platform for a long-term growth factor delivery. Moreover, the foam scaffolding of the nanocarrier system is a potential therapeutic three-dimensional matrix for cell culture and tissue engineering.
Collapse
|
33
|
Newton HB. Molecular neuro-oncology and development of targeted therapeutic strategies for brain tumors. Part 2: PI3K/Akt/PTEN, mTOR, SHH/PTCH and angiogenesis. Expert Rev Anticancer Ther 2014; 4:105-28. [PMID: 14748662 DOI: 10.1586/14737140.4.1.105] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Brain tumors are a diverse group of malignancies that remain refractory to conventional treatment approaches. Molecular neuro-oncology has now begun to clarify the transformed phenotype of brain tumors and identify oncogenic pathways that might be amenable to targeted therapy. Activity of the phosphoinositide 3; kinase (PI3K)/Akt pathway is often upregulated in brain tumors due to excessive stimulation by growth factor receptors and Ras. Loss of function of the tumor suppressor gene PTEN also frequently contributes to upregulation of PI3K/Akt. Several compounds, such as wortmannin and LY-294002, can target PI3K and inhibit activity of this pathway. The mammalian target of rapamycin (mTOR) is an important regulator of cell growth and metabolism and is often upregulated by Akt. Clinical trials of CCI-779, an inhibitor of mTOR, are ongoing in recurrent malignant glioma patients. The sonic hedgehog/PTCH pathway is involved in the tumorigenesis of some familial and sporadic medulloblastomas. This pathway can be targeted by cyclopamine, which is under evaluation in preclinical studies. Angiogenesis is a critical process for development and progression of brain tumors. Targeted approaches to inhibit angiogenesis include monoclonal antibodies, receptor tyrosine kinase inhibitors, antisense oligonucleotides and gene therapy. Clinical trials are ongoing for numerous angiogenesis inhibitors, including thalidomide, CC-5103 and PTK 787/ZK 222584. Further development of targeted therapies and evaluation of these new agents in clinical trials will be needed to improve survival and quality of life of patients with brain tumors.
Collapse
Affiliation(s)
- Herbert B Newton
- Dardinger Neuro-Oncology Center, Department of Neurology, Ohio State University Hospitals, 465 Means Hall, 1654 Upham Drive, Columbus, OH 43210, USA.
| |
Collapse
|
34
|
Ma X, Pietsch J, Wehland M, Schulz H, Saar K, Hübner N, Bauer J, Braun M, Schwarzwälder A, Segerer J, Birlem M, Horn A, Hemmersbach R, Waβer K, Grosse J, Infanger M, Grimm D. Differential gene expression profile and altered cytokine secretion of thyroid cancer cells in space. FASEB J 2013; 28:813-35. [DOI: 10.1096/fj.13-243287] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Xiao Ma
- Institute of BiomedicineDepartment of PharmacologyAarhus UniversityAarhusDenmark
| | - Jessica Pietsch
- Clinic for Plastic, Aesthetic, and Hand Surgery, Otto von Guericke University MagdeburgMagdeburgGermany
| | - Markus Wehland
- Clinic for Plastic, Aesthetic, and Hand Surgery, Otto von Guericke University MagdeburgMagdeburgGermany
| | - Herbert Schulz
- Max Delbrück Center for Molecular MedicineBerlin‐BuchGermany
| | - Katrin Saar
- Max Delbrück Center for Molecular MedicineBerlin‐BuchGermany
| | - Norbert Hübner
- Max Delbrück Center for Molecular MedicineBerlin‐BuchGermany
| | - Johann Bauer
- Max Planck Institute for BiochemistryMartinsriedGermany
| | - Markus Braun
- Institute for Molecular Physiology and Biotechnology of Plants (IMBIO)Gravitational Biology GroupUniversity of BonnBonnGermany
| | - Achim Schwarzwälder
- Life Science, Orbital Systems, and Space Exploration, Astrium/European Aeronautic Defense and Space (EADS)ImmenstaadGermany
| | - Jürgen Segerer
- Life Science, Orbital Systems, and Space Exploration, Astrium/European Aeronautic Defense and Space (EADS)ImmenstaadGermany
| | - Maria Birlem
- Life Science, Orbital Systems, and Space Exploration, Astrium/European Aeronautic Defense and Space (EADS)ImmenstaadGermany
| | - Astrid Horn
- Life Science, Orbital Systems, and Space Exploration, Astrium/European Aeronautic Defense and Space (EADS)ImmenstaadGermany
| | - Ruth Hemmersbach
- Institute of Aerospace MedicineDeutsches Zentrum für Luft‐ und Raumfahrt (DLR)CologneGermany
| | - Kai Waβer
- Institute of Aerospace MedicineDeutsches Zentrum für Luft‐ und Raumfahrt (DLR)CologneGermany
| | - Jirka Grosse
- Department of Nuclear MedicineUniversity of RegensburgRegensburgGermany
| | - Manfred Infanger
- Clinic for Plastic, Aesthetic, and Hand Surgery, Otto von Guericke University MagdeburgMagdeburgGermany
| | - Daniela Grimm
- Institute of BiomedicineDepartment of PharmacologyAarhus UniversityAarhusDenmark
- Clinic for Plastic, Aesthetic, and Hand Surgery, Otto von Guericke University MagdeburgMagdeburgGermany
| |
Collapse
|
35
|
Electrosprayed Microparticles with Loaded pDNA-Calcium Phosphate Nanoparticles to Promote the Regeneration of Mature Blood Vessels. Pharm Res 2013; 31:874-86. [DOI: 10.1007/s11095-013-1209-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 09/12/2013] [Indexed: 01/08/2023]
|
36
|
Chen F, Wan H, Xia T, Guo X, Wang H, Liu Y, Li X. Promoted regeneration of mature blood vessels by electrospun fibers with loaded multiple pDNA-calcium phosphate nanoparticles. Eur J Pharm Biopharm 2013; 85:699-710. [PMID: 23891771 DOI: 10.1016/j.ejpb.2013.07.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 07/10/2013] [Accepted: 07/15/2013] [Indexed: 01/08/2023]
Abstract
Vascularization is one of the capital challenges in the establishment of tissue engineering constructs and recovery of ischemic and wounded tissues. The aim of this study was to assess electrospun fibers with loadings of multiple pDNA to allow a localized delivery for an efficient regeneration of mature blood vessels. To induce sufficient protein expression, a reverse microemulsion process was adopted to load pDNA into calcium phosphate nanoparticles (CP-pDNA), which were electrospun into fibers to achieve a sustained release for 4 weeks. Compared with pDNA-infiltrated fibers, the localized and gradual release of pDNA facilitated cell proliferation, gene transfection, and extracellular matrix secretion and enhanced the generation of blood vessels after subcutaneous implantation. Compared with commonly used pDNA polyplexes with poly(ethyleneimine), CP-pDNA nanoparticles induced significantly lower cytotoxicity and less inflammation reaction after implantation into animals. Fibers with encapsulated nanoparticles containing plasmids encoding vascular endothelial growth factor (pVEGF) and basic fibroblast growth factors (pbFGF) led to significantly higher density of mature blood vessels than those containing individual plasmid. It is suggested that the integration of CP-pDNA nanoparticles with loadings of multiple plasmids into fibrous scaffolds should provide clinical relevance for therapeutic vascularization, getting fully vascularized in engineered tissues and regeneration of blood vessel substitutes.
Collapse
Affiliation(s)
- Fang Chen
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education of China, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, PR China
| | | | | | | | | | | | | |
Collapse
|
37
|
Chim SM, Tickner J, Chow ST, Kuek V, Guo B, Zhang G, Rosen V, Erber W, Xu J. Angiogenic factors in bone local environment. Cytokine Growth Factor Rev 2013; 24:297-310. [DOI: 10.1016/j.cytogfr.2013.03.008] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Accepted: 03/26/2013] [Indexed: 01/11/2023]
|
38
|
Bai Y, Yin G, Huang Z, Liao X, Chen X, Yao Y, Pu X. Localized delivery of growth factors for angiogenesis and bone formation in tissue engineering. Int Immunopharmacol 2013; 16:214-23. [PMID: 23587487 DOI: 10.1016/j.intimp.2013.04.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 04/01/2013] [Accepted: 04/01/2013] [Indexed: 01/14/2023]
Abstract
Angiogenesis is a key component of bone formation. Delivery of growth factors for both angiogenesis and osteogenesis is about to gain important potential as a future therapeutic tool. This review focuses on these growth factors that have dual functions in angiogenesis and osteogenesis, and their localized application. A major hurdle in the clinical development of growth factor therapy so far is how to assure safe and efficacious therapeutic use of such factors and avoid unwanted side effects and toxicity. It is now firmly established from the available information that the type, dose, combinations and delivery kinetics of growth factors all play a decisive role for the success of growth factor therapy. All of these parameters have to be adapted and optimized for each animal model or clinical case. In this review we discuss some important parameters associated with growth factor therapy and present an overview of selected preclinical studies, followed by a conceptual description of both established and proposed delivery strategies meeting therapeutic needs.
Collapse
Affiliation(s)
- Yan Bai
- College of Materials Science and Engineering, Sichuan University, Chengdu 610064, PR China
| | | | | | | | | | | | | |
Collapse
|
39
|
SAITO A, SAITO E, KUBOKI Y, KIMURA M, NAKAJIMA T, YUGE F, KATO T, HONMA Y, TAKAHASHI T, OHATA N. Periodontal regeneration following application of basic fibroblast growth factor-2 in combination with beta tricalcium phosphate in class III furcation defects in dogs. Dent Mater J 2013; 32:256-62. [DOI: 10.4012/dmj.2012-171] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
40
|
Muyal JP, Muyal V, Kotnala S, Kumar D, Bhardwaj H. Therapeutic potential of growth factors in pulmonary emphysematous condition. Lung 2012; 191:147-63. [PMID: 23161370 DOI: 10.1007/s00408-012-9438-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 11/04/2012] [Indexed: 02/02/2023]
Abstract
Pulmonary emphysema is a major manifestation of chronic obstructive pulmonary disease (COPD), which is characterized by progressive destruction of alveolar parenchyma with persistent inflammation of the small airways. Such destruction in the distal respiratory tract is irreversible and irreparable. All-trans-retinoic acid was suggested as a novel therapy for regeneration of lost alveoli in emphysema. However, profound discrepancies were evident between studies. At present, no effective therapeutic options are available that allow for the regeneration of lost alveoli in emphysematous human lungs. Recently, some reports on rodent's models have suggested the beneficial effects of various growth factors toward alveolar maintenance and repair processes.
Collapse
Affiliation(s)
- Jai Prakash Muyal
- Department of Biotechnology, School of Biotechnology, Gautam Buddha University, Greater Noida, 201308, India.
| | | | | | | | | |
Collapse
|
41
|
Wu Y, Lu CT, Li WF, Sun CZ, Yang W, Zhang Y, Su ZX, Zhang Y, Fu HX, Huang PT, Lv HF, Dai DD, Li X, Lin GY, Luo SM, Zhao YZ. Preparation and antitumor activity of bFGF-mediated active targeting doxorubicin microbubbles. Drug Dev Ind Pharm 2012; 39:1712-9. [DOI: 10.3109/03639045.2012.730527] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
42
|
Maina JN. Comparative molecular developmental aspects of the mammalian- and the avian lungs, and the insectan tracheal system by branching morphogenesis: recent advances and future directions. Front Zool 2012; 9:16. [PMID: 22871018 PMCID: PMC3502106 DOI: 10.1186/1742-9994-9-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 06/18/2012] [Indexed: 02/07/2023] Open
Abstract
Gas exchangers fundamentally form by branching morphogenesis (BM), a mechanistically profoundly complex process which derives from coherent expression and regulation of multiple genes that direct cell-to-cell interactions, differentiation, and movements by signaling of various molecular morphogenetic cues at specific times and particular places in the developing organ. Coordinated expression of growth-instructing factors determines sizes and sites where bifurcation occurs, by how much a part elongates before it divides, and the angle at which branching occurs. BM is essentially induced by dualities of factors where through feedback- or feed forward loops agonists/antagonists are activated or repressed. The intricate transactions between the development orchestrating molecular factors determine the ultimate phenotype. From the primeval time when the transformation of unicellular organisms to multicellular ones occurred by systematic accretion of cells, BM has been perpetually conserved. Canonical signalling, transcriptional pathways, and other instructive molecular factors are commonly employed within and across species, tissues, and stages of development. While much still remain to be elucidated and some of what has been reported corroborated and reconciled with rest of existing data, notable progress has in recent times been made in understanding the mechanism of BM. By identifying and characterizing the morphogenetic drivers, and markers and their regulatory dynamics, the elemental underpinnings of BM have been more precisely explained. Broadening these insights will allow more effective diagnostic and therapeutic interventions of developmental abnormalities and pathologies in pre- and postnatal lungs. Conservation of the molecular factors which are involved in the development of the lung (and other branched organs) is a classic example of nature's astuteness in economically utilizing finite resources. Once purposefully formed, well-tested and tried ways and means are adopted, preserved, and widely used to engineer the most optimal phenotypes. The material and time costs of developing utterly new instruments and routines with every drastic biological change (e.g. adaptation and speciation) are circumvented. This should assure the best possible structures and therefore functions, ensuring survival and evolutionary success.
Collapse
Affiliation(s)
- John N Maina
- Department of Zoology, University of Johannesburg, Auckland Park 2006, P,O, Box 524, Johannesburg, South Africa.
| |
Collapse
|
43
|
He S, Xia T, Wang H, Wei L, Luo X, Li X. Multiple release of polyplexes of plasmids VEGF and bFGF from electrospun fibrous scaffolds towards regeneration of mature blood vessels. Acta Biomater 2012; 8:2659-69. [PMID: 22484697 DOI: 10.1016/j.actbio.2012.03.044] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 03/12/2012] [Accepted: 03/28/2012] [Indexed: 11/18/2022]
Abstract
Key challenges associated with the outcomes of vascular grafting (for example, to fully vascularize engineered tissues and promptly regenerate blood vessel substitutes) remain unsolved. The local availability of angiogenic growth factors is highly desirable for tissue regeneration, and plasmid loading in scaffolds represents a powerful alternative for local production of tissue-inductive factors. No attempt has been made so far to clarify the efficacy of electrospun fibers with the loading of multiple plasmids to promote tissue regeneration. In the present study, core-sheath electrospun fibers with the encapsulation of polyplexes of basic fibroblast growth factor-encoding plasmid (pbFGF) and vascular endothelial growth factor-encoding plasmid (pVEGF) were evaluated to promote the generation of mature blood vessels. In vitro release indicated a sustained release of pDNA for ∼4 weeks with as low as ∼10% initial burst release, and the release patterns from the single and twofold plasmid-loaded systems coincided. In vitro investigations on human umbilical vein endothelial cells showed that the sustained release of pDNA from fibrous mats promoted cell attachment and viability, cell transfection and protein expression, and extracellular secretion of collagen IV and laminin. The acceleration of angiogenesis was assessed in vivo after subcutaneous implantation of fibrous scaffolds, and the explants were evaluated after 1, 2 and 4 weeks' treatment by histological and immunohistochemical staining. Compared with pDNA polyplex infiltrated fibrous mats, the pDNA polyplex encapsulated fibers alleviated the inflammation reaction and enhanced the generation of microvessels. Although there was no significant difference in the total number of microvessels, the density of mature vessels was significantly enhanced by the combined treatment with both pbFGF and pVEGF compared with those incorporating individual pDNA. The integration of the core-sheath structure, DNA condensation and multiple delivery strategies provided a potential platform for scaffold fabrication to regenerate functional tissues.
Collapse
Affiliation(s)
- Shuhui He
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education of China, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | | | | | | | | | | |
Collapse
|
44
|
Khalili S, Liu Y, Kornete M, Roescher N, Kodama S, Peterson A, Piccirillo CA, Tran SD. Mesenchymal stromal cells improve salivary function and reduce lymphocytic infiltrates in mice with Sjögren's-like disease. PLoS One 2012; 7:e38615. [PMID: 22685592 PMCID: PMC3369846 DOI: 10.1371/journal.pone.0038615] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 05/07/2012] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Non-obese diabetic (NOD) mice develop Sjögren's-like disease (SS-like) with loss of saliva flow and increased lymphocytic infiltrates in salivary glands (SGs). There are recent reports using multipotent mesenchymal stromal cells (MSCs) as a therapeutic strategy for autoimmune diseases due to their anti-inflammatory and immunomodulatory capabilities. This paper proposed a combined immuno- and cell-based therapy consisting of: A) an injection of complete Freund's adjuvant (CFA) to eradicate autoreactive T lymphocytes, and B) transplantations of MSCs to reselect lymphocytes. The objective of this was to test the effectiveness of CD45(-)/TER119(-) cells (MSCs) in re-establishing salivary function and in reducing the number of lymphocytic infiltrates (foci) in SGs. The second objective was to study if the mechanisms underlying a decrease in inflammation (focus score) was due to CFA, MSCs, or CFA+MSCs combined. METHODOLOGY/PRINCIPAL FINDINGS Donor MSCs were isolated from bones of male transgenic eGFP mice. Eight week-old female NOD mice received one of the following treatments: insulin, CFA, MSC, or CFA+MSC (combined therapy). Mice were followed for 14 weeks post-therapy. CD45(-)/TER119(-) cells demonstrated characteristics of MSCs as they were positive for Sca-1, CD106, CD105, CD73, CD29, CD44, negative for CD45, TER119, CD11b, had high number of CFU-F, and differentiated into osteocytes, chondrocytes and adipocytes. Both MSC and MSC+CFA groups prevented loss of saliva flow and reduced lymphocytic infiltrations in SGs. Moreover, the influx of T and B cells decreased in all foci in MSC and MSC+CFA groups, while the frequency of Foxp3(+) (T(reg)) cell was increased. MSC-therapy alone reduced inflammation (TNF-α, TGF-β), but the combination of MSC+CFA reduced inflammation and increased the regenerative potential of SGs (FGF-2, EGF). CONCLUSIONS/SIGNIFICANCE The combined use of MSC+CFA was effective in both preventing saliva secretion loss and reducing lymphocytic influx in salivary glands.
Collapse
Affiliation(s)
- Saeed Khalili
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| | - Younan Liu
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| | - Mara Kornete
- Department of Microbiology and Immunology, and FOCIS Centre of Excellence, Centre, Montreal, Quebec, Canada
| | - Nienke Roescher
- Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Shohta Kodama
- Department of Regenerative Medicine and Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Alan Peterson
- Molecular Oncology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Ciriaco A. Piccirillo
- Department of Microbiology and Immunology, and FOCIS Centre of Excellence, Centre, Montreal, Quebec, Canada
| | - Simon D. Tran
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
45
|
Weltrowski A, da Silva Almeida ML, Peschel D, Zhang K, Fischer S, Groth T. Mitogenic Activity of Sulfated Chitosan and Cellulose Derivatives is Related to Protection of FGF-2 from Proteolytic Cleavage. Macromol Biosci 2012; 12:740-50. [DOI: 10.1002/mabi.201100518] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 01/31/2012] [Indexed: 12/18/2022]
|
46
|
Anitua E, Prado R, Sánchez M, Orive G. Platelet-Rich Plasma: Preparation and Formulation. ACTA ACUST UNITED AC 2012. [DOI: 10.1053/j.oto.2012.01.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
47
|
Florkiewicz RZ, Ahluwalia A, Sandor Z, Szabo S, Tarnawski AS. Gastric mucosal injury activates bFGF gene expression and triggers preferential translation of high molecular weight bFGF isoforms through CUG-initiated, non-canonical codons. Biochem Biophys Res Commun 2011; 409:494-9. [PMID: 21600881 DOI: 10.1016/j.bbrc.2011.05.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 05/05/2011] [Indexed: 02/06/2023]
Abstract
Basic fibroblast growth factor (bFGF or FGF-2) is a pleiotropic growth factor that promotes growth of mesenchymal and epithelial cells, stimulates angiogenesis and neuroprotection. Moreover, exogenous bFGF by stimulating angiogenesis promotes healing of gastroduodenal ulcers and cardiac and brain injury. All these actions were demonstrated in regard to 18kDa bFGF isoform that is secreted by cells via an ER/Golgi-independent pathway and activates FGF receptors. However in some transformed and stressed cells and in some tissues (e.g. brain) the single copy bFGF gene encodes multiple gene products: 18 kDa and also higher molecular weight (HMW) bFGF isoforms: ∼21 and ∼22 kDa in rodents, and ∼22, ∼23 and ∼24 kDa in humans. The biologic roles of these HMW bFGF isoforms in vivo remain unknown. In this study we demonstrated that in normal, uninjured gastric mucosa, bFGF is almost exclusively expressed as 18kDa isoform translated through a classical AUG (methionine) codon. In contrast, in injured gastric mucosa of rat, bFGF gene is preferentially translated to HMW bFGF isoforms through alternative CUG (leucine) initiation codon. Gastric mucosal injury caused in rats a significant increase in bFGF mRNA at 8 and 24h vs. normal mucosa and a significant increase in bFGF protein at 24-72h, mainly due to increased expression of ∼21 and ∼22 kDa HMW bFGF isoforms. This is first demonstration that gastric mucosal injury and repair triggers local activation of bFGF gene with preferential translation of HMW bFGF isoforms through a non-canonical CUG codon. This study uncovered CUG-initiated HMW bFGF translation as a novel regulatory mechanism operating in vivo during gastric injury repair.
Collapse
Affiliation(s)
- Robert Z Florkiewicz
- Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
48
|
Nugent MA, Chen OS, Edelman ER. Controlled Release of Fibroblast Growth Factor: Activity in Cell Culture. ACTA ACUST UNITED AC 2011. [DOI: 10.1557/proc-252-273] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
ABSTRACTBasic fibroblast growth factor is a multi-potent cell regulatory factor that stimulates proliferation and angiogenesis. Controlled studies of basic fibroblast growth factor in animals have been hindered by the instability of this protein. In addition, many cells appear to require the continuous addition of basic fibroblast growth factor for optimal growth and function in culture. A system for the sustained delivery of active basic fibroblast growth factor might provide both a means to conduct log-term studies on activity and provide a practical alternative to multiple growth factor additions to cell cultures. Basic fibroblast growth factor was incorporated into standard polymer matrices, but the released growth factor had lost over 99% of its bioactivity. Loss of basic fibroblast growth factor activity was found to result from both physical inactivation and adsorption of the protein to surfaces. These problems were avoided by incorporating the growth factor into calcium cross-linked alginate microspheres containing heparinsepharose beads. Basic fibroblast growth factor was incorporated into these microspheres with 71% efficiency and active growth factor was released with predictable kinetics for up to 7 weeks. Release from these microspheres was controlled by the amount of heparin within the device and could be manipulated by simply altering the heparin content during fabrication. Alginate/heparin-sepharose microspheres were placed into growing cultures of bovine aortic endothelial cells and no cytotoxic effects were observed. Furthermore, microspheres containing growth factor provided long-term stimulation of cell proliferation and maintenance of endothelial cell morphology.
Collapse
|
49
|
|
50
|
Huang MC, Lo MJ, Lin YL, Chang SE, Huang WC, Kuo WC, Tsai MJ, Kuo HS, Shih YH, Cheng H. Functional recovery after the repair of transected cervical roots in the chronic stage of injury. J Neurotrauma 2010; 26:1795-804. [PMID: 19548814 DOI: 10.1089/neu.2008.0529] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The treatment of root injury is typically performed at the more chronic stages post injury, by which time a substantial number of neurons have died. Therefore, before being applied in the clinical setting, a treatment strategy for these lesions should prove to be as effective in the chronic stages of injury as it is in the acute stage. In this study, we simulated the most severe clinical scenarios to establish an optimal time window for repair at a chronic stage. The sixth to eighth cervical roots on the left side of female SD rats were cut at their junction with the spinal cord. One or three weeks later, the wound was reopened and these roots were repaired with intercostal nerve grafts, with subsequent application of aFGF and fibrin glue. In the control group, the wound was closed after re-exploration without further repair procedures. Sensory and motor functions were measured after the surgery. Spinal cord morphology, neuron survival, and nerve fiber regeneration were traced by CTB-HRP. Results showed that both the sensory and motor functions had significant recovery in the 1-week repair group, but not in the 3-week repair group. By CTB-HRP tracing, we found that the architecture of the spinal cords was relatively preserved in the 1-week repair group, while those of the control group showed significant atrophic change. There were regenerating nerve fibers in the dorsal horn and more motor neuron survival in the 1-week repair group compared to that of the 3-week group. It was concluded that treating transected cervical roots at a chronic stage with microsurgical nerve grafting and application of aFGF and fibrin glue can lead to significant functional recovery, as long as the repair is done before too many neurons die.
Collapse
Affiliation(s)
- Ming-Chao Huang
- Center for Neural Regeneration, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|