1
|
Sirois JP, Heinz A. Matrikines in the skin: Origin, effects, and therapeutic potential. Pharmacol Ther 2024; 260:108682. [PMID: 38917886 DOI: 10.1016/j.pharmthera.2024.108682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/31/2024] [Accepted: 06/21/2024] [Indexed: 06/27/2024]
Abstract
The extracellular matrix (ECM) represents a complex multi-component environment that has a decisive influence on the biomechanical properties of tissues and organs. Depending on the tissue, ECM components are subject to a homeostasis of synthesis and degradation, a subtle interplay that is influenced by external factors and the intrinsic aging process and is often disturbed in pathologies. Upon proteolytic cleavage of ECM proteins, small bioactive peptides termed matrikines can be formed. These bioactive peptides play a crucial role in cell signaling and contribute to the dynamic regulation of both physiological and pathological processes such as tissue remodeling and repair as well as inflammatory responses. In the skin, matrikines exert an influence for instance on cell adhesion, migration, and proliferation as well as vasodilation, angiogenesis and protein expression. Due to their manifold functions, matrikines represent promising leads for developing new therapeutic options for the treatment of skin diseases. This review article gives a comprehensive overview on matrikines in the skin, including their origin in the dermal ECM, their biological effects and therapeutic potential for the treatment of skin pathologies such as melanoma, chronic wounds and inflammatory skin diseases or for their use in anti-aging cosmeceuticals.
Collapse
Affiliation(s)
- Jonathan P Sirois
- Department of Pharmacy, LEO Foundation Center for Cutaneous Drug Delivery, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Heinz
- Department of Pharmacy, LEO Foundation Center for Cutaneous Drug Delivery, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
2
|
Wang Z, Shi H, Silveira PA, Mithieux SM, Wong WC, Liu L, Pham NTH, Hawkett BS, Wang Y, Weiss AS. Tropoelastin modulates systemic and local tissue responses to enhance wound healing. Acta Biomater 2024; 184:54-67. [PMID: 38871204 DOI: 10.1016/j.actbio.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/13/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Wound healing is facilitated by biomaterials-based grafts and substantially impacted by orchestrated inflammatory responses that are essential to the normal repair process. Tropoelastin (TE) based materials are known to shorten the period for wound repair but the mechanism of anti-inflammatory performance is not known. To explore this, we compared the performance of the gold standard Integra Dermal Regeneration Template (Integra), polyglycerol sebacate (PGS), and TE blended with PGS, in a murine full-thickness cutaneous wound healing study. Systemically, blending with TE favorably increased the F4/80+ macrophage population by day 7 in the spleen and contemporaneously induced elevated plasma levels of anti-inflammatory IL-10. In contrast, the PGS graft without TE prompted prolonged inflammation, as evidenced by splenomegaly and greater splenic granulocyte and monocyte fractions at day 14. Locally, the inclusion of TE in the graft led to increased anti-inflammatory M2 macrophages and CD4+T cells at the wound site, and a rise in Foxp3+ regulatory T cells in the wound bed by day 7. We conclude that the TE-incorporated skin graft delivers a pro-healing environment by modulating systemic and local tissue responses. STATEMENT OF SIGNIFICANCE: Tropoelastin (TE) has shown significant benefits in promoting the repair and regeneration of damaged human tissues. In this study, we show that TE promotes an anti-inflammatory environment that facilitates cutaneous wound healing. In a mouse model, we find that inserting a TE-containing material into a full-thickness wound results in defined, pro-healing local and systemic tissue responses. These findings advance our understanding of TE's restorative value in tissue engineering and regenerative medicine, and pave the way for clinical applications.
Collapse
Affiliation(s)
- Ziyu Wang
- School of Life and Environmental Sciences, the University of Sydney, NSW 2006, Australia; Charles Perkins Centre, the University of Sydney, NSW 2006, Australia
| | - Huaikai Shi
- Burns Research and Reconstructive Surgery, Anzac Research Institute, NSW 2139, Australia; Asbestos and Dust Disease Research Institute, Concord Hospital, Sydney, NSW 2139, Australia
| | - Pablo A Silveira
- Dendritic Cell Group, ANZAC Research Institute, Concord Hospital, Sydney, NSW 2139, Australia
| | - Suzanne M Mithieux
- School of Life and Environmental Sciences, the University of Sydney, NSW 2006, Australia; Charles Perkins Centre, the University of Sydney, NSW 2006, Australia
| | - Wai Cheng Wong
- Charles Perkins Centre, the University of Sydney, NSW 2006, Australia
| | - Linyang Liu
- School of Life and Environmental Sciences, the University of Sydney, NSW 2006, Australia; Charles Perkins Centre, the University of Sydney, NSW 2006, Australia
| | - Nguyen T H Pham
- Key Centre for Polymers and Colloids, School of Chemistry, the University of Sydney, NSW 2006, Australia
| | - Brian S Hawkett
- Key Centre for Polymers and Colloids, School of Chemistry, the University of Sydney, NSW 2006, Australia
| | - Yiwei Wang
- Burns Research and Reconstructive Surgery, Anzac Research Institute, NSW 2139, Australia; Jiangsu Provincial Engineering Research Centre of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China.
| | - Anthony S Weiss
- School of Life and Environmental Sciences, the University of Sydney, NSW 2006, Australia; Charles Perkins Centre, the University of Sydney, NSW 2006, Australia; The University of Sydney Nano Institute, the University of Sydney, NSW 2006, Australia.
| |
Collapse
|
3
|
Wozny MR, Nelea V, Siddiqui IFS, Wanga S, de Waard V, Strauss M, Reinhardt DP. Microfibril-associated glycoprotein 4 forms octamers that mediate interactions with elastogenic proteins and cells. Nat Commun 2024; 15:4015. [PMID: 38740766 PMCID: PMC11091212 DOI: 10.1038/s41467-024-48377-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
Microfibril-associated glycoprotein 4 (MFAP4) is a 36-kDa extracellular matrix glycoprotein with critical roles in organ fibrosis, chronic obstructive pulmonary disease, and cardiovascular disorders, including aortic aneurysms. MFAP4 multimerises and interacts with elastogenic proteins, including fibrillin-1 and tropoelastin, and with cells via integrins. Structural details of MFAP4 and its potential interfaces for these interactions are unknown. Here, we present a cryo-electron microscopy structure of human MFAP4. In the presence of calcium, MFAP4 assembles as an octamer, where two sets of homodimers constitute the top and bottom halves of each octamer. Each homodimer is linked together by an intermolecular disulphide bond. A C34S missense mutation prevents disulphide-bond formation between monomers but does not prevent octamer assembly. The atomic model, built into the 3.55 Å cryo-EM map, suggests that salt-bridge interactions mediate homodimer assembly, while non-polar residues form the interface between octamer halves. In the absence of calcium, an MFAP4 octamer dissociates into two tetramers. Binding studies with fibrillin-1, tropoelastin, LTBP4, and small fibulins show that MFAP4 has multiple surfaces for protein-protein interactions, most of which depend upon MFAP4 octamer assembly. The C34S mutation does not affect these protein interactions or cell interactions. MFAP4 assemblies with fibrillin-1 abrogate MFAP4 interactions with cells.
Collapse
Affiliation(s)
- Michael R Wozny
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Valentin Nelea
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | | | - Shaynah Wanga
- Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Vivian de Waard
- Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Mike Strauss
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.
| | - Dieter P Reinhardt
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada.
| |
Collapse
|
4
|
Liu D, Billington CJ, Raja N, Wong ZC, Levin MD, Resch W, Alba C, Hupalo DN, Biamino E, Bedeschi MF, Digilio MC, Squeo GM, Villa R, Parrish PCR, Knutsen RH, Osgood S, Freeman JA, Dalgard CL, Merla G, Pober BR, Mervis CB, Roberts AE, Morris CA, Osborne LR, Kozel BA. Matrisome and Immune Pathways Contribute to Extreme Vascular Outcomes in Williams-Beuren Syndrome. J Am Heart Assoc 2024; 13:e031377. [PMID: 38293922 PMCID: PMC11056152 DOI: 10.1161/jaha.123.031377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/28/2023] [Indexed: 02/01/2024]
Abstract
BACKGROUND Supravalvar aortic stenosis (SVAS) is a characteristic feature of Williams-Beuren syndrome (WBS). Its severity varies: ~20% of people with Williams-Beuren syndrome have SVAS requiring surgical intervention, whereas ~35% have no appreciable SVAS. The remaining individuals have SVAS of intermediate severity. Little is known about genetic modifiers that contribute to this variability. METHODS AND RESULTS We performed genome sequencing on 473 individuals with Williams-Beuren syndrome and developed strategies for modifier discovery in this rare disease population. Approaches include extreme phenotyping and nonsynonymous variant prioritization, followed by gene set enrichment and pathway-level association tests. We next used GTEx v8 and proteomic data sets to verify expression of candidate modifiers in relevant tissues. Finally, we evaluated overlap between the genes/pathways identified here and those ascertained through larger aortic disease/trait genome-wide association studies. We show that SVAS severity in Williams-Beuren syndrome is associated with increased frequency of common and rarer variants in matrisome and immune pathways. Two implicated matrisome genes (ACAN and LTBP4) were uniquely expressed in the aorta. Many genes in the identified pathways were previously reported in genome-wide association studies for aneurysm, bicuspid aortic valve, or aortic size. CONCLUSIONS Smaller sample sizes in rare disease studies necessitate new approaches to detect modifiers. Our strategies identified variation in matrisome and immune pathways that are associated with SVAS severity. These findings suggest that, like other aortopathies, SVAS may be influenced by the balance of synthesis and degradation of matrisome proteins. Leveraging multiomic data and results from larger aorta-focused genome-wide association studies may accelerate modifier discovery for rare aortopathies like SVAS.
Collapse
Affiliation(s)
- Delong Liu
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Charles J. Billington
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
- Department of PediatricsUniversity of MinnesotaMinneapolisMN
| | - Neelam Raja
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Zoe C. Wong
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Mark D. Levin
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Wulfgang Resch
- The High Performance Computing FacilityCenter for Information Technology, National Institutes of HealthBethesdaMD
| | - Camille Alba
- Henry M Jackson Foundation for the Advancement of Military MedicineBethesdaMD
| | - Daniel N. Hupalo
- Henry M Jackson Foundation for the Advancement of Military MedicineBethesdaMD
| | | | | | | | - Gabriella Maria Squeo
- Laboratory of Regulatory and Functional GenomicsFondazione IRCCS Casa Sollievo della SofferenzaSan Giovanni Rotondo (Foggia)Italy
| | - Roberta Villa
- Fondazione IRCCS Ca Granda Ospedale Maggiore Policlinico Medical Genetic UnitMilanItaly
| | - Pheobe C. R. Parrish
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
- Department of Genome SciencesUniversity of WashingtonSeattleWA
| | - Russell H. Knutsen
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Sharon Osgood
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Joy A. Freeman
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Clifton L. Dalgard
- Department of Anatomy, Physiology and Genetics, School of Medicinethe Uniformed Services University of the Health SciencesBethesdaMD
| | - Giuseppe Merla
- Laboratory of Regulatory and Functional GenomicsFondazione IRCCS Casa Sollievo della SofferenzaSan Giovanni Rotondo (Foggia)Italy
- Department of Molecular Medicine and Medical BiotechnologyUniversity of Naples Federico IINaplesItaly
| | - Barbara R. Pober
- Section of Genetics, Department of PediatricsMassachusetts General HospitalBostonMA
| | - Carolyn B. Mervis
- Department of Psychological and Brain SciencesUniversity of LouisvilleLouisvilleKY
| | - Amy E. Roberts
- Department of Cardiology and Division of Genetics and Genomics, Department of PediatricsBoston Children’s HospitalBostonMA
| | - Colleen A. Morris
- Department of PediatricsKirk Kerkorian School of Medicine at UNLVLas VegasNV
| | - Lucy R. Osborne
- Departments of Medicine and Molecular GeneticsUniversity of TorontoCanada
| | - Beth A. Kozel
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| |
Collapse
|
5
|
Morwood AJ, El-Karim IA, Clarke SA, Lundy FT. The Role of Extracellular Matrix (ECM) Adhesion Motifs in Functionalised Hydrogels. Molecules 2023; 28:4616. [PMID: 37375171 DOI: 10.3390/molecules28124616] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 06/29/2023] Open
Abstract
To create functional tissue engineering scaffolds, biomaterials should mimic the native extracellular matrix of the tissue to be regenerated. Simultaneously, the survival and functionality of stem cells should also be enhanced to promote tissue organisation and repair. Hydrogels, but in particular, peptide hydrogels, are an emerging class of biocompatible scaffolds which act as promising self-assembling biomaterials for tissue engineering and regenerative therapies, ranging from articular cartilage regeneration at joint defects, to regenerative spinal cord injury following trauma. To enhance hydrogel biocompatibility, it has become imperative to consider the native microenvironment of the site for regeneration, where the use of functionalised hydrogels with extracellular matrix adhesion motifs has become a novel, emerging theme. In this review, we will introduce hydrogels in the context of tissue engineering, provide insight into the complexity of the extracellular matrix, investigate specific adhesion motifs that have been used to generate functionalised hydrogels and outline their potential applications in a regenerative medicine setting. It is anticipated that by conducting this review, we will provide greater insight into functionalised hydrogels, which may help translate their use towards therapeutic roles.
Collapse
Affiliation(s)
- Anna J Morwood
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Ikhlas A El-Karim
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Susan A Clarke
- Medical Biology Centre, School of Nursing and Midwifery, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Fionnuala T Lundy
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| |
Collapse
|
6
|
Wu K, Liu Z, Wang W, Zhou F, Cheng Q, Bian Y, Su W, Liu B, Zha J, Zhao J, Zheng X. An artificially designed elastin-like recombinant polypeptide improves aging skin. Am J Transl Res 2022; 14:8562-8571. [PMID: 36628198 PMCID: PMC9827338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 11/13/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND As a substrate for cell growth, elastin can promote the regeneration and remodeling of the epidermis, which plays an important role in delaying skin aging. However, elastin proteins are more than 700 amino acids long and cannot be absorbed through the skin, which prevents the direct utilization of elastin in the prevention and treatment of aging skin. METHODS We designed an elastin-like recombinant polypeptide (ELR) which could be absorbed through the skin based on the property of hexapeptide VGVAPG. Thirty healthy Chinese Han female participants which met the criteria were enrolled in this study and all of them completed the tests including elasticity, tightness, and wrinkle detection. The participants used this polypeptide for 4 weeks and were tested in three visits: one day before trial started (D0), and 14 and 28 days after the trial (D14 and D28, respectively). Paired t-tests or Wilcoxon signed-rank tests for non-parametric measures were used to determine the difference between D0 and D14, or D0 and D28. RESULTS The skin elasticity level in the thirty participants was significantly increased after using ELR for 28 days (P=0.024), and the average value of skin firmness (Uf) declined from 3.313 (D0) to 3.292 (D14) and 3.265 (D28), although there was no statistically significant difference between treatment and pre-treatment. Furthermore, the wrinkle count (D14: P<0.001; D28: P<0.001), wrinkles volume (D14: P<0.001; D28: P=0.008), and wrinkles area (D14: P<0.001; D28: P<0.001) of Crow's feet were significantly improved by using ELR for 14 days or 28 days. CONCLUSION Continuous use of ELR could significantly improve skin elasticity and reduce wrinkles.
Collapse
Affiliation(s)
- Kejia Wu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, P. R. China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of EducationHefei, Anhui, P. R. China
- Anhui Province Laboratory of Inflammation and Immune Mediated DiseasesHefei 230031, Anhui, P. R. China
- Anhui Provincial Institute of Translational MedicineHefei 230031, Anhui, P. R. China
- First Clinical Medical College, Anhui Medical UniversityHefei 230022, Anhui, P. R. China
| | - Zhong Liu
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan UniversityGuangzhou, Guangdong, P. R. China
| | - Wanrong Wang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, P. R. China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of EducationHefei, Anhui, P. R. China
- Anhui Province Laboratory of Inflammation and Immune Mediated DiseasesHefei 230031, Anhui, P. R. China
- Anhui Provincial Institute of Translational MedicineHefei 230031, Anhui, P. R. China
- First Clinical Medical College, Anhui Medical UniversityHefei 230022, Anhui, P. R. China
| | - Feiran Zhou
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, P. R. China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of EducationHefei, Anhui, P. R. China
- Anhui Province Laboratory of Inflammation and Immune Mediated DiseasesHefei 230031, Anhui, P. R. China
- Anhui Provincial Institute of Translational MedicineHefei 230031, Anhui, P. R. China
- First Clinical Medical College, Anhui Medical UniversityHefei 230022, Anhui, P. R. China
| | - Qianhui Cheng
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, P. R. China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of EducationHefei, Anhui, P. R. China
- Anhui Province Laboratory of Inflammation and Immune Mediated DiseasesHefei 230031, Anhui, P. R. China
- Anhui Provincial Institute of Translational MedicineHefei 230031, Anhui, P. R. China
- First Clinical Medical College, Anhui Medical UniversityHefei 230022, Anhui, P. R. China
| | - Yannan Bian
- Yunnan Botanee Bio-technology Groip Co., Ltd.Kunming 650000, Yunnan, P. R. China
| | - Wenrou Su
- Yunnan Botanee Bio-technology Groip Co., Ltd.Kunming 650000, Yunnan, P. R. China
| | - Baoyu Liu
- Department of Microbiology, Anhui Medical UniversityHefei 230032, Anhui, P. R. China
| | - Jindian Zha
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, P. R. China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of EducationHefei, Anhui, P. R. China
- Anhui Province Laboratory of Inflammation and Immune Mediated DiseasesHefei 230031, Anhui, P. R. China
- Anhui Provincial Institute of Translational MedicineHefei 230031, Anhui, P. R. China
- Department of Health Management, Anhui Medical UniversityHefei 230032, Anhui, P. R. China
| | - Jun Zhao
- Department of Microbiology, Anhui Medical UniversityHefei 230032, Anhui, P. R. China
- Wuhu Interferon Bio-products Industry Research Institute Co., Ltd.Wuhu 241007, Anhui, P. R. China
| | - Xiaodong Zheng
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, P. R. China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of EducationHefei, Anhui, P. R. China
- Anhui Province Laboratory of Inflammation and Immune Mediated DiseasesHefei 230031, Anhui, P. R. China
- Anhui Provincial Institute of Translational MedicineHefei 230031, Anhui, P. R. China
| |
Collapse
|
7
|
Boëté Q, Lo M, Liu KL, Vial G, Lemarié E, Rougelot M, Steuckardt I, Harki O, Couturier A, Gaucher J, Bouyon S, Demory A, Boutin-Paradis A, El Kholti N, Berthier A, Pépin JL, Briançon-Marjollet A, Lambert E, Debret R, Faury G. Physiological Impact of a Synthetic Elastic Protein in Arterial Diseases Related to Alterations of Elastic Fibers: Effect on the Aorta of Elastin-Haploinsufficient Male and Female Mice. Int J Mol Sci 2022; 23:13464. [PMID: 36362244 PMCID: PMC9656458 DOI: 10.3390/ijms232113464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 09/08/2024] Open
Abstract
Elastic fibers, made of elastin (90%) and fibrillin-rich microfibrils (10%), are the key extracellular components, which endow the arteries with elasticity. The alteration of elastic fibers leads to cardiovascular dysfunctions, as observed in elastin haploinsufficiency in mice (Eln+/-) or humans (supravalvular aortic stenosis or Williams-Beuren syndrome). In Eln+/+ and Eln+/- mice, we evaluated (arteriography, histology, qPCR, Western blots and cell cultures) the beneficial impact of treatment with a synthetic elastic protein (SEP), mimicking several domains of tropoelastin, the precursor of elastin, including hydrophobic elasticity-related domains and binding sites for elastin receptors. In the aorta or cultured aortic smooth muscle cells from these animals, SEP treatment induced a synthesis of elastin and fibrillin-1, a thickening of the aortic elastic lamellae, a decrease in wall stiffness and/or a strong trend toward a reduction in the elastic lamella disruptions in Eln+/- mice. SEP also modified collagen conformation and transcript expressions, enhanced the aorta constrictive response to phenylephrine in several animal groups, and, in female Eln+/- mice, it restored the normal vasodilatory response to acetylcholine. SEP should now be considered as a biomimetic molecule with an interesting potential for future treatments of elastin-deficient patients with altered arterial structure/function.
Collapse
Affiliation(s)
- Quentin Boëté
- Université Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, U1300, 38000 Grenoble, France
| | - Ming Lo
- Institut de Biologie et Chimie des Protéines UMR5305-LBTI, CNRS, Lyon-7, Passage du Vercors, CEDEX 07, 69367 Lyon, France
| | - Kiao-Ling Liu
- Institut de Biologie et Chimie des Protéines UMR5305-LBTI, CNRS, Lyon-7, Passage du Vercors, CEDEX 07, 69367 Lyon, France
| | - Guillaume Vial
- Université Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, U1300, 38000 Grenoble, France
| | - Emeline Lemarié
- Université Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, U1300, 38000 Grenoble, France
| | - Maxime Rougelot
- Université Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, U1300, 38000 Grenoble, France
| | - Iris Steuckardt
- Université Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, U1300, 38000 Grenoble, France
| | - Olfa Harki
- Université Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, U1300, 38000 Grenoble, France
| | - Axel Couturier
- Université Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, U1300, 38000 Grenoble, France
| | - Jonathan Gaucher
- Université Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, U1300, 38000 Grenoble, France
| | - Sophie Bouyon
- Université Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, U1300, 38000 Grenoble, France
| | - Alexandra Demory
- Université Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, U1300, 38000 Grenoble, France
| | - Antoine Boutin-Paradis
- Université Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, U1300, 38000 Grenoble, France
| | - Naima El Kholti
- Institut de Biologie et Chimie des Protéines UMR5305-LBTI, CNRS, Lyon-7, Passage du Vercors, CEDEX 07, 69367 Lyon, France
| | - Aurore Berthier
- Institut de Biologie et Chimie des Protéines UMR5305-LBTI, CNRS, Lyon-7, Passage du Vercors, CEDEX 07, 69367 Lyon, France
| | - Jean-Louis Pépin
- Université Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, U1300, 38000 Grenoble, France
| | | | - Elise Lambert
- Institut de Biologie et Chimie des Protéines UMR5305-LBTI, CNRS, Lyon-7, Passage du Vercors, CEDEX 07, 69367 Lyon, France
| | - Romain Debret
- Institut de Biologie et Chimie des Protéines UMR5305-LBTI, CNRS, Lyon-7, Passage du Vercors, CEDEX 07, 69367 Lyon, France
| | - Gilles Faury
- Université Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, U1300, 38000 Grenoble, France
| |
Collapse
|
8
|
Tian DM, Wan HH, Chen JR, Ye YB, He Y, Liu Y, Tang LY, He ZY, Liu KZ, Gao CJ, Li SL, Xu Q, Yang Z, Lai C, Xu XJ, Ruan CS, Xu YS, Zhang C, Luo L, Yan LP. In-situ formed elastin-based hydrogels enhance wound healing via promoting innate immune cells recruitment and angiogenesis. Mater Today Bio 2022; 15:100300. [PMID: 35665231 PMCID: PMC9157562 DOI: 10.1016/j.mtbio.2022.100300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/03/2022] [Accepted: 05/17/2022] [Indexed: 01/06/2023]
Abstract
Harnessing the inflammation and angiogenesis is extremely important in wound healing. In this study, we developed bioactive elastin-based hydrogels which can recruit and modulate the innate immune cells and accelerate angiogenesis in the wound site and subsequently improve wound regeneration. These hydrogels were formed by visible-light cross-linking of acryloyl-(polyethylene glycol)-N-hydroxysuccinimide ester modified elastin with methacrylated gelatin, in order to mimic dermal microenvironment. These hydrogels showed highly tunable mechanical properties, swelling ratios and enzymatic degradation profiles, with moduli within the range of human skin. To mimic the in vivo degradation of the elastin by elastase from neutrophils, in vitro co-culture of the hydrogels and neutrophils was conducted. The derived conditioned medium containing elastin derived peptides (EDP-conditioned medium) promoted the expression of both M1 and M2 markers in M1 macrophages in vitro. Additionally, the EDP-conditioned medium induced superior tube formation of endothelia cells in Matrigel. In mice wound model, these elastin-based hydrogels attracted abundant neutrophils and predominant M2 macrophages to the wound and supported their infiltration into the hydrogels. The outstanding immunomodulatory effect of the elastin-based hydrogels resulted in superior angiogenesis, collagen deposition and dermal regeneration. Hence, these elastin-based hydrogels can be a promising regenerative platform to accelerate wound repair.
Collapse
|
9
|
Zhang X, Alanazi YF, Jowitt TA, Roseman AM, Baldock C. Elastic Fibre Proteins in Elastogenesis and Wound Healing. Int J Mol Sci 2022; 23:4087. [PMID: 35456902 PMCID: PMC9027394 DOI: 10.3390/ijms23084087] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 12/30/2022] Open
Abstract
As essential components of our connective tissues, elastic fibres give tissues such as major blood vessels, skin and the lungs their elasticity. Their formation is complex and co-ordinately regulated by multiple factors. In this review, we describe key players in elastogenesis: fibrillin-1, tropoelastin, latent TGFβ binding protein-4, and fibulin-4 and -5. We summarise their roles in elastogenesis, discuss the effect of their mutations on relevant diseases, and describe their interactions involved in forming the elastic fibre network. Moreover, we look into their roles in wound repair for a better understanding of their potential application in tissue regeneration.
Collapse
Affiliation(s)
- Xinyang Zhang
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (X.Z.); (T.A.J.)
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK;
| | - Yasmene F. Alanazi
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Thomas A. Jowitt
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (X.Z.); (T.A.J.)
| | - Alan M. Roseman
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK;
| | - Clair Baldock
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (X.Z.); (T.A.J.)
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK;
| |
Collapse
|
10
|
Hoareau M, El Kholti N, Debret R, Lambert E. Zebrafish as a Model to Study Vascular Elastic Fibers and Associated Pathologies. Int J Mol Sci 2022; 23:2102. [PMID: 35216218 PMCID: PMC8875079 DOI: 10.3390/ijms23042102] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/09/2022] [Accepted: 02/12/2022] [Indexed: 02/06/2023] Open
Abstract
Many extensible tissues such as skin, lungs, and blood vessels require elasticity to function properly. The recoil of elastic energy stored during a stretching phase is provided by elastic fibers, which are mostly composed of elastin and fibrillin-rich microfibrils. In arteries, the lack of elastic fibers leads to a weakening of the vessel wall with an increased risk to develop cardiovascular defects such as stenosis, aneurysms, and dissections. The development of new therapeutic molecules involves preliminary tests in animal models that recapitulate the disease and whose response to drugs should be as close as possible to that of humans. Due to its superior in vivo imaging possibilities and the broad tool kit for forward and reverse genetics, the zebrafish has become an important model organism to study human pathologies. Moreover, it is particularly adapted to large scale studies, making it an attractive model in particular for the first steps of investigations. In this review, we discuss the relevance of the zebrafish model for the study of elastic fiber-related vascular pathologies. We evidence zebrafish as a compelling alternative to conventional mouse models.
Collapse
Affiliation(s)
- Marie Hoareau
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Institut de Biologie et Chimie des Protéines, Université Lyon 1, 7, Passage du Vercors, CEDEX 07, F-69367 Lyon, France; (N.E.K.); (R.D.)
| | | | | | - Elise Lambert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Institut de Biologie et Chimie des Protéines, Université Lyon 1, 7, Passage du Vercors, CEDEX 07, F-69367 Lyon, France; (N.E.K.); (R.D.)
| |
Collapse
|
11
|
Tembely D, Henry A, Vanalderwiert L, Toussaint K, Bennasroune A, Blaise S, Sartelet H, Jaisson S, Galés C, Martiny L, Duca L, Romier-Crouzet B, Maurice P. The Elastin Receptor Complex: An Emerging Therapeutic Target Against Age-Related Vascular Diseases. Front Endocrinol (Lausanne) 2022; 13:815356. [PMID: 35222273 PMCID: PMC8873114 DOI: 10.3389/fendo.2022.815356] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/20/2022] [Indexed: 12/26/2022] Open
Abstract
The incidence of cardiovascular diseases is increasing worldwide with the growing aging of the population. Biological aging has major influence on the vascular tree and is associated with critical changes in the morphology and function of the arterial wall together with an extensive remodeling of the vascular extracellular matrix. Elastic fibers fragmentation and release of elastin degradation products, also known as elastin-derived peptides (EDPs), are typical hallmarks of aged conduit arteries. Along with the direct consequences of elastin fragmentation on the mechanical properties of arteries, the release of EDPs has been shown to modulate the development and/or progression of diverse vascular and metabolic diseases including atherosclerosis, thrombosis, type 2 diabetes and nonalcoholic steatohepatitis. Most of the biological effects mediated by these bioactive peptides are due to a peculiar membrane receptor called elastin receptor complex (ERC). This heterotrimeric receptor contains a peripheral protein called elastin-binding protein, the protective protein/cathepsin A, and a transmembrane sialidase, the neuraminidase-1 (NEU1). In this review, after an introductive part on the consequences of aging on the vasculature and the release of EDPs, we describe the composition of the ERC, the signaling pathways triggered by this receptor, and the current pharmacological strategies targeting ERC activation. Finally, we present and discuss new regulatory functions that have emerged over the last few years for the ERC through desialylation of membrane glycoproteins by NEU1, and its potential implication in receptor transactivation.
Collapse
Affiliation(s)
- Dignê Tembely
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Aubéri Henry
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Laetitia Vanalderwiert
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Kevin Toussaint
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Amar Bennasroune
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Sébastien Blaise
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Hervé Sartelet
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Stéphane Jaisson
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Céline Galés
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Université de Toulouse, Toulouse, France
| | - Laurent Martiny
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Béatrice Romier-Crouzet
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Pascal Maurice
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
- *Correspondence: Pascal Maurice, ; orcid.org0000-0003-2167-4808
| |
Collapse
|
12
|
Facchinello N, Astone M, Audano M, Oberkersch RE, Spizzotin M, Calura E, Marques M, Crisan M, Mitro N, Santoro MM. Oxidative pentose phosphate pathway controls vascular mural cell coverage by regulating extracellular matrix composition. Nat Metab 2022; 4:123-140. [PMID: 35102339 PMCID: PMC7612297 DOI: 10.1038/s42255-021-00514-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 12/06/2021] [Indexed: 12/11/2022]
Abstract
Vascular mural cells (vMCs) play an essential role in the development and maturation of the vasculature by promoting vessel stabilization through their interactions with endothelial cells. Whether endothelial metabolism influences mural cell recruitment and differentiation is unknown. Here, we show that the oxidative pentose phosphate pathway (oxPPP) in endothelial cells is required for establishing vMC coverage of the dorsal aorta during early vertebrate development in zebrafish and mice. We demonstrate that laminar shear stress and blood flow maintain oxPPP activity, which in turn, promotes elastin expression in blood vessels through production of ribose-5-phosphate. Elastin is both necessary and sufficient to drive vMC recruitment and maintenance when the oxPPP is active. In summary, our work demonstrates that endothelial cell metabolism regulates blood vessel maturation by controlling vascular matrix composition and vMC recruitment.
Collapse
Affiliation(s)
- Nicola Facchinello
- Laboratory of Angiogenesis and Cancer Metabolism, DiBio, University of Padua, Padua, Italy
| | - Matteo Astone
- Laboratory of Angiogenesis and Cancer Metabolism, DiBio, University of Padua, Padua, Italy
| | - Matteo Audano
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, Milan, Italy
| | - Roxana E Oberkersch
- Laboratory of Angiogenesis and Cancer Metabolism, DiBio, University of Padua, Padua, Italy
| | - Marianna Spizzotin
- Laboratory of Angiogenesis and Cancer Metabolism, DiBio, University of Padua, Padua, Italy
| | - Enrica Calura
- Department of Biology, University of Padua, Padua, Italy
| | - Madalena Marques
- Centre for Cardiovascular Science and Centre for Regenerative Medicine/Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Mihaela Crisan
- Centre for Cardiovascular Science and Centre for Regenerative Medicine/Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, Milan, Italy
| | - Massimo M Santoro
- Laboratory of Angiogenesis and Cancer Metabolism, DiBio, University of Padua, Padua, Italy.
| |
Collapse
|
13
|
Bocquet O, Tembely D, Rioult D, Terryn C, Romier B, Bennasroune A, Blaise S, Sartelet H, Martiny L, Duca L, Maurice P. Characterization of novel interactions with membrane NEU1 highlights new regulatory functions for the Elastin Receptor Complex in monocyte interaction with endothelial cells. Cell Biosci 2021; 11:206. [PMID: 34903296 PMCID: PMC8670255 DOI: 10.1186/s13578-021-00718-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/20/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Vascular aging is associated with remodeling of elastin, one of the main extracellular matrix component of the arterial wall, and production of elastin-derived peptides (EDP). These extracellular matrix degradation products have been shown to trigger biological activities through the elastin receptor complex (ERC) and data from the last decade have brought significant insights on the critical role played by its NEU1 subunit in the biological effects mediated by EDP and the ERC in vascular and metabolic diseases. RESULTS Using a proteomic approach, we previously identified new potential interaction partners of membrane NEU1. Here, we validated the interaction between NEU1 and the β2 integrin in human monocytes and show that binding of EDP to the ERC leads to desialylation of β2 integrin through NEU1. A similar action mechanism was identified in human umbilical vein endothelial cells (HUVEC) for intercellular cell adhesion molecule-1 (ICAM-1). Importantly, these effects were associated with a significant increase in monocyte adhesion to endothelial cells and monocyte transendothelial migration. CONCLUSIONS These results demonstrate that membrane NEU1 sialidase interacts and modulates the sialylation levels of the β2 integrin and ICAM-1 through the ERC in monocytes and endothelial cells, respectively, and suggest that EDP and the ERC, through this newly identified common mode of action governed by NEU1, may be important regulators of circulating monocyte recruitment to inflamed vascular sites. Moreover, by its ability to interact with and to modulate the sialylation of key membrane glycoproteins through NEU1, new biological functions are anticipated for EDP and the ERC in elastin remodeling-associated disorders.
Collapse
Affiliation(s)
- Olivier Bocquet
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Dignê Tembely
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Damien Rioult
- Plateau Technique Mobile de Cytométrie Environnementale MOBICYTE, URCA/INERIS, Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Reims, France
| | - Christine Terryn
- Plate-Forme Imagerie Cellulaire Et Tissulaire (PICT), Université de Reims Champagne Ardenne (URCA), UFR Médecine, Reims, France
| | - Béatrice Romier
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Amar Bennasroune
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Sébastien Blaise
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Hervé Sartelet
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Laurent Martiny
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Pascal Maurice
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France.
| |
Collapse
|
14
|
Singh M, Becker M, Godwin AR, Baldock C. Structural studies of elastic fibre and microfibrillar proteins. Matrix Biol Plus 2021; 12:100078. [PMID: 34355160 PMCID: PMC8322146 DOI: 10.1016/j.mbplus.2021.100078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 11/27/2022] Open
Abstract
Elastic tissues owe their functional properties to the composition of their extracellular matrices, particularly the range of extracellular, multidomain extensible elastic fibre and microfibrillar proteins. These proteins include elastin, fibrillin, latent TGFβ binding proteins (LTBPs) and collagens, where their biophysical and biochemical properties not only give the matrix structural integrity, but also play a vital role in the mechanisms that underlie tissue homeostasis. Thus far structural information regarding the structure and hierarchical assembly of these molecules has been challenging and the resolution has been limited due to post-translational modification and their multidomain nature leading to flexibility, which together result in conformational and structural heterogeneity. In this review, we describe some of the matrix proteins found in elastic fibres and the new emerging techniques that can shed light on their structure and dynamic properties.
Collapse
Affiliation(s)
- Mukti Singh
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK
| | - Mark Becker
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK
| | - Alan R.F. Godwin
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK
| | - Clair Baldock
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK
| |
Collapse
|
15
|
Beyens A, Pottie L, Sips P, Callewaert B. Clinical and Molecular Delineation of Cutis Laxa Syndromes: Paradigms for Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1348:273-309. [PMID: 34807425 DOI: 10.1007/978-3-030-80614-9_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Cutis laxa (CL) syndromes are a large and heterogeneous group of rare connective tissue disorders that share loose redundant skin as a hallmark clinical feature, which reflects dermal elastic fiber fragmentation. Both acquired and congenital-Mendelian- forms exist. Acquired forms are progressive and often preceded by inflammatory triggers in the skin, but may show systemic elastolysis. Mendelian forms are often pleiotropic in nature and classified upon systemic manifestations and mode of inheritance. Though impaired elastogenesis is a common denominator in all Mendelian forms of CL, the underlying gene defects are diverse and affect structural components of the elastic fiber or impair metabolic pathways interfering with cellular trafficking, proline synthesis, or mitochondrial functioning. In this chapter we provide a detailed overview of the clinical and molecular characteristics of the different cutis laxa types and review the latest insights on elastic fiber assembly and homeostasis from both human and animal studies.
Collapse
Affiliation(s)
- Aude Beyens
- Center for Medical Genetics Ghent, Department of Dermatology, Department of Biomolecular Medicine, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Lore Pottie
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Patrick Sips
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Bert Callewaert
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, Ghent University, Ghent, Belgium.
| |
Collapse
|
16
|
Bax DV, Nair M, Weiss AS, Farndale RW, Best SM, Cameron RE. Tailoring the biofunctionality of collagen biomaterials via tropoelastin incorporation and EDC-crosslinking. Acta Biomater 2021; 135:150-163. [PMID: 34454082 DOI: 10.1016/j.actbio.2021.08.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/13/2021] [Accepted: 08/18/2021] [Indexed: 11/17/2022]
Abstract
Recreating the cell niche of virtually all tissues requires composite materials fabricated from multiple extracellular matrix (ECM) macromolecules. Due to their wide tissue distribution, physical attributes and purity, collagen, and more recently, tropoelastin, represent two appealing ECM components for biomaterials development. Here we blend tropoelastin and collagen, harnessing the cell-modulatory properties of each biomolecule. Tropoelastin was stably co-blended into collagen biomaterials and was retained after EDC-crosslinking. We found that human dermal fibroblasts (HDF), rat glial cells (Rugli) and HT1080 fibrosarcoma cells ligate to tropoelastin via EDTA-sensitive and EDTA-insensitive receptors or do not ligate with tropoelastin, respectively. These differing elastin-binding properties allowed us to probe the cellular response to the tropoelastin-collagen composites assigning specific bioactivity to the collagen and tropoelastin component of the composite material. Tropoelastin addition to collagen increased total Rugli cell adhesion, spreading and proliferation. This persisted with EDC-crosslinking of the tropoelastin-collagen composite. Tropoelastin addition did not affect total HDF and HT1080 cell adhesion; however, it increased the contribution of cation-independent adhesion, without affecting the cell morphology or, for HT1080 cells, proliferation. Instead, EDC-crosslinking dictated the HDF and HT1080 cellular response. These data show that a tropoelastin component dominates the response of cells that possess non-integrin based tropoelastin receptors. EDC modification of the collagen component directs cell function when non-integrin tropoelastin receptors are not crucial for cell activity. Using this approach, we have assigned the biological contribution of each component of tropoelastin-collagen composites, allowing informed biomaterial design for directed cell function via more physiologically relevant mechanisms. STATEMENT OF SIGNIFICANCE: Biomaterials fabricated from multiple extracellular matrix (ECM) macromolecules are required to fully recreate the native tissue niche where each ECM macromolecule engages with a specific repertoire of cell-surface receptors. Here we investigate combining tropoelastin with collagen as they interact with cells via different receptors. We identified specific cell lines, which associate with tropoelastin via distinct classes of cell-surface receptor. These showed that tropoelastin, when combined with collagen, altered the cell behaviour in a receptor-usage dependent manner. Integrin-mediated tropoelastin interactions influenced cell proliferation and non-integrin receptors influenced cell spreading and proliferation. These data shed light on the interplay between biomaterial macromolecular composition, cell surface receptors and cell behaviour, advancing bespoke materials design and providing functionality to specific cell populations.
Collapse
Affiliation(s)
- Daniel V Bax
- Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Road, Cambridge, CB3 0FS, United Kingdom; Department of Biochemistry, University of Cambridge, Downing Site, Cambridge, CB2 1QW, United Kingdom.
| | - Malavika Nair
- Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Road, Cambridge, CB3 0FS, United Kingdom
| | - Anthony S Weiss
- Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia; Charles Perkins Centre, University of Sydney, NSW, 2006, Australia; Sydney Nano Institute, University of Sydney, NSW, 2006, Australia
| | - Richard W Farndale
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge, CB2 1QW, United Kingdom
| | - Serena M Best
- Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Road, Cambridge, CB3 0FS, United Kingdom
| | - Ruth E Cameron
- Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Road, Cambridge, CB3 0FS, United Kingdom
| |
Collapse
|
17
|
Wittig C, Szulcek R. Extracellular Matrix Protein Ratios in the Human Heart and Vessels: How to Distinguish Pathological From Physiological Changes? Front Physiol 2021; 12:708656. [PMID: 34421650 PMCID: PMC8371527 DOI: 10.3389/fphys.2021.708656] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/14/2021] [Indexed: 12/03/2022] Open
Abstract
Cardiovascular pathology is often accompanied by changes in relative content and/or ratios of structural extracellular matrix (ECM) proteins within the heart and elastic vessels. Three of these proteins, collagen-I, collagen-III, and elastin, make up the bulk of the ECM proteins in these tissues, forming a microenvironment that strongly dictates the tissue biomechanical properties and effectiveness of cardiac and vascular function. In this review, we aim to elucidate how the ratios of collagen-I to collagen-III and elastin to collagen are altered in cardiovascular diseases and the aged individuum. We elaborate on these major cardiovascular ECM proteins in terms of structure, tissue localization, turnover, and physiological function and address how their ratios change in aging, dilated cardiomyopathy, coronary artery disease with myocardial infarction, atrial fibrillation, aortic aneurysms, atherosclerosis, and hypertension. To the end of guiding in vitro modeling approaches, we focus our review on the human heart and aorta, discuss limitations in ECM protein quantification methodology, examine comparability between studies, and highlight potential in vitro applications. In summary, we found collagen-I relative concentration to increase or stay the same in cardiovascular disease, resulting in a tendency for increased collagen-I/collagen-III and decreased elastin/collagen ratios. These ratios were found to fall on a continuous scale with ranges defining distinct pathological states as well as a significant difference between the human heart and aortic ECM protein ratios.
Collapse
Affiliation(s)
- Corey Wittig
- Laboratory of in vitro Modeling Systems of Pulmonary Diseases, Institute of Physiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Robert Szulcek
- Laboratory of in vitro Modeling Systems of Pulmonary Diseases, Institute of Physiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
18
|
Yanagisawa H, Yokoyama U. Extracellular matrix-mediated remodeling and mechanotransduction in large vessels during development and disease. Cell Signal 2021; 86:110104. [PMID: 34339854 DOI: 10.1016/j.cellsig.2021.110104] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/08/2023]
Abstract
The vascular extracellular matrix (ECM) is synthesized and secreted during embryogenesis and facilitates the growth and remodeling of large vessels. Proper interactions between the ECM and vascular cells are pivotal for building the vasculature required for postnatal dynamic circulation. The ECM serves as a structural component by maintaining the integrity of the vessel wall while also regulating intercellular signaling, which involves cytokines and growth factors. The major ECM component in large vessels is elastic fibers, which include elastin and microfibrils. Elastin is predominantly synthesized by vascular smooth muscle cells (SMCs) and uses microfibrils as a scaffold to lay down and assemble cross-linked elastin. The absence of elastin causes developmental defects that result in the subendothelial proliferation of SMCs and inward remodeling of the vessel wall. Notably, elastic fiber formation is attenuated in the ductus arteriosus and umbilical arteries. These two vessels function during embryogenesis and close after birth via cellular proliferation, migration, and matrix accumulation. In dynamic postnatal mechano-environments, the elastic fibers in large vessels also serve an essential role in proper signal transduction as a component of elastin-contractile units. Disrupted mechanotransduction in SMCs leads to pathological conditions such as aortic aneurysms that exhibit outward remodeling. This review discusses the importance of the ECM-mainly the elastic fiber matrix-in large vessels during developmental remodeling and under pathological conditions. By dissecting the role of the ECM in large vessels, we aim to provide insights into the role of ECM-mediated signal transduction that can provide a basis for seeking new targets for intervention in vascular diseases.
Collapse
Affiliation(s)
- Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, The University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8577, Japan.
| | - Utako Yokoyama
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan.
| |
Collapse
|
19
|
Ozsvar J, Wang R, Tarakanova A, Buehler MJ, Weiss AS. Fuzzy binding model of molecular interactions between tropoelastin and integrin alphaVbeta3. Biophys J 2021; 120:3138-3151. [PMID: 34197806 DOI: 10.1016/j.bpj.2021.04.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 03/30/2021] [Accepted: 04/12/2021] [Indexed: 12/30/2022] Open
Abstract
Tropoelastin is the highly flexible monomer subunit of elastin, required for the resilience of the extracellular matrix in elastic tissues. To elicit biological signaling, multiple sites on tropoelastin bind to cell surface integrins in a poorly understood multifactorial process. We constructed a full atomistic molecular model of the interactions between tropoelastin and integrin αvβ3 using ensemble-based computational methodologies. Conformational changes of integrin αvβ3 associated with outside-in signaling were more frequently facilitated in an ensemble in which tropoelastin bound the integrin's α1 helix rather than the upstream canonical binding site. Our findings support a model of fuzzy binding, whereby many tropoelastin conformations and defined sites cooperatively interact with multiple αvβ3 regions. This model explains prior experimental binding to distinct tropoelastin regions, domains 17 and 36, and points to the cooperative participation of domain 20. Our study highlights the utility of ensemble-based approaches in helping to understand the interactive mechanisms of functionally significant flexible proteins.
Collapse
Affiliation(s)
- Jazmin Ozsvar
- Charles Perkins Centre, The University of Sydney, Sydney, Australia; School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Richard Wang
- Charles Perkins Centre, The University of Sydney, Sydney, Australia; School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Anna Tarakanova
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut; Department of Mechanical Engineering, University of Connecticut, Storrs, Connecticut
| | - Markus J Buehler
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Anthony S Weiss
- Charles Perkins Centre, The University of Sydney, Sydney, Australia; School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia; Sydney Nano Institute, The University of Sydney, Sydney, Australia.
| |
Collapse
|
20
|
Alshehri S, Susapto HH, Hauser CAE. Scaffolds from Self-Assembling Tetrapeptides Support 3D Spreading, Osteogenic Differentiation, and Angiogenesis of Mesenchymal Stem Cells. Biomacromolecules 2021; 22:2094-2106. [PMID: 33908763 PMCID: PMC8382244 DOI: 10.1021/acs.biomac.1c00205] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/15/2021] [Indexed: 01/01/2023]
Abstract
The apparent rise of bone disorders demands advanced treatment protocols involving tissue engineering. Here, we describe self-assembling tetrapeptide scaffolds for the growth and osteogenic differentiation of human mesenchymal stem cells (hMSCs). The rationally designed peptides are synthetic amphiphilic self-assembling peptides composed of four amino acids that are nontoxic. These tetrapeptides can quickly solidify to nanofibrous hydrogels that resemble the extracellular matrix and provide a three-dimensional (3D) environment for cells with suitable mechanical properties. Furthermore, we can easily tune the stiffness of these peptide hydrogels by just increasing the peptide concentration, thus providing a wide range of peptide hydrogels with different stiffnesses for 3D cell culture applications. Since successful bone regeneration requires both osteogenesis and vascularization, our scaffold was found to be able to promote angiogenesis of human umbilical vein endothelial cells (HUVECs) in vitro. The results presented suggest that ultrashort peptide hydrogels are promising candidates for applications in bone tissue engineering.
Collapse
Affiliation(s)
- Salwa Alshehri
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Hepi H. Susapto
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Charlotte A. E. Hauser
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| |
Collapse
|
21
|
Schmelzer CEH, Duca L. Elastic fibers: formation, function, and fate during aging and disease. FEBS J 2021; 289:3704-3730. [PMID: 33896108 DOI: 10.1111/febs.15899] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/16/2021] [Accepted: 03/22/2021] [Indexed: 01/09/2023]
Abstract
Elastic fibers are extracellular components of higher vertebrates and confer elasticity and resilience to numerous tissues and organs such as large blood vessels, lungs, and skin. Their formation and maturation take place in a complex multistage process called elastogenesis. It requires interactions between very different proteins but also other molecules and leads to the deposition and crosslinking of elastin's precursor on a scaffold of fibrillin-rich microfibrils. Mature fibers are exceptionally resistant to most influences and, under healthy conditions, retain their biomechanical function over the life of the organism. However, due to their longevity, they accumulate damages during aging. These are caused by proteolytic degradation, formation of advanced glycation end products, calcification, oxidative damage, aspartic acid racemization, lipid accumulation, carbamylation, and mechanical fatigue. The resulting changes can lead to diminution or complete loss of elastic fiber function and ultimately affect morbidity and mortality. Particularly, the production of elastokines has been clearly shown to influence several life-threatening diseases. Moreover, the structure, distribution, and abundance of elastic fibers are directly or indirectly influenced by a variety of inherited pathological conditions, which mainly affect organs and tissues such as skin, lungs, or the cardiovascular system. A distinction can be made between microfibril-related inherited diseases that are the result of mutations in diverse microfibril genes and indirectly affect elastogenesis, and elastinopathies that are linked to changes in the elastin gene. This review gives an overview on the formation, structure, and function of elastic fibers and their fate over the human lifespan in health and disease.
Collapse
Affiliation(s)
- Christian E H Schmelzer
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany.,Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Laurent Duca
- UMR CNRS 7369 MEDyC, SFR CAP-Sante, Université de Reims Champagne-Ardenne, France
| |
Collapse
|
22
|
Sharma A, Sharma P, Roy S. Elastin-inspired supramolecular hydrogels: a multifaceted extracellular matrix protein in biomedical engineering. SOFT MATTER 2021; 17:3266-3290. [PMID: 33730140 DOI: 10.1039/d0sm02202k] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The phenomenal advancement in regenerative medicines has led to the development of bioinspired materials to fabricate a biomimetic artificial extracellular matrix (ECM) to support cellular survival, proliferation, and differentiation. Researchers have diligently developed protein polymers consisting of functional sequences of amino acids evolved in nature. Nowadays, certain repetitive bioinspired polymers are treated as an alternative to synthetic polymers due to their unique properties like biodegradability, easy scale-up, biocompatibility, and non-covalent molecular associations which imparts tunable supramolecular architecture to these materials. In this direction, elastin has been identified as a potential scaffold that renders extensibility and elasticity to the tissues. Elastin-like polypeptides (ELPs) are artificial repetitive polymers that exhibit lower critical solution temperature (LCST) behavior in a particular environment than synthetic polymers and hence have gained extensive interest in the fabrication of stimuli-responsive biomaterials. This review discusses in detail the unique structural aspects of the elastin and its soluble precursor, tropoelastin. Furthermore, the versatility of elastin-like peptides is discussed through numerous examples that bolster the significance of elastin in the field of regenerative medicines such as wound care, cardiac tissue engineering, ocular disorders, bone tissue regeneration, etc. Finally, the review highlights the importance of exploring short elastin-mimetic peptides to recapitulate the structural and functional aspects of elastin for advanced healthcare applications.
Collapse
Affiliation(s)
- Archita Sharma
- Institute of Nano Science and Technology (INST), Sector 81, Knowledge City, Mohali, 140306, Punjab, India.
| | | | | |
Collapse
|
23
|
Bochicchio B, Yeo GC, Lee P, Emul D, Pepe A, Laezza A, Ciarfaglia N, Quaglino D, Weiss AS. Domains 12 to 16 of tropoelastin promote cell attachment and spreading through interactions with glycosaminoglycan and integrins alphaV and alpha5beta1. FEBS J 2021; 288:4024-4038. [DOI: 10.1111/febs.15702] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/20/2020] [Accepted: 01/04/2021] [Indexed: 11/27/2022]
Affiliation(s)
| | - Giselle C. Yeo
- Charles Perkins Centre The University of Sydney NSW Australia
- School of Life and Environmental Sciences The University of Sydney NSW Australia
| | - Pearl Lee
- Charles Perkins Centre The University of Sydney NSW Australia
- School of Life and Environmental Sciences The University of Sydney NSW Australia
| | - Deniz Emul
- Charles Perkins Centre The University of Sydney NSW Australia
- School of Life and Environmental Sciences The University of Sydney NSW Australia
| | - Antonietta Pepe
- Department of Science University of Basilicata Potenza Italy
| | - Antonio Laezza
- Department of Science University of Basilicata Potenza Italy
| | | | - Daniela Quaglino
- Department of Life Sciences University of Modena and Reggio Emilia Modena Italy
| | - Anthony S. Weiss
- Charles Perkins Centre The University of Sydney NSW Australia
- School of Life and Environmental Sciences The University of Sydney NSW Australia
- Sydney Nano Institute The University of Sydney NSW Australia
| |
Collapse
|
24
|
Reichheld SE, Muiznieks LD, Huynh Q, Wang N, Ing C, Miao M, Sitarz EE, Pomès R, Sharpe S, Keeley FW. The evolutionary background and functional consequences of the rs2071307 polymorphism in human tropoelastin. Biopolymers 2020; 112:e23414. [PMID: 33351193 DOI: 10.1002/bip.23414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 01/01/2023]
Abstract
Elastin is a major polymeric protein of the extracellular matrix, providing critical properties of extensibility and elastic recoil. The rs2071307 genomic polymorphism, resulting in the substitution of a serine for a glycine residue in a VPG motif in tropoelastin, has an unusually high minor allele frequency in humans. A consequence of such allelic heterozygosity would be the presence of a heterogeneous elastin polymer in up to 50% of the population, a situation which appears to be unique to Homo sapiens. VPG motifs are extremely common in hydrophobic domains of tropoelastins and are the sites of transient β-turns that are essential for maintaining the conformational flexibility required for its function as an entropic elastomer. Earlier data demonstrated that single amino acid substitutions in tropoelastin can have functional consequences for polymeric elastin, particularly when present in mixed polymers. Here, using NMR and molecular dynamics approaches, we show the rs2071307 polymorphism reduces local propensity for β-turn formation, with a consequent increase in polypeptide hydration and an expansion of the conformational ensemble manifested as an increased hydrodynamic radius, radius of gyration and asphericity. Furthermore, this substitution affects functional properties of polymeric elastin, particularly in heterogeneous polymers mimicking allelic heterozygosity. We discuss whether such effects, together with the unusually high minor allele frequency of the polymorphism, could imply some some evolutionary advantage for the heterozygous state.
Collapse
Affiliation(s)
- Sean E Reichheld
- Molecular Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, USA
| | - Lisa D Muiznieks
- Molecular Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, USA.,Elvesys Microfluidics Innovation Center, 172 rue de Charonne, 75011, Paris, France
| | - Quang Huynh
- Molecular Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, USA
| | - Nick Wang
- Molecular Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, USA.,135 W 52nd St. Apt 20A, 10019-7691, New York, New York, USA
| | - Christopher Ing
- Molecular Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, USA.,ProteinQure, Suite 304, 119 Spadina Avenue, M5V2L1, Toronto, Ontario, Canada
| | - Ming Miao
- Molecular Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, USA
| | - Eva E Sitarz
- Molecular Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, USA
| | - Régis Pomès
- Molecular Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, USA.,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Simon Sharpe
- Molecular Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, USA.,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Fred W Keeley
- Molecular Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, USA.,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Aghaei-Ghareh-Bolagh B, Mukherjee S, Lockley KM, Mithieux SM, Wang Z, Emmerson S, Darzi S, Gargett CE, Weiss AS. A novel tropoelastin-based resorbable surgical mesh for pelvic organ prolapse repair. Mater Today Bio 2020; 8:100081. [PMID: 33210083 PMCID: PMC7658716 DOI: 10.1016/j.mtbio.2020.100081] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/30/2020] [Accepted: 10/05/2020] [Indexed: 11/03/2022] Open
Abstract
Pelvic organ prolapse is a common condition that affects 1 in 4 women across all age groups. It is mainly caused by vaginal birth injury and can be exacerbated by obesity and increased age. Until recently, treatment strategies often used non-degradable synthetic meshes for reconstructive surgery. However, owing to their frequent, unacceptable rate of adverse events such as mesh erosion, transvaginal meshes have been banned in many countries. Recent reports have highlighted the urgent need for biocompatible design of meshes for a safe and effective treatment in the long term. This study reports the design and evaluation of a novel, elastin based degradable mesh using an ovine model of POP as a potential surgical treatment. Elastin is a protein component of the ECM and provides elasticity to tissues throughout the body. Tropoelastin, the monomer subunit of elastin, has been used with success in electrospun constructs as it is a naturally cell interactive polymer. Biomaterials that incorporate tropoelastin support cell attachment and proliferation, and have been proven to encourage elastogenesis and angiogenesis in vitro and in vivo. The biological properties of tropoelastin were combined with the physical properties of PCL, a degradable synthetic polymer, with the aim of producing, characterizing and assessing the performance of continuous tropoelastin:PCL electrospun yarns. Using a modified spinneret electrospinning system and adjusting settings based on relative humidity, four blends of tropoelastin:PCL yarns were fabricated with concentration ratios of 75:25, 50:50, 25:75 and 0:100. Yarns were assessed for ease of manufacture, fibrous architecture, protein/polymer content, yarn stability - including initial tropoelastin release, mechanical strength, and ability to support cell growth. Based on overall favorable properties, a mesh woven from the 50:50 tropoelastin:PCL yarn was implanted into the vagina of a parous ewe with vaginal wall weakness as a model of pelvic organ prolapse. This mesh showed excellent integration with new collagen deposition by SEM and a predominant M2 macrophage response with few pro-inflammatory M1 macrophages after 30 days. The woven tropoelastin:PCL electrospun mesh shows potential as an alternative to non-degradable, synthetic pelvic organ prolapse mesh products.
Collapse
Affiliation(s)
- B Aghaei-Ghareh-Bolagh
- Charles Perkins Centre, University of Sydney, NSW, 2006, Australia.,School of Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia
| | - S Mukherjee
- The Ritchie Centre, Hudson Institute of Medical Research, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Victoria, 3168, Australia
| | - K M Lockley
- Charles Perkins Centre, University of Sydney, NSW, 2006, Australia.,School of Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia
| | - S M Mithieux
- Charles Perkins Centre, University of Sydney, NSW, 2006, Australia.,School of Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia
| | - Z Wang
- Charles Perkins Centre, University of Sydney, NSW, 2006, Australia.,School of Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia
| | - S Emmerson
- The Ritchie Centre, Hudson Institute of Medical Research, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Victoria, 3168, Australia
| | - S Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Victoria, 3168, Australia
| | - C E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Victoria, 3168, Australia
| | - A S Weiss
- Charles Perkins Centre, University of Sydney, NSW, 2006, Australia.,School of Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia.,The University of Sydney Nano Institute, University of Sydney, NSW, 2006, Australia
| |
Collapse
|
26
|
Speziale P, Pietrocola G. The Multivalent Role of Fibronectin-Binding Proteins A and B (FnBPA and FnBPB) of Staphylococcus aureus in Host Infections. Front Microbiol 2020; 11:2054. [PMID: 32983039 PMCID: PMC7480013 DOI: 10.3389/fmicb.2020.02054] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/05/2020] [Indexed: 01/21/2023] Open
Abstract
Staphylococcus aureus, one of the most important human pathogens, is the causative agent of several infectious diseases including sepsis, pneumonia, osteomyelitis, endocarditis and soft tissue infections. This pathogenicity is due to a multitude of virulence factors including several cell wall-anchored proteins (CWA). CWA proteins have modular structures with distinct domains binding different ligands. The majority of S. aureus strains express two CWA fibronectin (Fn)-binding adhesins FnBPA and FnBPB (Fn-binding proteins A and B), which are encoded by closely related genes. The N-terminus of FnBPA and FnBPB comprises an A domain which binds ligands such as fibrinogen, elastin and plasminogen. The A domain of FnBPB also interacts with histones and this binding results in the neutralization of the antimicrobial activity of these molecules. The C-terminal moiety of these adhesins comprises a long, intrinsically disordered domain composed of 11/10 fibronectin-binding repeats. These repetitive motifs of FnBPs promote invasion of cells that are not usually phagocytic via a mechanism by which they interact with integrin α5β1 through a Fn mediated-bridge. The FnBPA and FnBPB A domains engage in homophilic cell-cell interactions and promote biofilm formation and enhance platelet aggregation. In this review we update the current understanding of the structure and functional properties of FnBPs and emphasize the role they may have in the staphylococcal infections.
Collapse
Affiliation(s)
- Pietro Speziale
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, Pavia, Italy
| | - Giampiero Pietrocola
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, Pavia, Italy
| |
Collapse
|
27
|
Heinz A. Elastases and elastokines: elastin degradation and its significance in health and disease. Crit Rev Biochem Mol Biol 2020; 55:252-273. [PMID: 32530323 DOI: 10.1080/10409238.2020.1768208] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Elastin is an important protein of the extracellular matrix of higher vertebrates, which confers elasticity and resilience to various tissues and organs including lungs, skin, large blood vessels and ligaments. Owing to its unique structure, extensive cross-linking and durability, it does not undergo significant turnover in healthy tissues and has a half-life of more than 70 years. Elastin is not only a structural protein, influencing the architecture and biomechanical properties of the extracellular matrix, but also plays a vital role in various physiological processes. Bioactive elastin peptides termed elastokines - in particular those of the GXXPG motif - occur as a result of proteolytic degradation of elastin and its non-cross-linked precursor tropoelastin and display several biological activities. For instance, they promote angiogenesis or stimulate cell adhesion, chemotaxis, proliferation, protease activation and apoptosis. Elastin-degrading enzymes such as matrix metalloproteinases, serine proteases and cysteine proteases slowly damage elastin over the lifetime of an organism. The destruction of elastin and the biological processes triggered by elastokines favor the development and progression of various pathological conditions including emphysema, chronic obstructive pulmonary disease, atherosclerosis, metabolic syndrome and cancer. This review gives an overview on types of human elastases and their action on human elastin, including the formation, structure and biological activities of elastokines and their role in common biological processes and severe pathological conditions.
Collapse
Affiliation(s)
- Andrea Heinz
- Department of Pharmacy, LEO Foundation Center for Cutaneous Drug Delivery, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Cao Y, Lee BH, Irvine SA, Wong YS, Bianco Peled H, Venkatraman S. Inclusion of Cross-Linked Elastin in Gelatin/PEG Hydrogels Favourably Influences Fibroblast Phenotype. Polymers (Basel) 2020; 12:polym12030670. [PMID: 32192137 PMCID: PMC7183321 DOI: 10.3390/polym12030670] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 12/16/2022] Open
Abstract
The capacity of a biomaterial to innately modulate cell behavior while meeting the mechanical property requirements of the implant is a much sought-after goal within bioengineering. Here we covalently incorporate soluble elastin into a gelatin–poly (ethylene glycol) (PEG) hydrogel for three-dimensional (3D) cell encapsulation to achieve these properties. The inclusion of elastin into a previously optimized gelatin–PEG hydrogel was then evaluated for effects on entrapped fibroblasts, with the aim to assess the hydrogel as an extracellular matrix (ECM)-mimicking 3D microenvironment for cellular guidance. Soluble elastin was incorporated both physically and covalently into novel gelatin/elastin hybrid PEG hydrogels with the aim to harness the cellular interactivity and mechanical tunability of both elastin and gelatin. This design allowed us to assess the benefits of elastin-containing hydrogels in guiding fibroblast activity for evaluation as a potential dermal replacement. It was found that a gelatin–PEG hydrogel with covalently conjugated elastin, supported neonatal fibroblast viability, promoted their proliferation from 7.3% to 13.5% and guided their behavior. The expression of collagen alpha-1(COL1A1) and elastin in gelatin/elastin hybrid gels increased 16-fold and 6-fold compared to control sample at day 9, respectively. Moreover, cells can be loaded into the hydrogel precursor solution, deposited, and the matrix cross-linked without affecting the incorporated cells adversely, thus enabling a potential injectable system for dermal wound healing.
Collapse
Affiliation(s)
- Ye Cao
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; (Y.C.); (B.H.L.); (S.A.I.); (Y.S.W.)
- The Inter-Departmental Program for Biotechnology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Bae Hoon Lee
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; (Y.C.); (B.H.L.); (S.A.I.); (Y.S.W.)
| | - Scott Alexander Irvine
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; (Y.C.); (B.H.L.); (S.A.I.); (Y.S.W.)
| | - Yee Shan Wong
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; (Y.C.); (B.H.L.); (S.A.I.); (Y.S.W.)
| | - Havazelet Bianco Peled
- Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel
- Correspondence: (H.B.P.); (S.V.)
| | - Subramanian Venkatraman
- Subramanian Venkatraman, Materials Science and Engineering, National University of Singapore, Singapore 119077, Singapore
- Correspondence: (H.B.P.); (S.V.)
| |
Collapse
|
29
|
Wen Q, Mithieux SM, Weiss AS. Elastin Biomaterials in Dermal Repair. Trends Biotechnol 2020; 38:280-291. [DOI: 10.1016/j.tibtech.2019.08.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/28/2019] [Accepted: 08/27/2019] [Indexed: 02/05/2023]
|
30
|
Han WM, Jang YC, García AJ. The Extracellular Matrix and Cell–Biomaterial Interactions. Biomater Sci 2020. [DOI: 10.1016/b978-0-12-816137-1.00045-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
31
|
Abstract
Elastic fibers are found in the extracellular matrix (ECM) of tissues requiring resilience and depend on elasticity. Elastin and its degradation products have multiple roles in the oncologic process. In many malignancies, the remodeled ECM expresses high levels of the elastin protein which may have either positive or negative effects on tumor growth. Elastin cross-linking with other ECM components and the enzymes governing this process all have effects on tumorigenesis. Elastases, and specifically neutrophil elastase, are key drivers of invasion and metastasis and therefore are important targets for inhibition. Elastin degradation leads to the generation of bioactive fragments and elastin-derived peptides that further modulate tumor growth and spread. Interestingly, elastin-like peptides (ELP) and elastin-derived peptides (EDP) may also be utilized as nano-carriers to combat tumor growth. EDPs drive tumor development in a variety of ways, and specifically targeting EDPs and their binding proteins are major objectives for ongoing and future anti-cancer therapies. Research on both the direct anti-cancer activity and the drug delivery capabilities of ELPs is another area likely to result in novel therapeutic agents in the near future.
Collapse
|
32
|
Lu X, Han L, Golts E, Baradarian S, Kassab GS. Homologous and heterologous assessment of a novel biomaterial for venous patch. J Vasc Surg Venous Lymphat Disord 2019; 8:458-469.e1. [PMID: 31837973 DOI: 10.1016/j.jvsv.2019.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/11/2019] [Indexed: 01/29/2023]
Abstract
OBJECTIVE This study evaluated swine and bovine pulmonary visceral pleura (PVP) as a vascular patch. Venous patches are frequently used in surgery for repair or reconstruction of veins. Autologous patches are often limited by the number and dimension of donor tissue and can result in donor complications. Bovine pericardium is the most common heterologous patch used by vascular surgeons. Researchers, however, are continually seeking to improve heterologous and synthetic patches for improved outcome. METHODS The PVP was peeled from swine and bovine lungs and cross-linked with glutaraldehyde. After sterilization and rinsing, the PVP patches were implanted in the jugular vein (10 × 35 mm) of pigs and dogs. Patency was evaluated by ultrasound, and animals were euthanized at 2 and 4 months. Neoendothelium and neomedia were evaluated by histologic analysis. RESULTS The jugular vein patched by PVP in pigs and dogs remained patent at 2 and 4 months with no adhesions, inflammation, or aneurysm in the patches. The biomarkers of endothelial cells-factor VIII, platelet/endothelial cell adhesion molecule 1, and endothelial nitric oxide synthase-were detected in the neoendothelial cells. The expression of vascular smooth muscle cell (VSMC) α-actin was robust in the neomedia at 2 and 4 months. Neomedia composed of VSMCs developed to nearly double the thickness of adjacent jugular vein. The circumferential orientation of VSMCs in neomedia further increased in the 4-month group. CONCLUSIONS The cross-linked swine and bovine PVP patch has a nonthrombogenic surface that maintains patency. The PVP patch may overcome the pitfall of compliance mismatch of synthetic patches. The proliferation of vascular cells assembled in the neoendothelium and neomedia in the patches may support long-term patency.
Collapse
Affiliation(s)
- Xiao Lu
- Division of Cardiovascular Bioengineering, California Medical Innovations Institute, San Diego, Calif
| | - Ling Han
- Division of Cardiovascular Bioengineering, California Medical Innovations Institute, San Diego, Calif
| | - Eugene Golts
- Division of Cardiovascular Surgery, University of California San Diego, San Diego, Calif
| | - Sam Baradarian
- Division of Cardiovascular Surgery, Scripps Clinic, San Diego, Calif
| | - Ghassan S Kassab
- Division of Cardiovascular Bioengineering, California Medical Innovations Institute, San Diego, Calif.
| |
Collapse
|
33
|
Sequence variants of human tropoelastin affecting assembly, structural characteristics and functional properties of polymeric elastin in health and disease. Matrix Biol 2019; 84:68-80. [DOI: 10.1016/j.matbio.2019.06.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/23/2019] [Accepted: 06/25/2019] [Indexed: 12/19/2022]
|
34
|
Vindin H, Mithieux SM, Weiss AS. Elastin architecture. Matrix Biol 2019; 84:4-16. [DOI: 10.1016/j.matbio.2019.07.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 07/08/2019] [Accepted: 07/08/2019] [Indexed: 11/15/2022]
|
35
|
Hedtke T, Schräder CU, Heinz A, Hoehenwarter W, Brinckmann J, Groth T, Schmelzer CEH. A comprehensive map of human elastin cross-linking during elastogenesis. FEBS J 2019; 286:3594-3610. [PMID: 31102572 DOI: 10.1111/febs.14929] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/09/2019] [Accepted: 05/15/2019] [Indexed: 01/05/2023]
Abstract
Elastin is an essential structural protein in the extracellular matrix of vertebrates. It is the core component of elastic fibers, which enable connective tissues such as those of the skin, lungs or blood vessels to stretch and recoil. This function is provided by elastin's exceptional properties, which mainly derive from a unique covalent cross-linking between hydrophilic lysine-rich motifs of units of the monomeric precursor tropoelastin. To date, elastin's cross-linking is poorly investigated. Here, we purified elastin from human tissue and cleaved it into soluble peptides using proteases with different specificities. We then analyzed elastin's molecular structure by identifying unmodified residues, post-translational modifications and cross-linked peptides by high-resolution mass spectrometry and amino acid analysis. The data revealed the presence of multiple isoforms in parallel and a complex and heterogeneous molecular interconnection. We discovered that the same lysine residues in different monomers were simultaneously involved in various cross-link types or remained unmodified. Furthermore, both types of cross-linking domains, Lys-Pro and Lys-Ala domains, participate not only in bifunctional inter- but also in intra-domain cross-links. We elucidated the sequences of several desmosine-containing peptides and the contribution of distinct domains such as 6, 14 and 25. In contrast to earlier assumptions proposing that desmosine cross-links are formed solely between two domains, we elucidated the structure of a peptide that proves a desmosine formation with participation of three Lys-Ala domains. In summary, these results provide new and detailed insights into the cross-linking process, which takes place within and between human tropoelastin units in a stochastic manner.
Collapse
Affiliation(s)
- Tobias Hedtke
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany.,Biomedical Materials Group, Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Germany
| | - Christoph U Schräder
- Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Germany
| | - Andrea Heinz
- Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Germany.,Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | - Wolfgang Hoehenwarter
- Proteome Analytics Research Group, Leibniz Institute for Plant Biochemistry, Halle (Saale), Germany
| | - Jürgen Brinckmann
- Institute of Virology and Cell Biology & Department of Dermatology, University of Lübeck, Germany
| | - Thomas Groth
- Biomedical Materials Group, Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Germany
| | - Christian E H Schmelzer
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany.,Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Germany.,Institute of Applied Dermatopharmacy at the Martin Luther University Halle-Wittenberg (IADP), Germany
| |
Collapse
|
36
|
Almeida H, Domingues RMA, Mithieux SM, Pires RA, Gonçalves AI, Gómez-Florit M, Reis RL, Weiss AS, Gomes ME. Tropoelastin-Coated Tendon Biomimetic Scaffolds Promote Stem Cell Tenogenic Commitment and Deposition of Elastin-Rich Matrix. ACS APPLIED MATERIALS & INTERFACES 2019; 11:19830-19840. [PMID: 31088069 DOI: 10.1021/acsami.9b04616] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Tendon tissue engineering strategies that recreate the biophysical and biochemical native microenvironment have a greater potential to achieve regeneration. Here, we developed tendon biomimetic scaffolds using mechanically competent yarns of poly-ε-caprolactone, chitosan, and cellulose nanocrystals to recreate the inherent tendon hierarchy from a nano-to-macro scale. These were then coated with tropoelastin (TROPO) through polydopamine (PDA) linking, to mimic the native extracellular matrix (ECM) composition and elasticity. Both PDA and TROPO coatings decreased surface stiffness without masking the underlying substrate. We found that human adipose-derived stem cells (hASCs) seeded onto these TROPO biomimetic scaffolds more rapidly acquired their spindle-shape morphology and high aspect ratio characteristic of tenocytes. Immunocytochemistry shows that the PDA and TROPO-coated surfaces boosted differentiation of hASCs toward the tenogenic lineage, with sustained expression of the tendon-related markers scleraxis and tenomodulin up to 21 days of culture. Furthermore, these surfaces enabled the deposition of a tendon-like ECM, supported by the expression of collagens type I and III, tenascin, and decorin. Gene expression analysis revealed a downregulation of osteogenic and fibrosis markers in the presence of TROPO when compared with the control groups, suggesting proper ECM deposition. Remarkably, differentiated cells exposed to TROPO acquired an elastogenic profile due to the evident elastin synthesis and deposition, contributing to the formation of a more mimetic matrix in comparison with the PDA-coated and uncoated conditions. In summary, our biomimetic substrates combining biophysical and biological cues modulate stem cell behavior potentiating their long-term tenogenic commitment and the production of an elastin-rich ECM.
Collapse
Affiliation(s)
- Helena Almeida
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics , University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra , Barco, 4805-017 Guimarães , Portugal
- ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães , Portugal
| | - Rui M A Domingues
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics , University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra , Barco, 4805-017 Guimarães , Portugal
- ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães , Portugal
- The Discoveries Centre for Regenerative and Precision Medicine , Headquarters at University of Minho , Avepark , Barco, 4805-017 Guimarães , Portugal
| | | | - Ricardo A Pires
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics , University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra , Barco, 4805-017 Guimarães , Portugal
- ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães , Portugal
- The Discoveries Centre for Regenerative and Precision Medicine , Headquarters at University of Minho , Avepark , Barco, 4805-017 Guimarães , Portugal
| | - Ana I Gonçalves
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics , University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra , Barco, 4805-017 Guimarães , Portugal
- ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães , Portugal
| | - Manuel Gómez-Florit
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics , University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra , Barco, 4805-017 Guimarães , Portugal
- ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães , Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics , University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra , Barco, 4805-017 Guimarães , Portugal
- ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães , Portugal
- The Discoveries Centre for Regenerative and Precision Medicine , Headquarters at University of Minho , Avepark , Barco, 4805-017 Guimarães , Portugal
| | | | - Manuela E Gomes
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics , University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra , Barco, 4805-017 Guimarães , Portugal
- ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães , Portugal
- The Discoveries Centre for Regenerative and Precision Medicine , Headquarters at University of Minho , Avepark , Barco, 4805-017 Guimarães , Portugal
| |
Collapse
|
37
|
Almouemen N, Kelly HM, O'Leary C. Tissue Engineering: Understanding the Role of Biomaterials and Biophysical Forces on Cell Functionality Through Computational and Structural Biotechnology Analytical Methods. Comput Struct Biotechnol J 2019; 17:591-598. [PMID: 31080565 PMCID: PMC6502738 DOI: 10.1016/j.csbj.2019.04.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 03/26/2019] [Accepted: 04/13/2019] [Indexed: 12/13/2022] Open
Abstract
Within the past 25 years, tissue engineering (TE) has grown enormously as a science and as an industry. Although classically concerned with the recapitulation of tissue and organ formation in our body for regenerative medicine, the evolution of TE research is intertwined with progress in other fields through the examination of cell function and behaviour in isolated biomimetic microenvironments. As such, TE applications now extend beyond the field of tissue regeneration research, operating as a platform for modifiable, physiologically-representative in vitro models with the potential to improve the translation of novel therapeutics into the clinic through a more informed understanding of the relevant molecular biology, structural biology, anatomy, and physiology. By virtue of their biomimicry, TE constructs incorporate features of extracellular macrostructure, molecular adhesive moieties, and biomechanical properties, converging with computational and structural biotechnology advances. Accordingly, this mini-review serves to contextualise TE for the computational and structural biotechnology reader and provides an outlook on how the disciplines overlap with respect to relevant advanced analytical applications.
Collapse
Affiliation(s)
- Nour Almouemen
- School of Pharmacy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
- Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland
| | - Helena M. Kelly
- School of Pharmacy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
- Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Cian O'Leary
- School of Pharmacy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
- Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
38
|
Ozsvar J, Tarakanova A, Wang R, Buehler MJ, Weiss AS. Allysine modifications perturb tropoelastin structure and mobility on a local and global scale. Matrix Biol Plus 2019; 2:100002. [PMID: 33543005 PMCID: PMC7852328 DOI: 10.1016/j.mbplus.2019.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/10/2019] [Accepted: 03/10/2019] [Indexed: 11/20/2022] Open
Abstract
Elastin provides elastic tissues with resilience through stretch and recoil cycles, and is primarily made of its extensively cross-linked monomer, tropoelastin. Here, we leverage the recently published full atomistic model of tropoelastin to assess how allysine modifications, which are essential to cross-linking, contribute to the dynamics and structural changes that occur in tropoelastin in the context of elastin assembly. We used replica exchange molecular dynamics to generate structural ensembles of allysine containing tropoelastin. We conducted principal component analysis on these ensembles and found that the molecule departs from the canonical structural ensemble. Furthermore, we showed that, while the canonical scissors-twist movement was retained, new movements emerged that deviated from those of the wild type protein, providing evidence for the involvement of a variety of molecular motions in elastin assembly. Additionally, we highlighted secondary structural changes and linked these perturbations to the longevity of specific salt bridges. We propose a model where allysines in tropoelastin contribute to hierarchical elastin assembly through global and local perturbations to molecular structure and dynamics. converting lysine to allysine by lysyl oxidases is needed to generate crosslinks between tropoelastin molecules in order to make elastin structural changes in the intact tropoelastin molecule ensue where modified tropoelastin molecules structurally depart from the canonical ensemble new molecular motions deviate from those of unmodified tropoelastin persistence times of specific salt bridges contribute to these perturbations allysines in tropoelastin contribute to hierarchical elastin assembly through global and local perturbations to molecular structure and dynamics
Collapse
Key Words
- 5ALK, tropoelastin containing 5 allysine residues
- ALK353, tropoelastin containing allysine at residue 353
- ALK353, tropoelastin containing allysine at residue 507
- ALL, allysine aldol
- ANM, anisotropic network model
- Assembly
- ECM, extracellular matrix
- Elastin
- LNL, lysinonorleucine
- MD, molecular dynamics
- Molecular dynamics
- NMA, normal mode analysis
- PCA, principal component analysis
- REMD, replica exchange molecular dynamics
- RMSD, root mean square deviation
- Replica exchange molecular dynamics
- SASA, solvent accessible surface area
- WT, wild type tropoelastin
Collapse
Affiliation(s)
- Jazmin Ozsvar
- Charles Perkins Centre, the University of Sydney, 2006 Sydney, NSW, Australia.,School of Life and Environmental Sciences, The University of Sydney, 2006 Sydney, NSW, Australia.,Cell Therapy Manufacturing Cooperative Research Centre, Adelaide, 5000, SA, Australia
| | - Anna Tarakanova
- Laboratory for Atomistic and Molecular Mechanics (LAMM), Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Richard Wang
- Charles Perkins Centre, the University of Sydney, 2006 Sydney, NSW, Australia.,School of Life and Environmental Sciences, The University of Sydney, 2006 Sydney, NSW, Australia
| | - Markus J Buehler
- Laboratory for Atomistic and Molecular Mechanics (LAMM), Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anthony S Weiss
- Charles Perkins Centre, the University of Sydney, 2006 Sydney, NSW, Australia.,School of Life and Environmental Sciences, The University of Sydney, 2006 Sydney, NSW, Australia.,Cell Therapy Manufacturing Cooperative Research Centre, Adelaide, 5000, SA, Australia.,Bosch Institute, The University of Sydney, 2006 Sydney, NSW, Australia.,Sydney Nano Institute, The University of Sydney, 2006 Sydney, NSW, Australia
| |
Collapse
|
39
|
Bax DV, Smalley HE, Farndale RW, Best SM, Cameron RE. Cellular response to collagen-elastin composite materials. Acta Biomater 2019; 86:158-170. [PMID: 30586647 DOI: 10.1016/j.actbio.2018.12.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/03/2018] [Accepted: 12/21/2018] [Indexed: 10/27/2022]
Abstract
Collagen is used extensively in tissue engineering due to its biocompatibility, near-universal tissue distribution, low cost and purity. However, native tissues are composites that include diverse extracellular matrix components, which influence strongly their mechanical and biological properties. Here, we provide important new findings on the differential regulation, by collagen and elastin, of the bio-response to the composite material. Soluble and insoluble elastin had differing effects on the stiffness and failure strength of the composite films. We established that Rugli cells bind elastin via EDTA-sensitive receptors, whilst HT1080 cells do not. These cells allowed us to probe the contribution of collagen alone (HT1080) and collagen plus elastin (Rugli) to the cellular response. In the presence of elastin, Rugli cell attachment, spreading and proliferation increased, presumably through elastin-binding receptors. By comparison, the attachment and spreading of HT1080 cells was modified by elastin inclusion, but without affecting their proliferation, indicating indirect modulation by elastin of the response of cells to collagen. These new insights highlight that access to elastin dominates the cellular response when elastin-binding receptors are present. In the absence of these receptors, modification of the collagen component and/or physical properties dictate the cellular response. Therefore, we can attribute the contribution of each constituent on the ultimate bioactivity of heterogeneous collagen-composite materials, permitting informed, systematic biomaterials design. STATEMENT OF SIGNIFICANCE: In recent years there has been a desire to replicate the complex extracellular matrix composition of tissues more closely, necessitating the need for composite protein-based materials. In this case both the physical and biochemical properties are altered with the addition of each component, with potential consequences on the cell. To date, the different contributions of each component have not been deconvolved, and instead the cell response to the scaffold as a whole has been observed. Instead, here, we have used specific cell lines, that are sensitive to specific components of an elastin-collagen composite, to resolve the bio-activity of each protein. This has shown that elastin-induced alteration of the collagen component can modulate early stage cell behaviour. By comparison the elastin component directly alters the cell response over the short and long term, but only where appropriate receptors are present on the cell. Due to the widespread use of collagen and elastin, we feel that this data permits, for the first time, the ability to systematically design collagen-composite materials to promote desired cell behaviour with associated advantages for biomaterials fabrication.
Collapse
|
40
|
Tumour cell blebbing and extracellular vesicle shedding: key role of matrikines and ribosomal protein SA. Br J Cancer 2019; 120:453-465. [PMID: 30739912 PMCID: PMC6461924 DOI: 10.1038/s41416-019-0382-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 12/06/2018] [Accepted: 12/20/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Carcinogenesis occurs in elastin-rich tissues and leads to local inflammation and elastolytic proteinase release. This contributes to bioactive matrix fragment (Matrikine) accumulation like elastin degradation products (EDP) stimulating tumour cell invasive and metastatic properties. We previously demonstrate that EDPs exert protumoural activities through Hsp90 secretion to stabilised extracellular proteinases. METHODS EDP influence on cancer cell blebbing and extracellular vesicle shedding were examined with a videomicroscope coupled with confocal Yokogawa spinning disk, by transmission electron microscopy, scanning electron microscopy and confocal microscopy. The ribosomal protein SA (RPSA) elastin receptor was identified after affinity chromatography by western blotting and cell immunolocalisation. mRNA expression was studied using real-time PCR. SiRNA were used to confirm the essential role of RPSA. RESULTS We demonstrate that extracellular matrix degradation products like EDPs induce tumour amoeboid phenotype with cell membrane blebbing and shedding of extracellular vesicle containing Hsp90 and proteinases in the extracellular space. EDPs influence intracellular calcium influx and cytoskeleton reorganisation. Among matrikines, VGVAPG and AGVPGLGVG peptides reproduced EDP effects through RPSA binding. CONCLUSIONS Our data suggests that matrikines induce cancer cell blebbing and extracellular vesicle release through RPSA binding, favouring dissemination, cell-to-cell communication and growth of cancer cells in metastatic sites.
Collapse
|
41
|
Deng S, Zhang H, Han W, Guo C, Deng C. Transforming Growth Factor-β-Neutralizing Antibodies Improve Alveolarization in the Oxygen-Exposed Newborn Mouse Lung. J Interferon Cytokine Res 2019; 39:106-116. [PMID: 30657417 DOI: 10.1089/jir.2018.0080] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Abnormal alveolar formation and excessive disordered elastin accumulation are key pathological features in bronchopulmonary dysplasia. Transforming growth factor (TGF)-β is an important regulator of the extracellular matrix in the developing lung. To determine if increased TGF-β would injure alveolar development by activating TGF-β signaling and by influencing the expression of elastogenesis-related protein, we performed intraperitoneal injection of newborn mice with the TGF-β-neutralizing antibody 1D11 and observed whether 1D11 had a protective role in the oxygen (O2)-exposed newborn mouse lung. The newborn mice were exposed to 85% O2 for 14 and 21 days. 1D11 was administered by intraperitoneal injection every day from postnatal days 3 to 20. Alveolar morphology was assessed by hematoxylin and eosin staining. The expression and distribution of elastin were evaluated by immunohistochemistry. The level of TGF-β signaling-related proteins were measured by immunohistochemistry, enzyme-linked immunosorbent assay, and Western blot. The expression levels of elastogenesis-related proteins, including tropoelastin, fibulin-5, and neutrophil elastase (NE), which participate in the synthesis, assembly, and degradation of elastin, were detected by real-time PCR and Western blot. In this research, impaired alveolar development and elastin deposition as well as the excessive activation of TGF-β signaling were observed in the newborn mouse lung exposed to hyperoxia. 1D11 improved alveolarization as well as the distribution of elastin in the newborn lung with hyperoxia exposure. The expression levels of tropoelastin, fibulin-5, and NE, which are important components of elastogenesis, were decreased by treatment with 1D11 in the injured newborn lung. These data demonstrate that 1D11 improved alveolarization by blocking the TGF-β signaling pathway and by reducing the abnormal expression of elastogenesis-related proteins in the O2-exposed newborn mouse lung. 1D11 may become a new therapeutic method to prevent the development of bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Sijun Deng
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,2 China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,3 Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Han Zhang
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,2 China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,3 Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Wenli Han
- 2 China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,3 Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China.,4 Laboratory Animal Center, Chongqing Medical University, Chongqing, China
| | - Chunbao Guo
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,2 China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,3 Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China.,5 Department of Hepatology and Liver Transplantation Center, Children's Hospital, Chongqing Medical University, Chongqing, China
| | - Chun Deng
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,2 China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,3 Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
42
|
Soluble matrix protein is a potent modulator of mesenchymal stem cell performance. Proc Natl Acad Sci U S A 2019; 116:2042-2051. [PMID: 30659152 DOI: 10.1073/pnas.1812951116] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We challenge the conventional designation of structural matrix proteins primarily as supporting scaffolds for resident cells. The extracellular matrix protein tropoelastin is classically regarded as a structural component that confers mechanical strength and resilience to tissues subject to repetitive elastic deformation. Here we describe how tropoelastin inherently induces a range of biological responses, even in cells not typically associated with elastic tissues and in a manner unexpected of typical substrate-dependent matrix proteins. We show that tropoelastin alone drives mesenchymal stem cell (MSC) proliferation and phenotypic maintenance, akin to the synergistic effects of potent growth factors such as insulin-like growth factor 1 and basic fibroblast growth factor. In addition, tropoelastin functionally surpasses these growth factors, as well as fibronectin, in allowing substantial media serum reduction without loss of proliferative potential. We further demonstrate that tropoelastin elicits strong mitogenic and cell-attractive responses, both as an immobilized substrate and as a soluble additive, via direct interactions with cell surface integrins αvβ3 and αvβ5. This duality of action converges the long-held mechanistic dichotomy between adhesive matrix proteins and soluble growth factors and uncovers the powerful, untapped potential of tropoelastin for clinical MSC expansion and therapeutic MSC recruitment. We propose that the potent, growth factor-like mitogenic and motogenic abilities of tropoelastin are biologically rooted in the need for rapid stem cell homing and proliferation during early development and/or wound repair.
Collapse
|
43
|
Magaz A, Faroni A, Gough JE, Reid AJ, Li X, Blaker JJ. Bioactive Silk-Based Nerve Guidance Conduits for Augmenting Peripheral Nerve Repair. Adv Healthc Mater 2018; 7:e1800308. [PMID: 30260575 DOI: 10.1002/adhm.201800308] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/22/2018] [Indexed: 02/03/2023]
Abstract
Repair of peripheral nerve injuries depends upon complex biology stemming from the manifold and challenging injury-healing processes of the peripheral nervous system. While surgical treatment options are available, they tend to be characterized by poor clinical outcomes for the injured patients. This is particularly apparent in the clinical management of a nerve gap whereby nerve autograft remains the best clinical option despite numerous limitations; in addition, effective repair becomes progressively more difficult with larger gaps. Nerve conduit strategies based on tissue engineering approaches and the use of silk as scaffolding material have attracted much attention in recent years to overcome these limitations and meet the clinical demand of large gap nerve repair. This review examines the scientific advances made with silk-based conduits for peripheral nerve repair. The focus is on enhancing bioactivity of the conduits in terms of physical guidance cues, inner wall and lumen modification, and imbuing novel conductive functionalities.
Collapse
Affiliation(s)
- Adrián Magaz
- Bio‐Active Materials GroupSchool of MaterialsMSS TowerThe University of Manchester Manchester M13 9PL UK
- Institute of Materials Research and Engineering (IMRE)Agency for Science Technology and Research (A*STAR) 2 Fusionopolis, Way, Innovis #08‐03 Singapore 138634 Singapore
| | - Alessandro Faroni
- Blond McIndoe LaboratoriesDivision of Cell Matrix Biology and Regenerative MedicineSchool of Biological SciencesFaculty of Biology, Medicine and HealthThe University of ManchesterManchester Academic Health Science Centre Manchester M13 9PL UK
| | - Julie E. Gough
- School of MaterialsThe University of Manchester Manchester M13 9PL UK
| | - Adam J. Reid
- Blond McIndoe LaboratoriesDivision of Cell Matrix Biology and Regenerative MedicineSchool of Biological SciencesFaculty of Biology, Medicine and HealthThe University of ManchesterManchester Academic Health Science Centre Manchester M13 9PL UK
- Department of Plastic Surgery and BurnsWythenshawe HospitalManchester University NHS Foundation TrustManchester Academic Health Science Centre Manchester M23 9LT UK
| | - Xu Li
- Institute of Materials Research and Engineering (IMRE)Agency for Science Technology and Research (A*STAR) 2 Fusionopolis, Way, Innovis #08‐03 Singapore 138634 Singapore
| | - Jonny J. Blaker
- Bio‐Active Materials GroupSchool of MaterialsMSS TowerThe University of Manchester Manchester M13 9PL UK
- School of MaterialsThe University of Manchester Manchester M13 9PL UK
| |
Collapse
|
44
|
Muiznieks LD, Sharpe S, Pomès R, Keeley FW. Role of Liquid–Liquid Phase Separation in Assembly of Elastin and Other Extracellular Matrix Proteins. J Mol Biol 2018; 430:4741-4753. [DOI: 10.1016/j.jmb.2018.06.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/29/2018] [Accepted: 06/01/2018] [Indexed: 10/14/2022]
|
45
|
Staiculescu MC, Cocciolone AJ, Procknow JD, Kim J, Wagenseil JE. Comparative gene array analyses of severe elastic fiber defects in late embryonic and newborn mouse aorta. Physiol Genomics 2018; 50:988-1001. [PMID: 30312140 PMCID: PMC6293116 DOI: 10.1152/physiolgenomics.00080.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/09/2018] [Accepted: 10/09/2018] [Indexed: 01/17/2023] Open
Abstract
Elastic fibers provide reversible elasticity to the large arteries and are assembled during development when hemodynamic forces are increasing. Mutations in elastic fiber genes are associated with cardiovascular disease. Mice lacking expression of the elastic fiber genes elastin ( Eln-/-), fibulin-4 ( Efemp2-/-), or lysyl oxidase ( Lox-/-) die at birth with severe cardiovascular malformations. All three genetic knockout models have elastic fiber defects, aortic wall thickening, and arterial tortuosity. However, Eln-/- mice develop arterial stenoses, while Efemp2-/- and Lox-/- mice develop ascending aortic aneurysms. We performed comparative gene array analyses of these three genetic models for two vascular locations and developmental stages to determine differentially expressed genes and pathways that may explain the common and divergent phenotypes. We first examined arterial morphology and wall structure in newborn mice to confirm that the lack of elastin, fibulin-4, or lysyl oxidase expression provided the expected phenotypes. We then compared gene expression levels for each genetic model by three-way ANOVA for genotype, vascular location, and developmental stage. We found three genes upregulated by genotype in all three models, Col8a1, Igfbp2, and Thbs1, indicative of a common response to severe elastic fiber defects in developing mouse aorta. Genes that are differentially regulated by vascular location or developmental stage in all three models suggest mechanisms for location or stage-specific disease pathology. Comparison of signaling pathways enriched in all three models shows upregulation of integrins and matrix proteins involved in early wound healing, but not of mature matrix molecules such as elastic fiber proteins or fibrillar collagens.
Collapse
Affiliation(s)
| | - Austin J Cocciolone
- Department of Biomedical Engineering, Washington University , St. Louis, Missouri
| | - Jesse D Procknow
- Department of Mechanical Engineering and Materials Science, Washington University , St. Louis, Missouri
| | - Jungsil Kim
- Department of Mechanical Engineering and Materials Science, Washington University , St. Louis, Missouri
| | - Jessica E Wagenseil
- Department of Mechanical Engineering and Materials Science, Washington University , St. Louis, Missouri
| |
Collapse
|
46
|
Abascal NC, Regan L. The past, present and future of protein-based materials. Open Biol 2018; 8:180113. [PMID: 30381364 PMCID: PMC6223211 DOI: 10.1098/rsob.180113] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/05/2018] [Indexed: 11/23/2022] Open
Abstract
Protein-based materials are finding new uses and applications after millennia of impacting the daily life of humans. Some of the earliest uses of protein-based materials are still evident in silk and wool textiles and leather goods. Today, even as silks, wools and leathers are still be used in traditional ways, these proteins are now seen as promising materials for biomaterials, vehicles of drug delivery and components of high-tech fabrics. With the advent of biosynthetic methods and streamlined means of protein purification, protein-based materials-recombinant and otherwise-are being used in a host of applications at the cutting edge of medicine, electronics, materials science and even fashion. This commentary aims to discuss a handful of these applications while taking a critical look at where protein-based materials may be used in the future.
Collapse
Affiliation(s)
- Nadia C Abascal
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Lynne Regan
- Department of Interdisciplinary Science, Centre for Synthetic and Systems Biology, Institute for Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
47
|
Tarakanova A, Yeo GC, Baldock C, Weiss AS, Buehler MJ. Tropoelastin is a Flexible Molecule that Retains its Canonical Shape. Macromol Biosci 2018; 19:e1800250. [DOI: 10.1002/mabi.201800250] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/03/2018] [Indexed: 11/07/2022]
Affiliation(s)
- Anna Tarakanova
- Laboratory for Atomistic and Molecular Mechanics Department of Civil and Environmental Engineering Massachusetts Institute of Technology 02139 Cambridge MA USA
| | - Giselle C. Yeo
- School of Life and Environmental Sciences The University of Sydney 2006 Sydney NSW Australia
- Charles Perkins Centre The University of Sydney 2006 Sydney NSW Australia
| | - Clair Baldock
- Wellcome Trust Centre for Cell‐Matrix Research Division of Cell Matrix Biology and Regenerative Medicine School of Biological Sciences Manchester Academic Health Science Centre The University of Manchester M13 9PL Manchester UK
| | - Anthony S. Weiss
- School of Life and Environmental Sciences The University of Sydney 2006 Sydney NSW Australia
- Charles Perkins Centre The University of Sydney 2006 Sydney NSW Australia
- Bosch Institute The University of Sydney 2006 Sydney NSW Australia
| | - Markus J. Buehler
- Laboratory for Atomistic and Molecular Mechanics Department of Civil and Environmental Engineering Massachusetts Institute of Technology 02139 Cambridge MA USA
| |
Collapse
|
48
|
Yeo GC, Kosobrodova E, Kondyurin A, McKenzie DR, Bilek MM, Weiss AS. Plasma‐Activated Substrate with a Tropoelastin Anchor for the Maintenance and Delivery of Multipotent Adult Progenitor Cells. Macromol Biosci 2018; 19:e1800233. [DOI: 10.1002/mabi.201800233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/19/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Giselle C. Yeo
- Charles Perkins CentreUniversity of Sydney NSW 2006 Australia
- School of Life and Environmental SciencesUniversity of Sydney NSW 2006 Australia
- Bosch InstituteUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| | - Elena Kosobrodova
- School of PhysicsUniversity of Sydney NSW 2006 Australia
- School of AerospaceMechanical and Mechatronic EngineeringUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| | - Alexey Kondyurin
- School of PhysicsUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| | - David R. McKenzie
- School of PhysicsUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| | - Marcela M. Bilek
- Charles Perkins CentreUniversity of Sydney NSW 2006 Australia
- School of PhysicsUniversity of Sydney NSW 2006 Australia
- School of AerospaceMechanical and Mechatronic EngineeringUniversity of Sydney NSW 2006 Australia
- Australian Institute of Nanoscale Science and TechnologyUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| | - Anthony S. Weiss
- Charles Perkins CentreUniversity of Sydney NSW 2006 Australia
- School of Life and Environmental SciencesUniversity of Sydney NSW 2006 Australia
- Bosch InstituteUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| |
Collapse
|
49
|
Soucy JR, Shirzaei Sani E, Portillo Lara R, Diaz D, Dias F, Weiss AS, Koppes AN, Koppes RA, Annabi N. Photocrosslinkable Gelatin/Tropoelastin Hydrogel Adhesives for Peripheral Nerve Repair. Tissue Eng Part A 2018; 24:1393-1405. [PMID: 29580168 PMCID: PMC6150941 DOI: 10.1089/ten.tea.2017.0502] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/21/2018] [Indexed: 12/29/2022] Open
Abstract
Suturing peripheral nerve transections is the predominant therapeutic strategy for nerve repair. However, the use of sutures leads to scar tissue formation, hinders nerve regeneration, and prevents functional recovery. Fibrin-based adhesives have been widely used for nerve reconstruction, but their limited adhesive and mechanical strength and inability to promote nerve regeneration hamper their utility as a stand-alone intervention. To overcome these challenges, we engineered composite hydrogels that are neurosupportive and possess strong tissue adhesion. These composites were synthesized by photocrosslinking two naturally derived polymers, gelatin-methacryloyl (GelMA) and methacryloyl-substituted tropoelastin (MeTro). The engineered materials exhibited tunable mechanical properties by varying the GelMA/MeTro ratio. In addition, GelMA/MeTro hydrogels exhibited 15-fold higher adhesive strength to nerve tissue ex vivo compared to fibrin control. Furthermore, the composites were shown to support Schwann cell (SC) viability and proliferation, as well as neurite extension and glial cell participation in vitro, which are essential cellular components for nerve regeneration. Finally, subcutaneously implanted GelMA/MeTro hydrogels exhibited slower degradation in vivo compared with pure GelMA, indicating its potential to support the growth of slowly regenerating nerves. Thus, GelMA/MeTro composites may be used as clinically relevant biomaterials to regenerate nerves and reduce the need for microsurgical suturing during nerve reconstruction.
Collapse
Affiliation(s)
- Jonathan R. Soucy
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
| | - Ehsan Shirzaei Sani
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
| | - Roberto Portillo Lara
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
- Tecnológico de Monterrey, Escuela de IngenierÍa y Ciencias, Zapopan, JAL, Mexico
| | - David Diaz
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
| | - Felipe Dias
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
| | - Anthony S. Weiss
- Charles Perkins Centre, School of Life and Environmental Sciences and Bosch Institute, University of Sydney, Sydney, Australia
| | - Abigail N. Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
- Department of Biology, Northeastern University, Boston, Massachusetts
| | - Ryan A. Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
| | - Nasim Annabi
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
50
|
Schräder CU, Heinz A, Majovsky P, Karaman Mayack B, Brinckmann J, Sippl W, Schmelzer CEH. Elastin is heterogeneously cross-linked. J Biol Chem 2018; 293:15107-15119. [PMID: 30108173 DOI: 10.1074/jbc.ra118.004322] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/12/2018] [Indexed: 01/30/2023] Open
Abstract
Elastin is an essential vertebrate protein responsible for the elasticity of force-bearing tissues such as those of the lungs, blood vessels, and skin. One of the key features required for the exceptional properties of this durable biopolymer is the extensive covalent cross-linking between domains of its monomer molecule tropoelastin. To date, elastin's exact molecular assembly and mechanical properties are poorly understood. Here, using bovine elastin, we investigated the different types of cross-links in mature elastin to gain insight into its structure. We purified and proteolytically cleaved elastin from a single tissue sample into soluble cross-linked and noncross-linked peptides that we studied by high-resolution MS. This analysis enabled the elucidation of cross-links and other elastin modifications. We found that the lysine residues within the tropoelastin sequence were simultaneously unmodified and involved in various types of cross-links with different other domains. The Lys-Pro domains were almost exclusively linked via lysinonorleucine, whereas Lys-Ala domains were found to be cross-linked via lysinonorleucine, allysine aldol, and desmosine. Unexpectedly, we identified a high number of intramolecular cross-links between lysine residues in close proximity. In summary, we show on the molecular level that elastin formation involves random cross-linking of tropoelastin monomers resulting in an unordered network, an unexpected finding compared with previous assumptions of an overall beaded structure.
Collapse
Affiliation(s)
- Christoph U Schräder
- From the Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Andrea Heinz
- From the Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany.,the Department of Pharmacy, University of Copenhagen, Copenhagen 2100, Denmark
| | - Petra Majovsky
- the Proteome Analytics Research Group, Leibniz Institute for Plant Biochemistry, Halle (Saale) 06120, Germany
| | - Berin Karaman Mayack
- From the Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Jürgen Brinckmann
- the Institute of Virology and Cell Biology, Department of Dermatology, University of Lübeck, Lübeck 23538, Germany, and
| | - Wolfgang Sippl
- From the Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Christian E H Schmelzer
- From the Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany, .,the Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale) 06120, Germany
| |
Collapse
|