1
|
Wang S, Li F, Feng X, Feng M, Niu X, Jiang X, Chen W, Bai R. Promoting collagen synthesis: a viable strategy to combat skin ageing. J Enzyme Inhib Med Chem 2025; 40:2488821. [PMID: 40213810 PMCID: PMC11995770 DOI: 10.1080/14756366.2025.2488821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
Skin ageing is a complex physiological process primarily characterised by the deepening of wrinkles and the sagging of the skin. Collagen is essential for maintaining skin elasticity and firmness. As skin ages, it experiences structural and functional changes in collagen, including a decrease in collagen synthesis and an increase in collagen hydrolysis. Thus, promoting collagen synthesis represents a practical strategy for mitigating skin ageing. This review systematically described the functions, classifications and biosynthesis process of collagen, as well as its role in skin ageing. Additionally, the major signalling pathways and targets associated with collagen synthesis were also discussed. More importantly, the review provided a detailed summary of natural products with collagen synthesis-promoting effects and highlighted small molecule compounds with potential anti-ageing activity, especially PPARδ agonists. The relevant content offers potential targets and lead compounds for the development of anti-skin ageing therapies.
Collapse
Affiliation(s)
- Shan Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
| | - Feifan Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
| | - Xilong Feng
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
| | - Meiling Feng
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
| | - Xiaotian Niu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
| | - Xiaoying Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
| | - Wenchao Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
| | - Renren Bai
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
| |
Collapse
|
2
|
Srinivasan S, Sherwood DR. The life cycle of type IV collagen. Matrix Biol 2025; 139:14-28. [PMID: 40306374 PMCID: PMC12146070 DOI: 10.1016/j.matbio.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/21/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Type IV collagen is a large triple helical molecule that forms a covalently cross-linked network within basement membranes (BMs). Type IV collagen networks play key roles in mechanically supporting tissues, shaping organs, filtering blood, and cell signaling. To ensure tissue health and function, all aspects of the type IV collagen life cycle must be carried out accurately. However, the large triple helical structure and complex life-cycle of type IV collagen, poses many challenges to cells and tissues. Type IV collagen predominantly forms heterotrimers and to ensure proper construction, expression of the distinct α-chains that comprise a heterotrimer needs tight regulation. The α-chains must also be accurately modified by several enzymes, some of which are specific to collagens, to build and stabilize the triple helical trimer. In addition, type IV collagen is exceptionally long (400 nm) and thus the packaging and trafficking of the triple helical trimer from the ER to the Golgi must be modified to accommodate the large type IV collagen molecule. During ER-to-Golgi trafficking, as well as during secretion and transport in the extracellular space, type IV collagen also associates with specific chaperone molecules that maintain the structure and solubility of collagen IV. Type IV collagen trimers are then delivered to BMs from local and distant sources where they are integrated into BMs by interactions with cell surface receptors and many diverse BM resident proteins. Within BMs type IV collagen self-associates into a network and is crosslinked by BM resident enzymes. Finally, homeostatic type IV collagen levels in BMs are maintained by poorly understood mechanisms involving proteolysis and endocytosis. Here, we provide an overview of the life cycle of collagen IV, highlighting unique mechanisms and poorly understood aspects of type IV collagen regulation.
Collapse
Affiliation(s)
- Sandhya Srinivasan
- Department of Biology, Duke University, 130 Science Drive, Box 90338, Durham, NC 27708, USA
| | - David R Sherwood
- Department of Biology, Duke University, 130 Science Drive, Box 90338, Durham, NC 27708, USA.
| |
Collapse
|
3
|
Srinivasan S, Ramos-Lewis W, Morais MR, Chi Q, Soh AW, Williams E, Lennon R, Sherwood DR. A collagen IV fluorophore knock-in toolkit reveals trimer diversity in C. elegans basement membranes. J Cell Biol 2025; 224:e202412118. [PMID: 40100062 PMCID: PMC11917169 DOI: 10.1083/jcb.202412118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025] Open
Abstract
The type IV collagen triple helix, composed of three ⍺-chains, is a core basement membrane (BM) component that assembles into a network within BMs. Endogenous tagging of all ⍺-chains with genetically encoded fluorophores has remained elusive, limiting our understanding of this crucial BM component. Through genome editing, we show that the C termini of the C. elegans type IV collagen ⍺-chains EMB-9 and LET-2 can be fused to a variety of fluorophores to create a strain toolkit with wild-type health. Using quantitative imaging, our results suggest a preference for LET-2-LET-2-EMB-9 trimer construction, but also tissue-specific flexibility in trimers assembled driven by differences in ⍺-chain expression levels. By tagging emb-9 and let-2 mutants that model human Gould syndrome, a complex multitissue disorder, we further discover defects in extracellular accumulation and turnover that might help explain disease pathology. Together, our findings identify a permissive tagging site in C. elegans that will allow diverse studies on type IV collagen regulation and function in animals.
Collapse
Affiliation(s)
| | | | - Mychel R.P.T. Morais
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Qiuyi Chi
- Department of Biology, Duke University, Durham, NC, USA
| | - Adam W.J. Soh
- Department of Biology, Duke University, Durham, NC, USA
| | - Emily Williams
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Rachel Lennon
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | | |
Collapse
|
4
|
Wang J, Xiang ZX, Luan MF, Gong JS, Su C, Li H, Xu ZH, Shi JS. High-level secretory expression of recombinant type III human-like collagen α1 in Pichia pastoris via multilevel systematic optimization. Int J Biol Macromol 2025; 313:144270. [PMID: 40381768 DOI: 10.1016/j.ijbiomac.2025.144270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Collagen is the main component that makes up the internal structure of animals and is extensively used in several industrial fields including food, materials, chemicals, and pharmaceuticals. Despite the variety of preparation methods available, there is significant potential for enhancing the yield of recombinant collagen produced through engineered Pichia pastoris (P. pastoris). Increasing the copy number of the recombinant type III human collagen α1 (hlCOLIII) gene to improve the level of expression of the recombinant protein and co-expression of molecular chaperones to alleviate the resulting endoplasmic reticulum stress further promotes hlCOLIII secretion. By optimizing transcription driven by the AOX1 promoter and improving translation efficiency, a strain of P. pastoris expressing hlCOLIII efficiently was constructed, achieving a yield of 10.3 g/L in a 5 L fermenter. Further, hlCOLIII demonstrated notable antioxidant capacity and performed well in bioactivity analyses, including cell proliferation, migration, and adhesion. This study lays a solid foundation for the scalable industrial production of recombinant collagen and opens new avenues for its exploration in advanced biomedical materials.
Collapse
Affiliation(s)
- Jing Wang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, School of Biotechnology, Jiangnan University, Wuxi 214122, PR China
| | - Zhi-Xiang Xiang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China
| | - Meng-Fan Luan
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China
| | - Jin-Song Gong
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China.
| | - Chang Su
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China
| | - Heng Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China
| | - Zheng-Hong Xu
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, School of Biotechnology, Jiangnan University, Wuxi 214122, PR China; Innovation Center for Advanced Brewing Science and Technology, College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China
| | - Jin-Song Shi
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China.
| |
Collapse
|
5
|
Li J, Yin F, Wang J, Du H, Xu F, Meskers S, Li Y, Wijker S, Peng Y, Bellan R, Vantomme G, Song J, Liu CS, Meijer EW. Self-Regulating Hydrogel with Reversible Optical Activity in Its Gel-to-Gel Transformation. J Am Chem Soc 2025; 147:17361-17371. [PMID: 40344185 PMCID: PMC12100726 DOI: 10.1021/jacs.5c03844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
This study reports a supramolecular gel system capable of dynamic gel-to-gel transformations and reversible inversion of optical activity between superhelical and single-helical structures without passing through a sol phase. Inspired by collagen-like adaptability, the system utilizes 4-pyridinylboronic acid and guanosine as building blocks. Hierarchical assembly is achieved through pH-responsive boronic ester formation and guanosine-mediated G-quadruplex stacking, enabling transitions between superhelices and single helices with opposite optical activity. The system employs three regulatory pathways: bidirectional pH modulation, monotonic pH increase, and monotonic pH decrease, demonstrating programmable and reversible control over chirality, morphology, and mechanical properties. In the autonomous pH regulation, we have created an out-of-equilibrium hydrogel system with controlled switching of optical activity. Unlike traditional gel-sol-gel systems, this gel maintains macroscopic stability during transformations. Our remarkable finding bridges the gap between static supramolecular assemblies and dynamic soft materials, offering a platform for designing functional, biomimetic systems. The combination of hierarchical organization, dynamic chirality control, and robust programmability positions this gel for applications in adaptive optics, responsive biomaterials, and programmable soft matter.
Collapse
Affiliation(s)
- Jingjing Li
- College
of New Energy, Zhengzhou University of Light
Industry, Zhengzhou450002, China
- Institute
for Complex Molecular Systems and Laboratory of Macromolecular and
Organic Chemistry, Eindhoven University
of Technology, Eindhoven5600 MB, Netherlands
- School
of Chemistry and Chemical Engineering, Henan
University of Technology, Zhengzhou450001, China
| | - Fang Yin
- Institute
for Complex Molecular Systems and Laboratory of Macromolecular and
Organic Chemistry, Eindhoven University
of Technology, Eindhoven5600 MB, Netherlands
| | - Jianhong Wang
- Institute
for Complex Molecular Systems and Laboratory of Macromolecular and
Organic Chemistry, Eindhoven University
of Technology, Eindhoven5600 MB, Netherlands
| | - Huachuan Du
- Institute
for Complex Molecular Systems and Laboratory of Macromolecular and
Organic Chemistry, Eindhoven University
of Technology, Eindhoven5600 MB, Netherlands
| | - Fan Xu
- Institute
for Complex Molecular Systems and Laboratory of Macromolecular and
Organic Chemistry, Eindhoven University
of Technology, Eindhoven5600 MB, Netherlands
| | - Stefan Meskers
- Institute
for Complex Molecular Systems and Laboratory of Macromolecular and
Organic Chemistry, Eindhoven University
of Technology, Eindhoven5600 MB, Netherlands
| | - Yudong Li
- Institute
for Complex Molecular Systems and Laboratory of Macromolecular and
Organic Chemistry, Eindhoven University
of Technology, Eindhoven5600 MB, Netherlands
| | - Stefan Wijker
- Institute
for Complex Molecular Systems and Laboratory of Macromolecular and
Organic Chemistry, Eindhoven University
of Technology, Eindhoven5600 MB, Netherlands
| | - Yu Peng
- College
of New Energy, Zhengzhou University of Light
Industry, Zhengzhou450002, China
| | - Riccardo Bellan
- Institute
for Complex Molecular Systems and Laboratory of Macromolecular and
Organic Chemistry, Eindhoven University
of Technology, Eindhoven5600 MB, Netherlands
| | - Ghislaine Vantomme
- Institute
for Complex Molecular Systems and Laboratory of Macromolecular and
Organic Chemistry, Eindhoven University
of Technology, Eindhoven5600 MB, Netherlands
| | - Jian Song
- School
of Chemical Engineering and Technology, Tianjin University, Tianjin300350, China
| | - Chun-Sen Liu
- College
of New Energy, Zhengzhou University of Light
Industry, Zhengzhou450002, China
| | - E. W. Meijer
- Institute
for Complex Molecular Systems and Laboratory of Macromolecular and
Organic Chemistry, Eindhoven University
of Technology, Eindhoven5600 MB, Netherlands
| |
Collapse
|
6
|
Dai Q, Lemke S, Lu Y, Taylor S, Li H, Fu S, Wu X, Wang N, Xue T, He X. Food Safety Evaluation of Recombinant Humanized Type III Collagen Produced by Komagataella phaffii SMD1168-2COL3. J Appl Toxicol 2025; 45:808-829. [PMID: 39746781 DOI: 10.1002/jat.4741] [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: 10/13/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 01/04/2025]
Abstract
Collagens are biofunctional proteins that have been widely used in many fields, including biomedical, cosmetics, and skin care for their value in maintaining the integrity of cellular membranes. Collagens are also commonly consumed in foods and provide a source of protein and amino acids. As part of the safety assessment for this particular recombinant humanized type III (RHTypeIII) collagen produced by Komagataella phaffii SMD1168-2COL3, a series of toxicological tests were conducted. This collagen has ≥ 90% amino acid sequence homology to bovine and porcine collagen. The RHTypeIII collagen showed no evidence of genotoxic potential in a battery of tests. It was not toxic in an acute oral study, with no effects at 10 g/kg BW. The RHTypeIII collagen was not developmentally toxic in Sprague Dawley (SD) rat, and the NOAEL was 4.5 g/kg BW/day. In a 90-day oral gavage study in rats, there were no adverse findings observed; therefore, the high dose level (4.5 g/kg BW/day) was considered the NOAEL. The protein sequence was subjected to homology searches against the AllergenOnline database (sliding 80-amino acid windows and full sequence searches). From the 80-amino acid alignment searches, 23 significant matches were identified (> 35% identity and E value < 1 × 10-7) to allergens of bovine, fish, anisakis, feverfew pollen, ragweed pollen, and wheat origin. Although matches were identified, further assessment of the in silico results combined with a literature review demonstrates that the risk of allergenic cross-reactivity for this collagen is low. These results demonstrate RHTypeIII collagen is not toxic and unlikely to present a risk of allergy when used as a food ingredient.
Collapse
Affiliation(s)
- Qiu Dai
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Shawna Lemke
- Hjelle Consulting Group, St. Louis, Missouri, USA
| | - Yuemei Lu
- Blotting Consultancy (Beijing) Co., Ltd, Beijing, China
| | | | - Haihang Li
- Jiangsu Trautec Medical Technology Co., Ltd, Changzhou, China
| | - Shengwei Fu
- Jiangsu Trautec Medical Technology Co., Ltd, Changzhou, China
| | - Xiaowen Wu
- Jiangsu Trautec Medical Technology Co., Ltd, Changzhou, China
| | - Nan Wang
- CAIQ Test (Beijing) Co., Ltd, Beijing, China
| | - Tian Xue
- Blotting Consultancy (Beijing) Co., Ltd, Beijing, China
| | - Xiaoyun He
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
7
|
Page-McCaw PS, Pokidysheva EN, Darris CE, Chetyrkin S, Fidler AL, Gallup J, Aspirnaut Team, Murawala P, Hudson JK, Boudko SP, Hudson BG. Collagen IV of basement membranes: I. Origin and diversification of COL4 genes enabling metazoan multicellularity, evolution, and adaptation. J Biol Chem 2025; 301:108496. [PMID: 40220996 PMCID: PMC12141075 DOI: 10.1016/j.jbc.2025.108496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
Collagen IV (Col-IV) is a major component of basement membranes, a specialized form of extracellular matrix that enabled the assembly of multicellular epithelial tissues. In mammals, Col-IV assembles from a family of six α-chains (α1-α6), forming three supramolecular scaffolds: Col-IVα121, Col-IVα345, and Col-IVα121-α556. The α-chains are encoded by six genes (COL4A1-6) that occur in pairs in a head-to-head arrangement. In Alport syndrome, variants in COL4A3, 4, or 5 genes, encoding Col-IVα345 scaffold in glomerular basement membrane (GBM), the kidney ultrafilter, cause progressive renal failure in millions of people worldwide. The molecular mechanisms of how variants cause dysfunction remain obscure. Here, we gained insights into Col-IVα345 function by determining its evolutionary lineage, as revealed from phylogenetic analyses and tissue expression of COL4 gene pairs. We found that the COL4A⟨1|2⟩ gene pair emerged in basal Ctenophores and Cnidaria phyla and is highly conserved across metazoans. The COL4A⟨1|2⟩ duplicated and arose as the progenitor to the COL4A⟨3|4⟩ gene pair in cyclostomes, coinciding with emergence of kidney GBM, and expressed and conserved in jawed vertebrates, except for amphibians, and a second duplication as the progenitor to the COL4A⟨5|6⟩ gene pair and conserved in jawed vertebrates. These findings revealed that Col-IVα121 is the progenitor scaffold, expressed ubiquitously in metazoan basement membranes, and which evolved into vertebrate Col-IVα345 and expressed in GBM. The Col-IVα345 scaffold, in comparison, has an increased number of cysteine residues, varying in number with osmolarity of the environment. Cysteines mediate disulfide crosslinks between protomers, an adaptation enabling a compact GBM that withstands the high hydrostatic pressure associated with glomerular ultrafiltration.
Collapse
Affiliation(s)
- Patrick S Page-McCaw
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Aspirnaut Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Elena N Pokidysheva
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Aspirnaut Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Carl E Darris
- Aspirnaut Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sergei Chetyrkin
- Aspirnaut Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Aaron L Fidler
- Aspirnaut Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Julianna Gallup
- Aspirnaut Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Prayag Murawala
- Mount Desert Island Biological Laboratory, Bar Harbor, Maine, USA; Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Julie K Hudson
- Aspirnaut Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Medical Education and Administration, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sergei P Boudko
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Aspirnaut Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Billy G Hudson
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Aspirnaut Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA; Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee, USA.
| |
Collapse
Collaborators
Julianna Gallup, Jillian Balser, Cullen Curbow, Ricard Harris, Cody Stothers, Keith Wade,
Collapse
|
8
|
De Marco M, Rai SR, Scietti L, Mattoteia D, Liberi S, Moroni E, Pinnola A, Vetrano A, Iacobucci C, Santambrogio C, Colombo G, Forneris F. Molecular structure and enzymatic mechanism of the human collagen hydroxylysine galactosyltransferase GLT25D1/COLGALT1. Nat Commun 2025; 16:3624. [PMID: 40240392 PMCID: PMC12003778 DOI: 10.1038/s41467-025-59017-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 04/08/2025] [Indexed: 04/18/2025] Open
Abstract
During collagen biosynthesis, lysine residues undergo extensive post-translational modifications through the alternate action of two distinct metal ion-dependent enzyme families (i.e., LH/PLODs and GLT25D/COLGALT), ultimately producing the highly conserved α-(1,2)-glucosyl-β-(1,O)-galactosyl-5-hydroxylysine pattern. Malfunctions in these enzymes are linked to developmental pathologies and extracellular matrix alterations associated to enhanced aggressiveness of solid tumors. Here, we characterized human GLT25D1/COLGALT1, revealing an elongated head-to-head homodimeric assembly. Each monomer encompasses two domains (named GT1 and GT2), both unexpectedly capable of binding metal ion cofactors and UDP-α-galactose donor substrates, resulting in four candidate catalytic sites per dimer. We identify the catalytic site in GT2, featuring an unusual Glu-Asp-Asp motif critical for Mn2+ binding, ruling out direct catalytic roles for the GT1 domain, but showing that in this domain the unexpectedly bound Ca2+ and UDP-α-galactose cofactors are critical for folding stability. Dimerization, albeit not essential for GLT25D1/COLGALT1 activity, provides a critical molecular contact site for multi-enzyme assembly interactions with partner multifunctional LH/PLOD lysyl hydroxylase-glycosyltransferase enzymes.
Collapse
Grants
- MFAG 20075, BRIDGE 27004 Associazione Italiana per la Ricerca sul Cancro (Italian Association for Cancer Research)
- Rarer Types EDS Grant 2022 Ehlers-Danlos Society (EDS)
- CDA 2013 Giovanni Armenise-Harvard Foundation
- NextGeneration-EU PNRR MUR M4C2 PE00000007 INF-ACT Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- PRIN PNRR 2022 P20224WAME Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- PRIN PNRR 2022 P20224WAME Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- Piano Operativo Salute, IMMUNO-HUB Ministero della Salute (Ministry of Health, Italy)
- regional law n° 9/2020, resolution n° 3776/2020 Regione Lombardia (Region of Lombardy)
- Please update "Ministero dell'Istruzione, dell'Università e della Ricerca" with "Ministero dell'Università e della Ricerca (MUR)"
Collapse
Affiliation(s)
- Matteo De Marco
- The Armenise-Harvard Laboratory of Structural Biology, Dept. Biology and Biotechnology, University of Pavia, Via Ferrata 9A, 27100, Pavia, Italy
| | - Sristi Raj Rai
- The Armenise-Harvard Laboratory of Structural Biology, Dept. Biology and Biotechnology, University of Pavia, Via Ferrata 9A, 27100, Pavia, Italy
| | - Luigi Scietti
- The Armenise-Harvard Laboratory of Structural Biology, Dept. Biology and Biotechnology, University of Pavia, Via Ferrata 9A, 27100, Pavia, Italy
- Biochemistry and Structural Biology Unit, Department of Experimental Oncology, IRCCS European Institute of Oncology (IEO), Via Adamello 16, 20139, Milan, Italy
| | - Daiana Mattoteia
- The Armenise-Harvard Laboratory of Structural Biology, Dept. Biology and Biotechnology, University of Pavia, Via Ferrata 9A, 27100, Pavia, Italy
| | - Stefano Liberi
- The Armenise-Harvard Laboratory of Structural Biology, Dept. Biology and Biotechnology, University of Pavia, Via Ferrata 9A, 27100, Pavia, Italy
| | | | - Alberta Pinnola
- BioPhotoLab, Dept. Biology and Biotechnology, University of Pavia, Via Ferrata 9A, 27100, Pavia, Italy
| | - Alice Vetrano
- Department of Physical and Chemical Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Claudio Iacobucci
- Department of Physical and Chemical Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Carlo Santambrogio
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Giorgio Colombo
- Department of Chemistry, University of Pavia, Via Taramelli 12, Pavia, Italy
| | - Federico Forneris
- The Armenise-Harvard Laboratory of Structural Biology, Dept. Biology and Biotechnology, University of Pavia, Via Ferrata 9A, 27100, Pavia, Italy.
- Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| |
Collapse
|
9
|
Ullah S, Zainol I. Fabrication and applications of biofunctional collagen biomaterials in tissue engineering. Int J Biol Macromol 2025; 298:139952. [PMID: 39824416 DOI: 10.1016/j.ijbiomac.2025.139952] [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: 10/23/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
Collagen is extensively used in tissue engineering for various organ tissue regeneration due to the main component of human organ extracellular matrix (ECM) and their inherent nature bioactivity. Collagen various types naturally exist in different organ ECMs. Collagen fabricated with natural ECM mimics architecture, composition and mechanical properties for various organ tissue regeneration. Collagen fabrication with organ-specific biofunctionality facilitated organ tissue engineering as compared to unmodified collagen biomaterials. Collagen biofunctionality improved by subjecting collagen to synthesis, fibers and surface modifications, and blending with other components. Furthermore, collagen is loaded with bioactive molecules, growth factors, drugs and cells also enhancing the biofunctionality of collagen biomaterials. In this review, we will explore the recent advancements in biofunctional collagen biomaterials fabrication with organ-specific biofunctionality in tissue engineering to resolve various organ tissue engineering issues and regeneration challenges. Biofunctional collagen biomaterials stimulate microenvironments inside and around the implants to excellently regulate cellular activities, differentiate cells into organ native cells, enhanced ECM production and remodeling to regenerate organ tissues with native structure, function and maturation. This review critically explored biofunctional collagen biomaterials fabrication in resolving various organ tissue engineering issues and regeneration challenges, and opening new directions of biofunctional collagen biomaterials fabrication, design and applications.
Collapse
Affiliation(s)
- Saleem Ullah
- Polymer Lab, Chemistry Department, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, 35900 Tanjung Malim, Perak, Darul Ridzuan, Malaysia.
| | - Ismail Zainol
- Polymer Lab, Chemistry Department, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, 35900 Tanjung Malim, Perak, Darul Ridzuan, Malaysia.
| |
Collapse
|
10
|
Nyström A. Dystrophic epidermolysis bullosa - From biochemistry to interventions. Matrix Biol 2025; 136:111-126. [PMID: 39922469 DOI: 10.1016/j.matbio.2025.02.001] [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: 12/06/2024] [Revised: 01/20/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
The skin, as a barrier organ meeting constant mechanical challenges, is equipped with multiple adhesive structures that collectively support resilient, yet flexible attachment of its epithelium -the epidermis to its mesenchyme - the dermis. One such structure is the collagen VII-composed anchoring fibril, which provides firm anchorage of the epidermal basement membrane to the underlying interstitial extracellular matrix. Blistering and wider tissue fragility in the genetic disease dystrophic epidermolysis bullosa (DEB) caused by collagen VII deficiency illustrate the essential function of collagen VII in supporting skin integrity. DEB is also a progressive inflammatory fibrotic disease with multi-organ involvement, indicating that collagen VII has broader functions than simply providing epithelial anchorage. This review explores the reciprocal relationship between collagen VII biology and DEB pathophysiology. A deeper understanding of collagen VII biology - spanning its synthesis, assembly into suprastructures, and regulatory roles - enhances our understanding of DEB. Conversely, detailed insights into DEB through analysis of disease progression or therapeutic interventions offer valuable information on the broader tissue and organismal roles of collagen VII in maintaining homeostasis. This review focuses on such knowledge exchange in advancing our understanding of collagen VII, the extracellular matrix in general, and inspiring potential strategies for treatment of DEB. Importantly, in a broader sense, the discussed themes are applicable to other conditions driven by compromised extracellular matrix instruction and integrity, leading to progressive damage and inflammation.
Collapse
Affiliation(s)
- Alexander Nyström
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, Hauptstrasse 7, 79140 Freiburg, Germany.
| |
Collapse
|
11
|
Mohan T, Gürer F, Bračič D, Lackner F, Nagaraj C, Maver U, Gradišnik L, Finšgar M, Kargl R, Kleinschek KS. Functionalization of Polycaprolactone 3D Scaffolds with Hyaluronic Acid Glycine-Peptide Conjugates and Endothelial Cell Adhesion. Biomacromolecules 2025; 26:1771-1787. [PMID: 39988967 PMCID: PMC11898084 DOI: 10.1021/acs.biomac.4c01559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/25/2025]
Abstract
This study enhances the bioactivity of polycaprolactone (PCL) scaffolds for tissue engineering by functionalizing them with oxidized hyaluronic acid glycine-peptide conjugates to improve endothelial cell adhesion and growth. Hyaluronic acid was conjugated with a glycine-peptide to create a bioactive interface on PCL (static water contact angle, SCA(H2O): 98°). The scaffolds were fabricated using a melt extrusion 3D printing technique. The HA-glycine peptide conjugates were oxidized and immobilized on aminolyzed PCL via Schiff-base chemistry, introducing hydrophilicity (SCA(H2O): 21°), multiple functional groups, and a negative zeta potential (-12.04 mV at pH 7.4). A quartz crystal microbalance confirmed chemical conjugation and quantified the mass (8.5-10.3 mg m-2) of oxidized HA-glycine on PCL. The functionalized scaffolds showed enhanced swelling, improved mechanical properties (2-fold increase in strength, from 26 to 51 MPa), and maintained integrity during degradation. In-vitro experiments demonstrated improved endothelial cell adhesion, proliferation and viability, suggesting the potential for vascularized tissue constructs.
Collapse
Affiliation(s)
- Tamilselvan Mohan
- Graz
University of Technology, Institute of Chemistry
and Technology of Biobased System, Stremayrgasse 9, 8010 Graz, Austria
- University
of Maribor, Faculty of Mechanical Engineering,
Laboratory for Characterisation and Processing of Polymers, Smetanova ulica17, 2000 Maribor, Slovenia
- Members
of the European Polysaccharide Network of Excellence (EPNOE), Celestijnenlaan 200 F, 3001 Leuven, Belgium
| | - Fazilet Gürer
- University
of Maribor, Faculty of Mechanical Engineering,
Laboratory for Characterisation and Processing of Polymers, Smetanova ulica17, 2000 Maribor, Slovenia
- Members
of the European Polysaccharide Network of Excellence (EPNOE), Celestijnenlaan 200 F, 3001 Leuven, Belgium
| | - Doris Bračič
- University
of Maribor, Faculty of Mechanical Engineering,
Laboratory for Characterisation and Processing of Polymers, Smetanova ulica17, 2000 Maribor, Slovenia
- Members
of the European Polysaccharide Network of Excellence (EPNOE), Celestijnenlaan 200 F, 3001 Leuven, Belgium
| | - Florian Lackner
- Graz
University of Technology, Institute of Chemistry
and Technology of Biobased System, Stremayrgasse 9, 8010 Graz, Austria
| | - Chandran Nagaraj
- Department
of Internal Medicine, Division of Pulmonology, Medical University of Graz, 8010 Graz, Austria
| | - Uroš Maver
- University
of Maribor, Faculty of Medicine,
Institute of Biomedical Sciences, Taborska Ulica 8, 2000 Maribor, Slovenia
- Members
of the European Polysaccharide Network of Excellence (EPNOE), Celestijnenlaan 200 F, 3001 Leuven, Belgium
| | - Lidija Gradišnik
- University
of Maribor, Faculty of Medicine,
Institute of Biomedical Sciences, Taborska Ulica 8, 2000 Maribor, Slovenia
| | - Matjaž Finšgar
- University
of Maribor, Faculty of Chemistry
and Chemical Engineering, Laboratory for Analytical Chemistry and
Industrial Analysis, Smetanova
ulica 17, 2000 Maribor, Slovenia
| | - Rupert Kargl
- Graz
University of Technology, Institute of Chemistry
and Technology of Biobased System, Stremayrgasse 9, 8010 Graz, Austria
- University
of Maribor, Faculty of Mechanical Engineering,
Laboratory for Characterisation and Processing of Polymers, Smetanova ulica17, 2000 Maribor, Slovenia
| | - Karin Stana Kleinschek
- Graz
University of Technology, Institute of Chemistry
and Technology of Biobased System, Stremayrgasse 9, 8010 Graz, Austria
- University
of Maribor, Institute of Automation,
Faculty of Electrical Engineering and Computer Science, Koroska cesta 46, 2000 Maribor, Slovenia
- Members
of the European Polysaccharide Network of Excellence (EPNOE), Celestijnenlaan 200 F, 3001 Leuven, Belgium
| |
Collapse
|
12
|
Ahmad AA, Ghim M, Kukreja G, Neishabouri A, Zhang Z, Li J, Salarian M, Toczek J, Gona K, Hedayatyanfard K, Morrison T, Zhang J, Huang YH, Liu C, Yu SM, Sadeghi MM. Collagen Hybridizing Peptide-Based Radiotracers for Molecular Imaging of Collagen Turnover in Pulmonary Fibrosis. J Nucl Med 2025; 66:425-433. [PMID: 39915119 PMCID: PMC11876730 DOI: 10.2967/jnumed.124.268832] [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: 09/20/2024] [Accepted: 01/06/2025] [Indexed: 02/12/2025] Open
Abstract
Pulmonary fibrosis is a characteristic feature of interstitial lung disease. Current clinical diagnostic methods provide a snapshot of the lung structure without information on disease activity. Collagen hybridizing peptides offer the opportunity to detect collagen remodeling through their hybridization with denatured collagen. Here, we sought to develop a 99mTc-labeled collagen hybridizing tracer to track denatured collagen in vivo and validate it in a murine model of pulmonary fibrosis. Methods: Imaging agents consisting of a polyhistidine or a poly-histidine-glutamic acid [(HE)3] peptide connected to an N-terminal targeting moiety with 9 glycine-proline-hydroxyproline repeats [(GPO)9] through a 3-glycine linker were synthesized. After radiolabeling with 99mTc-tricarbonyl, the labeled products' purity and stability were evaluated by high-performance liquid chromatography and γ-well counting, and their biodistributions were compared in mice. To induce pulmonary fibrosis, the lungs of 8- to 10-wk-old mice were exposed to bleomycin (or saline as control). At 3 wk after induction, SPECT/CT imaging with 99mTc-(HE)3-(GPO)9 was performed 1 h after injection and was followed by tissue collection to assess 99mTc-(HE)3-(GPO)9 biodistribution by γ-well counting and to evaluate lung histology. The specificity of the tracer uptake was assessed using a scrambled homolog. A group of animals underwent serial imaging 3 and 8-10 wk after induction. Results: The specific activity of the final radiolabeled product was 70.3 ± 14.8 GBq/µmol. Radiolabeled tracers were stable in blood for at least 2 h and showed rapid blood clearance. 99mTc-(HE)3-(GPO)9 showed lower liver uptake in biodistribution studies and was selected for in vivo imaging studies. SPECT/CT imaging of bleomycin-treated mice 3 wk after induction showed higher specific 99mTc-(HE)3-(GPO)9 lung uptake than that of control mice (P < 0.01) and that of bleomycin-treated mice 8-10 wk after induction, when fibrosis was resolved (P < 0.05). There was a significant correlation between lung uptake quantified by SPECT/CT and γ-well counting (Pearson R = 0.83, P < 0.001) and significant correlations between tracer uptake and indices of tissue fibrosis. Conclusion: 99mTc-(HE)3-(GPO)9 enables SPECT imaging of collagen turnover in pulmonary fibrosis. This approach expands the scope of existing diagnostic tools in fibrosis and can lead to better patient management by monitoring the effect of antifibrotic therapies.
Collapse
Affiliation(s)
- Azmi A Ahmad
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Mean Ghim
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Gunjan Kukreja
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Afarin Neishabouri
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Zhengxing Zhang
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Jie Li
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Mani Salarian
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Jakub Toczek
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Kiran Gona
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Keshvad Hedayatyanfard
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Tian Morrison
- Department of Biomedical Engineering and Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah; and
| | - Jiasheng Zhang
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Yiyun Henry Huang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Chi Liu
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - S Michael Yu
- Department of Biomedical Engineering and Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah; and
| | - Mehran M Sadeghi
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut;
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
13
|
De Gregorio V, Barua M, Lennon R. Collagen formation, function and role in kidney disease. Nat Rev Nephrol 2025; 21:200-215. [PMID: 39548215 DOI: 10.1038/s41581-024-00902-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2024] [Indexed: 11/17/2024]
Abstract
Highly abundant in mammals, collagens define the organization of tissues and participate in cell signalling. Most of the 28 vertebrate collagens, with the exception of collagens VI, VII, XXVI and XXVIII, can be categorized into five subgroups: fibrillar collagens, network-forming collagens, fibril-associated collagens with interrupted triple helices, membrane-associated collagens with interrupted triple helices and multiple triple-helix domains with interruptions. Collagen peptides are synthesized from the ribosome and enter the rough endoplasmic reticulum, where they undergo numerous post-translational modifications. The collagen chains form triple helices that can be secreted to form a diverse array of supramolecular structures in the extracellular matrix. Collagens are ubiquitously expressed and have been linked to a broad spectrum of disorders, including genetic disorders with kidney phenotypes. They also have an important role in kidney fibrosis and mass spectrometry-based proteomic studies have improved understanding of the composition of fibrosis in kidney disease. A wide range of therapeutics are in development for collagen and kidney disorders, including genetic approaches, chaperone therapies, protein degradation strategies and anti-fibrotic therapies. Improved understanding of collagens and their role in disease is needed to facilitate the development of more specific treatments for collagen and kidney disorders.
Collapse
Affiliation(s)
- Vanessa De Gregorio
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Moumita Barua
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
- Toronto General Hospital Research Institute, Toronto, Ontario, Canada.
- Division of Nephrology, University Health Network, Toronto, Ontario, Canada.
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
- Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester, UK.
| |
Collapse
|
14
|
de Moura Campos S, Dos Santos Costa G, Karp SG, Thomaz-Soccol V, Soccol CR. Innovations and challenges in collagen and gelatin production through precision fermentation. World J Microbiol Biotechnol 2025; 41:63. [PMID: 39910024 DOI: 10.1007/s11274-025-04276-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025]
Abstract
Collagen and gelatin are essential biomaterials widely used in industries such as food, cosmetics, healthcare, and pharmaceuticals. Traditionally derived from animal tissues, these proteins are facing growing demand for more sustainable and ethical production methods. Precision fermentation (PF) offers a promising alternative by using genetically engineered microorganisms to produce recombinant collagen and gelatin. This technology not only reduces environmental impact but also ensures consistent quality and higher yields. In this review, we provide a comprehensive overview of collagen and gelatin production through PF destined for the food sector, exploring key advances in recombinant technologies, synthetic biology, and bioprocess optimization. Challenges such as scaling production, cost-efficiency, and market integration are addressed, alongside emerging solutions for enhancing industrial competitiveness. We also highlight leading companies leveraging PF to drive innovation in the food industry. As PF continues to evolve, future developments are expected to improve efficiency, reduce costs, and expand the applications of recombinant collagen and gelatin, particularly in the food and supplement sectors.
Collapse
Affiliation(s)
- Sofia de Moura Campos
- Department of Bioprocess and Biotechnology Engineering, Federal University of Paraná, Curitiba, Brazil
| | - Gabriela Dos Santos Costa
- Department of Bioprocess and Biotechnology Engineering, Federal University of Paraná, Curitiba, Brazil
| | - Susan Grace Karp
- Department of Bioprocess and Biotechnology Engineering, Federal University of Paraná, Curitiba, Brazil
| | - Vanete Thomaz-Soccol
- Department of Bioprocess and Biotechnology Engineering, Federal University of Paraná, Curitiba, Brazil
| | - Carlos Ricardo Soccol
- Department of Bioprocess and Biotechnology Engineering, Federal University of Paraná, Curitiba, Brazil.
| |
Collapse
|
15
|
Wuergezhen D, Gindroz E, Morita R, Hashimoto K, Abe T, Kiyonari H, Fujiwara H. An eGFP-Col4a2 mouse model reveals basement membrane dynamics underlying hair follicle morphogenesis. J Cell Biol 2025; 224:e202404003. [PMID: 39656438 PMCID: PMC11629887 DOI: 10.1083/jcb.202404003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 10/26/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Precisely controlled remodeling of the basement membrane (BM) is crucial for morphogenesis, but its molecular and tissue-level dynamics, underlying mechanisms, and functional significance in mammals remain largely unknown due to limited visualization tools. We developed mouse lines in which the endogenous collagen IV gene (Col4a2) was fused with a fluorescent tag. Through live imaging of developing hair follicles, we reveal a spatial gradient in the turnover rate of COL4A2 that is closely coupled with both the BM expansion rate and the proliferation rate of epithelial progenitors. Epithelial progenitors are displaced with directionally expanding BMs but do not actively migrate on stationary BM. The addition of a matrix metalloproteinase inhibitor delays COL4A2 turnover, restrains BM expansion, and increases perpendicular divisions of epithelial progenitors, altering hair follicle morphology. Our findings highlight the spatially distinct dynamics of BM and their key roles in orchestrating progenitor cell behavior and organ shape during development.
Collapse
Affiliation(s)
- Duligengaowa Wuergezhen
- Laboratory for Tissue Microenvironment, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Graduate School of Medicine, Osaka University, Suita, Japan
| | - Eleonore Gindroz
- Laboratory for Tissue Microenvironment, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Ritsuko Morita
- Laboratory for Tissue Microenvironment, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Kei Hashimoto
- Laboratory for Tissue Microenvironment, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Hironobu Fujiwara
- Laboratory for Tissue Microenvironment, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
16
|
Page-McCaw PS, Pokidysheva EN, Darris CE, Chetyrkin S, Fidler AL, Gallup J, Murawala P, Hudson JK, Boudko S, Hudson BG. Collagen IV of basement membranes: I. Origin and diversification of COL4 genes enabling animal evolution and adaptation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.18.563013. [PMID: 37905027 PMCID: PMC10614949 DOI: 10.1101/2023.10.18.563013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Collagen IV is a major component of basement membranes, a specialized form of extracellular matrix that enabled the assembly of multicellular epithelial tissues. In mammals, collagen IV assembles from a family of six α-chains (α1 to α6), forming three supramolecular scaffolds: Col-IVα121, Col-IVα345 and Col-IVα121-α556. The α-chains are encoded by six genes (COL4A1-6) that occur in pairs in a head-to-head arrangement. In Alport syndrome, variants in COL4A3, 4 or 5 genes, encoding Col-IVα345 scaffold in glomerular basement membrane (GBM), the kidney ultrafilter, cause progressive renal failure in millions of people worldwide. How variants cause dysfunction remains obscure. Here, we gained insights into Col-IVα345 function by determining its evolutionary lineage, as revealed from phylogenetic analyses and tissue expression of COL4 gene-pairs. We found that the COL4A⟨1|2⟩ gene-pair emerged in basal Ctenophores and Cnidaria phyla and is highly conserved across metazoans. The COL4A⟨1|2⟩ duplicated and arose as the progenitor to the COL4A⟨3|4⟩ gene-pair in cyclostomes, coinciding with emergence of kidney GBM, and expressed and conserved in jawed-vertebrates, except for amphibians, and a second duplication as the progenitor to the COL4A⟨5|6⟩ gene-pair and conserved in jawed-vertebrates. These findings revealed that Col-IVα121 is the progenitor scaffold, expressed ubiquitously in metazoan basement membranes, and which evolved into vertebrate Col-IVα345 and expressed in GBM. The Col-IVα345 scaffold, in comparison, has an increased number of cysteine residues, varying in number with osmolarity of the environment. Cysteines mediate disulfide crosslinks between protomers, an adaptation enabling a compact GBM that withstands the high hydrostatic pressure associated with glomerular ultrafiltration.
Collapse
|
17
|
Cao H, Zeng Y, Yuan X, Wang JK, Tay CY. Waste-to-resource: Extraction and transformation of aquatic biomaterials for regenerative medicine. BIOMATERIALS ADVANCES 2025; 166:214023. [PMID: 39260186 DOI: 10.1016/j.bioadv.2024.214023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/16/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024]
Abstract
The fisheries and aquaculture industry are known for generating substantial waste or by-products, often underutilized, or relegated to low-value purposes. However, this overlooked segment harbors a rich repository of valuable bioactive materials of which have a broad-spectrum of high-value applications. As the blue economy gains momentum and fisheries expand, sustainable exploitation of these aquatic resources is increasingly prioritized. In this review, we present a comprehensive overview of technology-enabled methods for extracting and transforming aquatic waste into valuable biomaterials and their recent advances in regenerative medicine applications, focusing on marine collagen, chitin/chitosan, calcium phosphate and bioactive-peptides. We discuss the inherent bioactive qualities of these "waste-to-resource" aquatic biomaterials and identify opportunities for their use in regenerative medicine to advance healthcare while achieving the Sustainable Development Goals.
Collapse
Affiliation(s)
- Huaqi Cao
- China-Singapore International Joint Research Institute (CSIJRI), China Singapore Guangzhou Knowledge City, Huangpu District, Guangzhou, PR China
| | - Yuanjin Zeng
- China-Singapore International Joint Research Institute (CSIJRI), China Singapore Guangzhou Knowledge City, Huangpu District, Guangzhou, PR China
| | - Xueyu Yuan
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, PR China; School of Materials Science and Engineering, Nanyang Technological University, N4.1, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Jun Kit Wang
- School of Materials Science and Engineering, Nanyang Technological University, N4.1, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Chor Yong Tay
- China-Singapore International Joint Research Institute (CSIJRI), China Singapore Guangzhou Knowledge City, Huangpu District, Guangzhou, PR China; School of Materials Science and Engineering, Nanyang Technological University, N4.1, 50 Nanyang Avenue, Singapore 639798, Singapore; Center for Sustainable Materials (SusMat), Nanyang Technological University, Singapore 637553, Singapore; Nanyang Environment & Water Research Institute, 1 CleanTech Loop, CleanTech One, Singapore 637141, Singapore.
| |
Collapse
|
18
|
Wright SA, Lennon R, Greenhalgh AD. Basement membranes' role in immune cell recruitment to the central nervous system. J Inflamm (Lond) 2024; 21:53. [PMID: 39707430 DOI: 10.1186/s12950-024-00426-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024] Open
Abstract
Basement membranes form part of the extracellular matrix (ECM), which is the structural basis for all tissue. Basement membranes are cell-adherent sheets found between cells and vascular endothelia, including those of the central nervous system (CNS). There is exceptional regional specialisation of these structures, both in tissue organisation and regulation of tissue-specific cellular processes. Due to their location, basement membranes perform a key role in immune cell trafficking and therefore are important in inflammatory processes causing or resulting from CNS disease and injury. This review will describe basement membranes in detail, with special focus on the brain. We will cover how genetic changes drive brain pathology, describe basement membranes' role in immune cell recruitment and how they respond to various brain diseases. Understanding how basement membranes form the junction between the immune and central nervous systems will be a major advance in understanding brain disease.
Collapse
Affiliation(s)
- Shaun A Wright
- Lydia Becker Institute of Immunology and Inflammation, Division, Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Science, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Rachel Lennon
- Cell Matrix Biology & Regenerative Medicine and Wellcome Centre for Cell-Matrix Research, School of Biological Science, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Andrew D Greenhalgh
- Lydia Becker Institute of Immunology and Inflammation, Division, Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Science, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
19
|
Labelle-Dumais C, Mazur C, Kaya S, Obata Y, Lee B, Acevedo C, Alliston T, Gould DB. Skeletal pathology in mouse models of Gould syndrome is partially alleviated by genetically reducing TGFβ signaling. Matrix Biol 2024; 133:1-13. [PMID: 39097038 PMCID: PMC12032920 DOI: 10.1016/j.matbio.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
Skeletal defects are hallmark features of many extracellular matrix (ECM) and collagen-related disorders. However, a biological function in bone has never been defined for the highly evolutionarily conserved type IV collagen. Collagen type IV alpha 1 (COL4A1) and alpha 2 (COL4A2) form α1α1α2 (IV) heterotrimers that represent a fundamental basement membrane constituent present in every organ of the body, including the skeleton. COL4A1 and COL4A2 mutations cause Gould syndrome, a variable and clinically heterogenous multisystem disorder generally characterized by the presence of cerebrovascular disease with ocular, renal, and muscular manifestations. We have previously identified elevated TGFβ signaling as a pathological insult resulting from Col4a1 mutations and demonstrated that reducing TGFβ signaling ameliorate ocular and cerebrovascular phenotypes in Col4a1 mutant mouse models of Gould syndrome. In this study, we describe the first characterization of skeletal defects in Col4a1 mutant mice that include a developmental delay in osteogenesis and structural, biomechanical and vascular alterations of mature bones. Using distinct mouse models, we show that allelic heterogeneity influences the presentation of skeletal pathology resulting from Col4a1 mutations. Importantly, we found that TGFβ target gene expression is elevated in developing bones from Col4a1 mutant mice and show that genetically reducing TGFβ signaling partially ameliorates skeletal manifestations. Collectively, these findings identify a novel and unsuspected role for type IV collagen in bone biology, expand the spectrum of manifestations associated with Gould syndrome to include skeletal abnormalities, and implicate elevated TGFβ signaling in skeletal pathogenesis in Col4a1 mutant mice.
Collapse
Affiliation(s)
- Cassandre Labelle-Dumais
- Departments of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Courtney Mazur
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, 94143, USA; UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, CA 94143, USA
| | - Serra Kaya
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Yoshihiro Obata
- Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA 92093, USA
| | - Bryson Lee
- Departments of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Claire Acevedo
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, 94143, USA; Materials Science Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA 92093, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, 94143, USA; UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, CA 94143, USA
| | - Douglas B Gould
- Departments of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA; Department of Anatomy, Institute for Human Genetics, Bakar Aging Research Institute, and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
20
|
Shaik MI, Rahman SHA, Yusri AS, Ismail-Fitry MR, Kumar NSS, Sarbon NM. A review on the processing technique, physicochemical, and bioactive properties of marine collagen. J Food Sci 2024; 89:5205-5229. [PMID: 39126690 DOI: 10.1111/1750-3841.17273] [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: 03/25/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 08/12/2024]
Abstract
Collagens are conventionally derived from bovine and porcine sources. However, these sources were commonly associated with infectious diseases such as bovine spongiform encephalopathy, foot and mouth disease, autoimmune and allergic reactions, and religious constraints. The significant amount of collagen available in marine species, especially fish skins, scales, fins, and bones, shows that marine species can be a potential alternative source to mammalian collagen. Therefore, this review aims to give a clearer outlook on the processing techniques of marine collagen and its physicochemical and bioactive properties as a potential alternative to mammalian collagen. The two most suitable extraction methods for marine collagen are pepsin-soluble extraction and ultrasound-assisted extraction. Additionally, marine collagen's physicochemical and bioactive properties, such as antioxidants, wound healing, tissue engineering, and cosmetic biomaterial have been thoroughly discussed in this review. PRACTICAL APPLICATION: Collagen extracted from marine sources showed its potential in physicochemical and bioactive properties, including antioxidants and wound-healing capabilities, as an alternative to mammalian collagen. The significant amount of collagen found in marine species, particularly in fish skins, scales, bones, and sea cucumbers, suggests that marine sources could be a viable alternative to land mammal collagen due to their abundance and accessibility. The ultrasound-assisted extraction technique has improved the extracted marine collagen's physicochemical and bioactivity properties and quality properties.
Collapse
Affiliation(s)
- Mannur Ismail Shaik
- Faculty of Fisheries and Food Science, Universiti Malaysia Terengganu, Kuala Nerus, Terengganu, Malaysia
| | - Siti Hajar Abdul Rahman
- Faculty of Fisheries and Food Science, Universiti Malaysia Terengganu, Kuala Nerus, Terengganu, Malaysia
| | - Anis Syafiqah Yusri
- Faculty of Fisheries and Food Science, Universiti Malaysia Terengganu, Kuala Nerus, Terengganu, Malaysia
| | - Mohammad Rashedi Ismail-Fitry
- Department of Food Technology, Faculty of Food Science and Technology, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Nune Satya Sampath Kumar
- Department of Biotechnology, Vignan's Foundation for Science, Technology and Research, Guntur, Andhra Pradesh, India
| | - Norizah Mhd Sarbon
- Faculty of Fisheries and Food Science, Universiti Malaysia Terengganu, Kuala Nerus, Terengganu, Malaysia
| |
Collapse
|
21
|
Sundaram MV, Pujol N. The Caenorhabditis elegans cuticle and precuticle: a model for studying dynamic apical extracellular matrices in vivo. Genetics 2024; 227:iyae072. [PMID: 38995735 PMCID: PMC11304992 DOI: 10.1093/genetics/iyae072] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/25/2024] [Indexed: 07/14/2024] Open
Abstract
Apical extracellular matrices (aECMs) coat the exposed surfaces of animal bodies to shape tissues, influence social interactions, and protect against pathogens and other environmental challenges. In the nematode Caenorhabditis elegans, collagenous cuticle and zona pellucida protein-rich precuticle aECMs alternately coat external epithelia across the molt cycle and play many important roles in the worm's development, behavior, and physiology. Both these types of aECMs contain many matrix proteins related to those in vertebrates, as well as some that are nematode-specific. Extensive differences observed among tissues and life stages demonstrate that aECMs are a major feature of epithelial cell identity. In addition to forming discrete layers, some cuticle components assemble into complex substructures such as ridges, furrows, and nanoscale pillars. The epidermis and cuticle are mechanically linked, allowing the epidermis to sense cuticle damage and induce protective innate immune and stress responses. The C. elegans model, with its optical transparency, facilitates the study of aECM cell biology and structure/function relationships and all the myriad ways by which aECM can influence an organism.
Collapse
Affiliation(s)
- Meera V Sundaram
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Nathalie Pujol
- Aix Marseille University, INSERM, CNRS, CIML, Turing Centre for Living Systems, 13009 Marseille, France
| |
Collapse
|
22
|
Szalus K, Trzeciak M. The Role of Collagens in Atopic Dermatitis. Int J Mol Sci 2024; 25:7647. [PMID: 39062889 PMCID: PMC11276735 DOI: 10.3390/ijms25147647] [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: 06/01/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease affecting both children and adults. The clinical picture of AD manifests in typical skin lesions, such as localized eczema and dry skin, with dominant, persistent itching that leads to sleep disturbances. The pathophysiology of AD has been extensively investigated with respect to epigenetic and genetic factors, skin barrier defects, as well as immunological and microbial disorders. However, to date, the involvement of extracellular matrix (ECM) elements has received limited attention. Collagen, a major component of the ECM, may serve as a therapeutic target for the future treatment of AD. This paper summarizes the role of collagens, which are the most abundant components of the extracellular matrix in AD.
Collapse
Affiliation(s)
| | - Magdalena Trzeciak
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland
| |
Collapse
|
23
|
Yurchenco PD, Kulczyk AW. Polymerizing laminins in development, health, and disease. J Biol Chem 2024; 300:107429. [PMID: 38825010 PMCID: PMC11260871 DOI: 10.1016/j.jbc.2024.107429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/12/2024] [Accepted: 05/26/2024] [Indexed: 06/04/2024] Open
Abstract
Polymerizing laminins are multi-domain basement membrane (BM) glycoproteins that self-assemble into cell-anchored planar lattices to establish the initial BM scaffold. Nidogens, collagen-IV and proteoglycans then bind to the scaffold at different domain loci to create a mature BM. The LN domains of adjacent laminins bind to each other to form a polymer node, while the LG domains attach to cytoskeletal-anchoring integrins and dystroglycan, as well as to sulfatides and heparan sulfates. The polymer node, the repeating unit of the polymer scaffold, is organized into a near-symmetrical triskelion. The structure, recently solved by cryo-electron microscopy in combination with AlphaFold2 modeling and biochemical studies, reveals how the LN surface residues interact with each other and how mutations cause failures of self-assembly in an emerging group of diseases, the LN-lamininopathies, that include LAMA2-related dystrophy and Pierson syndrome.
Collapse
Affiliation(s)
- Peter D Yurchenco
- Department of Pathology & Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA.
| | - Arkadiusz W Kulczyk
- Department of Biochemistry and Microbiology, Institute for Quantitative Biomedicine, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
24
|
Ding S, Chen Y, Huang C, Song L, Liang Z, Wei B. Perception and response of skeleton to mechanical stress. Phys Life Rev 2024; 49:77-94. [PMID: 38564907 DOI: 10.1016/j.plrev.2024.03.011] [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: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
Mechanical stress stands as a fundamental factor in the intricate processes governing the growth, development, morphological shaping, and maintenance of skeletal mass. The profound influence of stress in shaping the skeletal framework prompts the assertion that stress essentially births the skeleton. Despite this acknowledgment, the mechanisms by which the skeleton perceives and responds to mechanical stress remain enigmatic. In this comprehensive review, our scrutiny focuses on the structural composition and characteristics of sclerotin, leading us to posit that it serves as the primary structure within the skeleton responsible for bearing and perceiving mechanical stress. Furthermore, we propose that osteocytes within the sclerotin emerge as the principal mechanical-sensitive cells, finely attuned to perceive mechanical stress. And a detailed analysis was conducted on the possible transmission pathways of mechanical stress from the extracellular matrix to the nucleus.
Collapse
Affiliation(s)
- Sicheng Ding
- Department of Minimally invasive spine surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Yiren Chen
- Department of Minimally invasive spine surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Chengshuo Huang
- Department of Minimally invasive spine surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Lijun Song
- Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Zhen Liang
- Department of Minimally invasive spine surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.
| | - Bo Wei
- Department of Minimally invasive spine surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.
| |
Collapse
|
25
|
Kuroda J, Hino H, Kondo S. Dynamics of actinotrichia, fibrous collagen structures in zebrafish fin tissues, unveiled by novel fluorescent probes. PNAS NEXUS 2024; 3:pgae266. [PMID: 39296332 PMCID: PMC11409509 DOI: 10.1093/pnasnexus/pgae266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 06/09/2024] [Indexed: 09/21/2024]
Abstract
Collagen fibers provide physical support to animal tissues by orienting in the correct position and at optimal density. Actinotrichia are thick collagen fibers that are present at the tips of fish fins and serve as scaffolds for bone formation. The arrangement and density of actinotrichia must be constantly maintained with a high degree of regularity to form spatial patterns in the fin bones, but the mechanisms of this process are largely unknown. To address this issue, we first identified two fluorescent probes that can stain actinotrichia clearly in vivo. Using these probes and time-lapse observation of actinotrichia synthesized at different growth stages, we revealed the following previously unknown dynamics of actinotrichia. (i) Actinotrichia do not stay stationary at the place where they are produced; instead, they move towards the dorsal area during the notochord bending and (ii) move towards the distal tip during the fin growth. (iii) Actinotrichia elongate asymmetrically as new collagen is added at the proximal side. (iv) Density is maintained by the insertion of new actinotrichia. (v) Actinotrichia are selectively degraded by osteoclasts. These findings suggest that the regular arrangement of actinotrichia is the outcome of multiple dynamic processes.
Collapse
Affiliation(s)
- Junpei Kuroda
- Laboratory of Pattern Formation, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiromu Hino
- Laboratory of Pattern Formation, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Shigeru Kondo
- Laboratory of Pattern Formation, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
26
|
Revert-Ros F, Ventura I, Prieto-Ruiz JA, Hernández-Andreu JM, Revert F. The Versatility of Collagen in Pharmacology: Targeting Collagen, Targeting with Collagen. Int J Mol Sci 2024; 25:6523. [PMID: 38928229 PMCID: PMC11203716 DOI: 10.3390/ijms25126523] [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: 05/08/2024] [Revised: 06/01/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Collagen, a versatile family of proteins with 28 members and 44 genes, is pivotal in maintaining tissue integrity and function. It plays a crucial role in physiological processes like wound healing, hemostasis, and pathological conditions such as fibrosis and cancer. Collagen is a target in these processes. Direct methods for collagen modulation include enzymatic breakdown and molecular binding approaches. For instance, Clostridium histolyticum collagenase is effective in treating localized fibrosis. Polypeptides like collagen-binding domains offer promising avenues for tumor-specific immunotherapy and drug delivery. Indirect targeting of collagen involves regulating cellular processes essential for its synthesis and maturation, such as translation regulation and microRNA activity. Enzymes involved in collagen modification, such as prolyl-hydroxylases or lysyl-oxidases, are also indirect therapeutic targets. From another perspective, collagen is also a natural source of drugs. Enzymatic degradation of collagen generates bioactive fragments known as matrikines and matricryptins, which exhibit diverse pharmacological activities. Overall, collagen-derived peptides present significant therapeutic potential beyond tissue repair, offering various strategies for treating fibrosis, cancer, and genetic disorders. Continued research into specific collagen targeting and the application of collagen and its derivatives may lead to the development of novel treatments for a range of pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | - Fernando Revert
- Mitochondrial and Molecular Medicine Research Group, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain; (F.R.-R.); (I.V.); (J.A.P.-R.); (J.M.H.-A.)
| |
Collapse
|
27
|
Tvaroška I. Glycosylation Modulates the Structure and Functions of Collagen: A Review. Molecules 2024; 29:1417. [PMID: 38611696 PMCID: PMC11012932 DOI: 10.3390/molecules29071417] [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: 02/26/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Collagens are fundamental constituents of the extracellular matrix and are the most abundant proteins in mammals. Collagens belong to the family of fibrous or fiber-forming proteins that self-assemble into fibrils that define their mechanical properties and biological functions. Up to now, 28 members of the collagen superfamily have been recognized. Collagen biosynthesis occurs in the endoplasmic reticulum, where specific post-translational modification-glycosylation-is also carried out. The glycosylation of collagens is very specific and adds β-d-galactopyranose and β-d-Glcp-(1→2)-d-Galp disaccharide through β-O-linkage to hydroxylysine. Several glycosyltransferases, namely COLGALT1, COLGALT2, LH3, and PGGHG glucosidase, were associated the with glycosylation of collagens, and recently, the crystal structure of LH3 has been solved. Although not fully understood, it is clear that the glycosylation of collagens influences collagen secretion and the alignment of collagen fibrils. A growing body of evidence also associates the glycosylation of collagen with its functions and various human diseases. Recent progress in understanding collagen glycosylation allows for the exploitation of its therapeutic potential and the discovery of new agents. This review will discuss the relevant contributions to understanding the glycosylation of collagens. Then, glycosyltransferases involved in collagen glycosylation, their structure, and catalytic mechanism will be surveyed. Furthermore, the involvement of glycosylation in collagen functions and collagen glycosylation-related diseases will be discussed.
Collapse
Affiliation(s)
- Igor Tvaroška
- Institute of Chemistry, Slovak Academy of Sciences, 845 38 Bratislava, Slovakia
| |
Collapse
|
28
|
Duan C, Yan Z, Wu C, Zhou X, Bao W. DNA methylation characteristics associated with chemotherapy resistance in epithelial ovarian cancer. Heliyon 2024; 10:e27212. [PMID: 38468944 PMCID: PMC10926131 DOI: 10.1016/j.heliyon.2024.e27212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
Objective The high mortality rate of epithelial ovarian cancer (EOC) is often attributed to the frequent development of chemoresistance. DNA methylation is a predictive biomarker for chemoresistance. Methods This study utilized DNA methylation profiles and relevant information from GEO and TCGA to identify different methylated CpG sites (DMCs) between chemoresistant and chemosensitive patients. Subsequently, we constructed chemoresistance risk models with DMCs. The genes corresponding to candidate DMCs in chemoresistance risk models were further analyzed to identify different methylated gene symbols (DMGs) associated with chemoresistance. The DMGs that showed a strong correlation with the corresponding DMCs were analyzed through immunohistochemistry. Results Compared to chemosensitive EOC patients, chemoresistant patients showed 423 hypermethylated CpGs and 1445 hypomethylated CpGs. The chemoresistance risk models based on DMCs have shown the improved predictive ability for chemoresistance in EOC (AUC = 65.0-76.2%). The methylations of cg25510164, cg13154880, cg15362155 and cg08665359 were strongly associated with decreased risk of chemoresistance. Conversely, the methylation of cg08872590 and cg14739437 significantly increased the risk. We identified 13 DMGs, from 47 DMCs corresponding genes, between chemosensitive and chemoresistant samples. Among the DMGs, the expression levels of DDR2 and OPCML exhibited strong correlations with the corresponding DMCs. DDR2 and OPCML both showed enhanced expression in chemoresistant ovarian microarray tissue. Conclusions Hypomethylated CpGs may play a significant role in DNA methylation associated with chemoresistance in EOC. The epigenetic modification of DDR2 could have important implications for the development of chemoresistance. Our study provides valuable insights for future research on DNA methylation in the chemoresistance of EOC.
Collapse
Affiliation(s)
| | | | - Cailiang Wu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080, Shanghai, China
| | - Xuexin Zhou
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080, Shanghai, China
| | - Wei Bao
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080, Shanghai, China
| |
Collapse
|
29
|
Macdonald JK, Mehta AS, Drake RR, Angel PM. Molecular analysis of the extracellular microenvironment: from form to function. FEBS Lett 2024; 598:602-620. [PMID: 38509768 PMCID: PMC11049795 DOI: 10.1002/1873-3468.14852] [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: 11/29/2023] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024]
Abstract
The extracellular matrix (ECM) proteome represents an important component of the tissue microenvironment that controls chemical flux and induces cell signaling through encoded structure. The analysis of the ECM represents an analytical challenge through high levels of post-translational modifications, protease-resistant structures, and crosslinked, insoluble proteins. This review provides a comprehensive overview of the analytical challenges involved in addressing the complexities of spatially profiling the extracellular matrix proteome. A synopsis of the process of synthesizing the ECM structure, detailing inherent chemical complexity, is included to present the scope of the analytical challenge. Current chromatographic and spatial techniques addressing these challenges are detailed. Capabilities for multimodal multiplexing with cellular populations are discussed with a perspective on developing a holistic view of disease processes that includes both the cellular and extracellular microenvironment.
Collapse
Affiliation(s)
- Jade K Macdonald
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC
| | - Anand S Mehta
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC
| | - Richard R Drake
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC
| | - Peggi M. Angel
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
30
|
Subramanian D, Tjahjono N, Hernandez PA, Varner VD, Petroll WM, Schmidtke DW. Fabrication of Micropatterns of Aligned Collagen Fibrils. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:2551-2561. [PMID: 38277615 PMCID: PMC11001481 DOI: 10.1021/acs.langmuir.3c02676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2024]
Abstract
Many tissues in vivo contain aligned structures such as filaments, fibrils, and fibers, which expose cells to anisotropic structural and topographical cues that range from the nanometer to micrometer scales. Understanding how cell behavior is regulated by these cues during physiological and pathological processes (e.g., wound healing, cancer invasion) requires substrates that can expose cells to anisotropic cues over several length scales. In this study, we developed a novel method of fabricating micropatterns of aligned collagen fibrils of different geometry onto PDMS-coated glass coverslips that allowed us to investigate the roles of topography and confinement on corneal cell behavior. When corneal cells were cultured on micropatterns of aligned collagen fibrils in the absence of confinement, the degree of cell alignment increased from 40 ± 14 to 82 ± 5% as the size of the micropattern width decreased from 750 to 50 μm. Although the cell area (∼2500 μm2), cell length (∼160 μm), and projected nuclear area (∼175 μm2) were relatively constant on the different micropattern widths, cells displayed an increased aspect ratio as the width of the aligned collagen fibril micropatterns decreased. We also observed that the morphology of cells adhering to the surrounding uncoated PDMS was dependent upon both the size of the aligned collagen fibril micropattern and the distance from the micropatterns. When corneal cells were confined to the micropatterns of aligned collagen fibrils by a Pluronic coating to passivate the surrounding area, a similar trend in increasing cell alignment was observed (35 ± 10 to 89 ± 2%). However, the projected nuclear area decreased significantly (∼210 to 130 μm2) as the micropattern width decreased from 750 to 50 μm. The development of this method allows for the deposition of aligned collagen fibril micropatterns of different geometries on a transparent and elastic substrate and provides an excellent model system to investigate the role of anisotropic cues in cell behavior.
Collapse
Affiliation(s)
- Divya Subramanian
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX
| | - Nathaniel Tjahjono
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX
| | - Paula A. Hernandez
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center at Dallas, TX, 75390
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center at Dallas, TX, 75390
| | - Victor D. Varner
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX
- Department of Surgery, University of Texas Southwestern Medical Center at Dallas, TX, 75390
| | - W. Matthew Petroll
- Department of Ophthalmology, University of Texas Southwestern Medical Center at Dallas, TX, 75390
| | - David W. Schmidtke
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX
- Department of Surgery, University of Texas Southwestern Medical Center at Dallas, TX, 75390
| |
Collapse
|
31
|
Parker EA, Duchman KR, Meyer AM, Wolf BR, Westermann RW. Menstrual Cycle Hormone Relaxin and ACL Injuries in Female Athletes: A Systematic Review. THE IOWA ORTHOPAEDIC JOURNAL 2024; 44:113-123. [PMID: 38919370 PMCID: PMC11195904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Background Female athletes are at increased risk for anterior cruciate ligament (ACL) injuries. The influence of hormonal variation on female ACL injury risk remains ill-defined. Recent data suggests that the collagen-degrading menstrual hormone relaxin may cyclically impact female ACL tissue quality. This review aims to identify any correlation between menstrual relaxin peaks and rates of female ACL injury. Methods A systematic review was performed, utilizing the MEDLINE, EMBASE, and CINAHL databases. Included studies had to directly address relaxin/female ACL interactions. The primary outcome variable was relaxin proteolysis of the ACL, at cellular, tissue, joint, and whole-organism levels. The secondary outcome variable was any discussed method of moderating relaxin levels, and the clinical results if available. Results AllThe numerous relaxin receptors on female ACLs upregulate local collagenolysis and suppress local collagen production. Peak serum relaxin concentrations (SRC) occur during menstrual cycle days 21-24; a time phase associated with greater risk of ACL injury. Oral contraceptives (OCPs) reduce SRC, with a potential ACLprotective effect. Conclusion A reasonable correlative and plausible causative relationship exists between peak relaxin levels and increased risk of ACL injury in females, and further investigation is warranted. Level of Evidence: III.
Collapse
Affiliation(s)
- Emily A. Parker
- Department of Orthopedics and Rehabilitation, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
- Department of Physical Medicine and Rehabilitation, Case Western Reserve University, Cleveland, Ohio, USA
| | - Kyle R. Duchman
- Department of Orthopedics and Rehabilitation, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Alex M. Meyer
- Department of Orthopedics and Rehabilitation, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Brian R. Wolf
- Department of Orthopedics and Rehabilitation, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Robert W. Westermann
- Department of Orthopedics and Rehabilitation, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| |
Collapse
|
32
|
Salvatore L, Russo F, Natali ML, Rajabimashhadi Z, Bagheri S, Mele C, Lionetto F, Sannino A, Gallo N. On the effect of pepsin incubation on type I collagen from horse tendon: Fine tuning of its physico-chemical and rheological properties. Int J Biol Macromol 2024; 256:128489. [PMID: 38043667 DOI: 10.1016/j.ijbiomac.2023.128489] [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: 07/06/2023] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
Type I collagen is commonly recognized as the gold standard biomaterial for the manufacturing of medical devices for health-care related applications. In recent years, with the final aim of developing scaffolds with optimal bioactivity, even more studies focused on the influence of processing parameters on collagen properties, since processing can strongly affect the architecture of collagen at various length scales and, consequently, scaffolds macroscopic performances. The ability to finely tune scaffold properties in order to closely mimic the tissues' hierarchical features, preserving collagen's natural conformation, is actually of great interest. In this work, the effect of the pepsin-based extraction step on the material final properties was investigated. Thus, the physico-chemical properties of fibrillar type I collagens upon being extracted under various conditions were analyzed in depth. Correlations of collagen structure at the supramolecular scale with its microstructural properties were done, confirming the possibility of tuning rheological, viscoelastic and degradation properties of fibrillar type I collagen.
Collapse
Affiliation(s)
- Luca Salvatore
- Typeone Biomaterials Srl, Via Europa 167, Calimera, 73021 Lecce, Italy.
| | - Francesca Russo
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | | | - Zahra Rajabimashhadi
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Sonia Bagheri
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Claudio Mele
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Francesca Lionetto
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Alessandro Sannino
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Nunzia Gallo
- Typeone Biomaterials Srl, Via Europa 167, Calimera, 73021 Lecce, Italy; Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| |
Collapse
|
33
|
Czarnowska E, Ratajska A, Jankowska-Steifer E, Flaht-Zabost A, Niderla-Bielińska J. Extracellular matrix molecules associated with lymphatic vessels in health and disease. Histol Histopathol 2024; 39:13-34. [PMID: 37350542 DOI: 10.14670/hh-18-641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
Lymphatic vessels (LyVs), responsible for fluid, solute, and immune cell homeostasis in the body, are closely associated with the adjacent extracellular matrix (ECM) molecules whose structural and functional impact on LyVs is currently more appreciated, albeit not entirely elucidated. These molecules, serving as a platform for various connective tissue cell activities and affecting LyV biology should be considered also as an integral part of the lymphatic system. Any alterations and changes in ECM molecules over the course of disease impair the function and structure of the LyV network. Remodeling of LyV cells, which are components of lymphatic vessel walls, also triggers alterations in ECM molecules and interstitial tissue composition. Therefore, in this review we aimed to present the current knowledge on ECM in tissues and particularly on molecules surrounding lymphatics in normal conditions and in disease.
Collapse
Affiliation(s)
| | - Anna Ratajska
- Department of Pathology, Medical University of Warsaw, Warsaw, Poland.
| | - Ewa Jankowska-Steifer
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw, Poland
| | | | | |
Collapse
|
34
|
Aoun M, Coelho A, Krämer A, Saxena A, Sabatier P, Beusch CM, Lönnblom E, Geng M, Do NN, Xu Z, Zhang J, He Y, Romero Castillo L, Abolhassani H, Xu B, Viljanen J, Rorbach J, Fernandez Lahore G, Gjertsson I, Kastbom A, Sjöwall C, Kihlberg J, Zubarev RA, Burkhardt H, Holmdahl R. Antigen-presenting autoreactive B cells activate regulatory T cells and suppress autoimmune arthritis in mice. J Exp Med 2023; 220:e20230101. [PMID: 37695523 PMCID: PMC10494526 DOI: 10.1084/jem.20230101] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 05/31/2023] [Accepted: 08/16/2023] [Indexed: 09/12/2023] Open
Abstract
B cells undergo several rounds of selection to eliminate potentially pathogenic autoreactive clones, but in contrast to T cells, evidence of positive selection of autoreactive B cells remains moot. Using unique tetramers, we traced natural autoreactive B cells (C1-B) specific for a defined triple-helical epitope on collagen type-II (COL2), constituting a sizeable fraction of the physiological B cell repertoire in mice, rats, and humans. Adoptive transfer of C1-B suppressed arthritis independently of IL10, separating them from IL10-secreting regulatory B cells. Single-cell sequencing revealed an antigen processing and presentation signature, including induced expression of CD72 and CCR7 as surface markers. C1-B presented COL2 to T cells and induced the expansion of regulatory T cells in a contact-dependent manner. CD72 blockade impeded this effect suggesting a new downstream suppressor mechanism that regulates antigen-specific T cell tolerization. Thus, our results indicate that autoreactive antigen-specific naïve B cells tolerize infiltrating T cells against self-antigens to impede the development of tissue-specific autoimmune inflammation.
Collapse
Affiliation(s)
- Mike Aoun
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Ana Coelho
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Alexander Krämer
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Amit Saxena
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Pierre Sabatier
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Christian Michel Beusch
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Erik Lönnblom
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Manman Geng
- Precision Medicine Institute, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Nhu-Nguyen Do
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
- Fraunhofer Institute for Translational Medicine and Pharmacology, and Fraunhofer Cluster of Excellence for Immune-Mediated Diseases, Frankfurt am Main, Germany
| | - Zhongwei Xu
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Jingdian Zhang
- Max Planck Institute Biology of Ageing—Karolinska Institute Laboratory, Karolinska Institute, Solna, Sweden
- Division of Molecular Metabolism, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Yibo He
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Laura Romero Castillo
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Hassan Abolhassani
- Division of Clinical Immunology, Department of Biosciences and Nutrition, Karolinska Institutet, Karolinska University Hospital, Neo Building, Solna, Sweden
| | - Bingze Xu
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Johan Viljanen
- Department of Chemistry, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Joanna Rorbach
- Max Planck Institute Biology of Ageing—Karolinska Institute Laboratory, Karolinska Institute, Solna, Sweden
- Division of Molecular Metabolism, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Gonzalo Fernandez Lahore
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Inger Gjertsson
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Alf Kastbom
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Christopher Sjöwall
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Jan Kihlberg
- Department of Chemistry, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Roman A. Zubarev
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
- Department of Pharmacological and Technological Chemistry, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Harald Burkhardt
- Fraunhofer Institute for Translational Medicine and Pharmacology, and Fraunhofer Cluster of Excellence for Immune-Mediated Diseases, Frankfurt am Main, Germany
- Division of Rheumatology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Rikard Holmdahl
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
- Precision Medicine Institute, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
35
|
Boudko SP, Pedchenko VK, Pokidysheva EN, Budko AM, Baugh R, Coates PT, Fidler AL, Hudson HM, Ivanov SV, Luer C, Pedchenko T, Preston RL, Rafi M, Vanacore R, Bhave G, Hudson JK, Hudson BG. Collagen IV of basement membranes: III. Chloride pressure is a primordial innovation that drives and maintains the assembly of scaffolds. J Biol Chem 2023; 299:105318. [PMID: 37797699 PMCID: PMC10656227 DOI: 10.1016/j.jbc.2023.105318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/07/2023] Open
Abstract
Collagen IV scaffold is a primordial innovation enabling the assembly of a fundamental architectural unit of epithelial tissues-a basement membrane attached to polarized cells. A family of six α-chains (α1 to α6) coassemble into three distinct protomers that form supramolecular scaffolds, noted as collagen IVα121, collagen IVα345, and collagen IVα121-α556. Chloride ions play a pivotal role in scaffold assembly, based on studies of NC1 hexamers from mammalian tissues. First, Cl- activates a molecular switch within trimeric NC1 domains that initiates protomer oligomerization, forming an NC1 hexamer between adjoining protomers. Second, Cl- stabilizes the hexamer structure. Whether this Cl--dependent mechanism is of fundamental importance in animal evolution is unknown. Here, we developed a simple in vitro method of SDS-PAGE to determine the role of solution Cl- in hexamer stability. Hexamers were characterized from 34 animal species across 15 major phyla, including the basal Cnidarian and Ctenophora phyla. We found that solution Cl- stabilized the quaternary hexamer structure across all phyla except Ctenophora, Ecdysozoa, and Rotifera. Further analysis of hexamers from peroxidasin knockout mice, a model for decreasing hexamer crosslinks, showed that solution Cl- also stabilized the hexamer surface conformation. The presence of sufficient chloride concentration in solution or "chloride pressure" dynamically maintains the native form of the hexamer. Collectively, our findings revealed that chloride pressure on the outside of cells is a primordial innovation that drives and maintains the quaternary and conformational structure of NC1 hexamers of collagen IV scaffolds.
Collapse
Affiliation(s)
- Sergei P Boudko
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA.
| | - Vadim K Pedchenko
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Elena N Pokidysheva
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Rachel Baugh
- Department of Medical Education and Administration, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Patrick Toby Coates
- Central Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, Australia
| | - Aaron L Fidler
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Heather M Hudson
- Department of Rehabilitation Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sergey V Ivanov
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Carl Luer
- Mote Marine Laboratory, Sarasota, Florida, USA
| | - Tetyana Pedchenko
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Robert L Preston
- School of Biological Sciences, Illinois State University, Normal, Illinois, USA
| | - Mohamed Rafi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Roberto Vanacore
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Gautam Bhave
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Julie K Hudson
- Department of Medical Education and Administration, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Billy G Hudson
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA; Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
36
|
Abtahi S, Chen X, Shahabi S, Nasiri N. Resorbable Membranes for Guided Bone Regeneration: Critical Features, Potentials, and Limitations. ACS MATERIALS AU 2023; 3:394-417. [PMID: 38089090 PMCID: PMC10510521 DOI: 10.1021/acsmaterialsau.3c00013] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 03/22/2024]
Abstract
Lack of horizontal and vertical bone at the site of an implant can lead to significant clinical problems that need to be addressed before implant treatment can take place. Guided bone regeneration (GBR) is a commonly used surgical procedure that employs a barrier membrane to encourage the growth of new bone tissue in areas where bone has been lost due to injury or disease. It is a promising approach to achieve desired repair in bone tissue and is widely accepted and used in approximately 40% of patients with bone defects. In this Review, we provide a comprehensive examination of recent advances in resorbable membranes for GBR including natural materials such as chitosan, collagen, silk fibroin, along with synthetic materials such as polyglycolic acid (PGA), polycaprolactone (PCL), polyethylene glycol (PEG), and their copolymers. In addition, the properties of these materials including foreign body reaction, mechanical stability, antibacterial property, and growth factor delivery performance will be compared and discussed. Finally, future directions for resorbable membrane development and potential clinical applications will be highlighted.
Collapse
Affiliation(s)
- Sara Abtahi
- NanoTech
Laboratory, School of Engineering, Faculty of Science and Engineering, Macquarie University, Sydney 2109, Australia
- Department
of Dental Biomaterials, School of Dentistry, Tehran University of Medical Sciences, Tehran 1416753955, Iran
| | - Xiaohu Chen
- NanoTech
Laboratory, School of Engineering, Faculty of Science and Engineering, Macquarie University, Sydney 2109, Australia
| | - Sima Shahabi
- Department
of Dental Biomaterials, School of Dentistry, Tehran University of Medical Sciences, Tehran 1416753955, Iran
| | - Noushin Nasiri
- NanoTech
Laboratory, School of Engineering, Faculty of Science and Engineering, Macquarie University, Sydney 2109, Australia
| |
Collapse
|
37
|
Boudko SP, Konopka EH, Kim W, Taga Y, Mizuno K, Springer TA, Hudson BG, Moy TI, Lin FY. A recombinant technique for mapping functional sites of heterotrimeric collagen helices: Collagen IV CB3 fragment as a prototype for integrin binding. J Biol Chem 2023; 299:104901. [PMID: 37302550 PMCID: PMC10404678 DOI: 10.1016/j.jbc.2023.104901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023] Open
Abstract
Collagen superfamily of proteins is a major component of the extracellular matrix. Defects in collagens underlie the cause of nearly 40 human genetic diseases in millions of people worldwide. Pathogenesis typically involves genetic alterations of the triple helix, a hallmark structural feature that bestows exceptional mechanical resistance to tensile forces and a capacity to bind a plethora of macromolecules. Yet, there is a paramount knowledge gap in understanding the functionality of distinct sites along the triple helix. Here, we present a recombinant technique to produce triple helical fragments for functional studies. The experimental strategy utilizes the unique capacity of the NC2 heterotrimerization domain of collagen IX to drive three α-chain selection and registering the triple helix stagger. For proof of principle, we produced and characterized long triple helical fragments of collagen IV that were expressed in a mammalian system. The heterotrimeric fragments encompassed the CB3 trimeric peptide of collagen IV, which harbors the binding motifs for α1β1 and α2β1 integrins. Fragments were characterized and shown to have a stable triple helix, post-translational modifications, and high affinity and specific binding of integrins. The NC2 technique is a universal tool for the high-yield production of heterotrimeric fragments of collagens. Fragments are suitable for mapping functional sites, determining coding sequences of binding sites, elucidating pathogenicity and pathogenic mechanisms of genetic mutations, and production of fragments for protein replacement therapy.
Collapse
Affiliation(s)
- Sergei P Boudko
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA.
| | | | - Woojin Kim
- Morphic Therapeutic, Inc, Waltham, Massachusetts, USA
| | - Yuki Taga
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | - Kazunori Mizuno
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | - Timothy A Springer
- Department of Biological Chemistry and Molecular Pharmacology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Billy G Hudson
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Terence I Moy
- Morphic Therapeutic, Inc, Waltham, Massachusetts, USA
| | - Fu-Yang Lin
- Morphic Therapeutic, Inc, Waltham, Massachusetts, USA.
| |
Collapse
|
38
|
Añazco C, Riedelsberger J, Vega-Montoto L, Rojas A. Exploring the Interplay between Polyphenols and Lysyl Oxidase Enzymes for Maintaining Extracellular Matrix Homeostasis. Int J Mol Sci 2023; 24:10985. [PMID: 37446164 PMCID: PMC10342021 DOI: 10.3390/ijms241310985] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Collagen, the most abundant structural protein found in mammals, plays a vital role as a constituent of the extracellular matrix (ECM) that surrounds cells. Collagen fibrils are strengthened through the formation of covalent cross-links, which involve complex enzymatic and non-enzymatic reactions. Lysyl oxidase (LOX) is responsible for catalyzing the oxidative deamination of lysine and hydroxylysine residues, resulting in the production of aldehydes, allysine, and hydroxyallysine. These intermediates undergo spontaneous condensation reactions, leading to the formation of immature cross-links, which are the initial step in the development of mature covalent cross-links. Additionally, non-enzymatic glycation contributes to the formation of abnormal cross-linking in collagen fibrils. During glycation, specific lysine and arginine residues in collagen are modified by reducing sugars, leading to the creation of Advanced Glycation End-products (AGEs). These AGEs have been associated with changes in the mechanical properties of collagen fibers. Interestingly, various studies have reported that plant polyphenols possess amine oxidase-like activity and can act as potent inhibitors of protein glycation. This review article focuses on compiling the literature describing polyphenols with amine oxidase-like activity and antiglycation properties. Specifically, we explore the molecular mechanisms by which specific flavonoids impact or protect the normal collagen cross-linking process. Furthermore, we discuss how these dual activities can be harnessed to generate properly cross-linked collagen molecules, thereby promoting the stabilization of highly organized collagen fibrils.
Collapse
Affiliation(s)
- Carolina Añazco
- Laboratorio de Bioquímica Nutricional, Escuela de Nutrición y Dietética, Carrera de Nutrición y Dietética, Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, General Lagos #1190, Valdivia 5110773, Chile
| | - Janin Riedelsberger
- Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, 1 Poniente 1141, Talca 3462227, Chile;
| | - Lorenzo Vega-Montoto
- Chemical and Radiation Measurement, Idaho National Laboratory (INL), 1705 N. Yellowstone Hwy, Idaho Falls, ID 83415, USA;
| | - Armando Rojas
- Biomedical Research Laboratories, Medicine Faculty, Catholic University of Maule, Talca 3480112, Chile;
| |
Collapse
|
39
|
Santini S, Schenkelaars Q, Jourda C, Duchesne M, Belahbib H, Rocher C, Selva M, Riesgo A, Vervoort M, Leys SP, Kodjabachian L, Le Bivic A, Borchiellini C, Claverie JM, Renard E. The compact genome of the sponge Oopsacas minuta (Hexactinellida) is lacking key metazoan core genes. BMC Biol 2023; 21:139. [PMID: 37337252 DOI: 10.1186/s12915-023-01619-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 05/09/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Explaining the emergence of the hallmarks of bilaterians is a central focus of evolutionary developmental biology-evodevo-and evolutionary genomics. For this purpose, we must both expand and also refine our knowledge of non-bilaterian genomes, especially by studying early branching animals, in particular those in the metazoan phylum Porifera. RESULTS We present a comprehensive analysis of the first whole genome of a glass sponge, Oopsacas minuta, a member of the Hexactinellida. Studying this class of sponge is evolutionary relevant because it differs from the three other Porifera classes in terms of development, tissue organization, ecology, and physiology. Although O. minuta does not exhibit drastic body simplifications, its genome is among the smallest of animal genomes sequenced so far, and surprisingly lacks several metazoan core genes (including Wnt and several key transcription factors). Our study also provides the complete genome of a symbiotic Archaea dominating the associated microbial community: a new Thaumarchaeota species. CONCLUSIONS The genome of the glass sponge O. minuta differs from all other available sponge genomes by its compactness and smaller number of encoded proteins. The unexpected loss of numerous genes previously considered ancestral and pivotal for metazoan morphogenetic processes most likely reflects the peculiar syncytial tissue organization in this group. Our work further documents the importance of convergence during animal evolution, with multiple convergent evolution of septate-like junctions, electrical-signaling and multiciliated cells in metazoans.
Collapse
Affiliation(s)
- Sébastien Santini
- Aix Marseille Univ, CNRS, IGS, UMR 7256, IMM, IM2B, IOM, Marseille, France
| | - Quentin Schenkelaars
- Aix Marseille Univ, Avignon Univ, CNRS, IRD, IMBE, Marseille, France
- Institut Jacques Monod, CNRS, UMR 7592, Univ Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Cyril Jourda
- Aix Marseille Univ, CNRS, IGS, UMR 7256, IMM, IM2B, IOM, Marseille, France
- CIRAD, UMR PVBMT, La Réunion, France
| | - Marc Duchesne
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada
| | - Hassiba Belahbib
- Aix Marseille Univ, CNRS, IGS, UMR 7256, IMM, IM2B, IOM, Marseille, France
| | - Caroline Rocher
- Aix Marseille Univ, Avignon Univ, CNRS, IRD, IMBE, Marseille, France
| | - Marjorie Selva
- Aix Marseille Univ, Avignon Univ, CNRS, IRD, IMBE, Marseille, France
| | - Ana Riesgo
- Department of Biodiversity and Evolutionary Biology, Madrid, Spain
- Department of Life Sciences, Natural History Museum of London, London, SW7 5BD, UK
| | - Michel Vervoort
- Institut Jacques Monod, CNRS, UMR 7592, Univ Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Sally P Leys
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada
| | - Laurent Kodjabachian
- Aix Marseille Univ, CNRS, IBDM, UMR 7288, Turing Center for Living Systems, Marseille, France
| | - André Le Bivic
- Aix Marseille Univ, CNRS, IBDM, UMR 7288, Marseille, France
| | | | | | - Emmanuelle Renard
- Aix Marseille Univ, Avignon Univ, CNRS, IRD, IMBE, Marseille, France.
- Aix Marseille Univ, CNRS, IBDM, UMR 7288, Marseille, France.
| |
Collapse
|
40
|
Ponleitner M, Allmer DM, Hecking M, Gatterer C, Graf S, Smogavec M, Laccone F, Rommer PS, Sunder-Plassmann G. Phenotyping of a novel COL4A4 and novel GLA variant in a patient presenting with microhematuria and mildly impaired kidney function: a case report. Front Genet 2023; 14:1211858. [PMID: 37323669 PMCID: PMC10267447 DOI: 10.3389/fgene.2023.1211858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/18/2023] [Indexed: 06/17/2023] Open
Abstract
We describe the case of a 44-year-old male patient with a longstanding history of microhematuria and mildly impaired kidney function (CKD G2A1). The family history disclosed three females who also had microhematuria. Genetic testing by whole exome sequencing revealed two novel variants in COL4A4 (NM_000092.5: c.1181G>T, NP_000083.3: p.Gly394Val, heterozygous, likely pathogenic; Alport syndrome, OMIM# 141200, 203780) and GLA (NM_000169.3: c.460A>G, NP_000160.1: p.Ile154Val, hemizygous, variant of uncertain significance; Fabry disease, OMIM# 301500), respectively. Extensive phenotyping revealed no biochemical or clinical evidence for the presence of Fabry disease. Thus, the GLA c.460A>G, p.Ile154Val, is to be classified as a benign variant, whereas the COL4A4 c.1181G>T, p.Gly394Val confirms the diagnosis of autosomal dominant Alport syndrome in this patient.
Collapse
Affiliation(s)
- Markus Ponleitner
- Department of Neurology, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Daniela Maria Allmer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Manfred Hecking
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Constantin Gatterer
- Division of Cardiology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Senta Graf
- Division of Cardiology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Mateja Smogavec
- Institute for Human Genetics, Medical University of Vienna, Vienna, Austria
| | - Franco Laccone
- Institute for Human Genetics, Medical University of Vienna, Vienna, Austria
| | - Paulus Stefan Rommer
- Department of Neurology, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Gere Sunder-Plassmann
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
41
|
Park K, Jayadev R, Payne SG, Kenny-Ganzert IW, Chi Q, Costa DS, Ramos-Lewis W, Thendral SB, Sherwood DR. Reciprocal discoidin domain receptor signaling strengthens integrin adhesion to connect adjacent tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532639. [PMID: 36993349 PMCID: PMC10055161 DOI: 10.1101/2023.03.14.532639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Separate tissues connect through adjoining basement membranes to carry out molecular barrier, exchange, and organ support functions. Cell adhesion at these connections must be robust and balanced to withstand independent tissue movement. Yet, how cells achieve synchronized adhesion to connect tissues is unknown. Here, we have investigated this question using the C. elegans utse-seam tissue connection that supports the uterus during egg-laying. Through genetics, quantitative fluorescence, and cell specific molecular disruption, we show that type IV collagen, which fastens the linkage, also activates the collagen receptor discoidin domain receptor 2 (DDR-2) in both the utse and seam. RNAi depletion, genome editing, and photobleaching experiments revealed that DDR-2 signals through LET-60/Ras to coordinately strengthen an integrin adhesion in the utse and seam that stabilizes their connection. These results uncover a synchronizing mechanism for robust adhesion during tissue connection, where collagen both affixes the linkage and signals to both tissues to bolster their adhesion.
Collapse
Affiliation(s)
- Kieop Park
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA
| | - Ranjay Jayadev
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA
| | - Sara G. Payne
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27708, USA
| | | | - Qiuyi Chi
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA
| | - Daniel S. Costa
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA
| | | | | | - David R. Sherwood
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA
- Correspondence:
| |
Collapse
|
42
|
An Update on the Clinical Efficacy and Safety of Collagen Injectables for Aesthetic and Regenerative Medicine Applications. Polymers (Basel) 2023; 15:polym15041020. [PMID: 36850304 PMCID: PMC9963981 DOI: 10.3390/polym15041020] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/19/2023] [Accepted: 02/13/2023] [Indexed: 02/22/2023] Open
Abstract
Soft tissues diseases significantly affect patients quality of life and usually require targeted, costly and sometimes constant interventions. With the average lifetime increase, a proportional increase of age-related soft tissues diseases has been witnessed. Due to this, the last two decades have seen a tremendous demand for minimally invasive one-step resolutive procedures. Intensive scientific and industrial research has led to the recognition of injectable formulations as a new advantageous approach in the management of complex diseases that are challenging to treat with conventional strategies. Among them, collagen-based products are revealed to be one of the most promising among bioactive biomaterials-based formulations. Collagen is the most abundant structural protein of vertebrate connective tissues and, because of its structural and non-structural role, is one of the most widely used multifunctional biomaterials in the health-related sectors, including medical care and cosmetics. Indeed, collagen-based formulations are historically considered as the "gold standard" and from 1981 have been paving the way for the development of a new generation of fillers. A huge number of collagen-based injectable products have been approved worldwide for clinical use and have routinely been introduced in many clinical settings for both aesthetic and regenerative surgery. In this context, this review article aims to be an update on the clinical outcomes of approved collagen-based injectables for both aesthetic and regenerative medicine of the last 20 years with an in-depth focus on their safety and effectiveness for the treatment of diseases of the integumental, gastrointestinal, musculoskeletal, and urogenital apparatus.
Collapse
|
43
|
Pomerleau V, Nicolas VR, Jurkovic CM, Faucheux N, Lauzon MA, Boisvert FM, Perreault N. FOXL1+ Telocytes in mouse colon orchestrate extracellular matrix biodynamics and wound repair resolution. J Proteomics 2023; 271:104755. [PMID: 36272709 DOI: 10.1016/j.jprot.2022.104755] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
Abstract
Recent studies have identified FoxL1+-telocytes (TCFoxL1+) as key players in gut epithelial-mesenchymal interactions which can determine the colonic microenvironment. Bone morphogenetic protein signaling disruption in TCFoxL1+ alters the physical and cellular microenvironment and leads to colon pathophysiology. This suggests a role for TCFoxL1+ in stromagenesis, but it is hard to identify the specific contribution of TCFoxL1+ when analyzing whole tissue profiling studies. We performed ex vivo deconstruction of control and BmpR1a△FoxL1+ colon samples, isolated the mesenchyme-enriched fractions, and determined the protein composition of the in vivo extracellular matrix (ECM) to analyze microenvironment variation. Matrisomic analysis of mesenchyme fractions revealed modulations in ECM proteins with functions associated with innate immunity, epithelial wound healing, and the collagen network. These results show that TCFoxL1+ is critical in orchestrating the biodynamics of the colon ECM. TCFoxL1+ disfunction reprograms the gut's microenvironment and drives the intestinal epithelium toward colonic pathologies. SIGNIFICANCE: In this study, the method that was elected to isolate ECM proteins might not encompass the full extent of ECM proteins in a tissue, due to the protocol chosen, as this protocol by Naba et al., targets more the insoluble part of the matrisome and eliminates the more soluble components in the first steps. However, this ECM-enrichment strategy represents an improvement and interesting avenue to study ECM proteins in the colon compared to total tissue analysis with a background of abundant cellular protein. Thus, the matrisomic approach presented in this study, and its target validation delivered a broader evaluation of the matrix remodeling occurring in the colonic sub-epithelial mesenchyme of the BmpR1a△FoxL1+ mouse model.
Collapse
Affiliation(s)
- Véronique Pomerleau
- Département d'Immunologie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Vilcy Reyes Nicolas
- Département d'Immunologie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Carla-Marie Jurkovic
- Département d'Immunologie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Nathalie Faucheux
- Département de génie chimique et de génie biotechnologique, Faculté de Génie, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Marc-Antoine Lauzon
- Département de génie chimique et de génie biotechnologique, Faculté de Génie, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - François-Michel Boisvert
- Département d'Immunologie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Nathalie Perreault
- Département d'Immunologie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
44
|
Ma L, Liang X, Yu S, Zhou J. Expression, characterization, and application potentiality evaluation of recombinant human-like collagen in Pichia pastoris. BIORESOUR BIOPROCESS 2022; 9:119. [PMID: 38647896 PMCID: PMC10992492 DOI: 10.1186/s40643-022-00606-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/20/2022] [Indexed: 11/18/2022] Open
Abstract
Collagen is a biofunctional protein that has been widely used in many fields, including but not limited to biomedical, cosmetics and skin care, food, and novel materials. Recombinant collagen has great potential as an alternative to collagen extracted from animals because it avoids the immune response, and the yield and properties are stable. However, challenges remain in the industrial application of recombinant collagen, including improving the expression yield, reducing the cost of purification for industry and expanding applications. In this study, a cloning and recombination method was used to heterologously express the recombinant human-like collagen (RHLC) in Pichia pastoris GS115 using the pPIC9k expression vector. The RHLC expression titre was 2.33 g/L via a 5-L fermenter, and the purification was completed within 48 h and was 98% pure. The characteristics of RHLC were investigated. Furthermore, potential applications for RHLC were explored, such as basal collagen sponge preparation, forming films with chitosan and production of collagen hydrolysed peptides. RHLC has various potential applications due to its triple helical structure, thermostability, good biocompatibility and film-forming ability.
Collapse
Affiliation(s)
- Lingling Ma
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education and School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
| | - Xiaolin Liang
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education and School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
| | - Shiqin Yu
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education and School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
- Engineering Research Center of Ministry of Education On Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Jingwen Zhou
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China.
- Key Laboratory of Industrial Biotechnology, Ministry of Education and School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China.
- Engineering Research Center of Ministry of Education On Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China.
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
45
|
Zhang W, Zhao J, Yang D. Anion-Coordination-Driven Assembly: From Discrete Supramolecular Self-Assemblies to Functional Soft Materials. Chempluschem 2022; 87:e202200294. [PMID: 36410745 DOI: 10.1002/cplu.202200294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/18/2022] [Indexed: 01/31/2023]
Abstract
Anion templated assembly of supramolecular systems has been extensively explored in previous reports, whereas anions serve only as an auxiliary and spectator role. With the development of anion coordination chemistry in recent years, anion coordination-driven assembly (ACDA) has emerged as a new strategy for the construction of supramolecular self-assemblies. Anions are proved to exist as the main actors in the construction of supramolecular architectures, i. e., serve as the coordination center. This Review will focus on the recent progress in anion-coordination-driven assembly of discrete supramolecular architectures, such as helicates, polyhedrons and polygons, and the various applications of 'aniono'-systems. At the end of this Review, we highlight current challenges and opportunities for future research of anion-coordination-driven self-assembly.
Collapse
Affiliation(s)
- Wenyao Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710069, P. R. China.,Key Laboratory of Magnetic Molecules and Magnetic Information Materials (Ministry of Education), School of Chemistry and Material Science, Shanxi Normal University, Taiyuan, 030006, P. R. China
| | - Jie Zhao
- School of Chemistry and Chemical Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, P. R. China
| | - Dong Yang
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710069, P. R. China
| |
Collapse
|
46
|
Bahr JC, Li XY, Feinberg TY, Jiang L, Weiss SJ. Divergent regulation of basement membrane trafficking by human macrophages and cancer cells. Nat Commun 2022; 13:6409. [PMID: 36302921 PMCID: PMC9613642 DOI: 10.1038/s41467-022-34087-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/13/2022] [Indexed: 12/25/2022] Open
Abstract
Macrophages and cancer cells populations are posited to navigate basement membrane barriers by either mobilizing proteolytic enzymes or deploying mechanical forces. Nevertheless, the relative roles, or identity, of the proteinase -dependent or -independent mechanisms used by macrophages versus cancer cells to transmigrate basement membrane barriers harboring physiologically-relevant covalent crosslinks remains ill-defined. Herein, both macrophages and cancer cells are shown to mobilize membrane-anchored matrix metalloproteinases to proteolytically remodel native basement membranes isolated from murine tissues while infiltrating the underlying interstitial matrix ex vivo. In the absence of proteolytic activity, however, only macrophages deploy actomyosin-generated forces to transmigrate basement membrane pores, thereby providing the cells with proteinase-independent access to the interstitial matrix while simultaneously exerting global effects on the macrophage transcriptome. By contrast, cancer cell invasive activity is reliant on metalloproteinase activity and neither mechanical force nor changes in nuclear rigidity rescue basement membrane transmigration. These studies identify membrane-anchored matrix metalloproteinases as key proteolytic effectors of basement membrane remodeling by macrophages and cancer cells while also defining the divergent invasive strategies used by normal and neoplastic cells to traverse native tissue barriers.
Collapse
Affiliation(s)
- Julian C Bahr
- Cancer Biology Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xiao-Yan Li
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Tamar Y Feinberg
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Long Jiang
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Stephen J Weiss
- Cancer Biology Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA.
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
47
|
Gianakas CA, Keeley DP, Ramos-Lewis W, Park K, Jayadev R, Kenny IW, Chi Q, Sherwood DR. Hemicentin-mediated type IV collagen assembly strengthens juxtaposed basement membrane linkage. J Cell Biol 2022; 222:213571. [PMID: 36282214 PMCID: PMC9597354 DOI: 10.1083/jcb.202112096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 08/16/2022] [Accepted: 09/26/2022] [Indexed: 01/07/2023] Open
Abstract
Basement membrane (BM) matrices surround and separate most tissues. However, through poorly understood mechanisms, BMs of adjacent tissue can also stably link to support organ structure and function. Using endogenous knock-in fluorescent proteins, conditional RNAi, optogenetics, and quantitative live imaging, we identified extracellular matrix proteins mediating a BM linkage (B-LINK) between the uterine utse and epidermal seam cell BMs in Caenorhabditis elegans that supports the uterus during egg-laying. We found that hemicentin is secreted by the utse and promotes fibulin-1 assembly to jointly initiate the B-LINK. During egg-laying, however, both proteins' levels decline and are not required for B-LINK maintenance. Instead, we discovered that hemicentin recruits ADAMTS9/20, which facilitates the assembly of high levels of type IV collagen that sustains the B-LINK during the mechanically active egg-laying period. This work reveals mechanisms underlying BM-BM linkage maturation and identifies a crucial function for hemicentin and fibulin-1 in initiating attachment and type IV collagen in strengthening this specialized form of tissue linkage.
Collapse
Affiliation(s)
- Claire A. Gianakas
- Department of Biology, Duke University, Durham, NC,Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | | | | | - Kieop Park
- Department of Biology, Duke University, Durham, NC
| | | | | | - Qiuyi Chi
- Department of Biology, Duke University, Durham, NC
| | - David R. Sherwood
- Department of Biology, Duke University, Durham, NC,Department of Pharmacology and Cancer Biology, Duke University, Durham, NC,Correspondence to David R. Sherwood:
| |
Collapse
|
48
|
Discovering design principles of collagen molecular stability using a genetic algorithm, deep learning, and experimental validation. Proc Natl Acad Sci U S A 2022; 119:e2209524119. [PMID: 36161946 DOI: 10.1073/pnas.2209524119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Collagen is the most abundant structural protein in humans, providing crucial mechanical properties, including high strength and toughness, in tissues. Collagen-based biomaterials are, therefore, used for tissue repair and regeneration. Utilizing collagen effectively during materials processing ex vivo and subsequent function in vivo requires stability over wide temperature ranges to avoid denaturation and loss of structure, measured as melting temperature (Tm). Although significant research has been conducted on understanding how collagen primary amino acid sequences correspond to Tm values, a robust framework to facilitate the design of collagen sequences with specific Tm remains a challenge. Here, we develop a general model using a genetic algorithm within a deep learning framework to design collagen sequences with specific Tm values. We report 1,000 de novo collagen sequences, and we show that we can efficiently use this model to generate collagen sequences and verify their Tm values using both experimental and computational methods. We find that the model accurately predicts Tm values within a few degrees centigrade. Further, using this model, we conduct a high-throughput study to identify the most frequently occurring collagen triplets that can be directly incorporated into collagen. We further discovered that the number of hydrogen bonds within collagen calculated with molecular dynamics (MD) is directly correlated to the experimental measurement of triple-helical quality. Ultimately, we see this work as a critical step to helping researchers develop collagen sequences with specific Tm values for intended materials manufacturing methods and biomedical applications, realizing a mechanistic materials by design paradigm.
Collapse
|
49
|
Puszkarska AM, Frenkel D, Colwell LJ, Duer MJ. Using sequence data to predict the self-assembly of supramolecular collagen structures. Biophys J 2022; 121:3023-3033. [PMID: 35859421 PMCID: PMC9463645 DOI: 10.1016/j.bpj.2022.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/25/2022] [Accepted: 07/12/2022] [Indexed: 11/02/2022] Open
Abstract
Collagen fibrils are the major constituents of the extracellular matrix, which provides structural support to vertebrate connective tissues. It is widely assumed that the superstructure of collagen fibrils is encoded in the primary sequences of the molecular building blocks. However, the interplay between large-scale architecture and small-scale molecular interactions makes the ab initio prediction of collagen structure challenging. Here, we propose a model that allows us to predict the periodic structure of collagen fibers and the axial offset between the molecules, purely on the basis of simple predictive rules for the interaction between amino acid residues. With our model, we identify the sequence-dependent collagen fiber geometries with the lowest free energy and validate the predicted geometries against the available experimental data. We propose a procedure for searching for optimal staggering distances. Finally, we build a classification algorithm and use it to scan 11 data sets of vertebrate fibrillar collagens, and predict the periodicity of the resulting assemblies. We analyzed the experimentally observed variance of the optimal stagger distances across species, and find that these distances, and the resulting fibrillar phenotypes, are evolutionary well preserved. Moreover, we observed that the energy minimum at the optimal stagger distance is broad in all cases, suggesting a further evolutionary adaptation designed to improve the assembly kinetics. Our periodicity predictions are not only in good agreement with the experimental data on collagen molecular staggering for all collagen types analyzed, but also for synthetic peptides. We argue that, with our model, it becomes possible to design tailor-made, periodic collagen structures, thereby enabling the design of novel biomimetic materials based on collagen-mimetic trimers.
Collapse
Affiliation(s)
- Anna M Puszkarska
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Daan Frenkel
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Lucy J Colwell
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom; Google Research, Mountain View, California
| | - Melinda J Duer
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
50
|
The Role of the Extracellular Matrix (ECM) in Wound Healing: A Review. Biomimetics (Basel) 2022; 7:biomimetics7030087. [PMID: 35892357 PMCID: PMC9326521 DOI: 10.3390/biomimetics7030087] [Citation(s) in RCA: 167] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/23/2022] [Accepted: 06/29/2022] [Indexed: 12/27/2022] Open
Abstract
The extracellular matrix (ECM) is a 3-dimensional structure and an essential component in all human tissues. It is comprised of varying proteins, including collagens, elastin, and smaller quantities of structural proteins. Studies have demonstrated the ECM aids in cellular adherence, tissue anchoring, cellular signaling, and recruitment of cells. During times of integumentary injury or damage, either acute or chronic, the ECM is damaged. Through a series of overlapping events called the wound healing phases—hemostasis, inflammation, proliferation, and remodeling—the ECM is synthesized and ideally returned to its native state. This article synthesizes current and historical literature to demonstrate the involvement of the ECM in the varying phases of the wound healing cascade.
Collapse
|