1
|
Lodewijk GA, Kozuki S, Han CJ, Topacio BR, Lee S, Nixon L, Zargari A, Knight G, Ashton R, Qi LS, Shariati SA. Self-organization of mouse embryonic stem cells into reproducible pre-gastrulation embryo models via CRISPRa programming. Cell Stem Cell 2025:S1934-5909(25)00083-9. [PMID: 40118066 DOI: 10.1016/j.stem.2025.02.015] [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: 03/05/2024] [Revised: 12/17/2024] [Accepted: 02/26/2025] [Indexed: 03/23/2025]
Abstract
Embryonic stem cells (ESCs) can self-organize into structures with spatial and molecular similarities to natural embryos. During development, embryonic and extraembryonic cells differentiate through activation of endogenous regulatory elements while co-developing via cell-cell interactions. However, engineering regulatory elements to self-organize ESCs into embryo models remains underexplored. Here, we demonstrate that CRISPR activation (CRISPRa) of two regulatory elements near Gata6 and Cdx2 generates embryonic patterns resembling pre-gastrulation mouse embryos. Live single-cell imaging revealed that self-patterning occurs through orchestrated collective movement driven by cell-intrinsic fate induction. In 3D, CRISPRa-programmed embryo models (CPEMs) exhibit morphological and transcriptomic similarity to pre-gastrulation mouse embryos. CPEMs allow versatile perturbations, including dual Cdx2-Elf5 activation to enhance trophoblast differentiation and lineage-specific activation of laminin and matrix metalloproteinases, uncovering their roles in basement membrane remodeling and embryo model morphology. Our findings demonstrate that minimal intrinsic epigenome editing can self-organize ESCs into programmable pre-gastrulation embryo models with robust lineage-specific perturbation capabilities.
Collapse
Affiliation(s)
- Gerrald A Lodewijk
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA; Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA; Institute for The Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Sayaka Kozuki
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA; Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA; Institute for The Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Clara J Han
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA; Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA; Institute for The Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Benjamin R Topacio
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA; Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA; Institute for The Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Seungho Lee
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA; Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA; Institute for The Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Lily Nixon
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA; Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA; Institute for The Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Abolfazl Zargari
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Gavin Knight
- Neurosetta LLC, Madison, WI, USA; Wisconsin Institute for Discovery, Madison, WI, USA
| | - Randolph Ashton
- Neurosetta LLC, Madison, WI, USA; Wisconsin Institute for Discovery, Madison, WI, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Lei S Qi
- Department of Bioengineering, Stanford University, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Chan Zuckerberg Biohub, San Francisco, San Francisco, CA, USA
| | - S Ali Shariati
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA; Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA; Institute for The Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA, USA.
| |
Collapse
|
2
|
Belančić A, Janković T, Gkrinia EMM, Kristić I, Rajič Bumber J, Rački V, Pilipović K, Vitezić D, Mršić-Pelčić J. Glial Cells in Spinal Muscular Atrophy: Speculations on Non-Cell-Autonomous Mechanisms and Therapeutic Implications. Neurol Int 2025; 17:41. [PMID: 40137462 PMCID: PMC11944370 DOI: 10.3390/neurolint17030041] [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: 02/09/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by homozygous deletions or mutations in the SMN1 gene, leading to progressive motor neuron degeneration. While SMA has been classically viewed as a motor neuron-autonomous disease, increasing evidence indicates a significant role of glial cells-astrocytes, microglia, oligodendrocytes, and Schwann cells-in the disease pathophysiology. Astrocytic dysfunction contributes to motor neuron vulnerability through impaired calcium homeostasis, disrupted synaptic integrity, and neurotrophic factor deficits. Microglia, through reactive gliosis and complement-mediated synaptic stripping, exacerbate neurodegeneration and neuroinflammation. Oligodendrocytes exhibit impaired differentiation and metabolic support, while Schwann cells display abnormalities in myelination, extracellular matrix composition, and neuromuscular junction maintenance, further compromising motor function. Dysregulation of pathways such as NF-κB, Notch, and JAK/STAT, alongside the upregulation of complement proteins and microRNAs, reinforces the non-cell-autonomous nature of SMA. Despite the advances in SMN-restorative therapies, they do not fully mitigate glial dysfunction. Targeting glial pathology, including modulation of reactive astrogliosis, microglial polarization, and myelination deficits, represents a critical avenue for therapeutic intervention. This review comprehensively examines the multifaceted roles of glial cells in SMA and highlights emerging glia-targeted strategies to enhance treatment efficacy and improve patient outcomes.
Collapse
Affiliation(s)
- Andrej Belančić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Tamara Janković
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | | | - Iva Kristić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Jelena Rajič Bumber
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Valentino Rački
- Department of Neurology, Clinical Hospital Centre Rijeka, Krešimirova 42, 51000 Rijeka, Croatia;
| | - Kristina Pilipović
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Dinko Vitezić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Jasenka Mršić-Pelčić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| |
Collapse
|
3
|
Zhang L, Zheng G, Zhao W, He C, Huang Z. Maxing Yigan formula promotes cartilage regeneration by regulating chondrocyte autophagy in osteoarthritis. In Vitro Cell Dev Biol Anim 2025; 61:268-274. [PMID: 39718676 DOI: 10.1007/s11626-024-01006-x] [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: 10/24/2024] [Accepted: 12/06/2024] [Indexed: 12/25/2024]
Abstract
Maxing Yigan formula (MYF) is a traditional Chinese medicine (TCM) prescription used for the treatment of OA for decades in China. However, the mechanism remains unknown. In this study, we developed a MYF-incorporated collagen sponge (MYF@CS) and investigated its cartilage regeneration effect and the underlying mechanism. In vitro experiments revealed that MYF significantly promoted cell viability, proliferation, and autophagy of OA chondrocytes. Furthermore, MYF@CS significantly enhanced chondrogenesis and cartilage regeneration, as assessed by macroscopic observation, the International Cartilage Repair Society (ICRS) visual histological score, and histological examination. Our findings suggest that MYF@CS could represent a significant therapeutic strategy for the treatment of OA.
Collapse
Affiliation(s)
- Liqin Zhang
- The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China
| | - Guangping Zheng
- Ganzhou Municipal Key Laboratory of Bone and Joint Research, The Affiliated Ganzhou Hospital of Nanchang University, Meiguan Avenue No. 16, Ganzhou, 341000, China
| | - Weicheng Zhao
- The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China.
- Ganzhou Municipal Key Laboratory of Bone and Joint Research, The Affiliated Ganzhou Hospital of Nanchang University, Meiguan Avenue No. 16, Ganzhou, 341000, China.
| | - Chun He
- The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China.
| | - Zhongming Huang
- The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China.
- Ganzhou Municipal Key Laboratory of Bone and Joint Research, The Affiliated Ganzhou Hospital of Nanchang University, Meiguan Avenue No. 16, Ganzhou, 341000, China.
| |
Collapse
|
4
|
Sun L, Shen Y, Li M, Wang Q, Li R, Gong S. Comprehensive Assessment of Collagen/Sodium Alginate-Based Sponges as Hemostatic Dressings. Molecules 2024; 29:2999. [PMID: 38998951 PMCID: PMC11243721 DOI: 10.3390/molecules29132999] [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: 05/24/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
In our search for a biocompatible composite hemostatic dressing, we focused on the design of a novel biomaterial composed of two natural biological components, collagen and sodium alginate (SA), cross-linked using 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide/N-hydroxysuccinimide (EDC/NHS) and oxidized sodium alginate (OSA). We conducted a series of tests to evaluate the physicochemical properties, acute systemic toxicity, skin irritation, intradermal reaction, sensitization, cytotoxicity, and in vivo femoral artery hemorrhage model. The results demonstrated the excellent biocompatibility of the collagen/sodium alginate (C/SA)-based dressings before and after crosslinking. Specifically, the femoral artery hemorrhage model revealed a significantly shortened hemostasis time of 132.5 ± 12.82 s for the EDC/NHS cross-linked dressings compared to the gauze in the blank group (hemostasis time of 251.43 ± 10.69 s). These findings indicated that C/SA-based dressings exhibited both good biocompatibility and a significant hemostatic effect, making them suitable for biomedical applications.
Collapse
Affiliation(s)
- Leilei Sun
- College of Life Science, Yantai University, Yantai 264005, China; (Y.S.); (M.L.); (Q.W.); (R.L.); (S.G.)
| | | | | | | | | | | |
Collapse
|
5
|
Fooladi S, Nematollahi MH, Rabiee N, Iravani S. Bacterial Cellulose-Based Materials: A Perspective on Cardiovascular Tissue Engineering Applications. ACS Biomater Sci Eng 2023. [PMID: 37146213 DOI: 10.1021/acsbiomaterials.3c00300] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Today, a wide variety of bio- and nanomaterials have been deployed for cardiovascular tissue engineering (TE), including polymers, metal oxides, graphene/its derivatives, organometallic complexes/composites based on inorganic-organic components, among others. Despite several advantages of these materials with unique mechanical, biological, and electrical properties, some challenges still remain pertaining to their biocompatibility, cytocompatibility, and possible risk factors (e.g., teratogenicity or carcinogenicity), restricting their future clinical applications. Natural polysaccharide- and protein-based (nano)structures with the benefits of biocompatibility, sustainability, biodegradability, and versatility have been exploited in the field of cardiovascular TE focusing on targeted drug delivery, vascular grafts, engineered cardiac muscle, etc. The usage of these natural biomaterials and their residues offers several advantages in terms of environmental aspects such as alleviating emission of greenhouse gases as well as the production of energy as a biomass consumption output. In TE, the development of biodegradable and biocompatible scaffolds with potentially three-dimensional structures, high porosity, and suitable cellular attachment/adhesion still needs to be comprehensively studied. In this context, bacterial cellulose (BC) with high purity, porosity, crystallinity, unique mechanical properties, biocompatibility, high water retention, and excellent elasticity can be considered as promising candidate for cardiovascular TE. However, several challenges/limitations regarding the absence of antimicrobial factors and degradability along with the low yield of production and extensive cultivation times (in large-scale production) still need to be resolved using suitable hybridization/modification strategies and optimization of conditions. The biocompatibility and bioactivity of BC-based materials along with their thermal, mechanical, and chemical stability are crucial aspects in designing TE scaffolds. Herein, cardiovascular TE applications of BC-based materials are deliberated, with a focus on the most recent advancements, important challenges, and future perspectives. Other biomaterials with cardiovascular TE applications and important roles of green nanotechnology in this field of science are covered to better compare and comprehensively review the subject. The application of BC-based materials and the collective roles of such biomaterials in the assembly of sustainable and natural-based scaffolds for cardiovascular TE are discussed.
Collapse
Affiliation(s)
- Saba Fooladi
- Department of Clinical Biochemistry, Afzalipour Medical School, Kerman University of Medical Sciences, 76169-13555 Kerman, Iran
| | - Mohammad Hadi Nematollahi
- Department of Clinical Biochemistry, Afzalipour Medical School, Kerman University of Medical Sciences, 76169-13555 Kerman, Iran
- Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, 76169-13555 Kerman, Iran
| | - Navid Rabiee
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Western Australia 6150, Australia
- School of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Siavash Iravani
- Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, 81746-73461 Isfahan, Iran
| |
Collapse
|
6
|
Zhao P, Dang Z, Liu M, Guo D, Luo R, Zhang M, Xie F, Zhang X, Wang Y, Pan S, Ma X. Molecular hydrogen promotes wound healing by inducing early epidermal stem cell proliferation and extracellular matrix deposition. Inflamm Regen 2023; 43:22. [PMID: 36973725 PMCID: PMC10044764 DOI: 10.1186/s41232-023-00271-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/26/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Despite progress in developing wound care strategies, there is currently no treatment that promotes the self-tissue repair capabilities. H2 has been shown to effectively protect cells and tissues from oxidative and inflammatory damage. While comprehensive effects and how H2 functions in wound healing remains unknown, especially for the link between H2 and extracellular matrix (ECM) deposition and epidermal stem cells (EpSCs) activation. METHODS Here, we established a cutaneous aseptic wound model and applied a high concentration of H2 (66% H2) in a treatment chamber. Molecular mechanisms and the effects of healing were evaluated by gene functional enrichment analysis, digital spatial profiler analysis, blood perfusion/oxygen detection assay, in vitro tube formation assay, enzyme-linked immunosorbent assay, immunofluorescent staining, non-targeted metabonomic analysis, flow cytometry, transmission electron microscope, and live-cell imaging. RESULTS We revealed that a high concentration of H2 (66% H2) greatly increased the healing rate (3 times higher than the control group) on day 11 post-wounding. The effect was not dependent on O2 or anti-reactive oxygen species functions. Histological and cellular experiments proved the fast re-epithelialization in the H2 group. ECM components early (3 days post-wounding) deposition were found in the H2 group of the proximal wound, especially for the dermal col-I, epidermal col-III, and dermis-epidermis-junction col-XVII. H2 accelerated early autologous EpSCs proliferation (1-2 days in advance) and then differentiation into myoepithelial cells. These epidermal myoepithelial cells could further contribute to ECM deposition. Other beneficial outcomes include sustained moist healing, greater vascularization, less T-helper-1 and T-helper-17 cell-related systemic inflammation, and better tissue remodelling. CONCLUSION We have discovered a novel pattern of wound healing induced by molecular hydrogen treatment. This is the first time to reveal the direct link between H2 and ECM deposition and EpSCs activation. These H2-induced multiple advantages in healing may be related to the enhancement of cell viability in various cells and the maintenance of mitochondrial functions at a basic level in the biological processes of life.
Collapse
Affiliation(s)
- Pengxiang Zhao
- Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, People's Republic of China
- Beijing Molecular Hydrogen Research Center, Beijing, 100124, People's Republic of China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing, 100124, People's Republic of China
| | - Zheng Dang
- Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, People's Republic of China
- Beijing Molecular Hydrogen Research Center, Beijing, 100124, People's Republic of China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing, 100124, People's Republic of China
| | - Mengyu Liu
- Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, People's Republic of China
- Beijing Molecular Hydrogen Research Center, Beijing, 100124, People's Republic of China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing, 100124, People's Republic of China
| | - Dazhi Guo
- Department of Hyperbaric Oxygen, Sixth Medical Center of PLA General Hospital, Beijing, 100048, People's Republic of China
| | - Ruiliu Luo
- Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, People's Republic of China
- Beijing Molecular Hydrogen Research Center, Beijing, 100124, People's Republic of China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing, 100124, People's Republic of China
| | - Mingzi Zhang
- Department of Plastic Surgery, Peking Union Medical College Hospital (Dongdan campus), No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100730, People's Republic of China
| | - Fei Xie
- Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, People's Republic of China
- Beijing Molecular Hydrogen Research Center, Beijing, 100124, People's Republic of China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing, 100124, People's Republic of China
| | - Xujuan Zhang
- Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, People's Republic of China
- Beijing Molecular Hydrogen Research Center, Beijing, 100124, People's Republic of China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing, 100124, People's Republic of China
| | - Youbin Wang
- Department of Plastic Surgery, Peking Union Medical College Hospital (Dongdan campus), No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100730, People's Republic of China
| | - Shuyi Pan
- Department of Hyperbaric Oxygen, Sixth Medical Center of PLA General Hospital, Beijing, 100048, People's Republic of China
| | - Xuemei Ma
- Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, People's Republic of China.
- Beijing Molecular Hydrogen Research Center, Beijing, 100124, People's Republic of China.
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing, 100124, People's Republic of China.
| |
Collapse
|
7
|
Nissen NI, Johansen AZ, Chen IM, Jensen C, Madsen EA, Hansen CP, Thorlacius-Ussing J, Karsdal M, Johansen JS, Diab HMH, Jørgensen LN, Willumsen N. High serum levels of the C-propetide of type V collagen (PRO-C5) are prognostic for short overall survival in patients with pancreatic ductal adenocarcinoma. Front Mol Biosci 2023; 10:1158058. [PMID: 36968276 PMCID: PMC10036831 DOI: 10.3389/fmolb.2023.1158058] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 02/23/2023] [Indexed: 03/12/2023] Open
Abstract
Introduction: Pancreatic ductal adenocarcinoma (PDAC) is characterized by a pronounced fibrotic tumor microenvironment, which impairs treatment response. Type I and V collagens are responsible for the densely packed fibrils in the tumor fibrosis environment. While the role of the major type I collagen in cancer is well described, less is known about the minor type V collagen. Quantifying collagen propeptides in serum has been shown to have prognostic and predictive value. In this study, we evaluated the clinical utility of measuring the propeptide of type V collagen (PRO-C5) in serum from a discovery cohort and a validation cohort of patients with PDAC as well as in non-pancreatic solid tumor types to explore the relevance of the PRO-C5 biomarker in cancer.Methods: Serum PRO-C5 was measured in three cohorts: a discovery cohort (19 healthy controls, 12 patients with chronic pancreatitis and 33 patients with PDAC (stage I-IV)), a validation cohort (800 patients with PDAC (stage I-IV)), and a non-pancreatic solid tumor type cohort of 33 healthy controls and 200 patients with 10 different non-pancreatic solid tumor types. The levels of serum PRO-C5 in patients with cancer were compared to levels in healthy controls. The association between PRO-C5 levels and overall survival (OS) was evaluated in patients with PDAC after adjusting for established prognostic factors.Results: PRO-C5 was significantly increased in serum from patients with PDAC compared to healthy controls (p < 0.001). High PRO-C5 levels were significantly associated with short OS in both the discovery- and the validation cohort, especially in early stages of PDAC (validation cohort stage II, HR = 2.0, 95%CI1.2-3.4). The association was independent of other prognostic parameters including stage, performance status and CA19-9. Furthermore, serum levels of PRO-C5 were significantly increased in serum from patients with other non-pancreatic solid tumor types compared to healthy controls.Conclusion: High levels of serum PRO-C5 is prognostic for short OS in patients with PDAC and may provide clinical value in many other tumor types beyond PDAC. This underlines the importance of type V collagen in tumor fibrosis. PRO-C5 could have the potential to be used in several aspects within drug discovery, patient stratification and drug efficacy.
Collapse
Affiliation(s)
- Neel I. Nissen
- Nordic Bioscience A/S, Herlev, Denmark
- *Correspondence: Neel I. Nissen,
| | - Astrid Z. Johansen
- Department of Oncology, Copenhagen University Hospital, Gentofte, Denmark
| | - Inna M. Chen
- Department of Oncology, Copenhagen University Hospital, Gentofte, Denmark
| | | | | | - Carsten P. Hansen
- Department of Surgery, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
| | | | | | - Julia S. Johansen
- Department of Oncology, Copenhagen University Hospital, Gentofte, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Medicine, Copenhagen University Hospital, Gentofte, Denmark
| | - Hadi M. H. Diab
- Digestive Disease Center, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Lars N. Jørgensen
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Digestive Disease Center, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
8
|
Yang GH, Kang D, An S, Ryu JY, Lee K, Kim JS, Song MY, Kim YS, Kwon SM, Jung WK, Jeong W, Jeon H. Advances in the development of tubular structures using extrusion-based 3D cell-printing technology for vascular tissue regenerative applications. Biomater Res 2022; 26:73. [PMID: 36471437 PMCID: PMC9720982 DOI: 10.1186/s40824-022-00321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 11/13/2022] [Indexed: 12/11/2022] Open
Abstract
Until recent, there are no ideal small diameter vascular grafts available on the market. Most of the commercialized vascular grafts are used for medium to large-sized blood vessels. As a solution, vascular tissue engineering has been introduced and shown promising outcomes. Despite these optimistic results, there are limitations to commercialization. This review will cover the need for extrusion-based 3D cell-printing technique capable of mimicking the natural structure of the blood vessel. First, we will highlight the physiological structure of the blood vessel as well as the requirements for an ideal vascular graft. Then, the essential factors of 3D cell-printing including bioink, and cell-printing system will be discussed. Afterwards, we will mention their applications in the fabrication of tissue engineered vascular grafts. Finally, conclusions and future perspectives will be discussed.
Collapse
Affiliation(s)
- Gi Hoon Yang
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-Do 15588 South Korea
| | - Donggu Kang
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-Do 15588 South Korea
| | - SangHyun An
- Preclinical Research Center, K Medi-hub, 80 Cheombok-ro, Dong-gu, Daegu, 41061 South Korea
| | - Jeong Yeop Ryu
- grid.258803.40000 0001 0661 1556Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, 130 Dongdeok‑ro, Jung‑gu, Daegu, 41944 South Korea
| | - KyoungHo Lee
- Preclinical Research Center, K Medi-hub, 80 Cheombok-ro, Dong-gu, Daegu, 41061 South Korea
| | - Jun Sik Kim
- Preclinical Research Center, K Medi-hub, 80 Cheombok-ro, Dong-gu, Daegu, 41061 South Korea
| | - Moon-Yong Song
- Medical Safety Center, Bio Division, Korea Conformity Laboratories 8, Gaetbeol-ro 145beon-gil, Yeonsu-gu, Incheon, 21999 South Korea
| | - Young-Sik Kim
- Medical Safety Center, Bio Division, Korea Conformity Laboratories 8, Gaetbeol-ro 145beon-gil, Yeonsu-gu, Incheon, 21999 South Korea
| | - Sang-Mo Kwon
- grid.262229.f0000 0001 0719 8572Department of Physiology, School of Medicine, Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Immunoregulatory Therapeutics Group in Brain Busan 21 Project, Pusan National University, Yangsan, 626-870 South Korea
| | - Won-Kyo Jung
- grid.412576.30000 0001 0719 8994Division of Biomedical Engineering and Research Center for Marine Integrated Bionics Technology, Pukyong National University, Daeyeon-dong, Nam-gu, Busan, 48513 South Korea
| | - Woonhyeok Jeong
- grid.412091.f0000 0001 0669 3109Department of Plastic and Reconstructive Surgery, Dongsan Medical Center, Keimyung University College of Medicine, 1035 Dalgubeol-daero, Dalseo-gu, Daegu, 42601 South Korea
| | - Hojun Jeon
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-Do 15588 South Korea
| |
Collapse
|
9
|
Jiang W, Wang H, Zheng J, Zhao Y, Xu S, Zhuo S, Wang H, Yan J. Post-operative anastomotic leakage and collagen changes in patients with rectal cancer undergoing neoadjuvant chemotherapy vs chemoradiotherapy. Gastroenterol Rep (Oxf) 2022; 10:goac058. [PMID: 36324613 PMCID: PMC9619829 DOI: 10.1093/gastro/goac058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 06/24/2022] [Accepted: 09/28/2022] [Indexed: 11/04/2022] Open
Abstract
Background A significant difference in the anastomotic leakage (AL) rate has been observed between patients with locally advanced rectal cancer who have undergone preoperative chemotherapy and those undergoing preoperative chemoradiotherapy. This study aimed to quantitatively analyse collagen structural changes caused by preoperative chemoradiotherapy and illuminate the relationship between collagen changes and AL. Methods Anastomotic distal and proximal "doughnut" specimens from the Sixth Affiliated Hospital of Sun Yat-sen University (Guangzhou, China) were quantitatively assessed for collagen structural changes between patients with and without preoperative radiotherapy using multiphoton imaging. Then, patients treated with preoperative chemoradiotherapy were used as a training cohort to construct an AL-SVM classifier by the Mann-Whitney U test and support vector machine (SVM). An independent test cohort from the Fujian Province Cancer Hospital (Fuzhou, China) was used to validate the AL-SVM classifier. Results A total of 207 patients were included from the Sixth Affiliated Hospital of Sun Yat-sen University. The AL rate in the preoperative chemoradiotherapy group (n = 107) was significantly higher than that in the preoperative chemotherapy group (n = 100) (21.5% vs 7.0%, P = 0.003). A fully quantitative analysis showed notable morphological and spatial distribution feature changes in collagen in the preoperative chemoradiotherapy group. Then, the patients who received preoperative chemoradiotherapy were used as a training cohort to construct the AL-SVM classifier based on five collagen features and the tumor distance from the anus. The AL-SVM classifier showed satisfactory discrimination and calibration with areas under the curve of 0.907 and 0.856 in the training and test cohorts, respectively. Conclusions The collagen structure may be notably altered by preoperative radiotherapy. The AL-SVM classifier was useful for the individualized prediction of AL in rectal cancer patients undergoing preoperative chemoradiotherapy.
Collapse
Affiliation(s)
| | | | | | - Yandong Zhao
- Department of Pathology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Shuoyu Xu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, P. R. China,Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
| | - Shuangmu Zhuo
- Corresponding authors. Jun Yan, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China. Tel: +86-20-61641682; Fax: +86-20-61641683; ; Hui Wang, Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd, Guangzhou, Guangdong 510655, P. R. China. Tel: +86-20-61641682; Fax: +86-20-61641683; ; Shuangmu Zhuo, School of Science, Jimei University, Xiamen, Fujian 361021, P. R. China. Tel.: +86-592-6181893; Fax: +86-592-6181893;
| | - Hui Wang
- Corresponding authors. Jun Yan, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China. Tel: +86-20-61641682; Fax: +86-20-61641683; ; Hui Wang, Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd, Guangzhou, Guangdong 510655, P. R. China. Tel: +86-20-61641682; Fax: +86-20-61641683; ; Shuangmu Zhuo, School of Science, Jimei University, Xiamen, Fujian 361021, P. R. China. Tel.: +86-592-6181893; Fax: +86-592-6181893;
| | - Jun Yan
- Corresponding authors. Jun Yan, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China. Tel: +86-20-61641682; Fax: +86-20-61641683; ; Hui Wang, Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd, Guangzhou, Guangdong 510655, P. R. China. Tel: +86-20-61641682; Fax: +86-20-61641683; ; Shuangmu Zhuo, School of Science, Jimei University, Xiamen, Fujian 361021, P. R. China. Tel.: +86-592-6181893; Fax: +86-592-6181893;
| |
Collapse
|
10
|
Darvish DM. Collagen fibril formation in vitro: From origin to opportunities. Mater Today Bio 2022; 15:100322. [PMID: 35757034 PMCID: PMC9218154 DOI: 10.1016/j.mtbio.2022.100322] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 12/13/2022] Open
Abstract
Sometimes, to move forward, it is necessary to look back. Collagen type I is one of the most commonly used biomaterials in tissue engineering and regenerative medicine. There are a variety of collagen scaffolds and biomedical products based on collagen have been made, and the development of new ones is still ongoing. Materials, where collagen is in the fibrillar form, have some advantages: they have superior mechanical properties, higher degradation time and, what is most important, mimic the structure of the native extracellular matrix. There are some standard protocols for the formation of collagen fibrils in vitro, but if we look more carefully at those methods, we can see some controversies. For example, why is the formation of collagen gel commonly carried out at 37 °C, when it was well investigated that the temperature higher than 35 °C results in a formation of not well-ordered fibrils? Biomimetic collagen materials can be obtained both using culture medium or neutralizing solution, but it requires a deep understanding of all of the crucial points. One of this point is collagen extraction method, since not every method retains the ability of collagen to reconstitute native banded fibrils. Collagen polymorphism is also often overlooked in spite of the appearance of different polymorphic forms during fibril formation is possible, especially when collagen blends are utilized. In this review, we will not only pay attention to these issues, but we will overview the most prominent works related to the formation of collagen fibrils in vitro starting from the first approaches and moving to the up-to-date recipes.
Collapse
Affiliation(s)
- Diana M Darvish
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Prospekt, 4, Saint-Petersburg, 194064, Russia
| |
Collapse
|
11
|
Tafti MF, Aghamollaei H, Moghaddam MM, Jadidi K, Alio JL, Faghihi S. Emerging tissue engineering strategies for the corneal regeneration. J Tissue Eng Regen Med 2022; 16:683-706. [PMID: 35585479 DOI: 10.1002/term.3309] [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: 11/23/2021] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 11/10/2022]
Abstract
Cornea as the outermost layer of the eye is at risk of various genetic and environmental diseases that can damage the cornea and impair vision. Corneal transplantation is among the most applicable surgical procedures for repairing the defected tissue. However, the scarcity of healthy tissue donations as well as transplantation failure has remained as the biggest challenges in confront of corneal grafting. Therefore, alternative approaches based on stem-cell transplantation and classic regenerative medicine have been developed for corneal regeneration. In this review, the application and limitation of the recently-used advanced approaches for regeneration of cornea are discussed. Additionally, other emerging powerful techniques such as 5D printing as a new branch of scaffold-based technologies for construction of tissues other than the cornea are highlighted and suggested as alternatives for corneal reconstruction. The introduced novel techniques may have great potential for clinical applications in corneal repair including disease modeling, 3D pattern scheming, and personalized medicine.
Collapse
Affiliation(s)
- Mahsa Fallah Tafti
- Stem Cell and Regenerative Medicine Group, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Hossein Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Khosrow Jadidi
- Vision Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Jorge L Alio
- Department of Research and Development, VISSUM, Alicante, Spain.,Cornea, Cataract and Refractive Surgery Department, VISSUM, Alicante, Spain.,Department of Pathology and Surgery, Division of Ophthalmology, Faculty of Medicine, Miguel Hernández University, Alicante, Spain
| | - Shahab Faghihi
- Stem Cell and Regenerative Medicine Group, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
12
|
Iraninasab S, Sharifian S, Homaei A, Homaee MB, Sharma T, Nadda AK, Kennedy JF, Bilal M, Iqbal HMN. Emerging trends in environmental and industrial applications of marine carbonic anhydrase: a review. Bioprocess Biosyst Eng 2022; 45:431-451. [PMID: 34821989 DOI: 10.1007/s00449-021-02667-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023]
Abstract
Biocatalytic conversion of greenhouse gases such as carbon dioxide into commercial products is one of the promising key approaches to solve the problem of climate change. Microbial enzymes, including carbonic anhydrase, NAD-dependent formate dehydrogenase, ribulose bisphosphate carboxylase, and methane monooxygenase, have been exploited to convert atmospheric gases into industrial products. Carbonic anhydrases are Zn2+-dependent metalloenzymes that catalyze the reversible conversion of CO2 into bicarbonate. They are widespread in bacteria, algae, plants, and higher organisms. In higher organisms, they regulate the physiological pH and contribute to CO2 transport in the blood. In plants, algae, and photosynthetic bacteria carbonic anhydrases are involved in photosynthesis. Converting CO2 into bicarbonate by carbonic anhydrases can solidify gaseous CO2, thereby reducing global warming due to the burning of fossil fuels. This review discusses the three-dimensional structures of carbonic anhydrases, their physiological role in marine life, their catalytic mechanism, the types of inhibitors, and their medicine and industry applications.
Collapse
Affiliation(s)
- Sudabeh Iraninasab
- Department of Marine Biology, Faculty of Marine Science and Technology, University of Hormozgan, P.O. Box 3995, Bandar Abbas, Iran
| | - Sana Sharifian
- Department of Marine Biology, Faculty of Marine Science and Technology, University of Hormozgan, P.O. Box 3995, Bandar Abbas, Iran
| | - Ahmad Homaei
- Department of Marine Biology, Faculty of Marine Science and Technology, University of Hormozgan, P.O. Box 3995, Bandar Abbas, Iran.
| | | | - Tanvi Sharma
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, 173 234, India
| | - Ashok Kumar Nadda
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, 173 234, India
| | - John F Kennedy
- Chembiotech Laboratories, Advanced Science and Technology Institute, The Kyrewood Centre, Tenbury Wells, Worcs, WR15 8FF, UK
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, 64849, Monterrey, Mexico
| |
Collapse
|
13
|
Samiei M, Alipour M, Khezri K, Saadat YR, Forouhandeh H, Abdolahinia ED, Vahed SZ, Sharifi S, Dizaj SM. Application of collagen and mesenchymal stem cells in regenerative dentistry. Curr Stem Cell Res Ther 2021; 17:606-620. [PMID: 34931969 DOI: 10.2174/1574888x17666211220100521] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/28/2021] [Accepted: 11/10/2021] [Indexed: 11/22/2022]
Abstract
Collagen is an important macromolecule of extracellular matrix (ECM) in bones, teeth, and temporomandibular joints. Mesenchymal stem cells (MSCs) interact with the components of the ECM such as collagen, proteoglycans, glycosaminoglycans (GAGs), and several proteins on behalf of variable matrix elasticity and bioactive cues. Synthetic collagen-based biomaterials could be effective scaffolds for regenerative dentistry applications due to mimicking of host tissues' ECM. These biomaterials are biocompatible, biodegradable, readily available, and non-toxic to cells whose capability promotes cellular response and wound healing in the craniofacial region. Collagen could incorporate other biomolecules to induce mineralization in calcified tissues such as bone and tooth. Moreover, the addition of these molecules or other polymers to collagen-based biomaterials could enhance mechanical properties, which is important in load-bearing areas such as the mandible. A literature review was performed via reliable internet database (mainly PubMed) based on MeSH keywords. This review first describes the properties of collagen as a key protein in the structure of hard tissues. Then, it introduces different types of collagens, the correlation between collagen and MSCs, and the methods used to modify collagen in regenerative dentistry including recent progression on the regeneration of periodontium, dentin-pulp complex, and temporomandibular joint by applying collagen. Besides, the prospects and challenges of collagen-based biomaterials in the craniofacial region pointes out.
Collapse
Affiliation(s)
- Mohammad Samiei
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdieh Alipour
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khadijeh Khezri
- Deputy of Food and Drug Administration, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Haleh Forouhandeh
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elaheh Dalir Abdolahinia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Simin Sharifi
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Maleki Dizaj
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Rickelt S, Neyaz A, Condon C, Whittaker CA, Zaidi AH, Taylor MS, Abbruzzese G, Mattia AR, Zukerberg L, Shroff SG, Yilmaz OH, Yılmaz O, Wu EY, Choi WT, Jobe BA, Odze RD, Patil DT, Deshpande V, Hynes RO. Agrin loss in Barrett's esophagus-related neoplasia and its utility as a diagnostic and predictive biomarker. Clin Cancer Res 2021; 28:1167-1179. [PMID: 34785582 DOI: 10.1158/1078-0432.ccr-21-2822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/29/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE There is an unmet need for identifying novel biomarkers in Barrett's esophagus (BE) that could stratify patients with regards to neoplastic progression. We investigate the expression patterns of extracellular matrix (ECM) molecules in BE and BE-related neoplasia, and assess their value as biomarkers for the diagnosis of BE-related neoplasia and to predict neoplastic progression. EXPERIMENTAL DESIGN Gene expression analyses of ECM matrisome gene sets were performed using publicly available data on human BE, BE-related dysplasia, esophageal ADCA and normal esophagus. Immunohistochemical expression of basement membrane (BM) marker agrin (AGRN) and p53 was analyzed in biopsies of BE-related neoplasia from 321 patients in three independent cohorts. RESULTS Differential gene expression analysis revealed significant enrichment of ECM matrisome gene sets in dysplastic BE and ADCA compared with controls. Loss of BM AGRN expression was observed in both BE-related dysplasia and ADCA. The mean AGRN loss in BE glands was significantly higher in BErelated dysplasia and ADCA compared to non-dysplastic BE (NDBE; p<0.001; specificity=82.2% and sensitivity=96.4%). Loss of AGRN was significantly higher in NDBE samples from progressors compared to non-progressors (p<0.001) and identified patients who progressed to advanced neoplasia with a specificity of 80.2% and sensitivity of 54.8%. Moreover, the combination of AGRN loss and abnormal p53 staining identified progression to BE-related advanced neoplasia with a specificity and sensitivity of 86.5% and 58.7%. CONCLUSIONS We highlight ECM changes during BE progression to neoplasia. BM AGRN loss is a novel diagnostic biomarker that can identify NDBE patients at increased risk of developing advanced neoplasia.
Collapse
Affiliation(s)
- Steffen Rickelt
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology
| | - Azfar Neyaz
- Department of Pathology, Massachusetts General Hospital
| | - Charlene Condon
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology
| | - Charles A Whittaker
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology
| | - Ali H Zaidi
- Esophageal and Lung Institute, Allegheny Health Network
| | | | - Genevieve Abbruzzese
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology
| | | | | | | | - Omer H Yilmaz
- The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology
| | - Osman Yılmaz
- Pathology & Laboratory Medicine, Boston University School of Medicine
| | | | - Won-Tak Choi
- Department of Pathology, University of California, San Francisco
| | | | | | - Deepa T Patil
- Department of Pathology, Brigham and Women's Hospital
| | - Vikram Deshpande
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School
| | - Richard O Hynes
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology
| |
Collapse
|
15
|
Hong G, Kim J, Oh H, Yun S, Kim CM, Jeong Y, Yun W, Shim J, Jang I, Kim C, Jin S. Production of Multiple Cell-Laden Microtissue Spheroids with a Biomimetic Hepatic-Lobule-Like Structure. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102624. [PMID: 34286875 PMCID: PMC11469225 DOI: 10.1002/adma.202102624] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/07/2021] [Indexed: 05/11/2023]
Abstract
The construction of an in vitro 3D cellular model to mimic the human liver is highly desired for drug discovery and clinical applications, such as patient-specific treatment and cell-based therapy in regenerative medicine. However, current bioprinting strategies are limited in their ability to generate multiple cell-laden microtissues with biomimetic structures. This study presents a method for producing hepatic-lobule-like microtissue spheroids using a bioprinting system incorporating a precursor cartridge and microfluidic emulsification system. The multiple cell-laden microtissue spheroids can be successfully generated at a speed of approximately 45 spheroids min-1 and with a uniform diameter. Hepatic and endothelial cells are patterned in a microtissue spheroid with the biomimetic structure of a liver lobule. The spheroids allow long-term culture with high cell viability, and the structural integrity is maintained longer than that of non-structured spheroids. Furthermore, structured spheroids show high MRP2, albumin, and CD31 expression levels. In addition, the in vivo study reveals that structured microtissue spheroids are stably engrafted. These results demonstrate that the method provides a valuable 3D structured microtissue spheroid model with lobule-like constructs and liver functions.
Collapse
Affiliation(s)
- Gyusik Hong
- Department of Mechanical EngineeringKorea Polytechnic University237 Sangidaehak‐roSiheung‐si15073Republic of Korea
| | - Jin Kim
- Laboratory Animal MedicineCollege of Veterinary MedicineSeoul National University1, Gwanak‐roGwanak‐guSeoul08826Republic of Korea
- College of Veterinary MedicineKonkuk University120, Neungdong‐ro, Gwangjin‐guSeoul05029Republic of Korea
| | - Hyeongkwon Oh
- Department of Mechanical EngineeringKorea Polytechnic University237 Sangidaehak‐roSiheung‐si15073Republic of Korea
| | - Seokhwan Yun
- Department of Mechanical EngineeringKorea Polytechnic University237 Sangidaehak‐roSiheung‐si15073Republic of Korea
| | - Chul Min Kim
- Department of MechatronicsGyeongsang National University33, Dongjin‐roJinju52725Republic of Korea
| | - Yun‐Mi Jeong
- Department of Mechanical EngineeringKorea Polytechnic University237 Sangidaehak‐roSiheung‐si15073Republic of Korea
| | - Won‐Soo Yun
- Department of Mechanical EngineeringKorea Polytechnic University237 Sangidaehak‐roSiheung‐si15073Republic of Korea
- Research InstituteT&R Biofab. Co. Ltd242 Pangyo‐roSeongnamGyeonggi13487Republic of Korea
| | - Jin‐Hyung Shim
- Department of Mechanical EngineeringKorea Polytechnic University237 Sangidaehak‐roSiheung‐si15073Republic of Korea
- Research InstituteT&R Biofab. Co. Ltd242 Pangyo‐roSeongnamGyeonggi13487Republic of Korea
| | - Ilho Jang
- Research InstituteT&R Biofab. Co. Ltd242 Pangyo‐roSeongnamGyeonggi13487Republic of Korea
| | - C‐Yoon Kim
- College of Veterinary MedicineKonkuk University120, Neungdong‐ro, Gwangjin‐guSeoul05029Republic of Korea
| | - Songwan Jin
- Department of Mechanical EngineeringKorea Polytechnic University237 Sangidaehak‐roSiheung‐si15073Republic of Korea
- Research InstituteT&R Biofab. Co. Ltd242 Pangyo‐roSeongnamGyeonggi13487Republic of Korea
| |
Collapse
|
16
|
Jiang W, Feng M, Zheng J, Wang G, Xu S, Zhou L, Zhuo S, Yan J. Association of the collagen score with anastomotic leakage in rectal cancer patients after neoadjuvant chemoradiotherapy. Surgery 2021; 170:1331-1341. [PMID: 34116856 DOI: 10.1016/j.surg.2021.05.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/25/2021] [Accepted: 05/15/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Collagen changes in the extracellular matrix caused by neoadjuvant chemoradiotherapy are a potential mechanism of anastomotic leakage. We aimed to construct a fully quantitative collagen score to describe collagen structure changes in the extracellular matrix and then develop and validate a prediction model to identify patients who are at a high risk of postoperative anastomotic leakage. METHODS This is a retrospective study in which 372 patients were enrolled, and their baseline clinicopathological characteristics were collected. Anastomotic distal and proximal "doughnut" specimens underwent second harmonic generation imaging, and collagen features were extracted. A LASSO regression was used to select significant predictors, and the collagen score was constructed. A prediction model based on collagen score was developed and internally and externally validated. RESULTS The primary cohort included 214 consecutive patients, and the anastomotic leakage rate was 8.9%. The validation cohort comprised 158 consecutive patients, and the anastomotic leakage rate was 10.1%. The collagen score was significantly related to anastomotic leakage in both cohorts (P < .001). Multivariate analysis revealed that tumor location, preoperative albumin, and collagen score were independent predictors of anastomotic leakage. These 3 predictors were incorporated into the prediction model, and a nomogram was established. The model showed good discrimination in the primary (area under the curve: 0.954) and validation (area under the curve: 0.928) cohorts. Decision curve analysis demonstrated that the nomogram was clinically useful. CONCLUSION The collagen score is associated with anastomotic leakage, and the collagen nomogram based on the collagen score is useful for individualized prediction of anastomotic leakage in rectal cancer patients with neoadjuvant chemoradiotherapy after surgery.
Collapse
Affiliation(s)
- Wei Jiang
- Department of General Surgery, Nanfang Hospital, Southern Medical University & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, China; School of Science, Jimei University, Xiamen, Fujian, China. https://twitter.com/DocJiangwei
| | - Mingyuan Feng
- Department of General Surgery, Nanfang Hospital, Southern Medical University & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, China. https://twitter.com/FengmingyuanN
| | - Jixiang Zheng
- Department of General Surgery, Nanfang Hospital, Southern Medical University & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, China. https://twitter.com/ZhengjixiangN
| | - Guangxing Wang
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, China. https://twitter.com/PGuangxingwang
| | - Shuoyu Xu
- Department of General Surgery, Nanfang Hospital, Southern Medical University & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, China; Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong China. https://twitter.com/shevashuoyu
| | - Linghong Zhou
- Department of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, China. https://twitter.com/LinghongZhou
| | - Shuangmu Zhuo
- School of Science, Jimei University, Xiamen, Fujian, China.
| | - Jun Yan
- Department of General Surgery, Nanfang Hospital, Southern Medical University & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, China.
| |
Collapse
|
17
|
Asthmatic Eosinophils Promote Contractility and Migration of Airway Smooth Muscle Cells and Pulmonary Fibroblasts In Vitro. Cells 2021; 10:cells10061389. [PMID: 34199925 PMCID: PMC8229663 DOI: 10.3390/cells10061389] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/25/2022] Open
Abstract
Enhanced contractility and migration of airway smooth muscle cells (ASMC) and pulmonary fibroblasts (PF) are part of airway remodeling in asthma. Eosinophils are the central inflammatory cells that participate in airway inflammation. However, the role of asthmatic eosinophils in ASMC and PF contractility, migration, and differentiation to contractile phenotype has not yet been precisely described. A total of 38 individuals were included in this study: 13 steroid-free non-severe allergic asthma (AA) patients, 11 severe non-allergic eosinophilic asthma (SNEA) patients, and 14 healthy subjects (HS). For AA patients and HS groups, a bronchial allergen challenge with D. pteronyssinus was performed. Individual combined cell cultures were prepared from isolated peripheral blood eosinophils and immortalized ASMC or commercial PF cell lines separately. The migration of ASMC and PF was evaluated using wound healing assay and contractility using collagen gel assay. Gene expression of contractile apparatus proteins, COL1A1, COL5A1, and FN, in ASMC and PF was evaluated using qRT-PCR. We found that contractility and migration of ASMC and PF significantly increased after incubation with asthmatic eosinophils compared to HS eosinophils, p < 0.05, and SNEA eosinophils demonstrated the highest effect on contractility of ASMC and migration of both cell lines, p < 0.05. AA and SNEA eosinophils significantly increased gene expression of contractile apparatus proteins, COL1A1 and FN, in both cell lines, p < 0.05. Furthermore, the allergen-activated AA eosinophils significantly increased the contractility of ASMC, and migration and gene expression in ASMC and PF, p < 0.05. Thus, asthmatic eosinophils change ASMC and PF behavior by increasing their contractility and migration, contributing to airway remodeling.
Collapse
|
18
|
Yadav E, Yadav P, Verma A. In silico Study of Trianthema portulacastrum Embedded Iron Oxide Nanoparticles on Glycogen Synthase Kinase-3β: A Possible Contributor to its Enhanced in vivo Wound Healing Potential. Front Pharmacol 2021; 12:664075. [PMID: 34079461 PMCID: PMC8165444 DOI: 10.3389/fphar.2021.664075] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/04/2021] [Indexed: 01/12/2023] Open
Abstract
Rich amount of phenolic compounds are available in Trianthema portulacastrum L. (TP) leaves and are traditionally utilized as a wound dressing material. Oxidative stress and inflammation affect the Wnt/β-catenin pathway by modulating the glycogen synthase kinase-3β (GSK) activity subjected to delay in wound healing. The objective of the current study was to explore the wound healing effect of ferric oxide nanoparticles biosynthesized with fractionated TP extract (FeTP). The ability of TP active components (polyphenols) to inhibit the GSK was explored by using molecular docking studies. FeTP were synthesized, characterized, utilized to prepare an ointment and its efficacy was investigated against full-thickness dermal wounds. Different wound healing parameters, level of enzymatic antioxidants, hydroxyproline content and tissue cytokines level were analyzed. Histopathology was performed to confirm the healing by newly formed tissue architecture. Rats treated with FeTP showed significantly swift healing with faster wound contraction rate, high tensile strength and hydroxyproline content along with the utilization of less time for epithelialization. Histopathological study also validated the potential wound healing effect of FeTP with complete re-epithelialization. The results of the present study cumulatively revealed that the green synthesized FeTP ointment approach may serve as a potential tool for dermal wound healing.
Collapse
Affiliation(s)
- Ekta Yadav
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences (SHUATS), Prayagraj, India
| | - Pankajkumar Yadav
- Pharmaceutics Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences (SHUATS), Prayagraj, India
| | - Amita Verma
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences (SHUATS), Prayagraj, India
| |
Collapse
|
19
|
Azizipour E, Aghamollaei H, Halabian R, Poormoghadam D, Saffari M, Entezari M, Salimi A. A novel hydrogel scaffold contained bioactive glass nanowhisker (BGnW) for osteogenic differentiation of human mesenchymal stem cells (hMSCs) in vitro. Int J Biol Macromol 2021; 174:562-572. [PMID: 33434552 DOI: 10.1016/j.ijbiomac.2021.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/26/2020] [Accepted: 01/01/2021] [Indexed: 12/18/2022]
Abstract
Employing hydrogels as an alternative strategy for repairing bone defects has received great attention in bone tissue engineering. In this study, hydrogel scaffold based on collagen, gelatin, and glutaraldehyde was combined with bioactive glass nanowhiskers (BGnW) to differentiate human mesenchymal stem cells (hMSCs) into the osteogenic lineage and inducing biomineralization. Pure Gel-Glu-Col and bioactive glass nanowhiskers were used as control throughout the paper. Chemical, physical and morphological characteristics of the nanocomposite scaffold were assessed meticulously using Fourier transform infrared spectroscopy (FTIR), X-ray diffraction (XRD), porosity measurement, water uptake ability, tensile test, and scanning electron microscopy (SEM). To determine the cytotoxicity and cell viability of the hydrogel, MTT assay and Acridine orange (AO) staining were performed. hMSCs seeded on Gel-Glu-Col/BGnW were then incubated with osteogenic differentiation media for 14 days. Biomineralization assays (alkaline phosphatase (ALP) activity, calcium content assay, von Kossa, and Alizarin red staining) were carried out, and osteogenic genes and protein markers were examined using real time-PCR and immunocytochemistry. Results showed that the components of the hydrogel were properly integrated. The mechanical property of hydrogel was enhanced following the addition of BGnW. Cell viability assays confirmed the biocompatibility of the scaffold and increasing the proliferation after incorporating BGnW into pure Ge1-Glu-Col. Our nanocomposite maintained an enhanced ability of biomineralization as compared to its pure counterparts. Molecular investigations revealed an elevated level of osteogenic markers as compared to Ge1-Glu-Col and BGnW. All in all, Gel-Glu-Col/BGnW seems to be a potential candidate for the regeneration of bone tissue.
Collapse
Affiliation(s)
- Esmat Azizipour
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Aghamollaei
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Baqiyatallah University Medical of Sciences, Tehran, Iran
| | - Delaram Poormoghadam
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mostafa Saffari
- Department of Pharmaceutics, School of Pharmacy, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ali Salimi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Narayanan N, Calve S. Extracellular matrix at the muscle - tendon interface: functional roles, techniques to explore and implications for regenerative medicine. Connect Tissue Res 2021; 62:53-71. [PMID: 32856502 PMCID: PMC7718290 DOI: 10.1080/03008207.2020.1814263] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The muscle-tendon interface is an anatomically specialized region that is involved in the efficient transmission of force from muscle to tendon. Due to constant exposure to loading, the interface is susceptible to injury. Current treatment methods do not meet the socioeconomic demands of reduced recovery time without compromising the risk of reinjury, requiring the need for developing alternative strategies. The extracellular matrix (ECM) present in muscle, tendon, and at the interface of these tissues consists of unique molecules that play significant roles in homeostasis and repair. Better, understanding the function of the ECM during development, injury, and aging has the potential to unearth critical missing information that is essential for accelerating the repair at the muscle-tendon interface. Recently, advanced techniques have emerged to explore the ECM for identifying specific roles in musculoskeletal biology. Simultaneously, there is a tremendous increase in the scope for regenerative medicine strategies to address the current clinical deficiencies. Advancements in ECM research can be coupled with the latest regenerative medicine techniques to develop next generation therapies that harness ECM for treating defects at the muscle-tendon interface. The current work provides a comprehensive review on the role of muscle and tendon ECM to provide insights about the role of ECM in the muscle-tendon interface and discusses the latest research techniques to explore the ECM to gathered information for developing regenerative medicine strategies.
Collapse
Affiliation(s)
- Naagarajan Narayanan
- Paul M. Rady Department of Mechanical Engineering, University of Colorado – Boulder, 1111 Engineering Drive, Boulder, Colorado 80309 – 0427
| | - Sarah Calve
- Paul M. Rady Department of Mechanical Engineering, University of Colorado – Boulder, 1111 Engineering Drive, Boulder, Colorado 80309 – 0427
| |
Collapse
|
21
|
Ustunel S, Prévôt ME, Clements RJ, Hegmann E. Cradle-to-cradle: designing biomaterials to fit as truly biomimetic cell scaffolds– a review. LIQUID CRYSTALS TODAY 2020. [DOI: 10.1080/1358314x.2020.1855919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Senay Ustunel
- Materials Science Graduate Program, Kent State University, Kent, OH, USA
- Advanced Materials and Liquid Crystal Institute, Kent State University, Kent, OH, USA
| | - Marianne E. Prévôt
- Advanced Materials and Liquid Crystal Institute, Kent State University, Kent, OH, USA
| | - Robert J. Clements
- Department of Biological Sciences, Kent State University, Kent, OH, USA
- Biomedical Sciences Program, Kent State University, Kent, OH, USA
- Brain Health Research Institute, Kent State University, Kent, OH, USA
| | - Elda Hegmann
- Materials Science Graduate Program, Kent State University, Kent, OH, USA
- Advanced Materials and Liquid Crystal Institute, Kent State University, Kent, OH, USA
- Department of Biological Sciences, Kent State University, Kent, OH, USA
- Biomedical Sciences Program, Kent State University, Kent, OH, USA
- Brain Health Research Institute, Kent State University, Kent, OH, USA
| |
Collapse
|
22
|
Yu J, Park SA, Kim WD, Ha T, Xin YZ, Lee J, Lee D. Current Advances in 3D Bioprinting Technology and Its Applications for Tissue Engineering. Polymers (Basel) 2020; 12:E2958. [PMID: 33322291 PMCID: PMC7764360 DOI: 10.3390/polym12122958] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 12/25/2022] Open
Abstract
Three-dimensional (3D) bioprinting technology has emerged as a powerful biofabrication platform for tissue engineering because of its ability to engineer living cells and biomaterial-based 3D objects. Over the last few decades, droplet-based, extrusion-based, and laser-assisted bioprinters have been developed to fulfill certain requirements in terms of resolution, cell viability, cell density, etc. Simultaneously, various bio-inks based on natural-synthetic biomaterials have been developed and applied for successful tissue regeneration. To engineer more realistic artificial tissues/organs, mixtures of bio-inks with various recipes have also been developed. Taken together, this review describes the fundamental characteristics of the existing bioprinters and bio-inks that have been currently developed, followed by their advantages and disadvantages. Finally, various tissue engineering applications using 3D bioprinting are briefly introduced.
Collapse
Affiliation(s)
- JunJie Yu
- Department of Biomedical Engineering, School of Integrative Engineering, Chung-Ang University, 221 Heukseok-Dong, Dongjak-Gu, Seoul 06974, Korea;
- Department of Nature-Inspired System and Application, Korea Institute of Machinery & Materials, 156 Gajeongbuk-Ro, Yuseong-Gu, Daejeon 34103, Korea; (S.A.P.); (W.D.K.)
| | - Su A Park
- Department of Nature-Inspired System and Application, Korea Institute of Machinery & Materials, 156 Gajeongbuk-Ro, Yuseong-Gu, Daejeon 34103, Korea; (S.A.P.); (W.D.K.)
| | - Wan Doo Kim
- Department of Nature-Inspired System and Application, Korea Institute of Machinery & Materials, 156 Gajeongbuk-Ro, Yuseong-Gu, Daejeon 34103, Korea; (S.A.P.); (W.D.K.)
| | - Taeho Ha
- Department of 3D Printing, Korea Institute of Machinery & Materials, 156 Gajeongbuk-Ro, Yuseong-Gu, Daejeon 34103, Korea;
| | - Yuan-Zhu Xin
- Department of Engineering Mechanics, School of Mechanical and Aerospace Engineering, Jilin University, No. 5988, Renmin Street, Changchun 130025, China;
| | - JunHee Lee
- Department of Nature-Inspired System and Application, Korea Institute of Machinery & Materials, 156 Gajeongbuk-Ro, Yuseong-Gu, Daejeon 34103, Korea; (S.A.P.); (W.D.K.)
| | - Donghyun Lee
- Department of Biomedical Engineering, School of Integrative Engineering, Chung-Ang University, 221 Heukseok-Dong, Dongjak-Gu, Seoul 06974, Korea;
| |
Collapse
|
23
|
Rajan AM, Ma RC, Kocha KM, Zhang DJ, Huang P. Dual function of perivascular fibroblasts in vascular stabilization in zebrafish. PLoS Genet 2020; 16:e1008800. [PMID: 33104690 PMCID: PMC7644104 DOI: 10.1371/journal.pgen.1008800] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 11/05/2020] [Accepted: 09/28/2020] [Indexed: 12/22/2022] Open
Abstract
Blood vessels are vital to sustain life in all vertebrates. While it is known that mural cells (pericytes and smooth muscle cells) regulate vascular integrity, the contribution of other cell types to vascular stabilization has been largely unexplored. Using zebrafish, we identified sclerotome-derived perivascular fibroblasts as a novel population of blood vessel associated cells. In contrast to pericytes, perivascular fibroblasts emerge early during development, express the extracellular matrix (ECM) genes col1a2 and col5a1, and display distinct morphology and distribution. Time-lapse imaging reveals that perivascular fibroblasts serve as pericyte precursors. Genetic ablation of perivascular fibroblasts markedly reduces collagen deposition around endothelial cells, resulting in dysmorphic blood vessels with variable diameters. Strikingly, col5a1 mutants show spontaneous hemorrhage, and the penetrance of the phenotype is strongly enhanced by the additional loss of col1a2. Together, our work reveals dual roles of perivascular fibroblasts in vascular stabilization where they establish the ECM around nascent vessels and function as pericyte progenitors. Blood vessels are essential to sustain life in humans. Defects in blood vessels can lead to serious diseases, such as hemorrhage, tissue ischemia, and stroke. However, how blood vessel stability is maintained by surrounding support cells is still poorly understood. Using the zebrafish model, we identify a new population of blood vessel associated cells termed perivascular fibroblasts, which originate from the sclerotome, an embryonic structure that is previously known to generate the skeleton of the animal. Perivascular fibroblasts are distinct from pericytes, a known population of blood vessel support cells. They become associated with blood vessels much earlier than pericytes and express several collagen genes, encoding main components of the extracellular matrix. Loss of perivascular fibroblasts or mutations in collagen genes result in fragile blood vessels prone to damage. Using cell tracing in live animals, we find that a subset of perivascular fibroblasts can differentiate into pericytes. Together, our work shows that perivascular fibroblasts play two important roles in maintaining blood vessel integrity. Perivascular fibroblasts secrete collagens to stabilize newly formed blood vessels and a sub-population of these cells also functions as precursors to generate pericytes to provide additional vascular support.
Collapse
Affiliation(s)
- Arsheen M. Rajan
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Roger C. Ma
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Katrinka M. Kocha
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Dan J. Zhang
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Peng Huang
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
24
|
Abstract
The extracellular matrix (ECM) is needed to maintain the structural integrity of tissues and to mediate cellular dynamics. Its main components are fibrous proteins and glycosaminoglycans, which provide a suitable environment for biological functions. Thus, biomaterials with ECM-like properties have been extensively developed by modulating their key components and properties. In the field of cardiac tissue engineering, the use of biomaterials offers several advantages in that biophysical and biochemical cues can be designed to mediate cardiac cells, which is critical for maturation and regeneration. This suggests that understanding biomaterials and their use in vivo and in vitro is beneficial in terms of advancing cardiac engineering. The current review provides an overview of both natural and synthetic biomaterials and their use in cardiac engineering. In addition, we focus on different strategies to recapitulate the cardiac tissue in 2D and 3D approaches, which is an important step for the maturation of cardiac tissues toward regeneration of the adult heart.
Collapse
|
25
|
Chaudhury S, Okuda KS, Koltowska K, Lagendijk AK, Paterson S, Baillie GJ, Simons C, Smith KA, Hogan BM, Bower NI. Localised Collagen2a1 secretion supports lymphatic endothelial cell migration in the zebrafish embryo. Development 2020; 147:dev.190983. [PMID: 32839180 DOI: 10.1242/dev.190983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 08/07/2020] [Indexed: 01/12/2023]
Abstract
The lymphatic vasculature develops primarily from pre-existing veins. A pool of lymphatic endothelial cells (LECs) first sprouts from cardinal veins followed by migration and proliferation to colonise embryonic tissues. Although much is known about the molecular regulation of LEC fate and sprouting during early lymphangiogenesis, we know far less about the instructive and permissive signals that support LEC migration through the embryo. Using a forward genetic screen, we identified mbtps1 and sec23a, components of the COP-II protein secretory pathway, as essential for developmental lymphangiogenesis. In both mutants, LECs initially depart the cardinal vein but then fail in their ongoing migration. A key cargo that failed to be secreted in both mutants was a type II collagen (Col2a1). Col2a1 is normally secreted by notochord sheath cells, alongside which LECs migrate. col2a1a mutants displayed defects in the migratory behaviour of LECs and failed lymphangiogenesis. These studies thus identify Col2a1 as a key cargo secreted by notochord sheath cells and required for the migration of LECs. These findings combine with our current understanding to suggest that successive cell-to-cell and cell-matrix interactions regulate the migration of LECs through the embryonic environment during development.
Collapse
Affiliation(s)
- Smrita Chaudhury
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Kazuhide S Okuda
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia.,Peter MacCallum Cancer Centre, Organogenesis and Cancer Program, Melbourne, Victoria 3000, Australia
| | - Katarzyna Koltowska
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Anne K Lagendijk
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Scott Paterson
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia.,Peter MacCallum Cancer Centre, Organogenesis and Cancer Program, Melbourne, Victoria 3000, Australia
| | - Gregory J Baillie
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Cas Simons
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Kelly A Smith
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia.,Department of Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Benjamin M Hogan
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia .,Peter MacCallum Cancer Centre, Organogenesis and Cancer Program, Melbourne, Victoria 3000, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Neil I Bower
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
26
|
Ji W, Hou B, Tang H, Cai M, Zheng W. Investigation of the effects of laminin present in the basal lamina of the peripheral nervous system on axon regeneration and remyelination using the nerve acellular scaffold. J Biomed Mater Res A 2020; 108:1673-1687. [PMID: 32196907 DOI: 10.1002/jbm.a.36933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 03/03/2020] [Accepted: 03/09/2020] [Indexed: 12/14/2022]
Abstract
This study aimed to investigate the effects of laminin (LN) located in the basal lamina, which are important components of the peripheral nervous system-extracellular matrix, on axon regeneration and remyelination. Nerve acellular scaffolds (NASs) (S-untreated) were prepared using the acellular technique. The active component LN in the NASs was blocked (S-LN- ) or upregulated (S-LN+ ); S-LN+ contained seven times more LN than did the S-untreated group. The adhesion capacity of Schwann cells (SCs) to the three types of NAS (S-untreated, S-LN- , and S-LN+ ) was assessed in vitro. Our results showed that the adhesion of SCs to the NASs was significantly reduced in the S-LN- group, whereas no difference was observed between the S-LN+ and S-untreated groups. The pretreated NASs were used to repair nerves in a nerve injury mouse model with the animals divided into four groups (S-LN- group, S-untreated group, S-LN+ group, and autograft group). Two weeks after surgery, although there was no difference in the S-LN- group, S-untreated group and S-LN+ group, the newly formed basal lamina in the S-LN- group were significantly lower than those in the other two groups. Four weeks after surgery, the S-LN+ group had higher numbers of newly generated axons and their calibers, more myelinated fibers, thicker myelin sheaths, increased myelin basic protein expression, and improved recovery of neural function compared to those of the S-LN- and S-untreated groups, but all of these parameters were significantly worse than those of the autograft group. Downregulation of the LN level in the NAS leads to a reduction in all of the above parameters.
Collapse
Affiliation(s)
- Wanqing Ji
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Bo Hou
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Hengxin Tang
- Department of Neurosurgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Meiqin Cai
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Wenhan Zheng
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
27
|
Alagha A, Nourallah A, Alhariri S. Dexamethasone- loaded polymeric porous sponge as a direct pulp capping agent. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 31:1689-1705. [PMID: 32402228 DOI: 10.1080/09205063.2020.1769801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
This study aims to achieve the principles of tissue engineering using biopolymers to be applied in the field of vital endodontic treatment to stimulate stem cells and engineering and regeneration of dentin tissue. the polymer blend was loaded with the steroidal anti-inflammatory drug, dexamethasone, and the porous drug-loaded bio-sponge was produced by lyophilization. Bio-sponge, as a direct pulp capping agent, was histologically studied compared to calcium hydroxide Ca(OH)2 in an animal experiment. The results indicated the effectiveness of the bio-sponge as a direct pulp capping agent where the dentin bridge was formed faster than Ca(OH)2 treated samples. There was no inflammatory response in the pulp tissue throughout the follow-up period. The porous bio-sponge loaded with dexamethasone with a neutral pH resulted in enhancement of the odontoblast differentiation from stem cells, resulting in the formation of a renewed dentin bridge without the slightest inflammatory response in the pulp.
Collapse
Affiliation(s)
- Amjad Alagha
- Faculty of Dentistry, Department of Pediatric Dentistry, Tishreen University, Lattakia, Syria
| | - Abdulwahab Nourallah
- Faculty of Dentistry, Department of Pediatric Dentistry, Tishreen University, Lattakia, Syria
| | - Sahar Alhariri
- Faculty of Science, Department of Chemistry, Damascus University, Damascus, Syria
| |
Collapse
|
28
|
Terzi A, Gallo N, Bettini S, Sibillano T, Altamura D, Madaghiele M, De Caro L, Valli L, Salvatore L, Sannino A, Giannini C. Sub‐ and Supramolecular X‐Ray Characterization of Engineered Tissues from Equine Tendon, Bovine Dermis, and Fish Skin Type‐I Collagen. Macromol Biosci 2020; 20:e2000017. [DOI: 10.1002/mabi.202000017] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 01/23/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Alberta Terzi
- Institute of Crystallography (IC)National Research Council Bari 70126 Italy
| | - Nunzia Gallo
- Department of Engineering for InnovationUniversity of Salento Lecce 73100 Italy
| | - Simona Bettini
- Department of Engineering for InnovationUniversity of Salento Lecce 73100 Italy
| | - Teresa Sibillano
- Institute of Crystallography (IC)National Research Council Bari 70126 Italy
| | - Davide Altamura
- Institute of Crystallography (IC)National Research Council Bari 70126 Italy
| | - Marta Madaghiele
- Department of Engineering for InnovationUniversity of Salento Lecce 73100 Italy
| | - Liberato De Caro
- Institute of Crystallography (IC)National Research Council Bari 70126 Italy
| | - Ludovico Valli
- Department of Biological and Environmental Sciences and TechnologiesUniversity of Salento Lecce 73100 Italy
| | - Luca Salvatore
- Department of Engineering for InnovationUniversity of Salento Lecce 73100 Italy
| | - Alessandro Sannino
- Department of Engineering for InnovationUniversity of Salento Lecce 73100 Italy
| | - Cinzia Giannini
- Institute of Crystallography (IC)National Research Council Bari 70126 Italy
| |
Collapse
|
29
|
Vandghanooni S, Eskandani M. Natural polypeptides-based electrically conductive biomaterials for tissue engineering. Int J Biol Macromol 2020; 147:706-733. [PMID: 31923500 DOI: 10.1016/j.ijbiomac.2019.12.249] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/28/2019] [Accepted: 12/28/2019] [Indexed: 12/11/2022]
Abstract
Fabrication of an appropriate scaffold is the key fundamental step required for a successful tissue engineering (TE). The artificial scaffold as extracellular matrix in TE has noticeable role in the fate of cells in terms of their attachment, proliferation, differentiation, orientation and movement. In addition, chemical and electrical stimulations affect various behaviors of cells such as polarity and functionality. Therefore, the fabrication approach and materials used for the preparation of scaffold should be more considered. Various synthetic and natural polymers have been used extensively for the preparation of scaffolds. The electrically conductive polymers (ECPs), moreover, have been used in combination with other polymers to apply electric fields (EF) during TE. In this context, composites of natural polypeptides and ECPs can be taken into account as context for the preparation of suitable scaffolds with superior biological and physicochemical features. In this review, we overviewed the simultaneous usage of natural polypeptides and ECPs for the fabrication of scaffolds in TE.
Collapse
Affiliation(s)
- Somayeh Vandghanooni
- Research Center for Pharmaceutical Nanotechnology, Biomedicine institute, Tabriz University of Medical Sciences, Tabriz, Iran; Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
30
|
Martinez B, Yang Y, Harker DMR, Farrar C, Mukundan H, Nath P, Mascareñas D. YAP/TAZ Related BioMechano Signal Transduction and Cancer Metastasis. Front Cell Dev Biol 2019; 7:199. [PMID: 31637239 PMCID: PMC6788381 DOI: 10.3389/fcell.2019.00199] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 09/05/2019] [Indexed: 01/01/2023] Open
Abstract
Mechanoreciprocity refers to a cell's ability to maintain tensional homeostasis in response to various types of forces. Physical forces are continually being exerted upon cells of various tissue types, even those considered static, such as the brain. Through mechanoreceptors, cells sense and subsequently respond to these stimuli. These forces and their respective cellular responses are prevalent in regulating everything from embryogenic tissue-specific differentiation, programmed cell death, and disease progression, the last of which being the subject of extensive attention. Abnormal mechanical remodeling of cells can provide clues as to the pathological status of tissues. This becomes particularly important in cancer cells, where cellular stiffness has been recently accepted as a novel biomarker for cancer metastasis. Several studies have also elucidated the importance of cell stiffness in cancer metastasis, with data highlighting that a reversal of tumor stiffness has the capacity to revert the metastatic properties of cancer. In this review, we summarize our current understanding of extracellular matrix (ECM) homeostasis, which plays a prominent role in tissue mechanics. We also describe pathological disruption of the ECM, and the subsequent implications toward cancer and cancer metastasis. In addition, we highlight the most novel approaches toward understanding the mechanisms which generate pathogenic cell stiffness and provide potential new strategies which have the capacity to advance our understanding of one of human-kinds' most clinically significant medical pathologies. These new strategies include video-based techniques for structural dynamics, which have shown great potential for identifying full-field, high-resolution modal properties, in this case, as a novel application.
Collapse
Affiliation(s)
- Bridget Martinez
- Engineering Institute, Los Alamos National Laboratory, Los Alamos, NM, United States
- Applied Modern Physics, Los Alamos National Laboratory, Los Alamos, NM, United States
- Department of Medicine, St. George’s University School of Medicine, St. George’s, Grenada
- Chemistry Division, Physical Chemistry and Applied Spectroscopy, Los Alamos National Laboratory, Los Alamos, NM, United States
| | - Yongchao Yang
- Energy and Global Security, Argonne National Laboratory, Lemont, IL, United States
| | | | - Charles Farrar
- Engineering Institute, Los Alamos National Laboratory, Los Alamos, NM, United States
| | - Harshini Mukundan
- Engineering Institute, Los Alamos National Laboratory, Los Alamos, NM, United States
| | - Pulak Nath
- Applied Modern Physics, Los Alamos National Laboratory, Los Alamos, NM, United States
| | - David Mascareñas
- Engineering Institute, Los Alamos National Laboratory, Los Alamos, NM, United States
| |
Collapse
|
31
|
Park MH, Kim AK, Manandhar S, Oh SY, Jang GH, Kang L, Lee DW, Hyeon DY, Lee SH, Lee HE, Huh TL, Suh SH, Hwang D, Byun K, Park HC, Lee YM. CCN1 interlinks integrin and hippo pathway to autoregulate tip cell activity. eLife 2019; 8:46012. [PMID: 31429823 PMCID: PMC6726423 DOI: 10.7554/elife.46012] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 08/15/2019] [Indexed: 01/14/2023] Open
Abstract
CCN1 (CYR61) stimulates active angiogenesis in various tumours, although the mechanism is largely unknown. Here, we report that CCN1 is a key regulator of endothelial tip cell activity in angiogenesis. Microvessel networks and directional vascular cell migration patterns were deformed in ccn1-knockdown zebrafish embryos. CCN1 activated VEGFR2 and downstream MAPK/PI3K signalling pathways, YAP/TAZ, as well as Rho effector mDia1 to enhance tip cell activity and CCN1 itself. VEGFR2 interacted with integrin αvβ3 through CCN1. Integrin αvβ3 inhibitor repressed tip cell number and sprouting in postnatal retinas from endothelial cell-specific Ccn1 transgenic mice, and allograft tumours in Ccn1 transgenic mice showed hyperactive vascular sprouting. Cancer patients with high CCN1 expression have poor survival outcomes and positive correlation with ITGAV and ITGB3 and high YAP/WWTR1. Thus, our data underscore the positive feedback regulation of tip cells by CCN1 through integrin αvβ3/VEGFR2 and increased YAP/TAZ activity, suggesting a promising therapeutic intervention for pathological angiogenesis.
Collapse
Affiliation(s)
- Myo-Hyeon Park
- BK21 Plus KNU Multi-Omics Creative Drug Research Team, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Ae Kyung Kim
- BK21 Plus KNU Multi-Omics Creative Drug Research Team, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea.,School of Life Sciences and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Sarala Manandhar
- BK21 Plus KNU Multi-Omics Creative Drug Research Team, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Su-Young Oh
- BK21 Plus KNU Multi-Omics Creative Drug Research Team, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Gun-Hyuk Jang
- BK21 Plus KNU Multi-Omics Creative Drug Research Team, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea.,School of Life Sciences and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Li Kang
- BK21 Plus KNU Multi-Omics Creative Drug Research Team, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Dong-Won Lee
- Department of Biomedical Sciences, Korea University, Ansan Hospital, Ansan, Republic of Korea
| | - Do Young Hyeon
- School of Interdisciplinary Bioscience and Bioengineering, POSTECH, Pohang, Republic of Korea
| | - Sun-Hee Lee
- BK21 Plus KNU Multi-Omics Creative Drug Research Team, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Hye Eun Lee
- BK21 Plus KNU Multi-Omics Creative Drug Research Team, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Tae-Lin Huh
- School of Life Sciences and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Sang Heon Suh
- Department of Internal Medicine, Chonnam National University Hospital, Gwangju, Korea
| | - Daehee Hwang
- Department of New Biology and Center for Plant Aging Research, DGIST, Daegu, Republic of Korea
| | - Kyunghee Byun
- Gachon University, School of Medicine, Incheon, Republic of Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, Korea University, Ansan Hospital, Ansan, Republic of Korea
| | - You Mie Lee
- BK21 Plus KNU Multi-Omics Creative Drug Research Team, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea.,School of Life Sciences and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
32
|
Li J, Wu S, Kim E, Yan K, Liu H, Liu C, Dong H, Qu X, Shi X, Shen J, Bentley WE, Payne GF. Electrobiofabrication: electrically based fabrication with biologically derived materials. Biofabrication 2019; 11:032002. [PMID: 30759423 PMCID: PMC7025432 DOI: 10.1088/1758-5090/ab06ea] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
While conventional material fabrication methods focus on form and strength to achieve function, the fabrication of material systems for emerging life science applications will need to satisfy a more subtle set of requirements. A common goal for biofabrication is to recapitulate complex biological contexts (e.g. tissue) for applications that range from animal-on-a-chip to regenerative medicine. In these cases, the material systems will need to: (i) present appropriate surface functionalities over a hierarchy of length scales (e.g. molecular features that enable cell adhesion and topographical features that guide differentiation); (ii) provide a suite of mechanobiological cues that promote the emergence of native-like tissue form and function; and (iii) organize structure to control cellular ingress and molecular transport, to enable the development of an interconnected cellular community that is engaged in cell signaling. And these requirements are not likely to be static but will vary over time and space, which will require capabilities of the material systems to dynamically respond, adapt, heal and reconfigure. Here, we review recent advances in the use of electrically based fabrication methods to build material systems from biological macromolecules (e.g. chitosan, alginate, collagen and silk). Electrical signals are especially convenient for fabrication because they can be controllably imposed to promote the electrophoresis, alignment, self-assembly and functionalization of macromolecules to generate hierarchically organized material systems. Importantly, this electrically based fabrication with biologically derived materials (i.e. electrobiofabrication) is complementary to existing methods (photolithographic and printing), and enables access to the biotechnology toolbox (e.g. enzymatic-assembly and protein engineering, and gene expression) to offer exquisite control of structure and function. We envision that electrobiofabrication will emerge as an important platform technology for organizing soft matter into dynamic material systems that mimic biology's complexity of structure and versatility of function.
Collapse
Affiliation(s)
- Jinyang Li
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, United States of America
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
The CCN protein family is composed of six matricellular proteins, which serve regulatory roles rather than structural roles in the extracellular matrix. First identified as secreted proteins which are induced by oncogenes, the acronym CCN came from the names of the first three members: CYR61, CTGF, and NOV. All six members of the CCN family consist of four cysteine-rich modular domains. CCN proteins are known to regulate cell adhesion, proliferation, differentiation, and apoptosis. In addition, CCN proteins are associated with cardiovascular and skeletal development, injury repair, inflammation, and cancer. They function either through binding to integrin receptors or by regulating the expression and activity of growth factors and cytokines. Given their diverse roles related to the pathology of certain diseases such as fibrosis, arthritis, atherosclerosis, diabetic nephropathy, retinopathy, and cancer, there are many emerging studies targeting CCN protein signaling pathways in attempts to elucidate their potentials as therapeutic targets. [BMB Reports 2018; 51(10): 486-493].
Collapse
Affiliation(s)
- Hyungjoo Kim
- Department of Life Science, Hanyang University, Seoul 04763, Korea
| | - Seogho Son
- Department of Life Science, Hanyang University, Seoul 04763, Korea
| | - Incheol Shin
- Department of Life Science, Hanyang University, Seoul 04763, and Natural Science Institute, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
34
|
Han GH, Peng J, Liu P, Ding X, Wei S, Lu S, Wang Y. Therapeutic strategies for peripheral nerve injury: decellularized nerve conduits and Schwann cell transplantation. Neural Regen Res 2019; 14:1343-1351. [PMID: 30964052 PMCID: PMC6524503 DOI: 10.4103/1673-5374.253511] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In recent years, the use of Schwann cell transplantation to repair peripheral nerve injury has attracted much attention. Animal-based studies show that the transplantation of Schwann cells in combination with nerve scaffolds promotes the repair of injured peripheral nerves. Autologous Schwann cell transplantation in humans has been reported recently. This article reviews current methods for removing the extracellular matrix and analyzes its composition and function. The development and secretory products of Schwann cells are also reviewed. The methods for the repair of peripheral nerve injuries that use myelin and Schwann cell transplantation are assessed. This survey of the literature data shows that using a decellularized nerve conduit combined with Schwann cells represents an effective strategy for the treatment of peripheral nerve injury. This analysis provides a comprehensive basis on which to make clinical decisions for the repair of peripheral nerve injury.
Collapse
Affiliation(s)
- Gong-Hai Han
- Kunming Medical University, Kunming, Yunnan Province; Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Ping Liu
- Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Xiao Ding
- Shihezi University Medical College, Shihezi, Xinjiang Uygur Autonomous Region, China
| | - Shuai Wei
- Shihezi University Medical College, Shihezi, Xinjiang Uygur Autonomous Region, China
| | - Sheng Lu
- 920th Hospital of Joint Service Support Force, Kunming, Yunnan Province, China
| | - Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
35
|
Aravinthan A, Park JK, Hossain MA, Sharmila J, Kim HJ, Kang CW, Kim NS, Kim JH. Collagen-based sponge hastens wound healing via decrease of inflammatory cytokines. 3 Biotech 2018; 8:487. [PMID: 30467532 DOI: 10.1007/s13205-018-1497-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/01/2018] [Indexed: 11/30/2022] Open
Abstract
The objective of this study was to compare and evaluate the efficacy of collagen-based sponge compared to commercial collagen sponge as a potent open wound-dressing material. In this study, 10 mm diameter skin incision was made on lateral side of rats. The wound was monitored regularly until day 12. Histopathology results revealed the faster re-epithelialization and lesser inflammatory cells, and also masson's trichrome staining showed that collagen fibrils were horizontal and interwoven in collagen-based sponge group. The expression of growth factors such as VEGF and TGF-β1 was found to be upregulated in transcriptional and translational levels, suggesting the importance of collagen-based sponge as a potent wound-healing material. Furthermore, IL-6 and TNF-α in the wound tissue were significantly down-regulated in 2 and 6 days in collagen-based sponge group and anti-inflammatory cytokine IL-10 level was found to be upregulated throughout 12 days. These results cumulatively revealed that collagen-based sponge may serve as novel material for wound healing in the animal model.
Collapse
Affiliation(s)
- Adithan Aravinthan
- 1Department of Physiology, College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 79 Gobong-ro, Iksan, Jeollabuk-Do 54596 Republic of Korea
| | - Jeong-Kyu Park
- R&D Center, B.B HealthCare Co. Ltd., 991 Buil-ro, Guro-gu, Seoul, Republic of Korea
| | - Mohammad Amjad Hossain
- 1Department of Physiology, College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 79 Gobong-ro, Iksan, Jeollabuk-Do 54596 Republic of Korea
| | - Judith Sharmila
- 1Department of Physiology, College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 79 Gobong-ro, Iksan, Jeollabuk-Do 54596 Republic of Korea
| | - Han-Jong Kim
- R&D Center, B.B HealthCare Co. Ltd., 991 Buil-ro, Guro-gu, Seoul, Republic of Korea
| | - Chang-Won Kang
- 1Department of Physiology, College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 79 Gobong-ro, Iksan, Jeollabuk-Do 54596 Republic of Korea
| | - Nam Soo Kim
- 1Department of Physiology, College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 79 Gobong-ro, Iksan, Jeollabuk-Do 54596 Republic of Korea
| | - Jong-Hoon Kim
- 1Department of Physiology, College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 79 Gobong-ro, Iksan, Jeollabuk-Do 54596 Republic of Korea
| |
Collapse
|
36
|
Malandraki-Miller S, Lopez CA, Al-Siddiqi H, Carr CA. Changing Metabolism in Differentiating Cardiac Progenitor Cells-Can Stem Cells Become Metabolically Flexible Cardiomyocytes? Front Cardiovasc Med 2018; 5:119. [PMID: 30283788 PMCID: PMC6157401 DOI: 10.3389/fcvm.2018.00119] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/10/2018] [Indexed: 12/15/2022] Open
Abstract
The heart is a metabolic omnivore and the adult heart selects the substrate best suited for each circumstance, with fatty acid oxidation preferred in order to fulfill the high energy demand of the contracting myocardium. The fetal heart exists in an hypoxic environment and obtains the bulk of its energy via glycolysis. After birth, the "fetal switch" to oxidative metabolism of glucose and fatty acids has been linked to the loss of the regenerative phenotype. Various stem cell types have been used in differentiation studies, but most are cultured in high glucose media. This does not change in the majority of cardiac differentiation protocols. Despite the fact that metabolic state affects marker expression and cellular function and activity, the substrate composition is currently being overlooked. In this review we discuss changes in cardiac metabolism during development, the various protocols used to differentiate progenitor cells to cardiomyocytes, what is known about stem cell metabolism and how consideration of metabolism can contribute toward maturation of stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
| | | | | | - Carolyn A. Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
37
|
Antibodies and methods for immunohistochemistry of extracellular matrix proteins. Matrix Biol 2018; 71-72:10-27. [PMID: 29730502 DOI: 10.1016/j.matbio.2018.04.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 01/12/2023]
Abstract
The diversity of extracellular matrix (ECM) proteins encoded in mammalian genomes and detected by proteomic analyses generates a need for well validated antibodies against these proteins. We present characterization of a large number of antibodies against ECM proteins, from both commercial and academic sources, together with discussion of methods and strategies for their effective use in immunohistochemistry and illustrations of their efficacy. These data should be of value to investigators seeking well validated antibodies to ECM proteins of interest and save significant time and money tracking down effective reagents.
Collapse
|
38
|
Filipe EC, Chitty JL, Cox TR. Charting the unexplored extracellular matrix in cancer. Int J Exp Pathol 2018; 99:58-76. [PMID: 29671911 DOI: 10.1111/iep.12269] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 02/26/2018] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is present in all solid tissues and considered a master regulator of cell behaviour and phenotype. The importance of maintaining the correct biochemical and biophysical properties of the ECM, and the subsequent regulation of cell and tissue homeostasis, is illustrated by the simple fact that the ECM is highly dysregulated in many different types of disease, especially cancer. The loss of tissue ECM homeostasis and integrity is seen as one of the hallmarks of cancer and typically defines transitional events in progression and metastasis. The vast majority of cancer studies place an emphasis on exploring the behaviour and intrinsic signalling pathways of tumour cells. Their goal was to identify ways to target intracellular pathways regulating cancer. Cancer progression and metastasis are powerfully influenced by the ECM and thus present a vast, unexplored repository of anticancer targets that we are only just beginning to tap into. Deconstructing the complexity of the tumour ECM landscape and identifying the interactions between the many cell types, soluble factors and extracellular-matrix proteins have proved challenging. Here, we discuss some of the emerging tools and platforms being used to catalogue and chart the ECM in cancer.
Collapse
Affiliation(s)
- Elysse C Filipe
- Cancer Division, Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, New South Wales, Australia
| | - Jessica L Chitty
- Cancer Division, Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, New South Wales, Australia
| | - Thomas R Cox
- Cancer Division, Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, New South Wales, Australia.,Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
39
|
Udhayakumar S, Shankar KG, Sowndarya S, Rose C. Novel fibrous collagen-based cream accelerates fibroblast growth for wound healing applications: in vitro and in vivo evaluation. Biomater Sci 2018; 5:1868-1883. [PMID: 28676877 DOI: 10.1039/c7bm00331e] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The present study reports the development of a novel film-forming bovine collagenous cream (BCC) based on bovine collagen (BC). In this study, collagen was isolated from bovine forestomach tissue, a novel source, and a cream formulation was prepared using some other bioactive ingredients. The electrophoretic pattern of the BC was found to be similar to type I collagen. The purity of BC was examined by amino acid analysis, which confirmed the presence of atelocollagen. The physicochemical properties of BCC such as rheology, spreadability, and temperature stability were characterized. The antimicrobial activity was examined against Bacillus subtilis, Staphylococcus aureus, and Escherichia coli, and BCC displayed excellent inhibitory effect. In vitro biocompatibility studies using NIH 3T3 fibroblast cells showed enhanced cell viability. FACS analysis revealed the non-toxic nature of BCC toward cells. The cell morphology and proliferation on the BCC matrix was studied using SEM and fluorescence microscopy. The in vivo wound healing efficacy of the BCC as a topical wound dressing was demonstrated on full thickness excision wounds in rat models. The healing profile showed that the BCC significantly enhanced re-epithelialization, collagen deposition, and contraction in the wound healing process. The findings of this study provide a new opportunity for the utilization of the untapped byproducts of the meat industry for valorization. We expect that this kind of topical healing cream could be a potential candidate in wound management and future clinical needs.
Collapse
Affiliation(s)
- Sivalingam Udhayakumar
- Department of Biochemistry and Biotechnology, CSIR-Central Leather Research Institute, Chennai 600020, India.
| | | | | | | |
Collapse
|
40
|
Wiig H, Luft FC, Titze JM. The interstitium conducts extrarenal storage of sodium and represents a third compartment essential for extracellular volume and blood pressure homeostasis. Acta Physiol (Oxf) 2018; 222. [PMID: 29193764 DOI: 10.1111/apha.13006] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/31/2017] [Accepted: 11/23/2017] [Indexed: 12/15/2022]
Abstract
The role of salt in the pathogenesis of arterial hypertension is not well understood. According to the current understanding, the central mechanism for blood pressure (BP) regulation relies on classical studies linking BP and Na+ balance, placing the kidney at the very centre of long-term BP regulation. To maintain BP homeostasis, the effective circulating fluid volume and thereby body Na+ content has to be maintained within very narrow limits. From recent work in humans and rats, the notion has emerged that Na+ could be stored somewhere in the body without commensurate water retention to buffer free extracellular Na+ and that previously unidentified extrarenal, tissue-specific regulatory mechanisms are operative regulating the release and storage of Na+ from a kidney-independent reservoir. Moreover, immune cells from the mononuclear phagocyte system not only function as local on-site sensors of interstitial electrolyte concentration, but also, together with lymphatics, act as systemic regulators of body fluid volume and BP. These studies have established new and unexpected targets in studies of BP control and thus the pathophysiology of hypertension: the interstitium/extracellular matrix of the skin, its inherent interstitial fluid and the lymphatic vasculature forming a vessel network in the interstitium. Aspects of the interstitium in relation to Na+ balance and hypertension are the focus of this review. Taken together, observations of salt storage in the skin to buffer free extracellular Na+ and macrophage modulation of the extracellular matrix and lymphatics suggest that electrolyte homeostasis in the body cannot be achieved by renal excretion alone, but also relies on extrarenal regulatory mechanisms.
Collapse
Affiliation(s)
- H. Wiig
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - F. C. Luft
- Experimental and Clinical Research Center; Max-Delbrück Center for Molecular Medicine; Charité Medical Faculty; Berlin Germany
- Division of Clinical Pharmacology; Department of Medicine; Vanderbilt University School of Medicine; Nashville TN USA
| | - J. M. Titze
- Division of Clinical Pharmacology; Department of Medicine; Vanderbilt University School of Medicine; Nashville TN USA
| |
Collapse
|
41
|
Patra C, Boccaccini A, Engel F. Vascularisation for cardiac tissue engineering: the extracellular matrix. Thromb Haemost 2017; 113:532-47. [DOI: 10.1160/th14-05-0480] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 09/03/2014] [Indexed: 02/07/2023]
Abstract
SummaryCardiovascular diseases present a major socio-economic burden. One major problem underlying most cardiovascular and congenital heart diseases is the irreversible loss of contractile heart muscle cells, the cardiomyocytes. To reverse damage incurred by myocardial infarction or by surgical correction of cardiac malformations, the loss of cardiac tissue with a thickness of a few millimetres needs to be compensated. A promising approach to this issue is cardiac tissue engineering. In this review we focus on the problem of in vitro vascularisation as implantation of cardiac patches consisting of more than three layers of cardiomyocytes (> 100 μm thick) already results in necrosis. We explain the need for vascularisation and elaborate on the importance to include non-myocytes in order to generate functional vascularised cardiac tissue. We discuss the potential of extracellular matrix molecules in promoting vascularisation and introduce nephronectin as an example of a new promising candidate. Finally, we discuss current biomaterial- based approaches including micropatterning, electrospinning, 3D micro-manufacturing technology and porogens. Collectively, the current literature supports the notion that cardiac tissue engineering is a realistic option for future treatment of paediatric and adult patients with cardiac disease.
Collapse
|
42
|
de Oliveira LSDS, de Araújo AA, de Araújo Júnior RF, Barboza CAG, Borges BCD, da Silva JSP. Low-level laser therapy (780 nm) combined with collagen sponge scaffold promotes repair of rat cranial critical-size defects and increases TGF-β, FGF-2, OPG/RANK and osteocalcin expression. Int J Exp Pathol 2017; 98:75-85. [PMID: 28556971 DOI: 10.1111/iep.12226] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 02/09/2017] [Indexed: 12/15/2022] Open
Abstract
The aim of this study was to evaluate the effect of collagen sponge scaffold (CSS) implantation associated with low-level laser therapy (LLLT) on repairing bone defects. A single 5-mm cranial defect was surgically created in forty Wistar rats, which then received one of the following four interventions (n = 10 per group): no treatment (G0); bone defect implanted with collagen sponge scaffold (CSS) alone (G1); defect treated with low-level laser therapy (LLLT) (wavelength 780 nm; total energy density 120 J/cm2 ; power 50 mW) alone (G2); and CSS associated with LLLT treatment (G3). After surgery, animals in each group were euthanized at 21 days and 30 days (n = 5 per euthanasia time group). Bone formation was monitored by X-ray imaging analysis. Biopsies were collected and processed for histological analysis and immunohistochemical evaluation of transforming growth factor-beta (TGF-β), fibroblast growth factor-2 (FGF-2), osteoprotegerin (OPG) and receptor activator of nuclear factor ƙ (RANK). Osteocalcin (OCN) was detected by immunofluorescence analysis. Compared to the G0 group, defects in the 30-day G3 group exhibited increased bone formation, both by increase in radiopaque areas (P < 0.01) and by histomorphometric analysis (P < 0.001). The histopathological analysis showed a decreased number of inflammatory cells (P < 0.001). The combined CCS + LLLT (G3) treatment also resulted in the most intense immunostaining for OPG, RANK, FGF-2 and TGF-β, and the most intense and diffuse OCN immunofluorescent labelling at 30 days postsurgery (G3 vs. G0 group, P < 0.05). Therefore, the use of CCS associated with LLLT could offer a synergistic advantage in improving the healing of bone fractures.
Collapse
Affiliation(s)
| | - Aurigena Antunes de Araújo
- Department of Biophysics and Pharmacology, Post Graduation Program in Public Health/Post Graduation Program in Pharmaceutical Science, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Raimundo Fernandes de Araújo Júnior
- Department of Morphology, Post Graduation Program in Health Science/Post Graduation Program in Functional and Structural Biology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Carlos Augusto Galvão Barboza
- Department of Morphology, Post-Graduation Program in Oral Pathology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Boniek Castillo Dutra Borges
- Department of Dentistry, Post-Graduation Program in Public Health, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - José Sandro Pereira da Silva
- Department of Dentistry, Post-Graduation Program in Public Health, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| |
Collapse
|
43
|
Fontes-Oliveira CC, Steinz M, Schneiderat P, Mulder H, Durbeej M. Bioenergetic Impairment in Congenital Muscular Dystrophy Type 1A and Leigh Syndrome Muscle Cells. Sci Rep 2017; 7:45272. [PMID: 28367954 PMCID: PMC5377256 DOI: 10.1038/srep45272] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 02/23/2017] [Indexed: 12/26/2022] Open
Abstract
Skeletal muscle has high energy requirement and alterations in metabolism are associated with pathological conditions causing muscle wasting and impaired regeneration. Congenital muscular dystrophy type 1A (MDC1A) is a severe muscle disorder caused by mutations in the LAMA2 gene. Leigh syndrome (LS) is a neurometabolic disease caused by mutations in genes related to mitochondrial function. Skeletal muscle is severely affected in both diseases and a common feature is muscle weakness that leads to hypotonia and respiratory problems. Here, we have investigated the bioenergetic profile in myogenic cells from MDC1A and LS patients. We found dysregulated expression of genes related to energy production, apoptosis and proteasome in myoblasts and myotubes. Moreover, impaired mitochondrial function and a compensatory upregulation of glycolysis were observed when monitored in real-time. Also, alterations in cell cycle populations in myoblasts and enhanced caspase-3 activity in myotubes were observed. Thus, we have for the first time demonstrated an impairment of the bioenergetic status in human MDC1A and LS muscle cells, which could contribute to cell cycle disturbance and increased apoptosis. Our findings suggest that skeletal muscle metabolism might be a promising pharmacological target in order to improve muscle function, energy efficiency and tissue maintenance of MDC1A and LS patients.
Collapse
Affiliation(s)
- Cibely C Fontes-Oliveira
- Unit of Muscle Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Maarten Steinz
- Unit of Muscle Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Peter Schneiderat
- Friedrich-Baur-Institute, Department of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Hindrik Mulder
- Unit of Molecular Metabolism, Department of Clinical Sciences, Lund University Diabetes Centre, Malmö University Hospital, Malmö, Sweden
| | - Madeleine Durbeej
- Unit of Muscle Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
44
|
Huang C, Liu L, You Z, Wang B, Du Y, Ogawa R. Keloid progression: a stiffness gap hypothesis. Int Wound J 2016; 14:764-771. [PMID: 27995750 DOI: 10.1111/iwj.12693] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 11/06/2016] [Indexed: 12/19/2022] Open
Abstract
Keloids are fibroproliferative skin disorders characterised clinically by continuous horizontal progression and post-surgical recurrence and histologically by the accumulation of collagen and fibroblast ingredients. Till now, their aetiology remains clear, which may cover genetic, environmental and metabolic factors. Evidence in the involvement of local mechanics (e.g. predilection site and typical shape) and the progress in mechanobiology have incubated our stiffness gap hypotheses in illustrating the chronic but constant development in keloid. We put forward that the enlarged gap between extracellular matrix (ECM) stiffness and cellular stiffness potentiates keloid progression. Matrix stiffness itself provides organisational guidance cues to regulate the mechanosensitive resident cells (e.g. proliferation, migration and apoptosis). During this dynamic process, the ECM stiffness and cell stiffness are not well balanced, and the continuously enlarged stiffness gap between them potentiates keloid progression. The cushion factors, such as prestress for cell stiffness and topology for ECM stiffness, serve as compensations, the decompensation of which aggravates keloid development. It can well explain the typical shape of keloids, their progression in a horizontal but not vertical direction and the post-surgical recurrence, which were evidenced by our clinical cases. Such a stiffness gap hypothesis might be bridged to mechanotherapeutic approaches for keloid progression.
Collapse
Affiliation(s)
- Chenyu Huang
- Department of Dermatology Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Longwei Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Zhifeng You
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Bingjie Wang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
45
|
Migration and Proliferative Activity of Mesenchymal Stem Cells in 3D Polylactide Scaffolds Depends on Cell Seeding Technique and Collagen Modification. Bull Exp Biol Med 2016; 162:120-126. [PMID: 27882461 DOI: 10.1007/s10517-016-3560-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Indexed: 10/20/2022]
Abstract
We analyzed viability of mesenchymal stem cells seeded by static and dynamic methods to highly porous fibrous 3D poly-L-lactide scaffolds with similar physical and chemical properties, but different spatial organization modified with collagen. Standard collagen coating promoted protein adsorption on the scaffold surface and improved adhesive properties of 100 μ-thick scaffolds. Modification of 600-μ scaffolds with collagen under pressure increased proliferative activity of mesenchymal stem cells seeded under static and dynamic (delivery of 100,000 cells in 10 ml medium in a perfusion system at a rate of 1 ml/min) conditions by 47 and 648%, respectively (measured after 120-h culturing by MTT test). Dynamic conditions provide more uniform distribution of collagen on scaffold fibers and promote cell penetration into 3D poly-L-lactide scaffolds with thickness >600 μ.
Collapse
|
46
|
Jiang M, Qiu J, Zhang L, Lü D, Long M, Chen L, Luo X. Changes in tension regulates proliferation and migration of fibroblasts by remodeling expression of ECM proteins. Exp Ther Med 2016; 12:1542-1550. [PMID: 27588075 DOI: 10.3892/etm.2016.3497] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 05/18/2016] [Indexed: 12/27/2022] Open
Abstract
Wound healing is a complicated but highly organized process in which cell migration and proliferation are actively involved. However, the process by which mechanical stretch regulates the proliferation and migration of human skin fibroblasts (HFs) and keratinocytes is poorly understood. Using a house built mechanical stretch device, we examined the HFs extracellular matrix (ECM) components changes under non-stretch, static stretch or cyclic stretch conditions. We further investigated the changes in ECM component protein expression levels in keratinocytes and analyzed the effects of individual ECM component on keratinocyte proliferation and migration. Particularly, the roles of calcium/calmodulin-dependent serine protein kinase (CASK) in the HF proliferation under cyclic stretch were investigated. Cyclic stretch suppressed HF proliferation compared with HFs without stretch or with static stretch. Cyclic stretch also led to a significant reduction in the levels of collagen I and a marked increase of fibronectin in HFs ECM. By contrast, collagen I levels increased and fibronectin levels decreased in response to non-stretch and static stretch conditions. After cyclic stretch, the proliferation of keratinocytes was inhibited by the cyclic stretch-induced ECM in HFs. The inoculation of keratinocytes with single ECM component suggested that collagen I was more capable of inducing cell proliferation than fibronectin, while it had less impact on cell migration compared with fibronectin. Furthermore, cyclic stretch induced by proliferation inhibition was associated with altered integrin β1-CASK signal pathway. The present results demonstrated the existence of HF-ECM-keratinocyte 'cross-talk' in cutaneous tissues. Thus, the integrin β1-CASK signal pathway in HFs may be involved in the outside-in signal transduction of extracellular stretch and the altered ECM component expression.
Collapse
Affiliation(s)
- Minmin Jiang
- Burn Research Institute, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China; Central Laboratory, The People's Hospital of Guizhou Province, Guiyang, Guizhou 550002, P.R. China
| | - Juhui Qiu
- Burn Research Institute, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China; National Key Laboratory of Trauma and Burns and Chongqing Key Laboratory of Disease Proteomics, Chongqing 400038, P.R. China
| | - Lingling Zhang
- Burn Research Institute, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China; National Key Laboratory of Trauma and Burns and Chongqing Key Laboratory of Disease Proteomics, Chongqing 400038, P.R. China
| | - Dongyuan Lü
- Key Laboratory of Microgravity (National Microgravity Laboratory) and Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Mian Long
- Key Laboratory of Microgravity (National Microgravity Laboratory) and Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Li Chen
- Burn Research Institute, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China; National Key Laboratory of Trauma and Burns and Chongqing Key Laboratory of Disease Proteomics, Chongqing 400038, P.R. China; Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Xiangdong Luo
- Burn Research Institute, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China; National Key Laboratory of Trauma and Burns and Chongqing Key Laboratory of Disease Proteomics, Chongqing 400038, P.R. China
| |
Collapse
|
47
|
Dong C, Lv Y. Application of Collagen Scaffold in Tissue Engineering: Recent Advances and New Perspectives. Polymers (Basel) 2016; 8:polym8020042. [PMID: 30979136 PMCID: PMC6432532 DOI: 10.3390/polym8020042] [Citation(s) in RCA: 441] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 01/24/2016] [Accepted: 01/27/2016] [Indexed: 12/11/2022] Open
Abstract
Collagen is the main structural protein of most hard and soft tissues in animals and the human body, which plays an important role in maintaining the biological and structural integrity of the extracellular matrix (ECM) and provides physical support to tissues. Collagen can be extracted and purified from a variety of sources and offers low immunogenicity, a porous structure, good permeability, biocompatibility and biodegradability. Collagen scaffolds have been widely used in tissue engineering due to these excellent properties. However, the poor mechanical property of collagen scaffolds limits their applications to some extent. To overcome this shortcoming, collagen scaffolds can be cross-linked by chemical or physical methods or modified with natural/synthetic polymers or inorganic materials. Biochemical factors can also be introduced to the scaffold to further improve its biological activity. This review will summarize the structure and biological characteristics of collagen and introduce the preparation methods and modification strategies of collagen scaffolds. The typical application of a collagen scaffold in tissue engineering (including nerve, bone, cartilage, tendon, ligament, blood vessel and skin) will be further provided. The prospects and challenges about their future research and application will also be pointed out.
Collapse
Affiliation(s)
- Chanjuan Dong
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China.
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, China.
| | - Yonggang Lv
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China.
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
48
|
Zuber M, Zia F, Zia KM, Tabasum S, Salman M, Sultan N. Collagen based polyurethanes—A review of recent advances and perspective. Int J Biol Macromol 2015; 80:366-74. [DOI: 10.1016/j.ijbiomac.2015.07.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 06/29/2015] [Accepted: 07/01/2015] [Indexed: 12/13/2022]
|
49
|
Miotto M, Gouveia RM, Connon CJ. Peptide Amphiphiles in Corneal Tissue Engineering. J Funct Biomater 2015; 6:687-707. [PMID: 26258796 PMCID: PMC4598678 DOI: 10.3390/jfb6030687] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 07/29/2015] [Accepted: 08/03/2015] [Indexed: 12/22/2022] Open
Abstract
The increasing interest in effort towards creating alternative therapies have led to exciting breakthroughs in the attempt to bio-fabricate and engineer live tissues. This has been particularly evident in the development of new approaches applied to reconstruct corneal tissue. The need for tissue-engineered corneas is largely a response to the shortage of donor tissue and the lack of suitable alternative biological scaffolds preventing the treatment of millions of blind people worldwide. This review is focused on recent developments in corneal tissue engineering, specifically on the use of self-assembling peptide amphiphiles for this purpose. Recently, peptide amphiphiles have generated great interest as therapeutic molecules, both in vitro and in vivo. Here we introduce this rapidly developing field, and examine innovative applications of peptide amphiphiles to create natural bio-prosthetic corneal tissue in vitro. The advantages of peptide amphiphiles over other biomaterials, namely their wide range of functions and applications, versatility, and transferability are also discussed to better understand how these fascinating molecules can help solve current challenges in corneal regeneration.
Collapse
Affiliation(s)
- Martina Miotto
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK.
| | - Ricardo M Gouveia
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK.
| | - Che J Connon
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK.
| |
Collapse
|
50
|
Cha SR, Jeong HK, Kim SY, Kim EY, Song JE, Park CH, Kwon SY, Khang G. Effect of Duck's Feet Derived Collagen Sponge on Skin Regeneration: In Vitro Study. POLYMER KOREA 2015. [DOI: 10.7317/pk.2015.39.3.493] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|