1
|
Deng Q, Du F, Pan S, Xia Y, Zhu Y, Zhang J, Li C, Yu S. Activation of angiopoietin-1 signaling with engineering mesenchymal stem cells promoted efficient angiogenesis in diabetic wound healing. Stem Cell Res Ther 2025; 16:75. [PMID: 39985096 PMCID: PMC11846275 DOI: 10.1186/s13287-025-04207-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/29/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Vascular insufficiency is associated with the pathogenesis and therapeutic outcomes of diabetic foot ulcers (DFU). While mesenchymal stem cells (MSCs) hold potential for DFU treatment, further enhancement in promoting angiogenesis in the challenging DFU wounds is imperative. METHODS The differential expression of pro- and anti-angiogenic factors during both normal and diabetic wound healing was compared using quantitative PCR. MSCs derived from the umbilical cord was prepared, and the engineered MSC (MSCANG1) overexpressing both the candidate pro-angiogenic gene, angiopoietin-1 (ANG1), and green fluorescent protein (GFP) was constructed using a lentiviral system. The pro-vascular stabilizing effects of MSCANG1 were assessed in primary endothelial cell cultures. Subsequently, MSCANG1 was transplanted into streptozotocin (STZ)-induced diabetic wound models to evaluate therapeutic effects on angiogenesis and wound healing. The underlying mechanisms were further examined both in vitro and in vivo. RESULTS The comprehensive analysis of the temporal expression of pro- and anti-angiogenic factors revealed a consistent impairment in ANG1 expression throughout diabetic wound healing. MSCANG1 exhibited robust EGFP expression in 80% of cells, with overexpression and secretion of the ANG1 protein. MSCANG1 notably enhanced the survival and tubulogenesis of endothelial cells and promoted the expression of junction proteins, facilitating the establishment of functional vasculature with improved vascular leakage. Although MSCANG1 did not enhance the survival of engrafted MSCs in diabetic wounds, it significantly promoted angiogenesis in diabetic wound healing, fostering the establishment of stable vasculature during the healing process. Activation of the protein kinase B (Akt) pathway and suppression of proto-oncogene tyrosine kinase Src (Src) activity in MSCANG1-treated diabetic wounds confirmed efficient angiogenesis process. Consequently, epidermal and dermal reconstruction, as well as skin appendage regeneration were markedly accelerated in MSCANG1-treated diabetic wounds compared to MSC-treated wounds. CONCLUSION Treatment with MSCs alone promotes angiogenesis and DFU healing, while the engineering of MSCs with ANG1 provides substantial additional benefits to this therapeutic process. The engineering of MSCs with ANG1 presents a promising avenue for developing innovative strategies in managing DFU.
Collapse
Affiliation(s)
- Qiong Deng
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - Fangzhou Du
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - Shenzhen Pan
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yuchen Xia
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yuxin Zhu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jingzhong Zhang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China.
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Chenglong Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Shuang Yu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China.
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
2
|
Villari G, Gioelli N, Gino M, Zhang H, Hodge K, Cordero F, Zanivan S, Zhu J, Serini G. Luminescent sensing of conformational integrin activation in living cells. Cell Rep 2025; 44:115319. [PMID: 39964812 DOI: 10.1016/j.celrep.2025.115319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 11/29/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025] Open
Abstract
Integrins are major receptors for secreted extracellular matrix, playing crucial roles in physiological and pathological contexts, such as angiogenesis and cancer. Regulation of the transition between inactive and active conformation is key for integrins to fulfill their functions, and pharmacological control of those dynamics may have therapeutic applications. We create and validate a prototypic luminescent β1 integrin activation sensor (β1IAS) by introducing a split luciferase into an activation reporting site between the βI and the hybrid domains. As a recombinant protein in both solution and living cells, β1IAS accurately reports β1 integrin activation in response to (bio)chemical and physical stimuli. A short interfering RNA (siRNA) high-throughput screening on live β1IAS knockin endothelial cells unveils hitherto unknown regulators of β1 integrin activation, such as β1 integrin inhibitors E3 ligase Pja2 and vascular endothelial growth factor B (VEGF-B). This split-luciferase-based strategy provides an in situ label-free measurement of integrin activation and may be applicable to other β integrins and receptors.
Collapse
Affiliation(s)
- Giulia Villari
- Department of Oncology, University of Torino School of Medicine, Candiolo, TO, Italy; Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo, TO, Italy
| | - Noemi Gioelli
- Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo, TO, Italy
| | - Marta Gino
- Department of Oncology, University of Torino School of Medicine, Candiolo, TO, Italy; Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo, TO, Italy
| | - Heng Zhang
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Kelly Hodge
- Cancer Research UK Scotland Institute, Glasgow, UK
| | | | - Sara Zanivan
- Cancer Research UK Scotland Institute, Glasgow, UK; School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Jieqing Zhu
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA; Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Guido Serini
- Department of Oncology, University of Torino School of Medicine, Candiolo, TO, Italy; Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo, TO, Italy.
| |
Collapse
|
3
|
Gao D, Shipman WD, Sun Y, Yang W, Mathew AT, Beraki L, Glahn JZ, Kochen A, Kyriakides TR, Horsley V, Hsia HC. An Injectable Alginate Hydrogel Modified by Collagen and Fibronectin for Better Cellular Environment. ACS APPLIED BIO MATERIALS 2025; 8:1675-1683. [PMID: 39886738 DOI: 10.1021/acsabm.4c01853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Encapsulating fibroblasts in alginate hydrogels is a promising strategy to promote wound healing. However, improving the cell function within the alginate matrix remains a challenge. In this study, we engineer an injectable hydrogel through mixing alginate function with collagen and fibronectin, creating a better microenvironment for enhancing fibroblast function and cytokine secretion. We systematically analyze microstructure, mechanical properties, and fibroblast behavior of the developed hydrogel and compare it to alginate control. Our results demonstrate that inclusion collagen and fibronectin lead to the formation of fibrils on macroporous structures with pore sizes ranging from 100 to 500 μm. Compared to collagen hydrogel, the composite hydrogel shows approximately 12-fold increase in storage modulus. After encapsulating fibroblasts into the modified hydrogels, we observed increased fibroblast spreading, proliferation, and cytokine secretion when compared to neat alginate hydrogel. In addition, VEGF secretion of encapsulated fibroblasts is upregulated, indicating its pro-angiogenic potential. These findings suggest that the alginate/collagen/fibronectin hydrogel-encapsulated fibroblasts might serve as a promising therapeutic approach for wound healing.
Collapse
Affiliation(s)
- Daqian Gao
- Division of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, 310 Cedar Street, New Haven, Connecticut 06510, United States
- VA Connecticut Healthcare, 950 Campbell Ave, West Haven, Connecticut 06516, United States
| | - William D Shipman
- Department of Dermatology, Yale School of Medicine, Yale University, 333 Cedar Street, New Haven, Connecticut 06510, United States
| | - Yaping Sun
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, Yale University, 333 Cedar Street, New Haven, Connecticut 06510, United States
| | - Weijun Yang
- The Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Angelin Tresa Mathew
- Department of Molecular, Cellular, Developmental Biology, Yale University, 260 Whitney Ave, New Haven, Connecticut 06511, United States
| | - Leleda Beraki
- Department of Biomedical Engineering, Yale University, 17 Hillhouse Ave, New Haven, Connecticut 06511, United States
| | - Joshua Zev Glahn
- Division of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, 310 Cedar Street, New Haven, Connecticut 06510, United States
| | - Alejandro Kochen
- Division of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, 310 Cedar Street, New Haven, Connecticut 06510, United States
| | - Themis R Kyriakides
- Department of Biomedical Engineering, Yale University, 17 Hillhouse Ave, New Haven, Connecticut 06511, United States
- Vascular Biology and Therapeutics Program, Yale School of Medicine, Yale University, 10 Amistad Street, New Haven, Connecticut 06510, United States
| | - Valerie Horsley
- Department of Dermatology, Yale School of Medicine, Yale University, 333 Cedar Street, New Haven, Connecticut 06510, United States
- Department of Molecular, Cellular, Developmental Biology, Yale University, 260 Whitney Ave, New Haven, Connecticut 06511, United States
| | - Henry C Hsia
- Division of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, 310 Cedar Street, New Haven, Connecticut 06510, United States
- VA Connecticut Healthcare, 950 Campbell Ave, West Haven, Connecticut 06516, United States
- Department of Biomedical Engineering, Yale University, 17 Hillhouse Ave, New Haven, Connecticut 06511, United States
| |
Collapse
|
4
|
Erning K, Wilson KL, Smith CS, Nguyen L, Joesph NI, Irengo R, Cao LY, Cumaran M, Shi Y, Lyu S, Riley L, Dunn TW, Carmichael ST, Segura T. Clustered VEGF Nanoparticles in Microporous Annealed Particle (MAP) Hydrogel Accelerates Functional Recovery and Brain Tissue Repair after Stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635733. [PMID: 39974959 PMCID: PMC11838428 DOI: 10.1101/2025.01.30.635733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Ischemic stroke, a blockage in the vasculature of the brain that results in insufficient blood flow, is one of the world's leading causes of disability. The cascade of inflammation and cell death that occurs immediately following stroke drives vascular and functional loss that does not fully recover over time, and no FDA-approved therapies exist that stimulate regeneration post-stroke. We have previously developed a hydrogel scaffold that delivered heparin nanoparticles with and without VEGF bound to their surface to promote angiogenesis and reduce inflammation, respectively. However, the inclusion of the naked heparin nanoparticles warranted concern over the development of bleeding complications. Here, we explore how microporous annealed particle (MAP) scaffolds functionalized with VEGF coated heparin nanoparticles can both reduce inflammation and promote angiogenesis - without the inclusion of free heparin nanoparticles. We show that our updated design not only successfully promotes de novo tissue formation, including the development of mature vessels and neurite sprouting, but it also leads to functional improvement in a photothrombotic stroke model. In addition, we find increased astrocyte infiltration into the infarct site correlated with mature vessel formation. This work demonstrates how our biomaterial design can enhance endogenous regeneration post-stroke while eliminating the need for excess heparin.
Collapse
|
5
|
Zheng Z, Gan S, Yang S, Hou C, Zhu Z, Wang H, Yu D, Qian Z, Xu HHK, Chen W. Enhanced surface hydrophilicity improves osseointegration of titanium implants via integrin-mediated osteoimmunomodulation. J Mater Chem B 2025; 13:496-510. [PMID: 39688175 DOI: 10.1039/d4tb02360a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Titanium (Ti) implants have become widespread especially in dentistry and orthopedics, where macrophage-driven osteoimmunomodulation is crucial to their success. Hydrophilic modification of Ti represents a promising strategy to enhance its immune and osteogenic responses. Herein, the osteoimmunomodulatory performance and integrin-mediated mechanism of novel non-thermal atmospheric plasma (NTAP) treatment to induce a hydrophilic Ti were investigated for the first time. Compared to a hydrophobic surface, NTAP-modified Ti possessed a 3-fold increase of pro-healing M2 macrophage makers, and the doubled osteogenic differentiation of mesenchymal stem cells was demonstrated in this immune microenvironment, thus improving early osseointegration. Mechanistically, the ameliorative osteoimmunomodulatory properties of NTAP were attributed to its positive and negative modulation in macrophages' integrin β1 or β2, and the subsequent FAK-PI3K/Akt or NF-κB signaling pathway. Collectively, this study highlighted the role of integrins and related signaling pathways in hydrophilic implant-caused macrophage polarization, therefore inventively unveiling the underlying mechanism of NTAP-enhanced osteoimmunomodulation. Furthermore, it established a robust theoretical foundation for the clinical application of this cost-effective, versatile, and transformation-valuable surface engineering strategy for the development of next-generation Ti implants.
Collapse
Affiliation(s)
- Zheng Zheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Shuaiqi Gan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Shuhan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Chuping Hou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Zhimin Zhu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Hang Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Deping Yu
- School of Mechanical Engineering, Sichuan University, Chengdu 610041, China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, Sichuan, China
| | - Hockin H K Xu
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Wenchuan Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
6
|
Wen Z, Orduno M, Liang Z, Gong X, Mak M. Optimization of Vascularized Intestinal Organoid Model. Adv Healthc Mater 2024; 13:e2400977. [PMID: 39091070 PMCID: PMC11652258 DOI: 10.1002/adhm.202400977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/03/2024] [Indexed: 08/04/2024]
Abstract
Vasculature is crucial for maintaining organ homeostasis and metabolism. Although 3D organoids can mimic organ structures and patterns, they still lack vascular systems, limiting the recapitulation of physiological complexities. Although vascularization of organoids has been demonstrated by mixing Matrigel in fibrin, how the mixed gel niche affects endothelial cells (ECs) and organoids remains unclear. Existing protocols rely on fibroblasts to promote vascular network formation. This study explores how varying the ratio of Matrigel in fibrin-Matrigel co-gel affects vascular network formation and intestinal organoid growth. A fine-tuned hydrogel is developed by adding aprotinin and 15% Matrigel in fibrin. Medium for co-culturing ECs and organoids is modified with basic fibroblast growth factor (bFGF) and heparin. In combination with fine-tuned hydrogel and modified medium, vascular network formation and organoid vascularization are successfully generated in the absence of fibroblast. Furthermore, structural cues and pore architectures are critical for angiogenesis and vascularization. By incorporating engineered thick collagen fiber bundles into the system, vascular network formation is guided by bundle architectures, enhancing interactions between vascular networks and organoids. The results demonstrate an optimized system that advances tissue and organoid vascularization by combining fiber bundles with fine-tuned hydrogel and modified medium.
Collapse
Affiliation(s)
- Zhang Wen
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Mariabelen Orduno
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Zixie Liang
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Xiangyu Gong
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| |
Collapse
|
7
|
Ramaraj PK, Pol M, Scinto SL, Jia X, Fox JM. Covalent Attachment of Functional Proteins to Microfiber Surfaces via a General Strategy for Site-Selective Tetrazine Ligation. ACS APPLIED MATERIALS & INTERFACES 2024; 16:63195-63206. [PMID: 39503333 PMCID: PMC11824234 DOI: 10.1021/acsami.4c12609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Surface modification of materials with proteins has various biological applications, and hence the methodology for surface modification needs to accommodate a wide range of proteins that differ in structure, size, and function. Presented here is a methodology that uses the Affinity Bioorthogonal Chemistry (ABC) tag, 3-(2-pyridyl)-6-methyltetrazine (PyTz), for the site-selective modification and purification of proteins and subsequent attachment of the protein to trans-cyclooctene (TCO)-functionalized hydrogel microfibers. This method of surface modification is shown to maintain the functionality of the protein after conjugation with proteins of varying size and functionalities, namely, HaloTag, NanoLuc luciferase (NanoLuc), and fibronectin type III domains 9-10 (FNIII 9-10). The method also supports surface modification with multiple proteins, which is shown by the simultaneous conjugation of HaloTag and NanoLuc on the microfiber surface. The ability to control the relative concentrations of multiple proteins presented on the surface is shown with the use of HaloTag and superfolder GFP (sfGFP). This application of the ABC-tagging methodology expands on existing surface modification methods and provides flexibility in the site-selective protein conjugation methods used along with the rapid kinetics of tetrazine ligation.
Collapse
Affiliation(s)
- Paramesh K Ramaraj
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - Mugdha Pol
- Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, United States
| | - Samuel L Scinto
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - Xinqiao Jia
- Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, United States
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Joseph M Fox
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| |
Collapse
|
8
|
Pandit A, Indurkar A, Locs J, Haugen HJ, Loca D. Calcium Phosphates: A Key to Next-Generation In Vitro Bone Modeling. Adv Healthc Mater 2024; 13:e2401307. [PMID: 39175382 PMCID: PMC11582516 DOI: 10.1002/adhm.202401307] [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: 04/09/2024] [Revised: 08/06/2024] [Indexed: 08/24/2024]
Abstract
The replication of bone physiology under laboratory conditions is a prime target behind the development of in vitro bone models. The model should be robust enough to elicit an unbiased response when stimulated experimentally, giving reproducible outcomes. In vitro bone tissue generation majorly requires the availability of cellular components, the presence of factors promoting cellular proliferation and differentiation, efficient nutrient supply, and a supporting matrix for the cells to anchor - gaining predefined topology. Calcium phosphates (CaP) are difficult to ignore while considering the above requirements of a bone model. Therefore, the current review focuses on the role of CaP in developing an in vitro bone model addressing the prerequisites of bone tissue generation. Special emphasis is given to the physico-chemical properties of CaP that promote osteogenesis, angiogenesis and provide sufficient mechanical strength for load-bearing applications. Finally, the future course of action is discussed to ensure efficient utilization of CaP in the in vitro bone model development field.
Collapse
Affiliation(s)
- Ashish Pandit
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| | - Abhishek Indurkar
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| | - Janis Locs
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| | | | - Dagnija Loca
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| |
Collapse
|
9
|
Huang J, Bao C, Yang C, Qu Y. Dual-tDCS Ameliorates Cerebral Injury and Promotes Motor Function Recovery via cGAS-STING Signaling Pathway in a Rat Model of Ischemic Stroke. Mol Neurobiol 2024:10.1007/s12035-024-04574-x. [PMID: 39455539 DOI: 10.1007/s12035-024-04574-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024]
Abstract
Ischemic stroke is one of the leading causes of death and disability. Dual transcranial direct current stimulation (dual-tDCS) is a promising intervention to treat ischemic stroke, but its efficacy and underlying mechanism remain to be verified. Cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway has recently emerged as a key mediator in cerebral injury. However, little is known about the effect of cGAS-STING on neuronal damage in ischemic stroke, and it remains to be studied whether the cGAS-STING pathway is involved in tDCS intervention for ischemic stroke. Therefore, we aimed to investigate whether dual-tDCS can alleviate ischemic brain injury in a rat model of ischemic stroke and if so, whether via cGAS-STING pathway. Middle cerebral artery occlusion (MCAO) was employed to induce a rat model of ischemic stroke. Male SD rats weighing 250-280 g were randomly assigned to the Sham, MCAO, Dual-tDCS, Dual-tDCS + RU.521, and Dual-tDCS + 2'3'-cGAMP groups, with 10 rats in each group completing the experiment. Behavioral, morphological, MRI, and molecular biological methods were performed. We found that the cGAS-STING pathway was activated and expressed in neurons after MCAO. Dual-tDCS improved motor function and infarct volume, inhibited neuronal apoptosis, promoted the expression of neurotrophins (BDNF and NGF), CD31, and VEGF, and suppressed inflammation reaction after MCAO via the cGAS-STING pathway. Taken together, dual-tDCS may improve MCAO-induced brain injury and promote the recovery of motor function, resulting from the inhibition of neuronal apoptosis and inflammation reaction, as well as promotion of the expression of nerve plasticity- and angiogenesis-related proteins, via cGAS-STING pathway.
Collapse
Affiliation(s)
- Jiapeng Huang
- Clinical Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Research Laboratory of Neurorehabilitation, Research Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chuncha Bao
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chunlan Yang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Research Laboratory of Neurorehabilitation, Research Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yun Qu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Research Laboratory of Neurorehabilitation, Research Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
10
|
Li W, Moretti L, Su X, Yeh CR, Torres MP, Barker TH. Strain-dependent glutathionylation of fibronectin fibers impacts mechano-chemical behavior and primes an integrin switch. Nat Commun 2024; 15:8751. [PMID: 39384749 PMCID: PMC11479631 DOI: 10.1038/s41467-024-52742-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/18/2024] [Indexed: 10/11/2024] Open
Abstract
The extracellular matrix (ECM) is a protein polymer network that physically supports cells within a tissue. It acts as an important physical and biochemical stimulus directing cell behaviors. For fibronectin (Fn), a predominant component of the ECM, these physical and biochemical activities are inextricably linked as physical forces trigger conformational changes that impact its biochemical activity. Here, we analyze whether oxidative post-translational modifications, specifically glutathionylation, alter Fn's mechano-chemical characteristics through stretch-dependent protein modification. ECM post-translational modifications represent a potential for time- or stimulus-dependent changes in ECM structure-function relationships that could persist over time with potentially significant impacts on cell and tissue behaviors. In this study, we show evidence that glutathionylation of Fn ECM fibers is stretch-dependent and alters Fn fiber mechanical properties with implications on the selectivity of engaging integrin receptors. These data demonstrate the existence of multimodal post-translational modification mechanisms within the ECM with high relevance to the microenvironmental regulation of downstream cell behaviors.
Collapse
Affiliation(s)
- Wei Li
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA, USA
| | - Leandro Moretti
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA, USA
| | - Xinya Su
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Chiuan-Ren Yeh
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA, USA
| | - Matthew P Torres
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
11
|
Rust R, Nih LR, Liberale L, Yin H, El Amki M, Ong LK, Zlokovic BV. Brain repair mechanisms after cell therapy for stroke. Brain 2024; 147:3286-3305. [PMID: 38916992 PMCID: PMC11449145 DOI: 10.1093/brain/awae204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/04/2024] [Accepted: 06/08/2024] [Indexed: 06/27/2024] Open
Abstract
Cell-based therapies hold great promise for brain repair after stroke. While accumulating evidence confirms the preclinical and clinical benefits of cell therapies, the underlying mechanisms by which they promote brain repair remain unclear. Here, we briefly review endogenous mechanisms of brain repair after ischaemic stroke and then focus on how different stem and progenitor cell sources can promote brain repair. Specifically, we examine how transplanted cell grafts contribute to improved functional recovery either through direct cell replacement or by stimulating endogenous repair pathways. Additionally, we discuss recently implemented preclinical refinement methods, such as preconditioning, microcarriers, genetic safety switches and universal (immune evasive) cell transplants, as well as the therapeutic potential of these pharmacologic and genetic manipulations to further enhance the efficacy and safety of cell therapies. By gaining a deeper understanding of post-ischaemic repair mechanisms, prospective clinical trials may be further refined to advance post-stroke cell therapy to the clinic.
Collapse
Affiliation(s)
- Ruslan Rust
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
| | - Lina R Nih
- Department of Brain Health, University of Nevada, Las Vegas, NV 89154, USA
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Hao Yin
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Lin Kooi Ong
- School of Health and Medical Sciences & Centre for Health Research, University of Southern Queensland, Toowoomba, QLD 4350, Australia
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
12
|
Libby JR, Royce H, Walker SR, Li L. The role of extracellular matrix in angiogenesis: Beyond adhesion and structure. BIOMATERIALS AND BIOSYSTEMS 2024; 15:100097. [PMID: 39129826 PMCID: PMC11315062 DOI: 10.1016/j.bbiosy.2024.100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/19/2024] [Accepted: 07/06/2024] [Indexed: 08/13/2024] Open
Abstract
While the extracellular matrix (ECM) has long been recognized for its structural contributions, anchoring cells for adhesion, providing mechanical support, and maintaining tissue integrity, recent efforts have elucidated its dynamic, reciprocal, and diverse properties on angiogenesis. The ECM modulates angiogenic signaling and mechanical transduction, influences the extent and degree of receptor activation, controls cellular behaviors, and serves as a reservoir for bioactive macromolecules. Collectively, these factors guide the formation, maturation, and stabilization of a functional vascular network. This review aims to shed light on the versatile roles of the ECM in angiogenesis, transcending its traditional functions as a mere structural material. We will explore its engagement and synergy in signaling modulation, interactions with various angiogenic factors, and highlight its importance in both health and disease. By capturing the essence of the ECM's diverse functionalities, we highlight the significance in the broader context of vascular biology, enabling the design of novel biomaterials to engineer vascularized tissues and their potential therapeutic implications.
Collapse
Affiliation(s)
- Jaxson R. Libby
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Haley Royce
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, USA
| | - Sarah R. Walker
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Linqing Li
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, USA
- Department of Chemistry, University of New Hampshire, Durham, NH, USA
| |
Collapse
|
13
|
Oliverio R, Liberelle B, Patenaude V, Moreau V, Thomas E, Virgilio N, Banquy X, De Crescenzo G. Cofunctionalization of Macroporous Dextran Hydrogels with Adhesive Peptides and Growth Factors Enables Vascular Spheroid Sprouting. ACS Biomater Sci Eng 2024; 10:5080-5093. [PMID: 39038278 DOI: 10.1021/acsbiomaterials.4c00455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Ensuring good definition of scaffolds used for 3D cell culture is a prominent challenge that hampers the development of tissue engineering platforms. Since dextran repels cell adhesion, using dextran-based materials biofunctionalized through a bottom-up approach allows for precise control over material definition. Here, we report the design of dextran hydrogels displaying a fully interconnected macropore network for the culture of vascular spheroids in vitro. We biofunctionalized the hydrogels with the RGD peptide sequence to promote cell adhesion. We used an affinity peptide pair, the E/K coiled coil, to load the gels with epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF). Dual functionalization with adhesive and proliferative cues allows vascular spheroids to colonize naturally cell-repellant dextran. In supplement-depleted medium, we report improved colonization of the macropores compared to that of unmodified dextran. Altogether, we propose a well-defined and highly versatile platform for tissue engineering and tissue vascularization applications.
Collapse
Affiliation(s)
- Romane Oliverio
- Department of Chemical Engineering, Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
- Faculty of Pharmacy, Axe Formulation et Analyse du Médicament (AFAM), Université de Montréal, Montréal H3T 1J4, Québec, Canada
| | - Benoît Liberelle
- Department of Chemical Engineering, Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
| | - Victor Patenaude
- Department of Chemical Engineering, Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
| | - Vaiana Moreau
- Department of Chemical Engineering, Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
- Department of Chemical Engineering, Centre de Recherche sur les Systèmes Polymères et Composites à Haute Performance (CREPEC), Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
| | - Elian Thomas
- Department of Chemical Engineering, Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
| | - Nick Virgilio
- Department of Chemical Engineering, Centre de Recherche sur les Systèmes Polymères et Composites à Haute Performance (CREPEC), Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
| | - Xavier Banquy
- Faculty of Pharmacy, Axe Formulation et Analyse du Médicament (AFAM), Université de Montréal, Montréal H3T 1J4, Québec, Canada
| | - Gregory De Crescenzo
- Department of Chemical Engineering, Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
| |
Collapse
|
14
|
Xing T, Wang X, Xu Y, Sun F, Chen M, Yan Q, Ma Z, Jiang H, Chen X, Li X, Sultan R, Yan T, Wang Z, Jia J. Click method preserves but EDC method compromises the therapeutic activities of the peptide-activated hydrogels for critical ischemic vessel regeneration. Biomed Pharmacother 2024; 177:116959. [PMID: 38906023 DOI: 10.1016/j.biopha.2024.116959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/25/2024] [Accepted: 06/15/2024] [Indexed: 06/23/2024] Open
Abstract
Peptide-functionalized hydrogel is one of commonly used biomaterials to introduce hydrogel-induced vessel regeneration. Despite many reports about the discoveries of high-active peptides (or ligands) for regeneration, the study on the conjugating methods for the hydrogel functionalization with peptides is limited. Here, we compared the vasculogenic efficacy of the peptide-functionalized hydrogels prepared by two commonly used conjugating methods, 1-ethyl-3-(3-dimethylamino propyl) carbodiimide (EDC) and Click methods, through cell models, organ-on-chips models, animal models, and RNA sequencing analysis. Two vascular-related cell types, the human umbilical vein endothelial cells (HUVECs) and the adipose-derived stem cells (ADSCs), have been cultured on the hydrogel surfaces prepared by EDC/Click methods. It showed that the hydrogels prepared by Click method supported the higher vasculogenic activities while the ones made by EDC method compromised the peptide activities on hydrogels. The vasculogenesis assays further revealed that hydrogels prepared by Click method promoted a better vascular network formation. In a critical ischemic hindlimb model, only the peptide-functionalized hydrogels prepared by Click method successfully salvaged the ischemic limb, significantly improved blood perfusion, and enhanced the functional recoveries (through gait analysis and animal behavior studies). RNA sequencing studies revealed that the hydrogels prepared by Click method significantly promoted the PI3K-AKT pathway activation compared to the hydrogels prepared by EDC method. All the results suggested that EDC method compromised the functions of the peptides, while Click method preserved the vascular regenerating capacities of the peptides on the hydrogels, illustrating the importance of the conjugating method during the preparation of the peptide-functionalized hydrogels.
Collapse
Affiliation(s)
- Tongying Xing
- School of Life Sciences, Shanghai University, Shanghai, China; Sino-Swiss Institute of Advanced Technology, School of Micro-electronics, Shanghai University, Shanghai, China
| | - Xuelin Wang
- School of Life Sciences, Shanghai University, Shanghai, China; Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai, China
| | - Yongqiang Xu
- Department of colorectal surgery, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Fei Sun
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Min Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Qiang Yan
- Department of Surgery, Huzhou Central Hospital, Huzhou, Zhejiang, China; Department of Surgery, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang, China
| | - Zhihong Ma
- Department of Precision Medical Clinical Research Center, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Haihong Jiang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xingxing Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xueyi Li
- Sino-Swiss Institute of Advanced Technology, School of Micro-electronics, Shanghai University, Shanghai, China
| | - Rabia Sultan
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Tingdong Yan
- School of Life Sciences, Shanghai University, Shanghai, China.
| | - Zhimin Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai, China.
| | - Jia Jia
- School of Life Sciences, Shanghai University, Shanghai, China; Sino-Swiss Institute of Advanced Technology, School of Micro-electronics, Shanghai University, Shanghai, China.
| |
Collapse
|
15
|
Hosty L, Heatherington T, Quondamatteo F, Browne S. Extracellular matrix-inspired biomaterials for wound healing. Mol Biol Rep 2024; 51:830. [PMID: 39037470 PMCID: PMC11263448 DOI: 10.1007/s11033-024-09750-9] [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: 05/21/2024] [Accepted: 06/21/2024] [Indexed: 07/23/2024]
Abstract
Diabetic foot ulcers (DFU) are a debilitating and life-threatening complication of Diabetes Mellitus. Ulceration develops from a combination of associated diabetic complications, including neuropathy, circulatory dysfunction, and repetitive trauma, and they affect approximately 19-34% of patients as a result. The severity and chronic nature of diabetic foot ulcers stems from the disruption to normal wound healing, as a result of the molecular mechanisms which underly diabetic pathophysiology. The current standard-of-care is clinically insufficient to promote healing for many DFU patients, resulting in a high frequency of recurrence and limb amputations. Biomaterial dressings, and in particular those derived from the extracellular matrix (ECM), have emerged as a promising approach for the treatment of DFU. By providing a template for cell infiltration and skin regeneration, ECM-derived biomaterials offer great hope as a treatment for DFU. A range of approaches exist for the development of ECM-derived biomaterials, including the use of purified ECM components, decellularisation and processing of donor/ animal tissues, or the use of in vitro-deposited ECM. This review discusses the development and assessment of ECM-derived biomaterials for the treatment of chronic wounds, as well as the mechanisms of action through which ECM-derived biomaterials stimulate wound healing.
Collapse
Affiliation(s)
- Louise Hosty
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
| | - Thomas Heatherington
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
| | - Fabio Quondamatteo
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland.
| | - Shane Browne
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland.
- CÙRAM, Centre for Research in Medical Devices, University of Galway, Galway, H91 W2TY, Ireland.
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
16
|
Wei J, Sun Y, Wang H, Zhu T, Li L, Zhou Y, Liu Q, Dai Z, Li W, Yang T, Wang B, Zhu C, Shen X, Yao Q, Song G, Zhao Y, Pei H. Designer cellular spheroids with DNA origami for drug screening. SCIENCE ADVANCES 2024; 10:eado9880. [PMID: 39028810 PMCID: PMC11259176 DOI: 10.1126/sciadv.ado9880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 06/14/2024] [Indexed: 07/21/2024]
Abstract
Current in vitro models struggle to balance the complexity of human diseases with suitability for large-scale drug tests. While 3D cultures simulate human tissues, they lack cellular intricacy, and integrating these models with high-throughput drug screening remains a challenge. Here, we introduce a method that uses self-assembling nucleic acid nanostructures decorated living cells, termed NACs, to create spheroids with a customizable 3D layout. To demonstrate its uniqueness, our method effectively creates designer 3D spheroids by combining parenchymal cells, stromal cells, and immune cells, leading to heightened physiological relevance and detailed modeling of complex chronic diseases and immune-stromal interactions. Our approach achieves a high level of biological fidelity while being standardized and straightforward to construct with the potential for large-scale drug discovery applications. By merging the precision of DNA nanotechnology with advanced cell culture techniques, we are streamlining human-centric models, striking a balance between complexity and standardization, to boost drug screening efficiency.
Collapse
Affiliation(s)
- Jiayi Wei
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Liver Diseases, Shanghai 200032, China
| | - Yueyang Sun
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Center of Brain-inspired Intelligent Materials and Devices, East China Normal University, Shanghai 200241, China
| | - Heming Wang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Liver Diseases, Shanghai 200032, China
| | - Tong Zhu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Center of Brain-inspired Intelligent Materials and Devices, East China Normal University, Shanghai 200241, China
| | - Li Li
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Center of Brain-inspired Intelligent Materials and Devices, East China Normal University, Shanghai 200241, China
| | - Ying Zhou
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Center of Brain-inspired Intelligent Materials and Devices, East China Normal University, Shanghai 200241, China
| | - Quan Liu
- Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130117, China
| | - Zhen Dai
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Wenjuan Li
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Taihua Yang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China
| | - Bingmei Wang
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Changfeng Zhu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Liver Diseases, Shanghai 200032, China
| | - Xizhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Liver Diseases, Shanghai 200032, China
| | - Qunyan Yao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Liver Diseases, Shanghai 200032, China
- Shanghai Geriatric Medical Center, Shanghai 201104, China
- Department of Gastroenterology and Hepatology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, China
| | - Guangqi Song
- Joint Laboratory of Biomaterials and Translational Medicine, Puheng Technology, Suzhou 215000, China
| | - Yicheng Zhao
- Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130117, China
- China-Japan Union Hospital of Jilin University, 130012 Changchun, Jilin, China
| | - Hao Pei
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Center of Brain-inspired Intelligent Materials and Devices, East China Normal University, Shanghai 200241, China
| |
Collapse
|
17
|
Mukhopadhyay U, Mandal T, Chakraborty M, Sinha B. The Plasma Membrane and Mechanoregulation in Cells. ACS OMEGA 2024; 9:21780-21797. [PMID: 38799362 PMCID: PMC11112598 DOI: 10.1021/acsomega.4c01962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 05/29/2024]
Abstract
Cells inhabit a mechanical microenvironment that they continuously sense and adapt to. The plasma membrane (PM), serving as the boundary of the cell, plays a pivotal role in this process of adaptation. In this Review, we begin by examining well-studied processes where mechanoregulation proves significant. Specifically, we highlight examples from the immune system and stem cells, besides discussing processes involving fibroblasts and other cell types. Subsequently, we discuss the common molecular players that facilitate the sensing of the mechanical signal and transform it into a chemical response covering integrins YAP/TAZ and Piezo. We then review how this understanding of molecular elements is leveraged in drug discovery and tissue engineering alongside a discussion of the methodologies used to measure mechanical properties. Focusing on the processes of endocytosis, we discuss how cells may respond to altered membrane mechanics using endo- and exocytosis. Through the process of depleting/adding the membrane area, these could also impact membrane mechanics. We compare pathways from studies illustrating the involvement of endocytosis in mechanoregulation, including clathrin-mediated endocytosis (CME) and the CLIC/GEEC (CG) pathway as central examples. Lastly, we review studies on cell-cell fusion during myogenesis, the mechanical integrity of muscle fibers, and the reported and anticipated roles of various molecular players and processes like endocytosis, thereby emphasizing the significance of mechanoregulation at the PM.
Collapse
Affiliation(s)
- Upasana Mukhopadhyay
- Department of Biological
Sciences, Indian Institute of Science Education
and Research Kolkata, Mohanpur, West Bengal 741246, India
| | - Tithi Mandal
- Department of Biological
Sciences, Indian Institute of Science Education
and Research Kolkata, Mohanpur, West Bengal 741246, India
| | | | - Bidisha Sinha
- Department of Biological
Sciences, Indian Institute of Science Education
and Research Kolkata, Mohanpur, West Bengal 741246, India
| |
Collapse
|
18
|
Wang G, Li Z, Wang G, Sun Q, Lin P, Wang Q, Zhang H, Wang Y, Zhang T, Cui F, Zhong Z. Advances in Engineered Nanoparticles for the Treatment of Ischemic Stroke by Enhancing Angiogenesis. Int J Nanomedicine 2024; 19:4377-4409. [PMID: 38774029 PMCID: PMC11108071 DOI: 10.2147/ijn.s463333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/02/2024] [Indexed: 05/24/2024] Open
Abstract
Angiogenesis, or the formation of new blood vessels, is a natural defensive mechanism that aids in the restoration of oxygen and nutrition delivery to injured brain tissue after an ischemic stroke. Angiogenesis, by increasing vessel development, may maintain brain perfusion, enabling neuronal survival, brain plasticity, and neurologic recovery. Induction of angiogenesis and the formation of new vessels aid in neurorepair processes such as neurogenesis and synaptogenesis. Advanced nano drug delivery systems hold promise for treatment stroke by facilitating efficient transportation across the the blood-brain barrier and maintaining optimal drug concentrations. Nanoparticle has recently been shown to greatly boost angiogenesis and decrease vascular permeability, as well as improve neuroplasticity and neurological recovery after ischemic stroke. We describe current breakthroughs in the development of nanoparticle-based treatments for better angiogenesis therapy for ischemic stroke employing polymeric nanoparticles, liposomes, inorganic nanoparticles, and biomimetic nanoparticles in this study. We outline new nanoparticles in detail, review the hurdles and strategies for conveying nanoparticle to lesions, and demonstrate the most recent advances in nanoparticle in angiogenesis for stroke treatment.
Collapse
Affiliation(s)
- Guangtian Wang
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhihui Li
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Gongchen Wang
- Department of Vascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Qixu Sun
- Department of Gastroenterology, Penglai People’s Hospital, Yantai, Shandong, 265600, People’s Republic of China
| | - Peng Lin
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Qian Wang
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Huishu Zhang
- Teaching Center of Biotechnology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Yanyan Wang
- Teaching Center of Morphology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Tongshuai Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Feiyun Cui
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhaohua Zhong
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| |
Collapse
|
19
|
Li H, Shang Y, Zeng J, Matsusaki M. Technology for the formation of engineered microvascular network models and their biomedical applications. NANO CONVERGENCE 2024; 11:10. [PMID: 38430377 PMCID: PMC10908775 DOI: 10.1186/s40580-024-00416-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/15/2024] [Indexed: 03/03/2024]
Abstract
Tissue engineering and regenerative medicine have made great progress in recent decades, as the fields of bioengineering, materials science, and stem cell biology have converged, allowing tissue engineers to replicate the structure and function of various levels of the vascular tree. Nonetheless, the lack of a fully functional vascular system to efficiently supply oxygen and nutrients has hindered the clinical application of bioengineered tissues for transplantation. To investigate vascular biology, drug transport, disease progression, and vascularization of engineered tissues for regenerative medicine, we have analyzed different approaches for designing microvascular networks to create models. This review discusses recent advances in the field of microvascular tissue engineering, explores potential future challenges, and offers methodological recommendations.
Collapse
Affiliation(s)
- He Li
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yucheng Shang
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Jinfeng Zeng
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
20
|
Bao H, Zhang Y, Xin H, Gao Y, Hou Y, Yue G, Wang N, Wang Y, Li C, Liu F, Zhao Y, Kong L. The Construction of Three-Layered Biomimetic Arterial Graft Balances Biomechanics and Biocompatibility for Dynamic Biological Reconstruction. ACS OMEGA 2024; 9:7609-7620. [PMID: 38405546 PMCID: PMC10882685 DOI: 10.1021/acsomega.3c06628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 12/24/2023] [Accepted: 01/15/2024] [Indexed: 02/27/2024]
Abstract
The process of reconstructing an arterial graft is a complex and dynamic process that is subject to the influence of various mechanical factors, including tissue regeneration and blood pressure. The attainment of favorable remodeling outcomes is contingent upon the biocompatibility and biomechanical properties of the arterial graft. A promising strategy involves the emulation of the three-layer structure of the native artery, wherein the inner layer is composed of polycaprolactone (PCL) fibers aligned with blood flow, exhibiting excellent biocompatibility that fosters endothelial cell growth and effectively prevents platelet adhesion. The middle layer, consisting of PCL and polyurethane (PU), offers mechanical support and stability by forming a contractile smooth muscle ring and antiexpansion PU network. The outer layer, composed of PCL fibers with an irregular arrangement, promotes the growth of nerves and pericytes for long-term vascular function. Prioritizing the reconstruction of the inner and outer layers establishes a stable environment for intermediate smooth muscle growth. Our three-layer arterial graft is designed to provide the blood vessel with mechanical support and stability through nondegradable PU, while the incorporation of degradable PCL generates potential spaces for tissue ingrowth, thereby transforming our graft into a living implant.
Collapse
Affiliation(s)
- Han Bao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Yanyuan Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - He Xin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Ye Gao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Yan Hou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Guichu Yue
- Key Laboratory of Bio-inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, China
| | - Nü Wang
- Key Laboratory of Bio-inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, China
| | - Yaqiong Wang
- Key Laboratory of Bio-inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, China
| | - Chun Li
- Shandong Nafeibo Technology Development Co., Ltd, Yantai 264000, China
| | - Fuwei Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Yong Zhao
- Key Laboratory of Bio-inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, China
| | - Liang Kong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
21
|
Browne S, Petit N, Quondamatteo F. Functionalised biomaterials as synthetic extracellular matrices to promote vascularisation and healing of diabetic wounds. Cell Tissue Res 2024; 395:133-145. [PMID: 38051351 DOI: 10.1007/s00441-023-03849-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 11/24/2023] [Indexed: 12/07/2023]
Abstract
Diabetic foot ulcers (DFU) are a type of chronic wound that constitute one of the most serious and debilitating complications associated with diabetes. The lack of clinically efficacious treatments to treat these recalcitrant wounds can lead to amputations for those worst affected. Biomaterial-based approaches offer great hope in this regard, as they provide a template for cell infiltration and tissue repair. However, there is an additional need to treat the underlying pathophysiology of DFUs, in particular insufficient vascularization of the wound which significantly hampers healing. Thus, the addition of pro-angiogenic moieties to biomaterials is a promising strategy to promote the healing of DFUs and other chronic wounds. In this review, we discuss the potential of biomaterials as treatments for DFU and the approaches that can be taken to functionalise these biomaterials such that they promote vascularisation and wound healing in pre-clinical models.
Collapse
Affiliation(s)
- Shane Browne
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Dublin, Ireland.
- CÚRAM, Centre for Research in Medical Devices, University of Galway, H91 W2TY, Galway, Ireland.
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland.
| | - Noémie Petit
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Dublin, Ireland
| | - Fabio Quondamatteo
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Dublin, Ireland.
| |
Collapse
|
22
|
Shan BH, Wu FG. Hydrogel-Based Growth Factor Delivery Platforms: Strategies and Recent Advances. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2210707. [PMID: 37009859 DOI: 10.1002/adma.202210707] [Citation(s) in RCA: 79] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/25/2023] [Indexed: 06/19/2023]
Abstract
Growth factors play a crucial role in regulating a broad variety of biological processes and are regarded as powerful therapeutic agents in tissue engineering and regenerative medicine in the past decades. However, their application is limited by their short half-lives and potential side effects in physiological environments. Hydrogels are identified as having the promising potential to prolong the half-lives of growth factors and mitigate their adverse effects by restricting them within the matrix to reduce their rapid proteolysis, burst release, and unwanted diffusion. This review discusses recent progress in the development of growth factor-containing hydrogels for various biomedical applications, including wound healing, brain tissue repair, cartilage and bone regeneration, and spinal cord injury repair. In addition, the review introduces strategies for optimizing growth factor release including affinity-based delivery, carrier-assisted delivery, stimuli-responsive delivery, spatial structure-based delivery, and cellular system-based delivery. Finally, the review presents current limitations and future research directions for growth factor-delivering hydrogels.
Collapse
Affiliation(s)
- Bai-Hui Shan
- State Key Laboratory of Digital Medical Engineering Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Fu-Gen Wu
- State Key Laboratory of Digital Medical Engineering Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| |
Collapse
|
23
|
Xu F, Sun W, Ma W, Wang W, Kong D, Chan YK, Ma Q. All-aqueous microfluidic printing of multifunctional bioactive microfibers promote whole-stage wound healing. Colloids Surf B Biointerfaces 2024; 234:113720. [PMID: 38157763 DOI: 10.1016/j.colsurfb.2023.113720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/22/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
Wound healing involves multi-stages of physiological responses, including hemostasis, inflammation, cell proliferation, and tissue remodeling. Satisfying all demands throughout different stages remains a rarely addressed challenge. Here we introduce an innovative all-aqueous microfluidic printing technique for fabricating multifunctional bioactive microfibers, effectively contributing to all four phases of the healing process. The distinctive feature of the developed microfibers lies in their capacity to be printed in a free-form manner in the aqueous-two phase system (ATPS). This is achieved through interfacial coacervation between alkyl-chitosan and alginate, with enhanced structural integrity facilitated by simultaneous crosslinking with calcium ions and alginate. The all-aqueous printed microfibers exhibit exceptional performance in terms of cell recruitment, blood cell coagulation, and hemostasis. The inclusion of a dodecyl carbon chain and amino groups in alkyl-chitosan imparts remarkable antimicrobial properties by anchoring to bacteria, complemented by potent antibacterial effects of encapsulated silver nanoparticles. Moreover, microfibers can load bioactive drugs like epidermal growth factor (EGF), preserving their activity and enhancing therapeutic effects during cell proliferation and tissue remodeling. With these sequential functions to guide the whole-stage wound healing, this work offers a versatile and robust paradigm for comprehensive wound treatment, holding great potential for optimal healing outcomes.
Collapse
Affiliation(s)
- Fenglan Xu
- Department of Clinical Pharmacy, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, China
| | - Wentao Sun
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China
| | - Wenyuan Ma
- School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Weijiang Wang
- School of Pharmacy, Qingdao University, Qingdao 266071, China; The Shandong Consortium in the Yellow River Basin for Prevention, Treatment and Drug Development for Primary Diseases Related to Alcoholism, Qingdao University, Qingdao 266071, China
| | - Dejuan Kong
- Tongliao market detection and Testing Center, Tongliao 028000, China
| | - Yau Kei Chan
- Department of Ophthalmology, The University of Hong Kong, 999077 the Hong Kong Special Administrative Region of China
| | - Qingming Ma
- School of Pharmacy, Qingdao University, Qingdao 266071, China; The Shandong Consortium in the Yellow River Basin for Prevention, Treatment and Drug Development for Primary Diseases Related to Alcoholism, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
24
|
Słyk Ż, Wrzesień R, Barszcz S, Gawrychowski K, Małecki M. Adeno-associated virus vector hydrogel formulations for brain cancer gene therapy applications. Biomed Pharmacother 2024; 170:116061. [PMID: 38154269 DOI: 10.1016/j.biopha.2023.116061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 12/30/2023] Open
Abstract
Gelatin-based formulations are utilized in neurosurgical procedures, with Medisponge® serving as an illustration of a secure and biocompatible hemostatic formulation. Noteworthy are combined hemostatic products that integrate pharmacological agents with gelatin. Gelatin matrices, which host biologically active substances, provide a platform for a variety of molecules. Biopolymers function as carriers for chemicals and genes, a facet particularly pertinent in brain cancer therapy, as gene therapy complement conventional approaches. The registration of Zolgensma underscores the efficacy of rAAV vectors in therapeutic gene delivery to the CNS. rAAVs, renowned for their safety, stability, and neuron-targeting capabilities, predominate in CNS gene therapy studies. The effectiveness of rAAV vector therapy varies based on the serotype and administration route. Local gene therapy employing hydrogel (e.g., post-tumor resection) enables the circumvention of the blood-brain barrier and restricts formulation diffusion. This study formulates gelatin rAAV gene formulations and evaluates vector transduction potential. Transduction efficiency was assessed using ex vivo mouse brains and in vitro cancer cell lines. In vitro, the transduction of rAAV vectors in gelatin matrices was quantified through qPCR, measuring the itr and Gfp expression. rAAVDJ and rAAV2 demonstrated superior transduction in ex vivo and in vitro models. Among the cell lines tested (Hs683, B16-F10, NIH:OVCAR-3), gelatin matrix F1 exhibited selective transduction, particularly with Hs683 human glioma cells, surpassing the performance Medisponge®. This research highlights the exploration of local brain cancer therapy, emphasizing the potential of gelatin as an rAAV vector carrier for gene therapy. The functional transduction activity of gelatin rAAV formulations is demonstrated.
Collapse
Affiliation(s)
- Żaneta Słyk
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland; Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland.
| | - Robert Wrzesień
- Central Laboratory of Experimental Animals, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Sławomir Barszcz
- Department of Neurosurgery, Children's Clinical Hospital, University Clinical Centre of the Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof Gawrychowski
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Maciej Małecki
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland; Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
25
|
Margolis EA, Friend NE, Rolle MW, Alsberg E, Putnam AJ. Manufacturing the multiscale vascular hierarchy: progress toward solving the grand challenge of tissue engineering. Trends Biotechnol 2023; 41:1400-1416. [PMID: 37169690 PMCID: PMC10593098 DOI: 10.1016/j.tibtech.2023.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/05/2023] [Accepted: 04/14/2023] [Indexed: 05/13/2023]
Abstract
In human vascular anatomy, blood flows from the heart to organs and tissues through a hierarchical vascular tree, comprising large arteries that branch into arterioles and further into capillaries, where gas and nutrient exchange occur. Engineering a complete, integrated vascular hierarchy with vessels large enough to suture, strong enough to withstand hemodynamic forces, and a branching structure to permit immediate perfusion of a fluidic circuit across scales would be transformative for regenerative medicine (RM), enabling the translation of engineered tissues of clinically relevant size, and perhaps whole organs. How close are we to solving this biological plumbing problem? In this review, we highlight advances in engineered vasculature at individual scales and focus on recent strategies to integrate across scales.
Collapse
Affiliation(s)
- Emily A Margolis
- University of Michigan, Department of Biomedical Engineering, Ann Arbor, MI, USA
| | - Nicole E Friend
- University of Michigan, Department of Biomedical Engineering, Ann Arbor, MI, USA
| | - Marsha W Rolle
- Worcester Polytechnic Institute, Department of Biomedical Engineering, Worcester, MA, USA
| | - Eben Alsberg
- University of Illinois at Chicago, Department of Biomedical Engineering, Chicago, IL, USA
| | - Andrew J Putnam
- University of Michigan, Department of Biomedical Engineering, Ann Arbor, MI, USA.
| |
Collapse
|
26
|
Yousafzai MS, Hammer JA. Using Biosensors to Study Organoids, Spheroids and Organs-on-a-Chip: A Mechanobiology Perspective. BIOSENSORS 2023; 13:905. [PMID: 37887098 PMCID: PMC10605946 DOI: 10.3390/bios13100905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/13/2023] [Accepted: 09/19/2023] [Indexed: 10/28/2023]
Abstract
The increasing popularity of 3D cell culture models is being driven by the demand for more in vivo-like conditions with which to study the biochemistry and biomechanics of numerous biological processes in health and disease. Spheroids and organoids are 3D culture platforms that self-assemble and regenerate from stem cells, tissue progenitor cells or cell lines, and that show great potential for studying tissue development and regeneration. Organ-on-a-chip approaches can be used to achieve spatiotemporal control over the biochemical and biomechanical signals that promote tissue growth and differentiation. These 3D model systems can be engineered to serve as disease models and used for drug screens. While culture methods have been developed to support these 3D structures, challenges remain to completely recapitulate the cell-cell and cell-matrix biomechanical interactions occurring in vivo. Understanding how forces influence the functions of cells in these 3D systems will require precise tools to measure such forces, as well as a better understanding of the mechanobiology of cell-cell and cell-matrix interactions. Biosensors will prove powerful for measuring forces in both of these contexts, thereby leading to a better understanding of how mechanical forces influence biological systems at the cellular and tissue levels. Here, we discussed how biosensors and mechanobiological research can be coupled to develop accurate, physiologically relevant 3D tissue models to study tissue development, function, malfunction in disease, and avenues for disease intervention.
Collapse
Affiliation(s)
- Muhammad Sulaiman Yousafzai
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John A. Hammer
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
27
|
Nicosia A, Salamone M, Costa S, Ragusa MA, Ghersi G. Mimicking Molecular Pathways in the Design of Smart Hydrogels for the Design of Vascularized Engineered Tissues. Int J Mol Sci 2023; 24:12314. [PMID: 37569691 PMCID: PMC10418696 DOI: 10.3390/ijms241512314] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/21/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Biomaterials are pivotal in supporting and guiding vascularization for therapeutic applications. To design effective, bioactive biomaterials, understanding the cellular and molecular processes involved in angiogenesis and vasculogenesis is crucial. Biomaterial platforms can replicate the interactions between cells, the ECM, and the signaling molecules that trigger blood vessel formation. Hydrogels, with their soft and hydrated properties resembling natural tissues, are widely utilized; particularly synthetic hydrogels, known for their bio-inertness and precise control over cell-material interactions, are utilized. Naturally derived and synthetic hydrogel bases are tailored with specific mechanical properties, controlled for biodegradation, and enhanced for cell adhesion, appropriate biochemical signaling, and architectural features that facilitate the assembly and tubulogenesis of vascular cells. This comprehensive review showcases the latest advancements in hydrogel materials and innovative design modifications aimed at effectively guiding and supporting vascularization processes. Furthermore, by leveraging this knowledge, researchers can advance biomaterial design, which will enable precise support and guidance of vascularization processes and ultimately enhance tissue functionality and therapeutic outcomes.
Collapse
Affiliation(s)
- Aldo Nicosia
- Institute for Biomedical Research and Innovation-National Research Council (IRIB-CNR), Via Ugo la Malfa 153, 90146 Palermo, Italy;
| | - Monica Salamone
- Institute for Biomedical Research and Innovation-National Research Council (IRIB-CNR), Via Ugo la Malfa 153, 90146 Palermo, Italy;
| | - Salvatore Costa
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (S.C.); (M.A.R.); (G.G.)
| | - Maria Antonietta Ragusa
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (S.C.); (M.A.R.); (G.G.)
| | - Giulio Ghersi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (S.C.); (M.A.R.); (G.G.)
| |
Collapse
|
28
|
Xu KL, Mauck RL, Burdick JA. Modeling development using hydrogels. Development 2023; 150:dev201527. [PMID: 37387575 PMCID: PMC10323241 DOI: 10.1242/dev.201527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
The development of multicellular complex organisms relies on coordinated signaling from the microenvironment, including both biochemical and mechanical interactions. To better understand developmental biology, increasingly sophisticated in vitro systems are needed to mimic these complex extracellular features. In this Primer, we explore how engineered hydrogels can serve as in vitro culture platforms to present such signals in a controlled manner and include examples of how they have been used to advance our understanding of developmental biology.
Collapse
Affiliation(s)
- Karen L. Xu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert L. Mauck
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, USA
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| |
Collapse
|
29
|
Kuwar R, Wen X, Zhang N, Sun D. Integrin binding peptides facilitate growth and interconnected vascular-like network formation of rat primary cortical vascular endothelial cells in vitro. Neural Regen Res 2023; 18:1052-1056. [PMID: 36254992 PMCID: PMC9827785 DOI: 10.4103/1673-5374.355760] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/28/2022] [Accepted: 07/11/2022] [Indexed: 11/07/2022] Open
Abstract
Neovascularization and angiogenesis in the brain are important physiological processes for normal brain development and repair/regeneration following insults. Integrins are cell surface adhesion receptors mediating important function of cells such as survival, growth and development during tissue organization, differentiation and organogenesis. In this study, we used an integrin-binding array platform to identify the important types of integrins and their binding peptides that facilitate adhesion, growth, development, and vascular-like network formation of rat primary brain microvascular endothelial cells. Brain microvascular endothelial cells were isolated from rat brain on post-natal day 7. Cells were cultured in a custom-designed integrin array system containing short synthetic peptides binding to 16 types of integrins commonly expressed on cells in vertebrates. After 7 days of culture, the brain microvascular endothelial cells were processed for immunostaining with markers for endothelial cells including von Willibrand factor and platelet endothelial cell adhesion molecule. 5-Bromo-2'-dexoyuridine was added to the culture at 48 hours prior to fixation to assess cell proliferation. Among 16 integrins tested, we found that α5β1, αvβ5 and αvβ8 greatly promoted proliferation of endothelial cells in culture. To investigate the effect of integrin-binding peptides in promoting neovascularization and angiogenesis, the binding peptides to the above three types of integrins were immobilized to our custom-designed hydrogel in three-dimensional (3D) culture of brain microvascular endothelial cells with the addition of vascular endothelial growth factor. Following a 7-day 3D culture, the culture was fixed and processed for double labeling of phalloidin with von Willibrand factor or platelet endothelial cell adhesion molecule and assessed under confocal microscopy. In the 3D culture in hydrogels conjugated with the integrin-binding peptide, brain microvascular endothelial cells formed interconnected vascular-like network with clearly discernable lumens, which is reminiscent of brain microvascular network in vivo. With the novel integrin-binding array system, we identified the specific types of integrins on brain microvascular endothelial cells that mediate cell adhesion and growth followed by functionalizing a 3D hydrogel culture system using the binding peptides that specifically bind to the identified integrins, leading to robust growth and lumenized microvascular-like network formation of brain microvascular endothelial cells in 3D culture. This technology can be used for in vitro and in vivo vascularization of transplants or brain lesions to promote brain tissue regeneration following neurological insults.
Collapse
Affiliation(s)
- Ram Kuwar
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| | - Xuejun Wen
- Department of Chemical and Life Science Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Ning Zhang
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Dong Sun
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
30
|
Cell–scaffold interactions in tissue engineering for oral and craniofacial reconstruction. Bioact Mater 2023; 23:16-44. [DOI: 10.1016/j.bioactmat.2022.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/22/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022] Open
|
31
|
Namjoo AR, Abrbekoh FN, Saghati S, Amini H, Saadatlou MAE, Rahbarghazi R. Tissue engineering modalities in skeletal muscles: focus on angiogenesis and immunomodulation properties. Stem Cell Res Ther 2023; 14:90. [PMID: 37061717 PMCID: PMC10105969 DOI: 10.1186/s13287-023-03310-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/28/2023] [Indexed: 04/17/2023] Open
Abstract
Muscular diseases and injuries are challenging issues in human medicine, resulting in physical disability. The advent of tissue engineering approaches has paved the way for the restoration and regeneration of injured muscle tissues along with available conventional therapies. Despite recent advances in the fabrication, synthesis, and application of hydrogels in terms of muscle tissue, there is a long way to find appropriate hydrogel types in patients with congenital and/or acquired musculoskeletal injuries. Regarding specific muscular tissue microenvironments, the applied hydrogels should provide a suitable platform for the activation of endogenous reparative mechanisms and concurrently deliver transplanting cells and therapeutics into the injured sites. Here, we aimed to highlight recent advances in muscle tissue engineering with a focus on recent strategies related to the regulation of vascularization and immune system response at the site of injury.
Collapse
Affiliation(s)
- Atieh Rezaei Namjoo
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Sepideh Saghati
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hassan Amini
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- General and Vascular Surgery Department, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
32
|
O'Connor CE, Neufeld A, Fortin CL, Johansson F, Mene J, Saxton SH, Simmonds SP, Kopyeva I, Gregorio NE, DeForest CA, Witten DM, Stevens KR. Highly Parallel Tissue Grafting for Combinatorial In Vivo Screening. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.16.533029. [PMID: 36993278 PMCID: PMC10055160 DOI: 10.1101/2023.03.16.533029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Material- and cell-based technologies such as engineered tissues hold great promise as human therapies. Yet, the development of many of these technologies becomes stalled at the stage of pre-clinical animal studies due to the tedious and low-throughput nature of in vivo implantation experiments. We introduce a 'plug and play' in vivo screening array platform called Highly Parallel Tissue Grafting (HPTG). HPTG enables parallelized in vivo screening of 43 three-dimensional microtissues within a single 3D printed device. Using HPTG, we screen microtissue formations with varying cellular and material components and identify formulations that support vascular self-assembly, integration and tissue function. Our studies highlight the importance of combinatorial studies that vary cellular and material formulation variables concomitantly, by revealing that inclusion of stromal cells can "rescue" vascular self-assembly in manner that is material-dependent. HPTG provides a route for accelerating pre-clinical progress for diverse medical applications including tissue therapy, cancer biomedicine, and regenerative medicine.
Collapse
|
33
|
Liu X, Gaihre B, Li L, Rezaei A, Tilton M, Elder BD, Lu L. Bioorthogonal "Click Chemistry" Bone Cement with Bioinspired Natural Mimicking Microstructures for Bone Repair. ACS Biomater Sci Eng 2023; 9:1585-1597. [PMID: 36854041 PMCID: PMC10123962 DOI: 10.1021/acsbiomaterials.2c01482] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Current bone cement systems often demand free radical or metal-related initiators and/or catalysts for the crosslinking process, which may cause serious toxicity to the human body. In addition, the resultant dense scaffolds may have a prolonged degradation time and are difficult for cells to infiltrate and form new tissue. In this study, we developed a porous "click" organic-inorganic nanohybrid (PO-click-ON) cement that crosslinks via metal-free biorthogonal click chemistry and forms porous structures mimicking the native bone tissue via particulate leaching. Strain-promoted click reaction enables fast and efficient crosslinking of polymer chains with the exclusion of any toxic initiator or catalyst. The resulting PO-click-ON implants supported exceptional in vitro stem cell adhesion and osteogenic differentiation with a large portion of stem cells infiltrated deep into the scaffolds. In vivo study using a rat cranial defect model demonstrated that the PO-click-ON system achieved outstanding cell adsorption, neovascularization, and bone formation. The porous click cement developed in this study serves as a promising platform with multifunctionality for bone and other tissue engineering applications.
Collapse
Affiliation(s)
- Xifeng Liu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Bipin Gaihre
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Linli Li
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Asghar Rezaei
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Maryam Tilton
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Benjamin D Elder
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Lichun Lu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
| |
Collapse
|
34
|
Blatchley MR, Anseth KS. Middle-out methods for spatiotemporal tissue engineering of organoids. NATURE REVIEWS BIOENGINEERING 2023; 1:329-345. [PMID: 37168734 PMCID: PMC10010248 DOI: 10.1038/s44222-023-00039-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 02/03/2023] [Indexed: 05/13/2023]
Abstract
Organoids recapitulate many aspects of the complex three-dimensional (3D) organization found within native tissues and even display tissue and organ-level functionality. Traditional approaches to organoid culture have largely employed a top-down tissue engineering strategy, whereby cells are encapsulated in a 3D matrix, such as Matrigel, alongside well-defined biochemical cues that direct morphogenesis. However, the lack of spatiotemporal control over niche properties renders cellular processes largely stochastic. Therefore, bottom-up tissue engineering approaches have evolved to address some of these limitations and focus on strategies to assemble tissue building blocks with defined multi-scale spatial organization. However, bottom-up design reduces the capacity for self-organization that underpins organoid morphogenesis. Here, we introduce an emerging framework, which we term middle-out strategies, that relies on existing design principles and combines top-down design of defined synthetic matrices that support proliferation and self-organization with bottom-up modular engineered intervention to limit the degrees of freedom in the dynamic process of organoid morphogenesis. We posit that this strategy will provide key advances to guide the growth of organoids with precise geometries, structures and function, thereby facilitating an unprecedented level of biomimicry to accelerate the utility of organoids to more translationally relevant applications.
Collapse
Affiliation(s)
- Michael R. Blatchley
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO USA
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO USA
| |
Collapse
|
35
|
Hao D, Liu R, Fernandez TG, Pivetti C, Jackson JE, Kulubya ES, Jiang HJ, Ju HY, Liu WL, Panitch A, Lam KS, Leach JK, Farmer DL, Wang A. A bioactive material with dual integrin-targeting ligands regulates specific endogenous cell adhesion and promotes vascularized bone regeneration in adult and fetal bone defects. Bioact Mater 2023; 20:179-193. [PMID: 35663336 PMCID: PMC9160290 DOI: 10.1016/j.bioactmat.2022.05.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 12/18/2022] Open
Abstract
Significant progress has been made in designing bone materials capable of directing endogenous cells to promote vascularized bone regeneration. However, current strategies lack regulation of the specific endogenous cell populations for vascularized bone regeneration, thus leading to adverse tissue formation and decreased regenerative efficiency. Here, we engineered a biomaterial to regulate endogenous cell adhesion and promote vascularized bone regeneration. The biomaterial works by presenting two synthetic ligands, LLP2A and LXW7, explicitly targeting integrins α4β1 and αvβ3, respectively, expressed on the surfaces of the cells related to bone formation and vascularization, such as mesenchymal stem cells (MSCs), osteoblasts, endothelial progenitor cells (EPCs), and endothelial cells (ECs). In vitro, the LLP2A/LXW7 modified biomaterial improved the adhesion of MSCs, osteoblasts, EPCs, and ECs via integrin α4β1 and αvβ3, respectively. In an adult rat calvarial bone defect model, the LLP2A/LXW7 modified biomaterial enhanced bone formation and vascularization by synergistically regulating endogenous cells with osteogenic and angiogenic potentials, such as DLX5+ cells, osteocalcin+ cells, CD34+/CD45- cells and CD31+ cells. In a fetal sheep spinal bone defect model, the LLP2A/LXW7 modified biomaterial augmented bone formation and vascularization without any adverse effects. This innovative biomaterial offers an off-the-shelf, easy-to-use, and biologically safe product suitable for vascularized bone regeneration in both fetal and adult disease environments. Two integrin-binding ligands for constructing vascularized bone biomaterial. Extracellular matrix (ECM)-mimicking collagen-based biomaterial with specific integrin binding sites for cell adhesion. Biomaterial regulates adhesion of endogenous stem cells with osteogenic and angiogenic potentials. Biomaterial promotes vascularized bone formation in adult and fetal bone defects without safety issues. An easy-to-make and off-the-shelf biomaterial for treatment of clinical bone diseases.
Collapse
Affiliation(s)
- Dake Hao
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Tomas Gonzalez Fernandez
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, United States
| | - Christopher Pivetti
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
| | - Jordan Elizabeth Jackson
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Edwin Samuel Kulubya
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Hong-Jiang Jiang
- Wendeng Orthopaedic Hospital, No. 1 Fengshan Road, Wendeng, 264400, Shandong, China
| | - Hai-Yang Ju
- Wendeng Orthopaedic Hospital, No. 1 Fengshan Road, Wendeng, 264400, Shandong, China
| | - Wen-Liang Liu
- Wendeng Orthopaedic Hospital, No. 1 Fengshan Road, Wendeng, 264400, Shandong, China
| | - Alyssa Panitch
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, United States
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - J. Kent Leach
- Department of Orthopaedic Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Diana L. Farmer
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, United States
- Corresponding author. Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California, Davis, 4625 2nd Ave., Research II, Suite 3005, Sacramento, CA, 95817, USA.
| |
Collapse
|
36
|
Yu Q, Jian Z, Yang D, Zhu T. Perspective insights into hydrogels and nanomaterials for ischemic stroke. Front Cell Neurosci 2023; 16:1058753. [PMID: 36761147 PMCID: PMC9902513 DOI: 10.3389/fncel.2022.1058753] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/30/2022] [Indexed: 01/26/2023] Open
Abstract
Ischemic stroke (IS) is a neurological disorder prevalent worldwide with a high disability and mortality rate. In the clinic setting, tissue plasminogen activator (tPA) and thrombectomy could restore blood flow of the occlusion region and improve the outcomes of IS patients; however, these therapies are restricted by a narrow time window. Although several preclinical trials have revealed the molecular and cellular mechanisms underlying infarct lesions, the translatability of most findings is unsatisfactory, which contributes to the emergence of new biomaterials, such as hydrogels and nanomaterials, for the treatment of IS. Biomaterials function as structural scaffolds or are combined with other compounds to release therapeutic drugs. Biomaterial-mediated drug delivery approaches could optimize the therapeutic effects based on their brain-targeting property, biocompatibility, and functionality. This review summarizes the advances in biomaterials in the last several years, aiming to discuss the therapeutic potential of new biomaterials from the bench to bedside. The promising prospects of new biomaterials indicate the possibility of an organic combination between materialogy and medicine, which is a novel field under exploration.
Collapse
Affiliation(s)
- Qingbo Yu
- Laboratory of Anesthesia & Critical Care Medicine, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, China,Department of Anesthesiology, North Sichuan Medical College, Nanchong, China
| | - Zhang Jian
- Sichuan Provincial Maternity and Child Health Care Hospital, Women’s and Children’s Hospital Affiliated of Chengdu Medical College, Chengdu, China
| | - Dan Yang
- Department of Anesthesiology, North Sichuan Medical College, Nanchong, China
| | - Tao Zhu
- Laboratory of Anesthesia & Critical Care Medicine, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, China,*Correspondence: Tao Zhu,
| |
Collapse
|
37
|
Shakeel A, Corridon PR. Mitigating challenges and expanding the future of vascular tissue engineering-are we there yet? Front Physiol 2023; 13:1079421. [PMID: 36685187 PMCID: PMC9846051 DOI: 10.3389/fphys.2022.1079421] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023] Open
Affiliation(s)
- Adeeba Shakeel
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Peter R. Corridon
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
- Biomedical Engineering, Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
38
|
Single and multi-dose drug loaded electrospun fiber mats for wound healing applications. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
39
|
Belousov A, Patlay A, Silant’ev V, Kovalev VV, Kumeiko V. Preparation of Hydrogels Based on Modified Pectins by Tuning Their Properties for Anti-Glioma Therapy. Int J Mol Sci 2022; 24:ijms24010630. [PMID: 36614073 PMCID: PMC9820215 DOI: 10.3390/ijms24010630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
The extracellular matrix (ECM) of the central nervous system (CNS), characterized by low stiffness and predominance of carbohydrates on protein components, mediates limited cell proliferation and migration. Pectins are polysaccharides derived from plants and could be very promising for a tunable hydrogel design that mimics the neural ECM. Aiming to regulate gel structure and viscoelastic properties, we elaborated 10 variants of pectin-based hydrogels via tuning the concentration of the polymer and the number of free carboxyl groups expressed in the degree of esterification (DE). Viscoelastic properties of hydrogels varied in the range of 3 to 900 Pa for G' and were chosen as the first criteria for the selection of variants suitable for CNS remodeling. For extended reciprocal characterization, two pairs of hydrogels were taken to test pectins with opposite DEs close to 0% and 50%, respectively, but with a similar rheology exceeding 100 Pa (G'), which was achieved by adjusting the concentration of pectin. Hydrogel swelling properties and in vitro stability, together with structure characterization using SEM and FTIR spectroscopy, displayed some differences that may sense for biomedical application. Bioassays on C6 and U87MG glioblastoma cultures testified the potential prospects of the anti-glioma activity of hydrogels developed by decreasing cell proliferation and modulating migration but supporting the high viability of neural cells.
Collapse
Affiliation(s)
- Andrei Belousov
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok 690922, Russia
| | - Aleksandra Patlay
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok 690922, Russia
| | - Vladimir Silant’ev
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok 690922, Russia
- Laboratory of Electrochemical Processes, Institute of Chemistry, FEB RAS, Vladivostok 690022, Russia
| | - Valeri V. Kovalev
- A.V. Zhirmunsky National Scientific Center of Marine Biology, FEB RAS, Vladivostok 690041, Russia
| | - Vadim Kumeiko
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok 690922, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, FEB RAS, Vladivostok 690041, Russia
- Correspondence:
| |
Collapse
|
40
|
Moeinvaziri F, Zarkesh I, Pooyan P, Nunez DA, Baharvand H. Inner ear organoids: progress and outlook, with a focus on the vascularization. FEBS J 2022; 289:7368-7384. [PMID: 34331740 DOI: 10.1111/febs.16146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 06/11/2021] [Accepted: 07/30/2021] [Indexed: 01/13/2023]
Abstract
The inner ear is a complex organ that encodes sound, motion, and orientation in space. Given the complexity of the inner ear, it is not surprising that treatments are relatively limited despite the fact that, in 2015, hearing loss was the fourth leading cause of years lived with disability worldwide. Inner ear organoid models are a promising tool to advance the study of multiple aspects of the inner ear to aid the development of new treatments and validate drug-based therapies. The blood supply of the inner ear plays a pivotal role in growth, maturation, and survival of inner ear tissues and their physiological functions. This vasculature cannot be ignored in order to achieve a truly in vivo-like model that mimics the microenvironment and niches of organ development. However, this aspect of organoid development has remained largely absent in the generation of inner ear organoids. The current review focuses on three-dimensional inner ear organoid and how recent technical progress in generating in vitro vasculature can enhance the next generation of these models.
Collapse
Affiliation(s)
- Farideh Moeinvaziri
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ibrahim Zarkesh
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Paria Pooyan
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Desmond A Nunez
- Division of Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, Canada
| | - Hossein Baharvand
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
41
|
Liu C, Yang ZX, Zhou SQ, Ding D, Hu YT, Yang HN, Han D, Hu SQ, Zong XM. Overexpression of vascular endothelial growth factor enhances the neuroprotective effects of bone marrow mesenchymal stem cell transplantation in ischemic stroke. Neural Regen Res 2022; 18:1286-1292. [PMID: 36453413 PMCID: PMC9838145 DOI: 10.4103/1673-5374.358609] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although bone marrow mesenchymal stem cells (BMSCs) might have therapeutic potency in ischemic stroke, the benefits are limited. The current study investigated the effects of BMSCs engineered to overexpress vascular endothelial growth factor (VEGF) on behavioral defects in a rat model of transient cerebral ischemia, which was induced by middle cerebral artery occlusion. VEGF-BMSCs or control grafts were injected into the left striatum of the infarcted hemisphere 24 hours after stroke. We found that compared with the stroke-only group and the vehicle- and BMSCs-control groups, the VEGF-BMSCs treated animals displayed the largest benefits, as evidenced by attenuated behavioral defects and smaller infarct volume 7 days after stroke. Additionally, VEGF-BMSCs greatly inhibited destruction of the blood-brain barrier, increased the regeneration of blood vessels in the region of ischemic penumbra, and reducedneuronal degeneration surrounding the infarct core. Further mechanistic studies showed that among all transplant groups, VEGF-BMSCs transplantation induced the highest level of brain-derived neurotrophic factor. These results suggest that BMSCs transplantation with vascular endothelial growth factor has the potential to treat ischemic stroke with better results than are currently available.
Collapse
Affiliation(s)
- Cui Liu
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Emergency Department of Affiliated Lianyungang Hospital of Xuzhou Medical University/The First People’s Hospital of Lianyungang, Lianyungang, Lianyungang, Jiangsu Province, China
| | - Zhi-Xiang Yang
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Si-Qi Zhou
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Ding Ding
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yu-Ting Hu
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Hong-Ning Yang
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Laboratory of Emergency Medicine, Second Clinical Medical College of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Dong Han
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Laboratory of Emergency Medicine, Second Clinical Medical College of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Shu-Qun Hu
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Laboratory of Emergency Medicine, Second Clinical Medical College of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Correspondence to: Xue-Mei Zong, ; Shu-Qun Hu, .
| | - Xue-Mei Zong
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Key Laboratory of Brain Diseases Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Correspondence to: Xue-Mei Zong, ; Shu-Qun Hu, .
| |
Collapse
|
42
|
Integrin-specific hydrogels for growth factor-free vasculogenesis. NPJ Regen Med 2022; 7:57. [PMID: 36167724 PMCID: PMC9515164 DOI: 10.1038/s41536-022-00253-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
Integrin-binding biomaterials have been extensively evaluated for their capacity to enable de novo formation of capillary-like structures/vessels, ultimately supporting neovascularization in vivo. Yet, the role of integrins as vascular initiators in engineered materials is still not well understood. Here, we show that αvβ3 integrin-specific 3D matrices were able to retain PECAM1+ cells from the stromal vascular fraction (SVF) of adipose tissue, triggering vasculogenesis in vitro in the absence of extrinsic growth factors. Our results suggest that αvβ3-RGD-driven signaling in the formation of capillary-like structures prevents the activation of the caspase 8 pathway and activates the FAK/paxillin pathway, both responsible for endothelial cells (ECs) survival and migration. We also show that prevascularized αvβ3 integrin-specific constructs inosculate with the host vascular system fostering in vivo neovascularization. Overall, this work demonstrates the ability of the biomaterial to trigger vasculogenesis in an integrin-specific manner, by activating essential pathways for EC survival and migration within a self-regulatory growth factor microenvironment. This strategy represents an improvement to current vascularization routes for Tissue Engineering constructs, potentially enhancing their clinical applicability.
Collapse
|
43
|
Gao Z, Yu Y, Dai K, Zhang T, Ji L, Wang X, Wang J, Liu C. Engineering Neutrophil Immunomodulatory Hydrogels Promoted Angiogenesis. ACS APPLIED MATERIALS & INTERFACES 2022; 14:39746-39758. [PMID: 36006024 DOI: 10.1021/acsami.2c08600] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Timely restoration of blood supply following ischemia is critical to rescue damaged tissue. However, clinical efficacy is hampered by the inflammatory response after ischemia. Whether inflammation fine tunes the angiogenesis and the function of blood vessels via the heterogeneity of neutrophils remain poorly understood. Herein, the objective of this work is to incorporate the growth factors secreted by neutrophils into a porous gelatin methacrylate (GelMA) hydrogel, which subsequently is used as a novel regenerative scaffold with defined architecture for ischemia. We demonstrate that anti-inflammatory neutrophils (N2-polarized neutrophils) play an important role in promoting the migration of human umbilical vein endothelial cells (HUVECs) and formation of capillary-like networks in vitro. More importantly, vascular anastomosis can be achieved by modulating the neutrophils to N2 phenotype. In addition, N2-polarized composite hydrogel scaffolds can regulate inflammation, maintain the survival of exogenous cells, and promote angiogenesis in vivo. Notably, the composite hydrogel scaffolds promote neovascularization during exogenous introduction of endothelial cells by anastomosis. Taken together, this study highlights N2-polarized neutrophils composite hydrogels can achieve vascularization rapidly by regulating inflammation and promoting vascular anastomosis. This work lays the foundation for research into the treatment of ischemia and may inspire further research into novel treatment options.
Collapse
Affiliation(s)
- Zehua Gao
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Yuanman Yu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Kai Dai
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Tingting Zhang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Luli Ji
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Xuanlin Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| |
Collapse
|
44
|
Fowler EW, van Venrooy EJ, Witt RL, Jia X. A TGFβR inhibitor represses keratin-7 expression in 3D cultures of human salivary gland progenitor cells. Sci Rep 2022; 12:15008. [PMID: 36056161 PMCID: PMC9440137 DOI: 10.1038/s41598-022-19253-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/26/2022] [Indexed: 11/09/2022] Open
Abstract
Salivary gland tissue engineering offers an attractive alternative for the treatment of radiation-induced xerostomia. Key to the success of this approach is the maintenance and expansion of secretory acinar cells in vitro. However, recent studies revealed that in vitro culture of primary salivary gland epithelial cells led to undesirable upregulation of the expression of keratin-7 (K7), a marker of ductal phenotype and frequently associated with cellular stress. We have previously shown that hyaluronic acid (HA)-based, RGDSP-decorated hydrogels support the 3D growth and assembly of primary human salivary gland stem/progenitor cells (hS/PCs). Here, we investigate whether the RGDSP culture also promotes K7 expression, and if so, what factors govern the K7 expression. Compared to hS/PCs maintained in blank HA gels, those grown in RGDSP cultures expressed a significantly higher level of K7. In other tissues, various transforming growth factor-β (TGF-β) superfamily members are reported to regulate K7 expression. Similarly, our immunoblot array and ELISA experiments confirmed the increased expression of TGF-β1 and growth/differentiation factor-15 (GDF-15) in RGDSP cultures. However, 2D model studies show that only TGF-β1 is required to induce K7 expression in hS/PCs. Immunocytochemical analysis of the intracellular effectors of TGF-β signaling, SMAD 2/3, further confirmed the elevated TGF-β signaling in RGDSP cultures. To maximize the regenerative potential of h/SPCs, cultures were treated with a pharmacological inhibitor of TGF-β receptor, A83-01. Our results show that A83-01 treatment can repress K7 expression not only in 3D RGDSP cultures but also under 2D conditions with exogenous TGF-β1. Collectively, we provide a link between TGF-β signaling and K7 expression in hS/PC cultures and demonstrate the effectiveness of TGF-β inhibition to repress K7 expression while maintaining the ability of RGDSP-conjugated HA gels to facilitate the rapid development of amylase expressing spheroids. These findings represent an important step towards regenerating salivary function with a tissue-engineered salivary gland.
Collapse
Affiliation(s)
- Eric W Fowler
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, 19716, USA.
| | - Emmett J van Venrooy
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Robert L Witt
- Helen F. Graham Cancer Center and Research Institute, Christiana Care, Newark, DE, 19713, USA
| | - Xinqiao Jia
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, 19716, USA.
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA.
- Department of Biomedical Engineering, University of Delaware, Newark, DE, 19716, USA.
- Delaware Biotechnology Institute, 590 Avenue 1743, Newark, DE, 19713, USA.
| |
Collapse
|
45
|
Sideris E, Kioulaphides S, Wilson K, Yu A, Chen J, Carmichael ST, Segura T. Particle hydrogels decrease cerebral atrophy and attenuate astrocyte and microglia/macrophage reactivity after stroke. ADVANCED THERAPEUTICS 2022; 5:2200048. [PMID: 36589207 PMCID: PMC9797126 DOI: 10.1002/adtp.202200048] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Indexed: 01/05/2023]
Abstract
Increasing numbers of individuals live with stroke related disabilities. Following stroke, highly reactive astrocytes and pro-inflammatory microglia can release cytokines and lead to a cytotoxic environment that causes further brain damage and prevents endogenous repair. Paradoxically, these same cells also activate pro-repair mechanisms that contribute to endogenous repair and brain plasticity. Here, we show that the direct injection of a hyaluronic acid based microporous annealed particle (MAP) hydrogel into the stroke core in mice reduces the percent of highly reactive astrocytes, increases the percent of alternatively activated microglia, decreases cerebral atrophy and preserves NF200 axonal bundles. Further, we show that MAP hydrogel promotes reparative astrocyte infiltration into the lesion, which directly coincides with axonal penetration into the lesion. This work shows that the injection of a porous scaffold into the stroke core can lead to clinically relevant decrease in cerebral atrophy and modulates astrocytes and microglia towards a pro-repair phenotype.
Collapse
Affiliation(s)
- Elias Sideris
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, CA, United States
| | - Sophia Kioulaphides
- Departments of Biomedical Engineering, Neurology, and Dermatology, Duke University, Durham, NC, United States
| | - Katrina Wilson
- Departments of Biomedical Engineering, Neurology, and Dermatology, Duke University, Durham, NC, United States
| | - Aaron Yu
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, CA, United States
| | - Jun Chen
- Departments of Biomedical Engineering, Neurology, and Dermatology, Duke University, Durham, NC, United States
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Tatiana Segura
- Departments of Biomedical Engineering, Neurology, and Dermatology, Duke University, Durham, NC, United States
| |
Collapse
|
46
|
Tissue Engineering Approaches to Uncover Therapeutic Targets for Endothelial Dysfunction in Pathological Microenvironments. Int J Mol Sci 2022; 23:ijms23137416. [PMID: 35806421 PMCID: PMC9266895 DOI: 10.3390/ijms23137416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 02/07/2023] Open
Abstract
Endothelial cell dysfunction plays a central role in many pathologies, rendering it crucial to understand the underlying mechanism for potential therapeutics. Tissue engineering offers opportunities for in vitro studies of endothelial dysfunction in pathological mimicry environments. Here, we begin by analyzing hydrogel biomaterials as a platform for understanding the roles of the extracellular matrix and hypoxia in vascular formation. We next examine how three-dimensional bioprinting has been applied to recapitulate healthy and diseased tissue constructs in a highly controllable and patient-specific manner. Similarly, studies have utilized organs-on-a-chip technology to understand endothelial dysfunction's contribution to pathologies in tissue-specific cellular components under well-controlled physicochemical cues. Finally, we consider studies using the in vitro construction of multicellular blood vessels, termed tissue-engineered blood vessels, and the spontaneous assembly of microvascular networks in organoids to delineate pathological endothelial dysfunction.
Collapse
|
47
|
Ke W, Ma L, Wang B, Song Y, Luo R, Li G, Liao Z, Shi Y, Wang K, Feng X, Li S, Hua W, Yang C. N-cadherin mimetic hydrogel enhances MSC chondrogenesis through cell metabolism. Acta Biomater 2022; 150:83-95. [DOI: 10.1016/j.actbio.2022.07.050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 07/17/2022] [Accepted: 07/26/2022] [Indexed: 02/07/2023]
|
48
|
Wang Y, Keshavarz M, Barhouse P, Smith Q. Strategies for Regenerative Vascular Tissue Engineering. Adv Biol (Weinh) 2022; 7:e2200050. [PMID: 35751461 DOI: 10.1002/adbi.202200050] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/15/2022] [Indexed: 11/11/2022]
Abstract
Vascularization remains one of the key challenges in creating functional tissue-engineered constructs for therapeutic applications. This review aims to provide a developmental lens on the necessity of blood vessels in defining tissue function while exploring stem cells as a suitable source for vascular tissue engineering applications. The intersections of stem cell biology, material science, and engineering are explored as potential solutions for directing vascular assembly.
Collapse
Affiliation(s)
- Yao Wang
- Department of Chemical and Biomolecular Engineering University of California Irvine CA 92697 USA
- Sue & Bill Gross Stem Cell Research Center University of California Irvine CA 92697 USA
| | - Mozhgan Keshavarz
- Department of Chemical and Biomolecular Engineering University of California Irvine CA 92697 USA
- Sue & Bill Gross Stem Cell Research Center University of California Irvine CA 92697 USA
| | - Patrick Barhouse
- Department of Chemical and Biomolecular Engineering University of California Irvine CA 92697 USA
- Sue & Bill Gross Stem Cell Research Center University of California Irvine CA 92697 USA
| | - Quinton Smith
- Department of Chemical and Biomolecular Engineering University of California Irvine CA 92697 USA
- Sue & Bill Gross Stem Cell Research Center University of California Irvine CA 92697 USA
| |
Collapse
|
49
|
Macklin BL, Lin YY, Emmerich K, Wisniewski E, Polster BM, Konstantopoulos K, Mumm JS, Gerecht S. Intrinsic epigenetic control of angiogenesis in induced pluripotent stem cell-derived endothelium regulates vascular regeneration. NPJ Regen Med 2022; 7:28. [PMID: 35551465 PMCID: PMC9098630 DOI: 10.1038/s41536-022-00223-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 04/14/2022] [Indexed: 11/17/2022] Open
Abstract
Human-induced pluripotent stem cell-derived endothelial cells (iECs) provide opportunities to study vascular development and regeneration, develop cardiovascular therapeutics, and engineer model systems for drug screening. The differentiation and characterization of iECs are well established; however, the mechanisms governing their angiogenic phenotype remain unknown. Here, we aimed to determine the angiogenic phenotype of iECs and the regulatory mechanism controlling their regenerative capacity. In a comparative study with HUVECs, we show that iECs increased expression of vascular endothelial growth factor receptor 2 (VEGFR2) mediates their highly angiogenic phenotype via regulation of glycolysis enzymes, filopodia formation, VEGF mediated migration, and robust sprouting. We find that the elevated expression of VEGFR2 is epigenetically regulated via intrinsic acetylation of histone 3 at lysine 27 by histone acetyltransferase P300. Utilizing a zebrafish xenograft model, we demonstrate that the ability of iECs to promote the regeneration of the amputated fin can be modulated by P300 activity. These findings demonstrate how the innate epigenetic status of iECs regulates their phenotype with implications for their therapeutic potential.
Collapse
Affiliation(s)
- Bria L Macklin
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Ying-Yu Lin
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Kevin Emmerich
- Department of Ophthalmology, Wilmer Eye Institute and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Emily Wisniewski
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma, and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Jeff S Mumm
- Department of Ophthalmology, Wilmer Eye Institute and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA. .,Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
50
|
Lu G, Xu Y, Liu Q, Chen M, Sun H, Wang P, Li X, Wang Y, Li X, Hui X, Luo E, Liu J, Jiang Q, Liang J, Fan Y, Sun Y, Zhang X. An instantly fixable and self-adaptive scaffold for skull regeneration by autologous stem cell recruitment and angiogenesis. Nat Commun 2022; 13:2499. [PMID: 35523800 PMCID: PMC9076642 DOI: 10.1038/s41467-022-30243-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 04/20/2022] [Indexed: 02/08/2023] Open
Abstract
Limited stem cells, poor stretchability and mismatched interface fusion have plagued the reconstruction of cranial defects by cell-free scaffolds. Here, we designed an instantly fixable and self-adaptive scaffold by dopamine-modified hyaluronic acid chelating Ca2+ of the microhydroxyapatite surface and bonding type I collagen to highly simulate the natural bony matrix. It presents a good mechanical match and interface integration by appropriate calcium chelation, and responds to external stress by flexible deformation. Meanwhile, the appropriate matrix microenvironment regulates macrophage M2 polarization and recruits endogenous stem cells. This scaffold promotes the proliferation and osteogenic differentiation of BMSCs in vitro, as well as significant ectopic mineralization and angiogenesis. Transcriptome analysis confirmed the upregulation of relevant genes and signalling pathways was associated with M2 macrophage activation, endogenous stem cell recruitment, angiogenesis and osteogenesis. Together, the scaffold realized 97 and 72% bone cover areas after 12 weeks in cranial defect models of rabbit (Φ = 9 mm) and beagle dog (Φ = 15 mm), respectively. Limited stem cells and mismatched interface fusion have plagued biomaterial-mediated cranial reconstruction. Here, the authors engineer an instantly fixable and self-adaptive scaffold to promote calcium chelation and interface integration, regulate macrophage M2 polarization, and recruit endogenous stem cells.
Collapse
Affiliation(s)
- Gonggong Lu
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.,Department of Neurosurgery, West China Hospital, Sichuan University, 37# Guoxue Lane, Chengdu, Sichuan, 610041, P. R. China.,College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Yang Xu
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.,College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Quanying Liu
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.,College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Manyu Chen
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.,College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Huan Sun
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.,College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Peilei Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.,College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Xing Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.,College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Yuxiang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.,College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Xiang Li
- Department of Neurosurgery, West China Hospital, Sichuan University, 37# Guoxue Lane, Chengdu, Sichuan, 610041, P. R. China
| | - Xuhui Hui
- Department of Neurosurgery, West China Hospital, Sichuan University, 37# Guoxue Lane, Chengdu, Sichuan, 610041, P. R. China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, 14#, 3rd, Section of Renmin South Road, Chengdu, Sichuan, 610041, P.R. China
| | - Jun Liu
- School of Biological Science & Medical Engineering, Southeast University, 2# Sipai Building, Xuanwu District, Nanjing, Jiangsu, 210096, P. R. China
| | - Qing Jiang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.,College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Jie Liang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.,College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.,Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China. .,College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.
| | - Yong Sun
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China. .,College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.,College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| |
Collapse
|