1
|
Lam T, Quach HT, Hall L, Abou Chakra M, Wong AP. A multidisciplinary approach towards modeling of a virtual human lung. NPJ Syst Biol Appl 2025; 11:38. [PMID: 40251169 PMCID: PMC12008392 DOI: 10.1038/s41540-025-00517-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Abstract
Integrating biological data with in silico modeling offers the transformative potential to develop virtual human models, or "digital twins." These models hold immense promise for deepening our understanding of diseases and uncovering new therapeutic strategies. This approach is especially valuable for diseases lacking reliable models. Here we review current modelling efforts in of human lung development, highlighting the role of interdisciplinary collaboration and key advances toward a digital lung twin.
Collapse
Affiliation(s)
- Timothy Lam
- Program in Developmental, Stem cell and Cancer Biology, Hospital for Sick Children, PGCRL 16-9420, Toronto, ON, Canada
| | - Henry T Quach
- Program in Developmental, Stem cell and Cancer Biology, Hospital for Sick Children, PGCRL 16-9420, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Lauren Hall
- Program in Developmental, Stem cell and Cancer Biology, Hospital for Sick Children, PGCRL 16-9420, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Maria Abou Chakra
- Donnelly Centre for Cellular and Biomedical Research, University of Toronto, Toronto, ON, Canada
| | - Amy P Wong
- Program in Developmental, Stem cell and Cancer Biology, Hospital for Sick Children, PGCRL 16-9420, Toronto, ON, Canada.
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
2
|
Zhang Y, Zhang L, Li P, Qiu L, Qu Y, Wu Y, Song H. Extracellular vesicles from adipose-derived stem cell alleviate diabetic cardiomyopathy by regulating Chit1/NLRP3/Caspase-1-Mediated pyroptosis. Int Immunopharmacol 2025; 146:113860. [PMID: 39700960 DOI: 10.1016/j.intimp.2024.113860] [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: 09/30/2024] [Revised: 11/17/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
It is well-established that chronic hyperglycemia progressively destroys the heart structure, weakening function and leading to diabetic cardiomyopathy (DCM). Extracellular vesicles derived from adipose-derived stem cell (ADSC-EVs) have been reported to have anti-inflammatory and immune-modulating effects, but their role in DCM is still poorly understood. Therefore, this study investigated the impact of ADSC-EVs on DCM and potential mechanisms. ADSC-EVs were isolated from the conditioned media of ADSCs. DCM rat models were established using streptozotocin (STZ) in vivo, and high glucose (HG) stimulated H9c2 cardiomyocytes to establish in vitro model. Then mRNA sequencing identified Chit1 as a key gene. Both in vivo and in vitro experiments demonstrated that chitinase 1 (Chit1) and NLRP3/Caspase-1-mediated pyroptosis levels were significantly upregulated in myocardial tissue of rat diabetic cardiomyopathy and hyperglycemic cardiomyocytes, which was reversed by ADSC-EVs treatment. We next observed that in hyperglycemic cardiomyocytes, downregulating Chit1 also resulted in a decrease in NLRP3/Caspase-1-mediated pyroptosis proteins. To a certain extent, the inhibitory effect of ADSC-EVs on the NLRP3/Caspase-1 signaling pathway was reversed by Chit1 overexpression. Taken together, we identified a novel mechanism by which ADSC-EVs regulate NLRP3/Caspase-1-mediated pyroptosis through Chit1 to alleviate diabetic cardiomyopathy, offering an innovative strategy for DCM treatment.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Endocrinology, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, PR China
| | - Liao Zhang
- Department of Endocrinology, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, PR China
| | - Pengjie Li
- Department of Endocrinology, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, PR China
| | - Lili Qiu
- Department of Endocrinology, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, PR China
| | - Ying Qu
- Department of Endocrinology, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, PR China
| | - Yunhe Wu
- Department of Endocrinology, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, PR China
| | - Haiyan Song
- Department of Endocrinology, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, PR China.
| |
Collapse
|
3
|
Wolint P, Hofmann S, von Atzigen J, Böni R, Miescher I, Giovanoli P, Calcagni M, Emmert MY, Buschmann J. Standardization to Characterize the Complexity of Vessel Network Using the Aortic Ring Model. Int J Mol Sci 2024; 26:291. [PMID: 39796147 PMCID: PMC11719671 DOI: 10.3390/ijms26010291] [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] [Revised: 12/05/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Regeneration after ischemia requires to be promoted by (re)perfusion of the affected tissue, and, to date, there is no therapy that covers all needs. In treatment with mesenchymal stem cells (MSC), the secretome acts via paracrine mechanisms and has a positive influence on vascular regeneration via proangiogenic factors. A lack of standardization and the high complexity of vascular structures make it difficult to compare angiogenic readouts from different studies. This emphasizes the need for improved approaches and the introduction of an index in the preclinical setting. A characterization of human MSC secretomes obtained from one of the three formats-single cells, small, and large spheroids-was performed using the chicken aortic ring assay in combination with a modified angiogenic activity index (AAI) and an angiogenic profile. While the secretome of the small spheroid group showed an inhibitory effect on angiogenesis, the large spheroid group impressed with a fully pro-angiogenic response, and a higher AAI compared to the single cell group, underlying the suitability of these three-stem cell-derived secretomes with their distinct angiogenic properties to validate the AAI and the novel angiogenic profile established here.
Collapse
Affiliation(s)
- Petra Wolint
- Division of Surgical Research, University Hospital of Zurich, 8091 Zurich, Switzerland
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Silvan Hofmann
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Julia von Atzigen
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Roland Böni
- White House Center for Liposuction, 8044 Zurich, Switzerland;
| | - Iris Miescher
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Pietro Giovanoli
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Maurizio Calcagni
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Maximilian Y. Emmert
- Institute for Regenerative Medicine (IREM), University of Zurich, 8952 Zurich, Switzerland;
- Deutsches Herzzentrum der Charité (DHZC), Department of Cardiothoracic and Vascular Surgery, 13353 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Johanna Buschmann
- Division of Surgical Research, University Hospital of Zurich, 8091 Zurich, Switzerland
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| |
Collapse
|
4
|
Zhang Z, Ma X, La Y, Guo X, Chu M, Bao P, Yan P, Wu X, Liang C. Advancements in the Application of scRNA-Seq in Breast Research: A Review. Int J Mol Sci 2024; 25:13706. [PMID: 39769466 PMCID: PMC11677372 DOI: 10.3390/ijms252413706] [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: 11/14/2024] [Revised: 12/10/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Single-cell sequencing technology provides apparent advantages in cell population heterogeneity, allowing individuals to better comprehend tissues and organs. Sequencing technology is currently moving beyond the standard transcriptome to the single-cell level, which is likely to bring new insights into the function of breast cells. In this study, we examine the primary cell types involved in breast development, as well as achievements in the study of scRNA-seq in the microenvironment, stressing the finding of novel cell subsets using single-cell approaches and analyzing the problems and solutions to scRNA-seq. Furthermore, we are excited about the field's promising future.
Collapse
Affiliation(s)
- Zhenyu Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China;
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Xiaoming Ma
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Yongfu La
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Xian Guo
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Min Chu
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Pengjia Bao
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Ping Yan
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Xiaoyun Wu
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Chunnian Liang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China;
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| |
Collapse
|
5
|
İyisan N, Hausdörfer O, Wang C, Hiendlmeier L, Harder P, Wolfrum B, Özkale B. Mechanoactivation of Single Stem Cells in Microgels Using a 3D-Printed Stimulation Device. SMALL METHODS 2024; 8:e2400272. [PMID: 39011729 PMCID: PMC11672187 DOI: 10.1002/smtd.202400272] [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] [Received: 02/23/2024] [Revised: 07/03/2024] [Indexed: 07/17/2024]
Abstract
In this study, the novel 3D-printed pressure chamber for encapsulated single-cell stimulation (3D-PRESS) platform is introduced for the mechanical stimulation of single stem cells in 3D microgels. The custom-designed 3D-PRESS, allows precise pressure application up to 400 kPa at the single-cell level. Microfluidics is employed to encapsulate single mesenchymal stem cells within ionically cross-linked alginate microgels with cell adhesion RGD peptides. Rigorous testing affirms the leak-proof performance of the 3D-PRESS device up to 400 kPa, which is fully biocompatible. 3D-PRESS is implemented on mesenchymal stem cells for mechanotransduction studies, by specifically targeting intracellular calcium signaling and the nuclear translocation of a mechanically sensitive transcription factor. Applying 200 kPa pressure on individually encapsulated stem cells reveals heightened calcium signaling in 3D microgels compared to conventional 2D culture. Similarly, Yes-associated protein (YAP) translocation into the nucleus occurs at 200 kPa in 3D microgels with cell-binding RGD peptides unveiling the involvement of integrin-mediated mechanotransduction in singly encapsulated stem cells in 3D microgels. Combining live-cell imaging with precise mechanical control, the 3D-PRESS platform emerges as a versatile tool for exploring cellular responses to pressure stimuli, applicable to various cell types, providing novel insights into single-cell mechanobiology.
Collapse
Affiliation(s)
- Nergishan İyisan
- Microrobotic Bioengineering Lab (MRBL)School of Computation, Information, and TechnologyDepartment of Electrical EngineeringTechnical University of Munich (TUM)Hans‐Piloty‐Straße 185748GarchingGermany
- Munich Institute of Robotics and Machine IntelligenceTechnical University of MunichGeorg‐Brauchle‐Ring 6080992MünchenGermany
- Munich Institute of Biomedical EngineeringTechnical University of MunichBoltzmannstraße 1185748GarchingGermany
| | - Oliver Hausdörfer
- Microrobotic Bioengineering Lab (MRBL)School of Computation, Information, and TechnologyDepartment of Electrical EngineeringTechnical University of Munich (TUM)Hans‐Piloty‐Straße 185748GarchingGermany
| | - Chen Wang
- Microrobotic Bioengineering Lab (MRBL)School of Computation, Information, and TechnologyDepartment of Electrical EngineeringTechnical University of Munich (TUM)Hans‐Piloty‐Straße 185748GarchingGermany
- Munich Institute of Robotics and Machine IntelligenceTechnical University of MunichGeorg‐Brauchle‐Ring 6080992MünchenGermany
- Munich Institute of Biomedical EngineeringTechnical University of MunichBoltzmannstraße 1185748GarchingGermany
| | - Lukas Hiendlmeier
- Munich Institute of Biomedical EngineeringTechnical University of MunichBoltzmannstraße 1185748GarchingGermany
- Neuroelectronics, School of Computation, Information, and TechnologyDepartment of Electrical EngineeringDepartment of Electrical EngineeringTechnical University of Munich (TUM)85748GarchingGermany
| | - Philipp Harder
- Microrobotic Bioengineering Lab (MRBL)School of Computation, Information, and TechnologyDepartment of Electrical EngineeringTechnical University of Munich (TUM)Hans‐Piloty‐Straße 185748GarchingGermany
- Munich Institute of Robotics and Machine IntelligenceTechnical University of MunichGeorg‐Brauchle‐Ring 6080992MünchenGermany
- Munich Institute of Biomedical EngineeringTechnical University of MunichBoltzmannstraße 1185748GarchingGermany
| | - Bernhard Wolfrum
- Munich Institute of Biomedical EngineeringTechnical University of MunichBoltzmannstraße 1185748GarchingGermany
- Neuroelectronics, School of Computation, Information, and TechnologyDepartment of Electrical EngineeringDepartment of Electrical EngineeringTechnical University of Munich (TUM)85748GarchingGermany
| | - Berna Özkale
- Microrobotic Bioengineering Lab (MRBL)School of Computation, Information, and TechnologyDepartment of Electrical EngineeringTechnical University of Munich (TUM)Hans‐Piloty‐Straße 185748GarchingGermany
- Munich Institute of Robotics and Machine IntelligenceTechnical University of MunichGeorg‐Brauchle‐Ring 6080992MünchenGermany
- Munich Institute of Biomedical EngineeringTechnical University of MunichBoltzmannstraße 1185748GarchingGermany
| |
Collapse
|
6
|
Wang Z, Lin M, Pan Y, Liu Y, Yang C, Wu J, Wang Y, Yan B, Zhou J, Chen R, Liu C. Periostin + myeloid cells improved long bone regeneration in a mechanosensitive manner. Bone Res 2024; 12:59. [PMID: 39406726 PMCID: PMC11480347 DOI: 10.1038/s41413-024-00361-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 07/06/2024] [Accepted: 08/01/2024] [Indexed: 10/19/2024] Open
Abstract
Myeloid cells are pivotal in the inflammatory and remodeling phases of fracture repair. Here, we investigate the effect of periostin expressed by myeloid cells on bone regeneration in a monocortical tibial defect (MTD) model. In this study, we show that periostin is expressed by periosteal myeloid cells, primarily the M2 macrophages during bone regeneration. Knockout of periostin in myeloid cells reduces cortical bone thickness, disrupts trabecular bone connectivity, impairs repair impairment, and hinders M2 macrophage polarization. Mechanical stimulation is a regulator of periostin in macrophages. By activating transforming growth factor-β (TGF-β), it increases periostin expression in macrophages and induces M2 polarization. This mechanosensitive effect also reverses the delayed bone repair induced by periostin deficiency in myeloid cells by strengthening the angiogenesis-osteogenesis coupling. In addition, transplantation of mechanically conditioned macrophages into the periosteum over a bone defect results in substantially enhanced repair, confirming the critical role of macrophage-secreted periostin in bone repair. In summary, our findings suggest that mechanical stimulation regulates periostin expression and promotes M2 macrophage polarization, highlighting the potential of mechanically conditioned macrophages as a therapeutic strategy for enhancing bone repair.
Collapse
Affiliation(s)
- Ziyan Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Minmin Lin
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Yonghao Pan
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Yang Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Chengyu Yang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Jianqun Wu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Yan Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Bingtong Yan
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Jingjing Zhou
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Rouxi Chen
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Chao Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
7
|
Soleymani H, Ghorbani M, Sedghi M, Allahverdi A, Naderi-Manesh H. Microfluidics single-cell encapsulation reveals that poly-l-lysine-mediated stem cell adhesion to alginate microgels is crucial for cell-cell crosstalk and its self-renewal. Int J Biol Macromol 2024; 274:133418. [PMID: 38936577 DOI: 10.1016/j.ijbiomac.2024.133418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 04/08/2024] [Accepted: 06/23/2024] [Indexed: 06/29/2024]
Abstract
Microfluidic cell encapsulation has provided a platform for studying the behavior of individual cells and has become a turning point in single-cell analysis during the last decade. The engineered microenvironment, along with protecting the immune response, has led to increasingly presenting the results of practical and pre-clinical studies with the goals of disease treatment, tissue engineering, intelligent control of stem cell differentiation, and regenerative medicine. However, the significance of cell-substrate interaction versus cell-cell communications in the microgel is still unclear. In this study, monodisperse alginate microgels were generated using a flow-focusing microfluidic device to determine how the cell microenvironment can control human bone marrow-derived mesenchymal stem cells (hBMSCs) viability, proliferation, and biomechanical features in single-cell droplets versus multi-cell droplets. Collected results show insufficient cell proliferation (234 % and 329 %) in both single- and multi-cell alginate microgels. Alginate hydrogels supplemented with poly-l-lysine (PLL) showed a better proliferation rate (514 % and 780 %) in a comparison of free alginate hydrogels. Cell stiffness data illustrate that hBMSCs cultured in alginate hydrogels have higher membrane flexibility and migration potency (Young's modulus equal to 1.06 kPa), whereas PLL introduces more binding sites for cell attachment and causes lower flexibility and migration potency (Young's modulus equal to 1.83 kPa). Considering that cell adhesion is the most important parameter in tissue engineering, in which cells do not run away from a 3D substrate, PLL enhances cell stiffness and guarantees cell attachments. In conclusion, cell attachment to PLL-mediated alginate hydrogels is crucial for cell viability and proliferation. It suggests that cell-cell signaling is good enough for stem cell viability, but cell-PLL attachment alongside cell-cell signaling is crucial for stem cell proliferation and self-renewal.
Collapse
Affiliation(s)
- Hossein Soleymani
- Biophysics Department, Faculty of Biological Sciences, Tarbiat Modares University, 14115-154 Tehran, Iran.
| | - Mohammad Ghorbani
- Faculty of Natural Sciences, University of Tabriz, 51666-16471 Tabriz, Iran
| | - Mosslim Sedghi
- Biophysics Department, Faculty of Biological Sciences, Tarbiat Modares University, 14115-154 Tehran, Iran
| | - Abdollah Allahverdi
- Biophysics Department, Faculty of Biological Sciences, Tarbiat Modares University, 14115-154 Tehran, Iran.
| | - Hossein Naderi-Manesh
- Biophysics Department, Faculty of Biological Sciences, Tarbiat Modares University, 14115-154 Tehran, Iran; Department of Nanobiotechnology, Faculty of Biological Science, Tarbiat Modares University, 14115-154 Tehran, Iran.
| |
Collapse
|
8
|
Chen B, Zhu Q, Duan M, Li Q, Wang G, Guan X, Yu P, Xu X, He Y, Xu Y. Optimal Treatment Parameters for Ultrasound-Stimulated Microbubbles in Upregulating Proliferation and Stemness of Bone Marrow Mesenchymal Stem Cells. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:1333-1342. [PMID: 38563453 DOI: 10.1002/jum.16457] [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] [Received: 09/17/2023] [Revised: 03/16/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES Ultrasound-targeted microbubble disruption (UTMD) is a widely used technique to improve the differentiation and proliferation capacity of mesenchymal stem cells (MSCs), but the optimal therapeutic parameters for UTMD are unclear. In this study, we aimed to find the appropriate peak negative pressure (PNP), which is a key parameter for enhancing the stemness properties and proliferation of MSCs. METHODS Experiments were performed in UTMD group, ultrasound (US) group under different PNP exposure conditions (0.5, 1.0, and 1.5 MPa), and control group. Apoptosis safety was analyzed by flow cytometry and MSC proliferation was measured at 12, 24, and 36 hours after irradiation by cell counting kit 8. The expression of the stemness genes NANOG, OCT-4, and SOX-2 were determined by enzyme-linked immunosorbent assay (ELISA) or reverse transcription polymerase chain reaction. RESULTS The results showed that the 1.5 MPa UTMD-treated group had the highest proliferation capacity of MSCs at 24 hours. ELISA or quantitative reverse transcription polymerase chain reaction results showed that UTMD treatment of the 1.5 MPa group significantly upregulated the expression of the stemness genes NANOG, SOX-2, and OCT-4. CONCLUSIONS In conclusion, the appropriate peak PNP value of UTMD was 1.5 MPa, and 1.5 MPa-mediated UTMD group obviously promoted MSCs proliferation and maintained stemness by upregulating the expression of stemness genes.
Collapse
Affiliation(s)
- Beibei Chen
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
- Department of Ultrasound, Postgraduate Training Basement of Jinzhou Medical University, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Qiong Zhu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
- Department of Ultrasound, 953th Hospital, Shigatse Branch, Xinqiao Hospital, Army Medical University, Shigatse, China
| | - Mao Duan
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Qinglong Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Gong Wang
- Department of Ultrasound, Postgraduate Training Basement of Jinzhou Medical University, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Xue Guan
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Pu Yu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiaoxun Xu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ying He
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yali Xu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
9
|
Massidda MW, Ashirov D, Demkov A, Sices A, Baker AB. A Computational Model of Mechanical Stretching of Cultured Cells on a Flexible Membrane. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597769. [PMID: 38895285 PMCID: PMC11185657 DOI: 10.1101/2024.06.06.597769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Mechanical forces applied to cells are known to regulate a wide variety of biological processes. Recent studies have supported that mechanical forces can cause nuclear deformation, leading to significant alterations in the gene expression and chromatin landscape of the cell. While the stresses and strains applied to cells is it is often known or controlled experimentally on a macroscopic length scale, it is often unclear what the actual forces and displacements are at the microscopic level of the cell. In this work, we created a model of cell deformation during application of mechanical stretch to cultured cells growth on a flexible membrane. This configuration is commonly used is in experimental studies as a means to apply controlled mechanical strains to adherent cultured cells. The parameters used in the study were used for application of strain to a mesenchymal stem cell stretched on a membrane. computational model was created to simulate the stresses and strains within the cell under a variety of stain amplitudes, waveforms and frequencies of mechanical loading with the range of commonly used experimental systems. The results demonstrate the connection between mechanical loading parameters applied through the flexible membrane and the resulting stresses and strains within the cell and nucleus. Using a viscoelastic model of chromatin, we connected the results provide to a rough model of resulting deformation within chromatin from the forces applied to the nucleus. Overall, the model is useful in providing insight between experimentally applied mechanical forces and the actual forces within the cell to better interpret the results of experimental studies. Statement of Significance In this work, we created a computational model of the mechanical stretching of cell on a flexible membrane under cyclic mechanical loading. This model provides insight into the forces and displacements inside of cell that result from that application of stretch. As many experiments use this set up, our work is relevant to interpreting many studies that use mechanical stretch to stimulate mechanotransduction.
Collapse
|
10
|
Massidda MW, Demkov A, Sices A, Lee M, Lee J, Paull TT, Kim J, Baker AB. Mechanical Rejuvenation of Mesenchymal Stem Cells from Aged Patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597781. [PMID: 38895474 PMCID: PMC11185588 DOI: 10.1101/2024.06.06.597781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Mesenchymal stem cells (MSC) are an appealing therapeutic cell type for many diseases. However, patients with poor health or advanced age often have MSCs with poor regenerative properties. A major limiter of MSC therapies is cellular senescence, which is marked by limited proliferation capability, diminished multipotency, and reduced regenerative properties. In this work, we explored the ability of applied mechanical forces to reduce cellular senescence in MSCs. Our studies revealed that mechanical conditioning caused a lasting enhancement in proliferation, overall cell culture expansion potential, multipotency, and a reduction of senescence in MSCs from aged donors. Mechanistic studies suggested that these functional enhancements were mediated by oxidative stress and DNA damage repair signaling with mechanical load altering the expression of proteins of the sirtuin pathway, the DNA damage repair protein ATM, and antioxidant proteins. In addition, our results suggest a biophysical mechanism in which mechanical stretch leads to improved recognition of damaged DNA in the nucleus. Analysis of the cells through RNA-seq and ATAC-seq, demonstrated that mechanical loading alters the cell's genetic landscape to cause broad shifts in transcriptomic patterns that related to senescence. Overall, our results demonstrate that mechanical conditioning can rejuvenate mesenchymal stem cells derived from aged patients and improve their potential as a therapeutic cell type. GRAPHICAL ABSTRACT
Collapse
|
11
|
Bai Y, Chen J, Zhang S, Xu G, Mao Z, Ding Y, Wang W. Inflammation-Responsive Cell Membrane-Camouflaged Nanoparticles against Liver Fibrosis via Regulating Endoplasmic Reticulum Stress and Oxidative Stress. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310443. [PMID: 38372054 DOI: 10.1002/adma.202310443] [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] [Received: 10/09/2023] [Revised: 02/02/2024] [Indexed: 02/20/2024]
Abstract
Liver fibrosis represents a reversible stage of various chronic liver diseases that progresses to cirrhosis. This condition is characterized by an imbalance between tissue damage and repair, and the production of fibers in the liver exceeds their degradation. Oxidative stress (OS) resulting from tissue injury and endoplasmic reticulum stress (ERS) triggered by the overproduction of proteins are pivotal factors in liver fibrosis. Melatonin demonstrates the capability to neutralize free radicals, shielding cells from oxidative harm. It is also a specific inhibitor of the ERS receptor transcription activating factor 6 (ATF6), indicating its great potential in ameliorating liver fibrosis. However, its limited water solubility and oral bioavailability of under 15% present hurdles in achieving therapeutic blood concentrations for treating liver fibrosis. The PLGA@Melatonin is constructed by loading melatonin with poly (lactic-co-glycolic acid) (PLGA). Platelet membranes (PM) and activated hepatic stellate cell membranes (HSCM) with high expression of the platelet-derived growth factor receptor (PDGFR) are extracted to successfully construct PM@PLGA@Melatonin and HSCM@PLGA@Melatonin, which are subsequently utilized to treat mice with liver fibrosis. The results illustrated the remarkable therapeutic effects of the two nanoparticles on liver fibrosis, along with their excellent targeting and biosafety properties.
Collapse
Affiliation(s)
- Yang Bai
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, 310058, China
| | - Jiaqi Chen
- Center for Plastic & Reconstructive Surgery, Department of Stomatology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Sitong Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, 310058, China
| | - Guangyu Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, 310058, China
| | - Zhengwei Mao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, 310058, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, 310058, China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, 310058, China
| |
Collapse
|
12
|
Liu X, Rong N, Tian Z, Rich J, Niu L, Li P, Huang L, Dong Y, Zhou W, Zhang P, Chen Y, Wang C, Meng L, Huang TJ, Zheng H. Acoustothermal transfection for cell therapy. SCIENCE ADVANCES 2024; 10:eadk1855. [PMID: 38630814 PMCID: PMC11023511 DOI: 10.1126/sciadv.adk1855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 03/19/2024] [Indexed: 04/19/2024]
Abstract
Transfected stem cells and T cells are promising in personalized cell therapy and immunotherapy against various diseases. However, existing transfection techniques face a fundamental trade-off between transfection efficiency and cell viability; achieving both simultaneously remains a substantial challenge. This study presents an acoustothermal transfection method that leverages acoustic and thermal effects on cells to enhance the permeability of both the cell membrane and nuclear envelope to achieve safe, efficient, and high-throughput transfection of primary T cells and stem cells. With this method, two types of plasmids were simultaneously delivered into the nuclei of mesenchymal stem cells (MSCs) with efficiencies of 89.6 ± 1.2%. CXCR4-transfected MSCs could efficiently target cerebral ischemia sites in vivo and reduce the infarct volume in mice. Our acoustothermal transfection method addresses a key bottleneck in balancing the transfection efficiency and cell viability, which can become a powerful tool in the future for cellular and gene therapies.
Collapse
Affiliation(s)
- Xiufang Liu
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Ning Rong
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Zhenhua Tian
- Department of Mechanical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Joseph Rich
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Lili Niu
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Pengqi Li
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Laixin Huang
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Yankai Dong
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wei Zhou
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Pengfei Zhang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Guangdong 518055, China
| | - Yizhao Chen
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Guangdong 518055, China
| | - Congzhi Wang
- National Innovation Center for Advanced Medical Devices, 385 Mintang Road, Shenzhen 518131, China
| | - Long Meng
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Tony Jun Huang
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Hairong Zheng
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| |
Collapse
|
13
|
Safaei S, Fadaee M, Farzam OR, Yari A, Poursaei E, Aslan C, Samemaleki S, Shanehbandi D, Baradaran B, Kazemi T. Exploring the dynamic interplay between exosomes and the immune tumor microenvironment: implications for breast cancer progression and therapeutic strategies. Breast Cancer Res 2024; 26:57. [PMID: 38553754 PMCID: PMC10981336 DOI: 10.1186/s13058-024-01810-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/18/2024] [Indexed: 04/01/2024] Open
Abstract
Breast cancer continues to pose a substantial worldwide health concern, demanding a thorough comprehension of the complex interaction between cancerous cells and the immune system. Recent studies have shown the significant function of exosomes in facilitating intercellular communication and their participation in the advancement of cancer. Tumor-derived exosomes have been identified as significant regulators in the context of breast cancer, playing a crucial role in modulating immune cell activity and contributing to the advancement of the illness. This study aims to investigate the many effects of tumor-derived exosomes on immune cells in the setting of breast cancer. Specifically, we will examine their role in influencing immune cell polarization, facilitating immunological evasion, and modifying the tumor microenvironment. Furthermore, we explore the nascent domain of exosomes produced from immune cells and their prospective involvement in the prevention of breast cancer. This paper focuses on new research that emphasizes the immunomodulatory characteristics of exosomes produced from immune cells. It also explores the possibility of these exosomes as therapeutic agents or biomarkers for the early identification and prevention of breast cancer. The exploration of the reciprocal connections between exosomes formed from tumors and immune cells, together with the rising significance of exosomes derived from immune cells, presents a potential avenue for the advancement of novel approaches in the field of breast cancer therapy and prevention.
Collapse
Affiliation(s)
- Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Manouchehr Fadaee
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Omid Rahbar Farzam
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Amirhossein Yari
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Elham Poursaei
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Cynthia Aslan
- Research Center for Integrative Medicine in Aging, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Samemaleki
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran.
| |
Collapse
|
14
|
Zheng Y, Li Y, Li M, Wang R, Jiang Y, Zhao M, Lu J, Li R, Li X, Shi S. COVID-19 cooling: Nanostrategies targeting cytokine storm for controlling severe and critical symptoms. Med Res Rev 2024; 44:738-811. [PMID: 37990647 DOI: 10.1002/med.21997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/16/2023] [Accepted: 10/29/2023] [Indexed: 11/23/2023]
Abstract
As severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants continue to wreak havoc worldwide, the "Cytokine Storm" (CS, also known as the inflammatory storm) or Cytokine Release Syndrome has reemerged in the public consciousness. CS is a significant contributor to the deterioration of infected individuals. Therefore, CS control is of great significance for the treatment of critically ill patients and the reduction of mortality rates. With the occurrence of variants, concerns regarding the efficacy of vaccines and antiviral drugs with a broad spectrum have grown. We should make an effort to modernize treatment strategies to address the challenges posed by mutations. Thus, in addition to the requirement for additional clinical data to monitor the long-term effects of vaccines and broad-spectrum antiviral drugs, we can use CS as an entry point and therapeutic target to alleviate the severity of the disease in patients. To effectively combat the mutation, new technologies for neutralizing or controlling CS must be developed. In recent years, nanotechnology has been widely applied in the biomedical field, opening up a plethora of opportunities for CS. Here, we put forward the view of cytokine storm as a therapeutic target can be used to treat critically ill patients by expounding the relationship between coronavirus disease 2019 (COVID-19) and CS and the mechanisms associated with CS. We pay special attention to the representative strategies of nanomaterials in current neutral and CS research, as well as their potential chemical design and principles. We hope that the nanostrategies described in this review provide attractive treatment options for severe and critical COVID-19 caused by CS.
Collapse
Affiliation(s)
- Yu Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuke Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mao Li
- Health Management Centre, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - Rujing Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Mengnan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
15
|
Bury MI, Fuller NJ, Wang X, Chan YY, Sturm RM, Oh SS, Sofer LA, Arora HC, Sharma TT, Nolan BG, Feng W, Rabizadeh RR, Barac M, Edassery SS, Goedegebuure MM, Wang LW, Ganesh B, Halliday LC, Seniw ME, Edassery SL, Mahmud NB, Hofer MD, McKenna KE, Cheng EY, Ameer GA, Sharma AK. Multipotent bone marrow cell-seeded polymeric composites drive long-term, definitive urinary bladder tissue regeneration. PNAS NEXUS 2024; 3:pgae038. [PMID: 38344009 PMCID: PMC10855019 DOI: 10.1093/pnasnexus/pgae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 01/16/2024] [Indexed: 03/02/2024]
Abstract
To date, there are no efficacious translational solutions for end-stage urinary bladder dysfunction. Current surgical strategies, including urinary diversion and bladder augmentation enterocystoplasty (BAE), utilize autologous intestinal segments (e.g. ileum) to increase bladder capacity to protect renal function. Considered the standard of care, BAE is fraught with numerous short- and long-term clinical complications. Previous clinical trials employing tissue engineering approaches for bladder tissue regeneration have also been unable to translate bench-top findings into clinical practice. Major obstacles still persist that need to be overcome in order to advance tissue-engineered products into the clinical arena. These include scaffold/bladder incongruencies, the acquisition and utility of appropriate cells for anatomic and physiologic tissue recapitulation, and the choice of an appropriate animal model for testing. In this study, we demonstrate that the elastomeric, bladder biomechanocompatible poly(1,8-octamethylene-citrate-co-octanol) (PRS; synthetic) scaffold coseeded with autologous bone marrow-derived mesenchymal stem cells and CD34+ hematopoietic stem/progenitor cells support robust long-term, functional bladder tissue regeneration within the context of a clinically relevant baboon bladder augmentation model simulating bladder trauma. Partially cystectomized baboons were independently augmented with either autologous ileum or stem-cell-seeded small-intestinal submucosa (SIS; a commercially available biological scaffold) or PRS grafts. Stem-cell synergism promoted functional trilayer bladder tissue regeneration, including whole-graft neurovascularization, in both cell-seeded grafts. However, PRS-augmented animals demonstrated fewer clinical complications and more advantageous tissue characterization metrics compared to ileum and SIS-augmented animals. Two-year study data demonstrate that PRS/stem-cell-seeded grafts drive bladder tissue regeneration and are a suitable alternative to BAE.
Collapse
Affiliation(s)
- Matthew I Bury
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Natalie J Fuller
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Xinlong Wang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Yvonne Y Chan
- Department of Urologic Surgery, University of California at Davis, Davis, CA 95817, USA
| | - Renea M Sturm
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Sang Su Oh
- Biologic Resources Laboratory, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Laurel A Sofer
- Department of Urology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Hans C Arora
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Tiffany T Sharma
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Bonnie G Nolan
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Wei Feng
- Flow Cytometry Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Rebecca R Rabizadeh
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Milica Barac
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Sonia S Edassery
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Madeleine M Goedegebuure
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Larry W Wang
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Balaji Ganesh
- Flow Cytometry Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Lisa C Halliday
- Biologic Resources Laboratory, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Mark E Seniw
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
| | - Seby L Edassery
- Center for Translational Research and Education, Loyola University Chicago, Chicago, IL 60153, USA
| | - Nadim B Mahmud
- Division of Hematology/Oncology, Department of Medicine, University of Illinois Cancer Center, Chicago, IL 60612, USA
| | | | - Kevin E McKenna
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60612, USA
| | - Earl Y Cheng
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Stanley Manne Children's Research Institute, Louis A. Simpson and Kimberly K. Querrey Biomedical Research Center, Chicago, IL 60611, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Guillermo A Ameer
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
- Vascular Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60612, USA
| | - Arun K Sharma
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Stanley Manne Children's Research Institute, Louis A. Simpson and Kimberly K. Querrey Biomedical Research Center, Chicago, IL 60611, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
16
|
Kaonis S, Aboellail Z, Forman J, Ghosh S. High-Throughput Multiparametric Quantification of Mechanics Driven Heterogeneity in Mesenchymal Stromal Cell Population. Adv Biol (Weinh) 2024; 8:e2300318. [PMID: 37840408 DOI: 10.1002/adbi.202300318] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Indexed: 10/17/2023]
Abstract
Mesenchymal stromal or stem cells (MSCs) are one of the most promising candidates for a myriad of cell therapy applications. Despite showing promise in numerous preclinical and clinical studies, MSC-based therapy is not yet a reality for regenerative medicine due to its suboptimal outcome at the clinical endpoint. The mechanical environment is a critical determinant of MSC gene expression and function. This study reports that MSC population becomes phenotypically heterogenous and commits to an unwanted osteoprogenitor pathway when it experiences an abnormal mechanically stiff environment, compared to its native softer environment. A method is developed to measure the heterogeneity using nuclear shape, chromatin state, and CD73 marker. Heterogeneity is shown to be associated with a larger spread in the nuclear shape parameters and a smaller spread in the chromatin openness. Subsequently, intervention strategies are investigated to create a more homogeneous MSC population. Culturing MSCs on soft surfaces or inhibiting actomyosin on stiff surfaces can make them more homogeneous, while inhibiting YAP, Runx2, and actin polymerization helps maintain but does not fully homogenize them. This study offers insights for cell and tissue engineers, aiding in the design of optimal conditions and materials for MSC culture, ultimately enhancing their therapeutic potential.
Collapse
Affiliation(s)
- Samantha Kaonis
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
| | - Zack Aboellail
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
- Department of Chemical and Biological Engineering, Colorado State University, 400 Isotope Dr, Fort Collins, CO, 80521, USA
| | - Jack Forman
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
- Department of Chemical and Biological Engineering, Colorado State University, 400 Isotope Dr, Fort Collins, CO, 80521, USA
| | - Soham Ghosh
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
- Department of Mechanical Engineering, Colorado State University, 400 Isotope Dr, Fort Collins, CO, 80521, USA
- Cell and Molecular Biology, Colorado State University, 1050 Libbie Coy Way, Fort Collins, CO, 80524, USA
| |
Collapse
|
17
|
Ward NA, Hanley S, Tarpey R, Schreiber LHJ, O'Dwyer J, Roche ET, Duffy GP, Dolan EB. Intermittent actuation attenuates fibrotic behaviour of myofibroblasts. Acta Biomater 2024; 173:80-92. [PMID: 37967693 DOI: 10.1016/j.actbio.2023.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/17/2023]
Abstract
The foreign body response (FBR) to implanted materials culminates in the deposition of a hypo-permeable, collagen rich fibrotic capsule by myofibroblast cells at the implant site. The fibrotic capsule can be deleterious to the function of some medical implants as it can isolate the implant from the host environment. Modulation of fibrotic capsule formation has been achieved using intermittent actuation of drug delivery implants, however the mechanisms underlying this response are not well understood. Here, we use analytical, computational, and in vitro models to understand the response of human myofibroblasts (WPMY-1 stromal cell line) to intermittent actuation using soft robotics and investigate how actuation can alter the secretion of collagen and pro/anti-inflammatory cytokines by these cells. Our findings suggest that there is a mechanical loading threshold that can modulate the fibrotic behaviour of myofibroblasts, by reducing the secretion of soluble collagen, transforming growth factor beta-1 and interleukin 1-beta, and upregulating the anti-inflammatory interleukin-10. By improving our understanding of how cells involved in the FBR respond to mechanical actuation, we can harness this technology to improve functional outcomes for a wide range of implanted medical device applications including drug delivery and cell encapsulation platforms. STATEMENT OF SIGNIFICANCE: A major barrier to the successful clinical translation of many implantable medical devices is the foreign body response (FBR) and resultant deposition of a hypo-permeable fibrotic capsule (FC) around the implant. Perturbation of the implant site using intermittent actuation (IA) of soft-robotic implants has previously been shown to modulate the FBR and reduce FC thickness. However, the mechanisms of action underlying this response were largely unknown. Here, we investigate how IA can alter the activity of myofibroblast cells, and ultimately suggest that there is a mechanical loading threshold within which their fibrotic behaviour can be modulated. These findings can be harnessed to improve functional outcomes for a wide range of medical implants, particularly drug delivery and cell encapsulation devices.
Collapse
Affiliation(s)
- Niamh A Ward
- Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, Ireland
| | - Shirley Hanley
- Flow Cytometry Core Facility, University of Galway, Galway, Ireland
| | - Ruth Tarpey
- Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, Ireland; Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Lucien H J Schreiber
- Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, Ireland; Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Joanne O'Dwyer
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Ellen T Roche
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
| | - Garry P Duffy
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland; Advanced Materials and BioEngineering Research Centre (AMBER), Trinity College Dublin, Dublin, Ireland; CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, Ireland
| | - Eimear B Dolan
- Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, Ireland; CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, Ireland.
| |
Collapse
|
18
|
Doron G, Wood LB, Guldberg RE, Temenoff JS. Poly(ethylene glycol)-Based Hydrogel Microcarriers Alter Secretory Activity of Genetically Modified Mesenchymal Stromal Cells. ACS Biomater Sci Eng 2023; 9:6282-6292. [PMID: 37906515 PMCID: PMC10646834 DOI: 10.1021/acsbiomaterials.3c00954] [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: 07/13/2023] [Revised: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 11/02/2023]
Abstract
In order to scale up culture therapeutic cells, such as mesenchymal stromal cells (MSCs), culture in suspension bioreactors using microcarriers (μCs) is preferred. However, the impact of microcarrier type on the resulting MSC secretory activity has not been investigated. In this study, two poly(ethylene glycol) hydrogel formulations with different swelling ratios (named "stiffer" and "softer") were fabricated as μC substrates to culture MSCs and MSCs genetically modified to express the interleukin-1 receptor antagonist (IL-1Ra-MSCs). Changes in cell number, secretory and angiogenic activity, and changes in MAPK signaling were evaluated when cultured on hydrogel μCs, as well as on tissue culture plastic-based Synthemax μCs. We demonstrated that culture on stiffer μCs increased secretion of IL-1Ra compared to culture on Synthemax μCs by IL-1Ra-MSCs by 1.2- to 1.6-fold, as well as their in vitro angiogenic activity, compared to culture on Synthemax μCs, while culture on both stiffer and softer μCs altered the secretion of several other factors compared to culture on Synthemax μCs. Changes in angiogenic activity corresponded with increased gene expression and secretion of hepatocyte growth factor by MSCs cultured on softer μCs by 2.5- to 6-fold compared to MSCs cultured on Synthemax μCs. Quantification of phosphoprotein signaling with the MAPK pathway revealed broad reduction of pathway activation by IL-1Ra-MSCs cultured on both stiffer and softer μCs compared to Synthemax, where phosphorylated c-Jun, ATF2, and MEK1 were reduced specifically on softer μCs. Overall, this study showed that μC surfaces can influence the secretory activity of genetically modified MSCs and identified associated changes in MAPK pathway signaling, which is a known central regulator of cytokine secretion.
Collapse
Affiliation(s)
- Gilad Doron
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr. NW, Atlanta, Georgia 30332, United States
| | - Levi B. Wood
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr. NW, Atlanta, Georgia 30332, United States
- George
W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Dr. NW, Atlanta, Georgia 30318, United States
- Parker
H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332, United States
| | - Robert E. Guldberg
- Knight
Campus for Accelerating Scientific Impact, University of Oregon, 6231 University of Oregon, Eugene, Oregon 97403, United States
| | - Johnna S. Temenoff
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr. NW, Atlanta, Georgia 30332, United States
- Parker
H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332, United States
| |
Collapse
|
19
|
Chen S, Wang Z, Lu H, Yang R, Wu J. Crucial Factors Influencing the Involvement of Odontogenic Exosomes in Dental Pulp Regeneration. Stem Cell Rev Rep 2023; 19:2632-2649. [PMID: 37578647 DOI: 10.1007/s12015-023-10597-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2023] [Indexed: 08/15/2023]
Abstract
Recent progress in exosome based studies has revealed that they possess several advantages over cells, including "cell-free" properties, low immunogenicity and ethical controversy, high biological safety and effective action. These characteristics confer exosomes significant advantages that allow them to overcome the limitations associated with traditional "cell therapy" by circumventing the issues of immune rejection, scarcity of donor cells, heterogeneity, and ethical concerns. Identification of a complete and effective radical treatment for irreversible pulpal disease, a common clinical problem, continues to pose challenges. Although traditional root canal therapy remains the primary clinical treatment, it does not fully restore the physiological functions of pulp. Although stem cell transplantation appears to be a relatively viable treatment strategy for pulp disease, issues such as cell heterogeneity and poor regeneration effects remain problematic. Dental pulp regeneration strategies based on "cell-free" exosome therapies explored by numerous studies appear to have shown significant advantages. In particular, exosomes derived from odontogenic stem cells have demonstrated considerable potential in tooth tissue regeneration engineering, and continue to exhibit superior therapeutic effects compared to non-odontogenic stem cell-derived exosomes. However, only a few studies have comprehensively summarised their research results, particularly regarding the critical factors involved in the process. Therefore, in this study, our purpose was to review the effects exerted by odontogenic exosomes on pulp regeneration and to analyse and discus crucial factors related to this process, thereby providing scholars with a feasible and manageable new concept with respect to regeneration schemes.
Collapse
Affiliation(s)
- San Chen
- Department of Endodontics, School of Stomatology/Affiliated Stomatological Hospital, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Zijie Wang
- Department of Endodontics, School of Stomatology/Affiliated Stomatological Hospital, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Hongqiao Lu
- Department of Endodontics, School of Stomatology/Affiliated Stomatological Hospital, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Runze Yang
- Department of Endodontics, School of Stomatology/Affiliated Stomatological Hospital, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Jiayuan Wu
- Department of Endodontics, School of Stomatology/Affiliated Stomatological Hospital, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| |
Collapse
|
20
|
Tang H, Sun W, Liu X, Gao Q, Chen Y, Xie C, Lin W, Chen J, Wang L, Fan Z, Zhang L, Ren Y, She Y, He Y, Chen C. A bioengineered trachea-like structure improves survival in a rabbit tracheal defect model. Sci Transl Med 2023; 15:eabo4272. [PMID: 37729433 DOI: 10.1126/scitranslmed.abo4272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
A practical strategy for engineering a trachea-like structure that could be used to repair or replace a damaged or injured trachea is an unmet need. Here, we fabricated bioengineered cartilage (BC) rings from three-dimensionally printed fibers of poly(ɛ-caprolactone) (PCL) and rabbit chondrocytes. The extracellular matrix (ECM) secreted by the chondrocytes combined with the PCL fibers formed a "concrete-rebar structure," with ECM deposited along the PCL fibers, forming a grid similar to that of native cartilage. PCL fiber-hydrogel rings were then fabricated and alternately stacked with BC rings on silicone tubes. This trachea-like structure underwent vascularization after heterotopic transplantation into rabbits for 4 weeks. The vascularized bioengineered trachea-like structure was then orthotopically transplanted by end-to-end anastomosis to native rabbit trachea after a segment of trachea had been resected. The bioengineered trachea-like structure displayed mechanical properties similar to native rabbit trachea and transmural angiogenesis between the rings. The 8-week survival rate in transplanted rabbits was 83.3%, and the respiratory rate of these animals was similar to preoperative levels. This bioengineered trachea-like structure may have potential for treating tracheal stenosis and other tracheal injuries.
Collapse
Affiliation(s)
- Hai Tang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Weiyan Sun
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Xiucheng Liu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Qing Gao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yi Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Chaoqi Xie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Weikang Lin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Jiafei Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Long Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Ziwen Fan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Lei Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| |
Collapse
|
21
|
Dhumale P, Nielsen JV, Hansen ACS, Burton M, Beck HC, Jørgensen MG, Toyserkani NM, Haahr MK, Hansen ST, Lund L, Thomassen M, Sørensen JA, Andersen DC, Jensen CH, Sheikh SP. CD31 defines a subpopulation of human adipose-derived regenerative cells with potent angiogenic effects. Sci Rep 2023; 13:14401. [PMID: 37658225 PMCID: PMC10474028 DOI: 10.1038/s41598-023-41535-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023] Open
Abstract
Cellular heterogeneity represents a major challenge for regenerative treatment using freshly isolated Adipose Derived Regenerative Cells (ADRCs). Emerging data suggest superior efficacy of ADRCs as compared to the ex vivo expanded and more homogeneous ADRCs (= ASCs) for indications involving (micro)vascular deficiency, however, it remains unknown which ADRC cell subtypes account for the improvement. Surprisingly, we found regarding erectile dysfunction (ED) that the number of injected CD31+ ADRCs correlated positively with erectile function 12 months after one bolus of autologous ADRCs. Comprehensive in vitro and ex vivo analyses confirmed superior pro-angiogenic and paracrine effects of human CD31+ enriched ADRCs compared to the corresponding CD31- and parent ADRCs. When CD31+, CD31- and ADRCs were co-cultured in aortic ring- and corpus cavernous tube formation assays, the CD31+ ADRCs induced significantly higher tube development. This effect was corroborated using conditioned medium (CM), while quantitative mass spectrometric analysis suggested that this is likely explained by secretory pro-angiogenic proteins including DKK3, ANGPT2, ANAX2 and VIM, all enriched in CD31+ ADRC CM. Single-cell RNA sequencing showed that transcripts of the upregulated and secreted proteins were present in 9 endothelial ADRC subsets including endothelial progenitor cells in the heterogenous non-cultured ADRCs. Our data suggest that the vascular benefit of using ADRCs in regenerative medicine is dictated by CD31+ ADRCs.
Collapse
Affiliation(s)
- Pratibha Dhumale
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital (OUH), Odense, Denmark
| | - Jakob Vennike Nielsen
- Department of Clinical Biochemistry, Odense University Hospital (OUH), Odense, Denmark
| | | | - Mark Burton
- Department of Clinical Genetics, OUH, Odense, Denmark
| | - Hans Christian Beck
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital (OUH), Odense, Denmark
| | - Mads Gustaf Jørgensen
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Research Unit for Plastic Surgery, Department of Clinical Research, SDU, Odense, Denmark
| | - Navid Mohamadpour Toyserkani
- Department of Plastic Surgery, OUH, Odense, Denmark
- Research Unit for Plastic Surgery, Department of Clinical Research, SDU, Odense, Denmark
| | | | - Sabrina Toft Hansen
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Urology, OUH, Odense, Denmark
| | - Lars Lund
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Urology, OUH, Odense, Denmark
| | - Mads Thomassen
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Clinical Genetics, OUH, Odense, Denmark
| | - Jens Ahm Sørensen
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Plastic Surgery, OUH, Odense, Denmark
- Research Unit for Plastic Surgery, Department of Clinical Research, SDU, Odense, Denmark
| | - Ditte Caroline Andersen
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital (OUH), Odense, Denmark
| | - Charlotte Harken Jensen
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital (OUH), Odense, Denmark
| | - Søren Paludan Sheikh
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark.
- Department of Clinical Biochemistry, Odense University Hospital (OUH), Odense, Denmark.
| |
Collapse
|
22
|
Beatty R, Mendez KL, Schreiber LHJ, Tarpey R, Whyte W, Fan Y, Robinson ST, O'Dwyer J, Simpkin AJ, Tannian J, Dockery P, Dolan EB, Roche ET, Duffy GP. Soft robot-mediated autonomous adaptation to fibrotic capsule formation for improved drug delivery. Sci Robot 2023; 8:eabq4821. [PMID: 37647382 DOI: 10.1126/scirobotics.abq4821] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 08/02/2023] [Indexed: 09/01/2023]
Abstract
The foreign body response impedes the function and longevity of implantable drug delivery devices. As a dense fibrotic capsule forms, integration of the device with the host tissue becomes compromised, ultimately resulting in device seclusion and treatment failure. We present FibroSensing Dynamic Soft Reservoir (FSDSR), an implantable drug delivery device capable of monitoring fibrotic capsule formation and overcoming its effects via soft robotic actuations. Occlusion of the FSDSR porous membrane was monitored over 7 days in a rodent model using electrochemical impedance spectroscopy. The electrical resistance of the fibrotic capsule correlated to its increase in thickness and volume. Our FibroSensing membrane showed great sensitivity in detecting changes at the abiotic/biotic interface, such as collagen deposition and myofibroblast proliferation. The potential of the FSDSR to overcome fibrotic capsule formation and maintain constant drug dosing over time was demonstrated in silico and in vitro. Controlled closed loop release of methylene blue into agarose gels (with a comparable fold change in permeability relating to 7 and 28 days in vivo) was achieved by adjusting the magnitude and frequency of pneumatic actuations after impedance measurements by the FibroSensing membrane. By sensing fibrotic capsule formation in vivo, the FSDSR will be capable of probing and adapting to the foreign body response through dynamic actuation changes. Informed by real-time sensor signals, this device offers the potential for long-term efficacy and sustained drug dosing, even in the setting of fibrotic capsule formation.
Collapse
Affiliation(s)
- Rachel Beatty
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
- SFI Centre for Advanced Materials and BioEngineering Research (AMBER), Trinity College Dublin, Dublin, Ireland
| | - Keegan L Mendez
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lucien H J Schreiber
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Ruth Tarpey
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, Ireland
- Biomedical Engineering, School of Engineering, University of Galway, Galway, Ireland
| | - William Whyte
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yiling Fan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Scott T Robinson
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
- SFI Centre for Advanced Materials and BioEngineering Research (AMBER), Trinity College Dublin, Dublin, Ireland
| | - Joanne O'Dwyer
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Andrew J Simpkin
- School of Mathematical and Statistical Sciences, University of Galway, Galway, Ireland
| | - Joseph Tannian
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Peter Dockery
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Eimear B Dolan
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, Ireland
- Biomedical Engineering, School of Engineering, University of Galway, Galway, Ireland
| | - Ellen T Roche
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Garry P Duffy
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
- SFI Centre for Advanced Materials and BioEngineering Research (AMBER), Trinity College Dublin, Dublin, Ireland
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, Ireland
| |
Collapse
|
23
|
Whisler J, Shahreza S, Schlegelmilch K, Ege N, Javanmardi Y, Malandrino A, Agrawal A, Fantin A, Serwinski B, Azizgolshani H, Park C, Shone V, Demuren OO, Del Rosario A, Butty VL, Holroyd N, Domart MC, Hooper S, Szita N, Boyer LA, Walker-Samuel S, Djordjevic B, Sheridan GK, Collinson L, Calvo F, Ruhrberg C, Sahai E, Kamm R, Moeendarbary E. Emergent mechanical control of vascular morphogenesis. SCIENCE ADVANCES 2023; 9:eadg9781. [PMID: 37566656 PMCID: PMC10421067 DOI: 10.1126/sciadv.adg9781] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 07/13/2023] [Indexed: 08/13/2023]
Abstract
Vascularization is driven by morphogen signals and mechanical cues that coordinately regulate cellular force generation, migration, and shape change to sculpt the developing vascular network. However, it remains unclear whether developing vasculature actively regulates its own mechanical properties to achieve effective vascularization. We engineered tissue constructs containing endothelial cells and fibroblasts to investigate the mechanics of vascularization. Tissue stiffness increases during vascular morphogenesis resulting from emergent interactions between endothelial cells, fibroblasts, and ECM and correlates with enhanced vascular function. Contractile cellular forces are key to emergent tissue stiffening and synergize with ECM mechanical properties to modulate the mechanics of vascularization. Emergent tissue stiffening and vascular function rely on mechanotransduction signaling within fibroblasts, mediated by YAP1. Mouse embryos lacking YAP1 in fibroblasts exhibit both reduced tissue stiffness and develop lethal vascular defects. Translating our findings through biology-inspired vascular tissue engineering approaches will have substantial implications in regenerative medicine.
Collapse
Affiliation(s)
- Jordan Whisler
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Somayeh Shahreza
- Department of Mechanical Engineering, University College London, London, UK
| | | | - Nil Ege
- Tumour Cell Biology Laboratory, Francis Crick Institute, London, UK
- Mnemo Therapeutics, 101 Boulevard Murat, 75016 Paris, France
| | - Yousef Javanmardi
- Department of Mechanical Engineering, University College London, London, UK
| | - Andrea Malandrino
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering and Research Center for Biomedical Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany, 10-14 08019 Barcelona, Spain
| | - Ayushi Agrawal
- Department of Mechanical Engineering, University College London, London, UK
| | - Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, London, UK
- Department of Biosciences, University of Milan, Via G. Celoria 26, 20133 Milan, Italy
| | - Bianca Serwinski
- Department of Mechanical Engineering, University College London, London, UK
- 199 Biotechnologies Ltd., Gloucester Road, London W2 6LD, UK
- Northeastern University London, London, E1W 1LP, UK
| | - Hesham Azizgolshani
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Clara Park
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Victoria Shone
- Experimental Histopathology Laboratory, Francis Crick Institute, London, UK
| | - Olukunle O. Demuren
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Amanda Del Rosario
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Vincent L. Butty
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Natalie Holroyd
- UCL Centre for Advanced Biomedical Imaging, Paul O'Gorman Building, 72 Huntley Street, London, UK
| | | | - Steven Hooper
- Tumour Cell Biology Laboratory, Francis Crick Institute, London, UK
| | - Nicolas Szita
- Department of Biochemical Engineering, University College London, London, UK
| | - Laurie A. Boyer
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Simon Walker-Samuel
- UCL Centre for Advanced Biomedical Imaging, Paul O'Gorman Building, 72 Huntley Street, London, UK
| | - Boris Djordjevic
- Department of Mechanical Engineering, University College London, London, UK
- 199 Biotechnologies Ltd., Gloucester Road, London W2 6LD, UK
| | - Graham K. Sheridan
- School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham, UK
| | - Lucy Collinson
- Electron Microscopy Laboratory, Francis Crick Institute, London, UK
| | - Fernando Calvo
- Instituto de Biomedicina y Biotecnología de Cantabria (Consejo Superior de Investigaciones Científicas, Universidad de Cantabria), Santander, Spain
| | | | - Erik Sahai
- Tumour Cell Biology Laboratory, Francis Crick Institute, London, UK
| | - Roger Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, London, UK
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- 199 Biotechnologies Ltd., Gloucester Road, London W2 6LD, UK
| |
Collapse
|
24
|
West TM, Howsmon DP, Massidda MW, Vo HN, Janobas AA, Baker AB, Sacks MS. The effects of strain history on aortic valve interstitial cell activation in a 3D hydrogel environment. APL Bioeng 2023; 7:026101. [PMID: 37035541 PMCID: PMC10076067 DOI: 10.1063/5.0138030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/13/2023] [Indexed: 04/05/2023] Open
Abstract
Aortic valves (AVs) undergo unique stretch histories that include high rates and magnitudes. While major differences in deformation patterns have been observed between normal and congenitally defective bicuspid aortic valves (BAVs), the relation to underlying mechanisms of rapid disease onset in BAV patients remains unknown. To evaluate how the variations in stretch history affect AV interstitial cell (AVIC) activation, high-throughput methods were developed to impart varied cyclical biaxial stretch histories into 3D poly(ethylene) glycol hydrogels seeded with AVICs for 48 h. Specifically, a physiologically mimicking stretch history was compared to two stretch histories with varied peak stretch and stretch rate. Post-conditioned AVICs were imaged for nuclear shape, alpha smooth muscle actin (αSMA) and vimentin (VMN) polymerization, and small mothers against decapentaplegic homologs 2 and 3 (SMAD 2/3) nuclear activity. The results indicated that bulk gel deformations were accurately transduced to the AVICs. Lower peak stretches lead to increased αSMA polymerization. In contrast, VMN polymerization was a function of stretch rate, with SMAD 2/3 nuclear localization and nuclear shape also trending toward stretch rate dependency. Lower than physiological levels of stretch rate led to higher SMAD 2/3 activity, higher VMN polymerization around the nucleus, and lower nuclear elongation. αSMA polymerization did not correlate with VMN polymerization, SMAD 2/3 activity, nor nuclear shape. These results suggest that a negative feedback loop may form between SMAD 2/3, VMN, and nuclear shape to maintain AVIC homeostatic nuclear deformations, which is dependent on stretch rate. These novel results suggest that AVIC mechanobiological responses are sensitive to stretch history and provide insight into the mechanisms of AV disease.
Collapse
Affiliation(s)
- Toni M. West
- James T. Willerson Center for Cardiovascular Modelling and Simulation, Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, Austin, Texas 78711, USA
| | - Daniel P. Howsmon
- James T. Willerson Center for Cardiovascular Modelling and Simulation, Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, Austin, Texas 78711, USA
| | - Miles W. Massidda
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas 78711, USA
| | | | | | - Aaron B. Baker
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas 78711, USA
| | - Michael S. Sacks
- James T. Willerson Center for Cardiovascular Modelling and Simulation, Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, Austin, Texas 78711, USA
| |
Collapse
|
25
|
Massidda MW, Im B, Lee J, Baker AB. Mechanical conditioning of human mesenchymal stem cells for enhancing vascular regeneration. STAR Protoc 2023; 4:102103. [PMID: 36853695 PMCID: PMC9929623 DOI: 10.1016/j.xpro.2023.102103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/14/2022] [Accepted: 01/20/2023] [Indexed: 02/10/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) are an appealing cell type for therapeutic applications but remain limited by poor efficacy in clinical trials. Here, we describe a conditioning technique that enhances the vascular regenerative properties of hMSCs and increases their expression of endothelial cell and pericyte markers. We also describe an alginate gel encapsulation protocol for delivering the conditioned cells. For complete details on the use and execution of this protocol, please refer to Lee et al. (2021).1.
Collapse
Affiliation(s)
- Miles W Massidda
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX, USA
| | - ByungGee Im
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX, USA
| | - Jason Lee
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX, USA
| | - Aaron B Baker
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA; The Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
26
|
Qiu J, Liu XJ, You BA, Ren N, Liu H. Application of Nanomaterials in Stem Cell-Based Therapeutics for Cardiac Repair and Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206487. [PMID: 36642861 DOI: 10.1002/smll.202206487] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/16/2022] [Indexed: 06/17/2023]
Abstract
Cardiovascular disease is a leading cause of disability and death worldwide. Although the survival rate of patients with heart diseases can be improved with contemporary pharmacological treatments and surgical procedures, none of these therapies provide a significant improvement in cardiac repair and regeneration. Stem cell-based therapies are a promising approach for functional recovery of damaged myocardium. However, the available stem cells are difficult to differentiate into cardiomyocytes, which result in the extremely low transplantation efficiency. Nanomaterials are widely used to regulate the myocardial differentiation of stem cells, and play a very important role in cardiac tissue engineering. This study discusses the current status and limitations of stem cells and cell-derived exosomes/micro RNAs based cardiac therapy, describes the cardiac repair mechanism of nanomaterials, summarizes the recent advances in nanomaterials used in cardiac repair and regeneration, and evaluates the advantages and disadvantages of the relevant nanomaterials. Besides discussing the potential clinical applications of nanomaterials in cardiac therapy, the perspectives and challenges of nanomaterials used in stem cell-based cardiac repair and regeneration are also considered. Finally, new research directions in this field are proposed, and future research trends are highlighted.
Collapse
Affiliation(s)
- Jie Qiu
- Medical Research Institute, Jinan Nanjiao Hospital, Jinan, 250002, P. R. China
| | - Xiang-Ju Liu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, 250012, P. R. China
| | - Bei-An You
- Department of Cardiovascular Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Jinan, 266035, P. R. China
| | - Na Ren
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, P. R. China
| | - Hong Liu
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, P. R. China
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| |
Collapse
|
27
|
Sun R, Song X, Zhou K, Zuo Y, Wang R, Rifaie-Graham O, Peeler DJ, Xie R, Leng Y, Geng H, Brachi G, Ma Y, Liu Y, Barron L, Stevens MM. Assembly of Fillable Microrobotic Systems by Microfluidic Loading with Dip Sealing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207791. [PMID: 36502366 PMCID: PMC7615483 DOI: 10.1002/adma.202207791] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/02/2022] [Indexed: 06/17/2023]
Abstract
Microrobots can provide spatiotemporally well-controlled cargo delivery that can improve therapeutic efficiency compared to conventional drug delivery strategies. Robust microfabrication methods to expand the variety of materials or cargoes that can be incorporated into microrobots can greatly broaden the scope of their functions. However, current surface coating or direct blending techniques used for cargo loading result in inefficient loading and poor cargo protection during transportation, which leads to cargo waste, degradation and non-specific release. Herein, a versatile platform to fabricate fillable microrobots using microfluidic loading and dip sealing (MLDS) is presented. MLDS enables the encapsulation of different types of cargoes within hollow microrobots and protection of cargo integrity. The technique is supported by high-resolution 3D printing with an integrated microfluidic loading system, which realizes a highly precise loading process and improves cargo loading capacity. A corresponding dip sealing strategy is developed to encase and protect the loaded cargo whilst maintaining the geometric and structural integrity of the loaded microrobots. This dip sealing technique is suitable for different materials, including thermal and light-responsive materials. The MLDS platform provides new opportunities for microrobotic systems in targeted drug delivery, environmental sensing, and chemically powered micromotor applications.
Collapse
Affiliation(s)
- Rujie Sun
- Department of Materials, Imperial College London, London SW7 2AZ, UK
| | - Xin Song
- Department of Materials, Imperial College London, London SW7 2AZ, UK
| | - Kun Zhou
- Department of Materials, Imperial College London, London SW7 2AZ, UK
| | - Yuyang Zuo
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - Richard Wang
- Department of Materials, Imperial College London, London SW7 2AZ, UK
| | | | - David J. Peeler
- Department of Materials, Imperial College London, London SW7 2AZ, UK
| | - Ruoxiao Xie
- Department of Materials, Imperial College London, London SW7 2AZ, UK
| | - Yixuan Leng
- Department of Materials, Imperial College London, London SW7 2AZ, UK
| | - Hongya Geng
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Giulia Brachi
- Department of Materials, Imperial College London, London SW7 2AZ, UK
| | - Yun Ma
- Department of Materials, Imperial College London, London SW7 2AZ, UK
| | - Yutong Liu
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK
| | - Lorna Barron
- Department of Materials, Imperial College London, London SW7 2AZ, UK
| | - Molly M. Stevens
- Department of Materials, Imperial College London, London SW7 2AZ, UK
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
28
|
Chen X, Yu W, Zhang J, Fan X, Liu X, Liu Q, Pan S, Dixon RAF, Li P, Yu P, Shi A. Therapeutic angiogenesis and tissue revascularization in ischemic vascular disease. J Biol Eng 2023; 17:13. [PMID: 36797776 PMCID: PMC9936669 DOI: 10.1186/s13036-023-00330-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023] Open
Abstract
Ischemic vascular disease is a major healthcare problem. The keys to treatment lie in vascular regeneration and restoration of perfusion. However, current treatments cannot satisfy the need for vascular regeneration to restore blood circulation. As biomedical research has evolved rapidly, a variety of potential alternative therapeutics has been explored widely, such as growth factor-based therapy, cell-based therapy, and material-based therapy including nanomedicine and biomaterials. This review will comprehensively describe the main pathogenesis of vascular injury in ischemic vascular disease, the therapeutic function of the above three treatment strategies, the corresponding potential challenges, and future research directions.
Collapse
Affiliation(s)
- Xinyue Chen
- grid.412455.30000 0004 1756 5980The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006 Jiangxi China
| | - Wenlu Yu
- grid.260463.50000 0001 2182 8825School of Ophthalmology and Optometry of Nanchang University, Nanchang, 330006 China
| | - Jing Zhang
- grid.412455.30000 0004 1756 5980Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006 Jiangxi China
| | - Xiao Fan
- grid.412455.30000 0004 1756 5980Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006 Jiangxi China
| | - Xiao Liu
- grid.412536.70000 0004 1791 7851Department of Cardiovascular Medicine, The Second Affiliated Hospital of Sun Yat Sen University, Guangzhou, 51000 Guangdong China
| | - Qi Liu
- grid.416470.00000 0004 4656 4290Wafic Said Molecular Cardiology Research Laboratory, The Texas Heart Institute, Houston, TX USA
| | - Su Pan
- grid.416470.00000 0004 4656 4290Wafic Said Molecular Cardiology Research Laboratory, The Texas Heart Institute, Houston, TX USA
| | - Richard A. F. Dixon
- grid.416470.00000 0004 4656 4290Wafic Said Molecular Cardiology Research Laboratory, The Texas Heart Institute, Houston, TX USA
| | - Pengyang Li
- grid.224260.00000 0004 0458 8737Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA USA
| | - Peng Yu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China. .,Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China.
| | - Ao Shi
- School of Medicine, St. George University of London, London, UK. .,School of Medicine, University of Nicosia, Nicosia, Cyprus.
| |
Collapse
|
29
|
Sun D, Mou S, Chen L, Yang J, Wang R, Zhong A, Wang W, Tong J, Wang Z, Sun J. High yield engineered nanovesicles from ADSC with enriched miR-21-5p promote angiogenesis in adipose tissue regeneration. Biomater Res 2022; 26:83. [PMID: 36528594 PMCID: PMC9758932 DOI: 10.1186/s40824-022-00325-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have been found to have a great potential for soft tissue repair due to various biological functions, including pro-angiogenesis and low immunogenicity. However, the low yield and heterogeneity of MSC-EVs limited their clinical transformation. This study was designed to develop a novel adipose-derived stem cell engineered nanovesicles (ADSC-NVs) with high production and explore its pro-angiogenetic effect and application in adipose tissue regeneration. METHODS Adipose-derived stem cell-derived extracellular vesicles (ADSC-EVs) were isolated from an EVs-free culture medium for human ADSCs (hADSCs). ADSC-NVs were prepared by sequentially extruding ADSCs followed by iodixanol density gradient ultracentrifugation and were compared with ADSC-EVs in morphology, size distribution, protein contents and yield. The pro-angiogenetic effect of ADSC-NVs in different doses (0, 5, 20 and 80 μg/mL) in vitro was determined using transwell assay, tube formation assay, western blot and qRT-PCR. In vivo, BALB/c nude mice were administered injection of a mixture of fat granules and different dose of ADSC-NVs and grafts were harvested at 12 weeks post-transplantation for further analysis. By analyzing the weight and volume of grafts and histological evaluation, we investigated the effect of ADSC-NVs in vessel formation and adipose tissue regeneration. RESULTS Our results showed yield of purified ADSC-NVs was approximately 20 times more than that of ADSC-EVs secreted by the same number of ADSCs. In vitro, both ADSC-NVs and ADSC-EVs exhibited a dose-dependent pro-angiogenetic effect, despite their distinct miRNA profiles. These effects of ADSC-NVs may be mediated by enriched miR-21-5p via PTEN inhibition and PI3K/p-Akt signaling activation. Furthermore, after a mixed injection of ADSC-NVs, vessel formation and adipose regeneration were observed in vivo in fat implants. CONCLUSIONS Our study developed a potent alternative of ADSC-EVs. ADSC-NVs have a high pro-angiogenesis potential and can be used as cell-free therapeutic biomaterials in soft tissue regeneration.
Collapse
Affiliation(s)
- Di Sun
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Shan Mou
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Lifeng Chen
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Jie Yang
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Rongrong Wang
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Aimei Zhong
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Wei Wang
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Jing Tong
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Zhenxing Wang
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Jiaming Sun
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| |
Collapse
|
30
|
Whyte W, Goswami D, Wang SX, Fan Y, Ward NA, Levey RE, Beatty R, Robinson ST, Sheppard D, O'Connor R, Monahan DS, Trask L, Mendez KL, Varela CE, Horvath MA, Wylie R, O'Dwyer J, Domingo-Lopez DA, Rothman AS, Duffy GP, Dolan EB, Roche ET. Dynamic actuation enhances transport and extends therapeutic lifespan in an implantable drug delivery platform. Nat Commun 2022; 13:4496. [PMID: 35922421 PMCID: PMC9349266 DOI: 10.1038/s41467-022-32147-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 07/18/2022] [Indexed: 12/03/2022] Open
Abstract
Fibrous capsule (FC) formation, secondary to the foreign body response (FBR), impedes molecular transport and is detrimental to the long-term efficacy of implantable drug delivery devices, especially when tunable, temporal control is necessary. We report the development of an implantable mechanotherapeutic drug delivery platform to mitigate and overcome this host immune response using two distinct, yet synergistic soft robotic strategies. Firstly, daily intermittent actuation (cycling at 1 Hz for 5 minutes every 12 hours) preserves long-term, rapid delivery of a model drug (insulin) over 8 weeks of implantation, by mediating local immunomodulation of the cellular FBR and inducing multiphasic temporal FC changes. Secondly, actuation-mediated rapid release of therapy can enhance mass transport and therapeutic effect with tunable, temporal control. In a step towards clinical translation, we utilise a minimally invasive percutaneous approach to implant a scaled-up device in a human cadaveric model. Our soft actuatable platform has potential clinical utility for a variety of indications where transport is affected by fibrosis, such as the management of type 1 diabetes. Drug delivery implants suffer from diminished release profiles due to fibrous capsule formation over time. Here, the authors use soft robotic actuation to modulate the immune response of the host to maintain drug delivery over the longer-term and to perform controlled release in vivo.
Collapse
Affiliation(s)
- William Whyte
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Debkalpa Goswami
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sophie X Wang
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Yiling Fan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Niamh A Ward
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biomedical Engineering, National University of Ireland Galway, Galway, Ireland
| | - Ruth E Levey
- Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Rachel Beatty
- Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Scott T Robinson
- Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland.,Advanced Materials and BioEngineering Research Centre (AMBER), Trinity College Dublin, Dublin, Ireland
| | - Declan Sheppard
- Department of Radiology, University Hospital, Galway, Ireland
| | - Raymond O'Connor
- Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - David S Monahan
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.,Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Lesley Trask
- Department of Biomedical Engineering, National University of Ireland Galway, Galway, Ireland
| | - Keegan L Mendez
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
| | - Claudia E Varela
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
| | - Markus A Horvath
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
| | - Robert Wylie
- Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Joanne O'Dwyer
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biomedical Engineering, National University of Ireland Galway, Galway, Ireland
| | - Daniel A Domingo-Lopez
- Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Arielle S Rothman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Garry P Duffy
- Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland.,Advanced Materials and BioEngineering Research Centre (AMBER), Trinity College Dublin, Dublin, Ireland
| | - Eimear B Dolan
- Department of Biomedical Engineering, National University of Ireland Galway, Galway, Ireland.
| | - Ellen T Roche
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA.
| |
Collapse
|
31
|
Li J, Zhang Y, Lou Z, Li M, Cui L, Yang Z, Zhang L, Zhang Y, Gu N, Yang F. Magnetic Nanobubble Mechanical Stress Induces the Piezo1-Ca 2+ -BMP2/Smad Pathway to Modulate Neural Stem Cell Fate and MRI/Ultrasound Dual Imaging Surveillance for Ischemic Stroke. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201123. [PMID: 35555970 DOI: 10.1002/smll.202201123] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/21/2022] [Indexed: 06/15/2023]
Abstract
Neural stem cells (NSCs) are used to treat various nervous system diseases because of their self-renewal ability and multidirectional differentiation potential. However, an insufficient ability to track their migration in vivo and poor control over their survival and differentiation efficiency are two major critical challenges for clinical application. Here, it is shown that when magnetic nanobubbles (MNBs), which are assembled from magnetic nanoparticles, are internalized by NSCs, intramembrane volumetric oscillation of the MNBs induces an increase in intracellular hydrostatic pressure and cytoskeleton force, resulting in the activation of the Piezo1-Ca2+ mechanosensory channel. This subsequently triggers the BMP2/Smad biochemical signaling pathway, leading to differentiation of NSCs into the neuronal phenotype. Signaling through the Piezo1-Ca2+ -BMP2/Smad pathway can be further accelerated by application of an external shear stress force using low-intensity pulsed ultrasound. More importantly, magnetic resonance imaging and ultrasound imaging surveillance of NSCs based on MNB labeling can be leveraged to provide NSC therapeutic outcomes. Both the in vitro and in vivo findings demonstrate that a bubble nanostructure-induced physical force can modulate and control the mechanical signaling pathway regulating stem cell development.
Collapse
Affiliation(s)
- Jing Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Yao Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
- College of Materials Science and Engineering, Nanjing Forestry University, Nanjing, 210037, P. R. China
| | - Zhichao Lou
- College of Materials Science and Engineering, Nanjing Forestry University, Nanjing, 210037, P. R. China
| | - Mingxi Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 210009, P. R. China
| | - Lin Cui
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Zhenrong Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Lijuan Zhang
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201204, P. R. China
| | - Yu Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
- College of Materials Science and Engineering, Nanjing Forestry University, Nanjing, 210037, P. R. China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Fang Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| |
Collapse
|
32
|
Wang ZB, Wang ZT, Sun Y, Tan L, Yu JT. The future of stem cell therapies of Alzheimer's disease. Ageing Res Rev 2022; 80:101655. [PMID: 35660003 DOI: 10.1016/j.arr.2022.101655] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/04/2022] [Accepted: 05/27/2022] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) places a heavy burden on the global economy. There is no effective disease-modifying treatment available at present. Since the advent of induced pluripotent stem cells (iPSCs) reprogrammed from human somatic cells, new approaches using iPSC-derived products provided novel insights into AD pathogenesis and drug candidates for the AD treatment. Multiple recent studies using animal models have increased the possibility of reducing pathology and improving cognitive function by cell replacement therapies. In this review, we summarized the advantages, limitations, and future directions of cell replacement therapy, discussed the safety and ethical concerns of this novel therapeutic approach and the possibility of translation to clinical practice.
Collapse
|
33
|
Liu Q, Zhang X, Zhang J. Exosome-Based Nanoplatforms: The Emerging Tools for Breast Cancer Therapy. Front Oncol 2022; 12:898605. [PMID: 35574366 PMCID: PMC9096132 DOI: 10.3389/fonc.2022.898605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/04/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer (BC) remains the leading malignant tumor type among females worldwide. The patients with BC are still faced with undesirable metastasis, relapse rate, and drug resistance. Exosomes are defined as naturally occurring extracellular vesicles (EVs) with typical biomarkers that reflect the characteristics of the parent cells. Exosomes are crucial mediators involved in intercellular communication. By transferring multiple cargoes, represented by proteins, nucleic acids, lipids, metabolites, exosomes contribute to reshaping the recipient cell function and fate. Growing evidence has documented that exosomes originating from BC cells are important participants involved in BC progression and treatments. Nanoparticle-based technology is the cutting-edge field for renewing pharmaceuticals and has endowed deep improvements in precise BC treatment. Additionally, due to their perfect features of the low immune prototype, limited adverse effects, prolongated circulation, and easy modification, exosomes have received much attention as candidates in nano-medicine of BC. The nanoplatforms constructed by exosomes have safety, intelligence, biomimetic, and controlled released advantages for combating BC. Here, we emphasize the multiple exosomes from a variety of cell sources in constructing nanoplatforms for BC therapy, mainly including exosomes and their cargoes, genetically engineered exosomes, and exosome-based carriers. This field would shed light on the promising exosome-based delivery system in BC therapy.
Collapse
Affiliation(s)
- Quan Liu
- Department of Thyroid and Breast Surgery, Xiantao First People's Hospital Affiliated to Yangtze University, Xiantao, China
| | - Xian Zhang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Jun Zhang
- Department of Thyroid and Breast Surgery, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| |
Collapse
|
34
|
Tian KK, Huang SC, Xia XX, Qian ZG. Fibrous Structure and Stiffness of Designer Protein Hydrogels Synergize to Regulate Endothelial Differentiation of Bone Marrow Mesenchymal Stem Cells. Biomacromolecules 2022; 23:1777-1788. [PMID: 35312276 DOI: 10.1021/acs.biomac.2c00032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Matrix stiffness and fibrous structure provided by the native extracellular matrix have been increasingly appreciated as important cues in regulating cell behaviors. Recapitulating these physical cues for cell fate regulation remains a challenge due to the inherent difficulties in making mimetic hydrogels with well-defined compositions, tunable stiffness, and structures. Here, we present two series of fibrous and porous hydrogels with tunable stiffness based on genetically engineered resilin-silk-like and resilin-like protein polymers. Using these hydrogels as substrates, the mechanoresponses of bone marrow mesenchymal stem cells to stiffness and fibrous structure were systematically studied. For both hydrogel series, increasing compression modulus from 8.5 to 14.5 and 23 kPa consistently promoted cell proliferation and differentiation. Nonetheless, the promoting effects were more pronounced on the fibrous gels than their porous counterparts at all three stiffness levels. More interestingly, even the softest fibrous gel (8.5 kPa) allowed the stem cells to exhibit higher endothelial differentiation capability than the toughest porous gel (23 kPa). The predominant role of fibrous structure on the synergistic regulation of endothelial differentiation was further explored. It was found that the stiffness signal activated Yes-associated protein (YAP), the main regulator of endothelial differentiation, via spreading of focal adhesions, whereas fibrous structure reinforced YAP activation by promoting the maturation of focal adhesions and associated F-actin alignment. Therefore, our results shed light on the interplay of physical cues in regulating stem cells and may guide the fabrication of designer proteinaceous matrices toward regenerative medicine.
Collapse
Affiliation(s)
- Kai-Kai Tian
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Sheng-Chen Huang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Xiao-Xia Xia
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Zhi-Gang Qian
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| |
Collapse
|
35
|
Phillips JW, Prominski A, Tian B. Recent advances in materials and applications for bioelectronic and biorobotic systems. VIEW 2022. [DOI: 10.1002/viw.20200157] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Jacob W. Phillips
- Department of Chemistry The University of Chicago Chicago Illinois USA
| | - Aleksander Prominski
- Department of Chemistry The University of Chicago Chicago Illinois USA
- The James Franck Institute The University of Chicago Chicago Illinois USA
- The Institute for Biophysical Dynamics The University of Chicago Chicago Illinois USA
| | - Bozhi Tian
- Department of Chemistry The University of Chicago Chicago Illinois USA
- The James Franck Institute The University of Chicago Chicago Illinois USA
- The Institute for Biophysical Dynamics The University of Chicago Chicago Illinois USA
| |
Collapse
|
36
|
Lu D, Chen M, Yu L, Chen Z, Guo H, Zhang Y, Han Z, Xu T, Wang H, Zhou X, Zhou Z, Teng G. Smart-Polypeptide-Coated Mesoporous Fe 3O 4 Nanoparticles: Non-Interventional Target-Embolization/Thermal Ablation and Multimodal Imaging Combination Theranostics for Solid Tumors. NANO LETTERS 2021; 21:10267-10278. [PMID: 34878286 DOI: 10.1021/acs.nanolett.1c03340] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Tumor theranostics hold great potential for personalized medicine in the future, and transcatheter arterial embolization (TAE) is an important clinical treatment for unresectable or hypervascular tumors. In order to break the limitation, simplify the procedure of TAE, and achieve ideal combinatorial theranostic capability, here, a kind of triblock-polypeptide-coated perfluoropentane-loaded mesoporous Fe3O4 nanocomposites (PFP-m-Fe3O4@PGTTCs) were prepared for non-interventional target-embolization, magnetic hyperthermia, and multimodal imaging combination theranostics of solid tumors. The results of systematic animal experiments by H22-tumor-bearing mice and VX2-tumor-bearing rabbits in vivo indicated that PFP-m-Fe3O4@PGTTC-6.3 has specific tumor accumulation and embolization effects. The tumors' growth has been inhibited and the tumors disappeared 4 weeks and ≤15 days post-injection with embolization and magnetic hyperthermia combination therapy, respectively. The results also showed an excellent effect of magnetic resonance/ultrasound/SPECT multimodal imaging. This pH-responsive non-interventional embolization combinatorial theranostics system provides a novel embolization and multifunctional theranostic candidate for solid tumors.
Collapse
Affiliation(s)
- Dedai Lu
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education, Key Laboratory of Eco-environmental Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Mingshu Chen
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education, Key Laboratory of Eco-environmental Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Lili Yu
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education, Key Laboratory of Eco-environmental Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Zhengpeng Chen
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education, Key Laboratory of Eco-environmental Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Hongyun Guo
- Institute of Gansu Medical Science Research, Gansu Provincial Cancer Hospital, Lanzhou, 730050, China
| | - Yongdong Zhang
- Institute of Gansu Medical Science Research, Gansu Provincial Cancer Hospital, Lanzhou, 730050, China
| | - Zhiming Han
- Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Tingting Xu
- Zhongda Hospital Southeast University, Jiangsu Key Laboratory of Molecular Imaging and Function Imaging, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Haijun Wang
- Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Xing Zhou
- Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Zubang Zhou
- Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Gaojun Teng
- Zhongda Hospital Southeast University, Jiangsu Key Laboratory of Molecular Imaging and Function Imaging, School of Medicine, Southeast University, Nanjing, 210009, China
| |
Collapse
|
37
|
Kumar N, Saraber P, Ding Z, Kusumbe AP. Diversity of Vascular Niches in Bones and Joints During Homeostasis, Ageing, and Diseases. Front Immunol 2021; 12:798211. [PMID: 34975909 PMCID: PMC8718446 DOI: 10.3389/fimmu.2021.798211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/19/2021] [Indexed: 12/29/2022] Open
Abstract
The bones and joints in the skeletal system are composed of diverse cell types, including vascular niches, bone cells, connective tissue cells and mineral deposits and regulate whole-body homeostasis. The capacity of maintaining strength and generation of blood lineages lies within the skeletal system. Bone harbours blood and immune cells and their progenitors, and vascular cells provide several immune cell type niches. Blood vessels in bone are phenotypically and functionally diverse, with distinct capillary subtypes exhibiting striking changes with age. The bone vasculature has a special impact on osteogenesis and haematopoiesis, and dysregulation of the vasculature is associated with diverse blood and bone diseases. Ageing is associated with perturbed haematopoiesis, loss of osteogenesis, increased adipogenesis and diminished immune response and immune cell production. Endothelial and perivascular cells impact immune cell production and play a crucial role during inflammation. Here, we discuss normal and maladapted vascular niches in bone during development, homeostasis, ageing and bone diseases such as rheumatoid arthritis and osteoarthritis. Further, we discuss the role of vascular niches during bone malignancy.
Collapse
Affiliation(s)
| | | | | | - Anjali P. Kusumbe
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), Tissue and Tumor Microenvironments Group, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
38
|
Wang Z, Liu Y, Zhang J, Lin M, Xiao C, Bai H, Liu C. Mechanical loading alleviated the inhibition of β2-adrenergic receptor agonist terbutaline on bone regeneration. FASEB J 2021; 35:e22033. [PMID: 34739146 DOI: 10.1096/fj.202101045rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 12/14/2022]
Abstract
The long-term use of adrenergic medication in treating various conditions, such as asthma, increases the chances of bone fracture. Dynamic mechanical loading at a specific time is a method for improving bone quality and promoting healing. Therefore, we hypothesized that precisely controlling the mechanical environment can contribute to the alleviation of the negative effects of chronic treatment with the common asthma drug terbutaline, which is a β2-adrenergic receptor agonist that facilitates bone homeostasis and defect repair through its anabolic effect on osteogenic cells. Our in vitro results showed that terbutaline can directly inhibit osteogenesis by impairing osteogenic differentiation and mineralization. Chronic treatment in vivo was simulated by administering terbutaline to C57BL/6J mice for 4 weeks before bone defect surgery and mechanical loading. We utilized a stabilized tibial defect model, which allowed the application of anabolic mechanical loading. During homeostasis, chronic terbutaline treatment reduced the bone formation rate, the fracture toughness of long bones, and the concentrations of bone formation markers in the sera. During defect repair, terbutaline decreased the bone volume, type H vessel, and total blood vessel volume. Terbutaline treatment reduced the number of osteogenic cells. Periostin, which was secreted mainly by Prrx1+ osteoprogenitors and F4/80+ macrophages, was inhibited by treating the bone defect with terbutaline. Interestingly, controlled mechanical loading facilitated the recovery of bone volume and periostin expression and the number of osteogenic cells within the defect. In conclusion, mechanical loading can rescue negative effects on new bone accrual and repair induced by chronic terbutaline treatment.
Collapse
Affiliation(s)
- Ziyan Wang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Jianing Zhang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Chufan Xiao
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Haoying Bai
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
39
|
Fang Y, Yang X, Lin Y, Shi J, Prominski A, Clayton C, Ostroff E, Tian B. Dissecting Biological and Synthetic Soft-Hard Interfaces for Tissue-Like Systems. Chem Rev 2021; 122:5233-5276. [PMID: 34677943 DOI: 10.1021/acs.chemrev.1c00365] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Soft and hard materials at interfaces exhibit mismatched behaviors, such as mismatched chemical or biochemical reactivity, mechanical response, and environmental adaptability. Leveraging or mitigating these differences can yield interfacial processes difficult to achieve, or inapplicable, in pure soft or pure hard phases. Exploration of interfacial mismatches and their associated (bio)chemical, mechanical, or other physical processes may yield numerous opportunities in both fundamental studies and applications, in a manner similar to that of semiconductor heterojunctions and their contribution to solid-state physics and the semiconductor industry over the past few decades. In this review, we explore the fundamental chemical roles and principles involved in designing these interfaces, such as the (bio)chemical evolution of adaptive or buffer zones. We discuss the spectroscopic, microscopic, (bio)chemical, and computational tools required to uncover the chemical processes in these confined or hidden soft-hard interfaces. We propose a soft-hard interaction framework and use it to discuss soft-hard interfacial processes in multiple systems and across several spatiotemporal scales, focusing on tissue-like materials and devices. We end this review by proposing several new scientific and engineering approaches to leveraging the soft-hard interfacial processes involved in biointerfacing composites and exploring new applications for these composites.
Collapse
Affiliation(s)
- Yin Fang
- The James Franck Institute, University of Chicago, Chicago, Illinois 60637, United States
| | - Xiao Yang
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Yiliang Lin
- The James Franck Institute, University of Chicago, Chicago, Illinois 60637, United States.,Department of Chemistry, University of Chicago, Chicago, Illinois 60637, United States.,The Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois 60637, United States
| | - Jiuyun Shi
- The James Franck Institute, University of Chicago, Chicago, Illinois 60637, United States.,Department of Chemistry, University of Chicago, Chicago, Illinois 60637, United States.,The Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois 60637, United States
| | - Aleksander Prominski
- The James Franck Institute, University of Chicago, Chicago, Illinois 60637, United States.,Department of Chemistry, University of Chicago, Chicago, Illinois 60637, United States.,The Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois 60637, United States
| | - Clementene Clayton
- Department of Chemistry, University of Chicago, Chicago, Illinois 60637, United States
| | - Ellie Ostroff
- Department of Chemistry, University of Chicago, Chicago, Illinois 60637, United States
| | - Bozhi Tian
- The James Franck Institute, University of Chicago, Chicago, Illinois 60637, United States.,Department of Chemistry, University of Chicago, Chicago, Illinois 60637, United States.,The Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
40
|
|
41
|
Boosting stem cell vascular regenerative capacity. Nat Rev Cardiol 2021; 18:306. [PMID: 33542521 DOI: 10.1038/s41569-021-00525-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
42
|
Lee JH, Yoon JY, Lee JH, Lee HH, Knowles JC, Kim HW. Emerging biogenesis technologies of extracellular vesicles for tissue regenerative therapeutics. J Tissue Eng 2021; 12:20417314211019015. [PMID: 34104388 PMCID: PMC8155774 DOI: 10.1177/20417314211019015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/02/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs), including exosomes, carry the genetic packages of RNA, DNA, and proteins and are heavily involved in cell-cell communications and intracellular signalings. Therefore, EVs are spotlighted as therapeutic mediators for the treatment of injured and dysfunctional tissues as well as biomarkers for the detection of disease status and progress. Several key issues in EVs, including payload content and bioactivity, targeting and bio-imaging ability, and mass-production, need to be improved to enable effective therapeutics and clinical translation. For this, significant efforts have been made recently, including genetic modification, biomolecular and chemical treatment, application of physical/mechanical cues, and 3D cultures. Here we communicate those recent technological advances made mainly in the biogenesis process of EVs or at post-collection stages, which ultimately aimed to improve the therapeutic efficacy in tissue healing and disease curing and the possibility of clinical translation. This communication will help tissue engineers and biomaterial scientists design and produce EVs optimally for tissue regenerative therapeutics.
Collapse
Affiliation(s)
- Jung-Hwan Lee
- Institute of Tissue Regeneration
Engineering (ITREN), Dankook University, Chungcheongnam-do, Cheonan, Republic of
Korea
- Department of Nanobiomedical Science
& BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook
University, Chungcheongnam-do, Cheonan, Republic of Korea
- Department of Biomaterials Science,
College of Dentistry, Dankook University, Chungcheongnam-do, Cheonan, Republic of
Korea
- Department of Regenerative Dental
Medicine, College of Dentistry, Dankook University, Chungcheongnam-do, Cheonan,
Republic of Korea
- Cell & Matter Institute, Dankook
University, Chungcheongnam-do, Cheonan, South Korea
- UCL Eastman-Korea Dental Medicine
Innovation Centre, Dankook University, Chungcheongnam-do, Cheonan, Republic of
Korea
| | - Ji-Young Yoon
- Institute of Tissue Regeneration
Engineering (ITREN), Dankook University, Chungcheongnam-do, Cheonan, Republic of
Korea
- Department of Nanobiomedical Science
& BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook
University, Chungcheongnam-do, Cheonan, Republic of Korea
| | - Jun Hee Lee
- Institute of Tissue Regeneration
Engineering (ITREN), Dankook University, Chungcheongnam-do, Cheonan, Republic of
Korea
- Department of Nanobiomedical Science
& BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook
University, Chungcheongnam-do, Cheonan, Republic of Korea
- Department of Regenerative Dental
Medicine, College of Dentistry, Dankook University, Chungcheongnam-do, Cheonan,
Republic of Korea
- Cell & Matter Institute, Dankook
University, Chungcheongnam-do, Cheonan, South Korea
- UCL Eastman-Korea Dental Medicine
Innovation Centre, Dankook University, Chungcheongnam-do, Cheonan, Republic of
Korea
| | - Hae-Hyoung Lee
- Institute of Tissue Regeneration
Engineering (ITREN), Dankook University, Chungcheongnam-do, Cheonan, Republic of
Korea
- Department of Biomaterials Science,
College of Dentistry, Dankook University, Chungcheongnam-do, Cheonan, Republic of
Korea
- UCL Eastman-Korea Dental Medicine
Innovation Centre, Dankook University, Chungcheongnam-do, Cheonan, Republic of
Korea
| | - Jonathan C Knowles
- UCL Eastman-Korea Dental Medicine
Innovation Centre, Dankook University, Chungcheongnam-do, Cheonan, Republic of
Korea
- Division of Biomaterials and Tissue
Engineering, Eastman Dental Institute, University College London, Royal Free
Hospital, London, UK
- The Discoveries Centre for Regenerative
and Precision Medicine, Eastman Dental Institute, University College London, London,
UK
| | - Hae-Won Kim
- Institute of Tissue Regeneration
Engineering (ITREN), Dankook University, Chungcheongnam-do, Cheonan, Republic of
Korea
- Department of Nanobiomedical Science
& BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook
University, Chungcheongnam-do, Cheonan, Republic of Korea
- Department of Biomaterials Science,
College of Dentistry, Dankook University, Chungcheongnam-do, Cheonan, Republic of
Korea
- Department of Regenerative Dental
Medicine, College of Dentistry, Dankook University, Chungcheongnam-do, Cheonan,
Republic of Korea
- Cell & Matter Institute, Dankook
University, Chungcheongnam-do, Cheonan, South Korea
- UCL Eastman-Korea Dental Medicine
Innovation Centre, Dankook University, Chungcheongnam-do, Cheonan, Republic of
Korea
| |
Collapse
|