1
|
Aminnezhad S, Hama NH, Hasan AH, Bagheri F, Alavi M. Applications of biocompatible polymeric nanomaterials in three-dimensional (3D) scaffolds: Bacterial infections and diabetes. Int J Biol Macromol 2025; 301:140331. [PMID: 39894115 DOI: 10.1016/j.ijbiomac.2025.140331] [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: 06/12/2024] [Revised: 01/12/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
Printed form of polymeric nanomaterials in formation, crosslinking, structure, properties, toxicity and biocompatibility refers to the application of nanotechnology and 3D printing techniques to fabricate polymeric nanomaterials with specific physicochemical and biomedical features. In this regard, applications of 3D printing techniques, specifically for production of 3D scaffold have received huge attention in diabetes and bacterial infections. This review has tried to address recent advances and challenges related to applications of biocompatible polymeric nanomaterials in 3D printing techniques to ameliorate bacterial infections and diabetes. The applications of metal/metal oxide such as silver, gold, zinc, and titanium dioxide, and polymeric nanoparticles can augment the antimicrobial and degradation characteristics of 3D-printed scaffolds. The rapid advancements in 3D bio-printed scaffolds, specifically by artificial intelligence (AI) present a transformative landscape for diabetes treatment, addressing the complex challenges associated with impaired wound healing and tissue regeneration in individuals with diabetes mellitus.
Collapse
Affiliation(s)
- Sargol Aminnezhad
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Nabaz Hamarashid Hama
- Medical Laboratory Science Department, College of Science, Komar University of Science and Technology, Sulaimani, Kurdistan Region, Iraq.
| | - Ayad H Hasan
- Department of Biomedical Sciences, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq; Department of Medical Microbiology, Faculty of Science and Health, Koya University, Koya, KOY45, Kurdistan Region, Iraq.
| | - Fatemeh Bagheri
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehran Alavi
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Kurdistan, Iran.
| |
Collapse
|
2
|
Song H, Li J, Yang H, Kong B, Xu Y, Li X, Li H. Enhancement of functional insulin-producing cell differentiation from embryonic stem cells through MST1-silencing. Diabetol Metab Syndr 2025; 17:93. [PMID: 40108649 PMCID: PMC11924671 DOI: 10.1186/s13098-025-01666-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/09/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Islet β-cell transplantation offers a promising treatment for repairing pancreatic damage in diabetes, with the transcription factor pancreatic duodenal homeobox-1 (PDX1) being crucial for β-cell function and insulin secretion. Mammalian threonine protein kinase (MST1) is recognized for its role in regulating PDX1 during cell apoptosis, yet its function in embryonic stem cell (ESC) differentiation into insulin-producing cells (IPCs) remain underexplored. This study investigated the effect of MST1-silencing on the differentiation of ESC into IPCs. METHODS ESCs were transfected utilizing a recombinant MST1-silencing lentiviral vector (shMST1). qRT-PCR, immunofluorescence, flow cytometry, western blot and ELISA assays were performed to examine function of IPCs in vitro. Furthermore, these IPCs were transplanted into type 1 diabetic mellitus (T1DM) rats. Measuring the changes in blood glucose concentration of animals before and after IPCs transplantation. Intraperitoneal glucose tolerance test (IPGT) was used to determine the regulatory effect of IPCs transplantation on blood glucose stimulation and immunohistochemistry was used to detect the expression of pancreatic Insulin protein in T1DM rats. RESULTS It was observed that IPCs from the shMST1 group exhibited notably improvement in insulin secretion and glucose responsiveness, suggesting MST1 suppression may enhance IPC maturity. The rats demonstrated significant normalization of blood sugar levels and increased insulin levels, akin to non-diabetic controls. This implies that MST1-silencing not only augments IPC function in vitro but also their therapeutic efficacy in vivo. CONCLUSIONS The findings indicate that targeting MST1 offers a novel approach for deriving functionally mature IPCs from ESCs, potentially advancing cell replacement therapies for diabetes. This research underscores the importance of developing IPCs with competent insulin secretion for diabetes treatment in vitro.
Collapse
Affiliation(s)
- Hui Song
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China
- Institute of Endocrinology, Ningxia Medical University, Yinchuan, 750004, China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China
| | - Jiarui Li
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China
- Institute of Endocrinology, Ningxia Medical University, Yinchuan, 750004, China
| | - Haohao Yang
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China
- Institute of Endocrinology, Ningxia Medical University, Yinchuan, 750004, China
| | - Bin Kong
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China
| | - Yu Xu
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China
- Institute of Endocrinology, Ningxia Medical University, Yinchuan, 750004, China
| | - Xiong Li
- People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750004, China.
| | - Hui Li
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China.
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
3
|
Rohban R, Martins CP, Esni F. Advanced therapy to cure diabetes: mission impossible is now possible? Front Cell Dev Biol 2024; 12:1484859. [PMID: 39629270 PMCID: PMC11611888 DOI: 10.3389/fcell.2024.1484859] [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: 08/22/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Cell and Gene therapy are referred to as advanced therapies that represent overlapping fields of regenerative medicine. They have similar therapeutic goals such as to modify cellular identity, improve cell function, or fight a disease. These two therapeutic avenues, however, possess major differences. While cell therapy involves introduction of new cells, gene therapy entails introduction or modification of genes. Furthermore, the aim of cell therapy is often to replace, or repair damaged tissue, whereas gene therapy is used typically as a preventive approach. Diabetes mellitus severely affects the quality of life of afflicted individuals and has various side effects including cardiovascular, ophthalmic disorders, and neuropathy while putting enormous economic pressure on both the healthcare system and the patient. In recent years, great effort has been made to develop cutting-edge therapeutic interventions for diabetes treatment, among which cell and gene therapies stand out. This review aims to highlight various cell- and gene-based therapeutic approaches leading to the generation of new insulin-producing cells as a topmost "panacea" for treating diabetes, while deliberately avoiding a detailed molecular description of these approaches. By doing so, we aim to target readers who are new to the field and wish to get a broad helicopter overview of the historical and current trends of cell- and gene-based approaches in β-cell regeneration.
Collapse
Affiliation(s)
- Rokhsareh Rohban
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Christina P. Martins
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Farzad Esni
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States
- UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- McGowan Institute for regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
4
|
Li Y, He C, Liu R, Xiao Z, Sun B. Stem cells therapy for diabetes: from past to future. Cytotherapy 2023; 25:1125-1138. [PMID: 37256240 DOI: 10.1016/j.jcyt.2023.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/05/2023] [Accepted: 04/24/2023] [Indexed: 06/01/2023]
Abstract
Diabetes mellitus is a chronic disease of carbohydrate metabolism characterized by uncontrolled hyperglycemia due to the body's impaired ability to produce or respond to insulin. Oral or injectable exogenous insulin and its analogs cannot mimic endogenous insulin secreted by healthy individuals, and pancreatic and islet transplants face a severe shortage of sources and transplant complications, all of which limit the widespread use of traditional strategies in diabetes treatment. We are now in the era of stem cells and their potential in ameliorating human disease. At the same time, the rapid development of gene editing and cell-encapsulation technologies has added to the wings of stem cell therapy. However, there are still many unanswered questions before stem cell therapy can be applied clinically to patients with diabetes. In this review, we discuss the progress of strategies to obtain insulin-producing cells from different types of stem cells, the application of gene editing in stem cell therapy for diabetes, as well as summarize the current advanced cell encapsulation technologies in diabetes therapy and look forward to the future development of stem cell therapy in diabetes.
Collapse
Affiliation(s)
- Yumin Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Cong He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China; Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital,The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Rui Liu
- Department of Genetic Engineering, College of Natural Science, University of Suwon, Kyunggi-Do, Republic of Korea
| | - Zhongdang Xiao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China.
| | - Bo Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China.
| |
Collapse
|
5
|
Okcu A, Yazir Y, Şimşek T, Mert S, Duruksu G, Öztürk A, Kiliç KC, Akpinar G, Kasap M. Investigation of the effect of pancreatic decellularized matrix on encapsulated Islets of Langerhans with mesenchymal stem cells. Tissue Cell 2023; 82:102110. [PMID: 37235912 DOI: 10.1016/j.tice.2023.102110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/02/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023]
Abstract
OBJECTIVE In this study, it was aimed to provide a therapeutic approach for T1DM by encapsulating the pancreatic islets with mesenchymal stem cells and decellularized pancreatic extracellular matrix to support the survival of islets while maintaining their cellular activity. METHOD Pancreatic extracellular matrix was decellularized using different concentrations of detergent series. After the preparation of the protein-based tissue extracellular matrix was shown to be free of cells or any genetic material by molecular, immunofluorescence and histochemical techniques. Following the homogenization of the decellularized pancreatic extracellular matrix and the analysis of its protein composition by LC-MS, the matrix proteins were incorporated with pancreatic islets and rat adipose tissue-derived MSCs (rAT-MSCs) in alginate microcapsules. Glucose-stimulated insulin secretion property of the islet cells in the microbeads was evaluated by insulin ELISA. The gene expression profile of the encapsulated cells was analyzed by Real-Time PCR. RESULTS Unlike the protein composition of whole pancreatic tissue, the decellularized pancreas matrix was free of histone proteins or proteins originated from mitochondria. The protein matrix derived from pancreatic tissue was shown to support the growth and maintenance of the islet cells. When compared to the non-encapsulated pancreatic islet, the encapsulated cells demonstrate to be more efficient in terms of insulin expression. CONCLUSION The extracellular pancreatic matrix obtained in this study was directly used as supplementary in the alginate-based microcapsule enhancing the cell survival. The tissue matrix protein and alginate had a synergistic effect on total insulin secretion, which might have the potential to overcome the insulin deficiency. Despite the improvement in the cell viability and the number, the efficiency of the insulin secretion in response to glucose stimulation from the alginate microcapsules did not meet the expectation when compared with the non-encapsulated pancreatic islets.
Collapse
Affiliation(s)
- Alparslan Okcu
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey
| | - Yusufhan Yazir
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey; Department of Histology and Embryology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey.
| | - Turgay Şimşek
- Department of General Surgery, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Serap Mert
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey; Department of Chemistry and Chemical Processing Technology, Kocaeli University, Turkey; Department of Polymer Science and Technology, Kocaeli University, Kocaeli, Turkey
| | - Gökhan Duruksu
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey
| | - Ahmet Öztürk
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey
| | - Kamil Can Kiliç
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey
| | - Gürler Akpinar
- Department of Medical Biology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Murat Kasap
- Department of Medical Biology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
6
|
Beydag-Tasöz BS, Yennek S, Grapin-Botton A. Towards a better understanding of diabetes mellitus using organoid models. Nat Rev Endocrinol 2023; 19:232-248. [PMID: 36670309 PMCID: PMC9857923 DOI: 10.1038/s41574-022-00797-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 01/22/2023]
Abstract
Our understanding of diabetes mellitus has benefited from a combination of clinical investigations and work in model organisms and cell lines. Organoid models for a wide range of tissues are emerging as an additional tool enabling the study of diabetes mellitus. The applications for organoid models include studying human pancreatic cell development, pancreatic physiology, the response of target organs to pancreatic hormones and how glucose toxicity can affect tissues such as the blood vessels, retina, kidney and nerves. Organoids can be derived from human tissue cells or pluripotent stem cells and enable the production of human cell assemblies mimicking human organs. Many organ mimics relevant to diabetes mellitus are already available, but only a few relevant studies have been performed. We discuss the models that have been developed for the pancreas, liver, kidney, nerves and vasculature, how they complement other models, and their limitations. In addition, as diabetes mellitus is a multi-organ disease, we highlight how a merger between the organoid and bioengineering fields will provide integrative models.
Collapse
Affiliation(s)
- Belin Selcen Beydag-Tasöz
- The Novo Nordisk Foundation Center for Stem Cell Biology, Copenhagen, Denmark
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Siham Yennek
- The Novo Nordisk Foundation Center for Stem Cell Biology, Copenhagen, Denmark
| | - Anne Grapin-Botton
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
- Paul Langerhans Institute Dresden, Dresden, Germany.
| |
Collapse
|
7
|
El Khoury R, Ramirez SP, Loyola CD, Joddar B. Demonstration of doxorubicin's cardiotoxicity and screening using a 3D bioprinted spheroidal droplet-based system. RSC Adv 2023; 13:8338-8351. [PMID: 36922946 PMCID: PMC10010162 DOI: 10.1039/d3ra00421j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/03/2023] [Indexed: 03/16/2023] Open
Abstract
Doxorubicin (DOX) is a highly effective anthracycline chemotherapy agent effective in treating a broad range of life-threatening malignancies but it causes cardiotoxicity in many subjects. While the mechanism of its cardiotoxic effects remains elusive, DOX-related cardiotoxicity can lead to heart failure in patients. In this study, we investigated the effects of DOX-induced cardiotoxicity on human cardiomyocytes (CMs) using a three-dimensional (3D) bioprinted cardiac spheroidal droplet based-system in comparison with the traditional two-dimensional cell (2D) culture model. The effects of DOX were alleviated with the addition of N-acetylcysteine (NAC) and Tiron. Caspase-3 activity was quantified, and reactive oxygen species (ROS) production was measured using dihydroethidium (DHE) staining. Application of varying concentrations of DOX (0.4 μM-1 μM) to CMs revealed a dose-specific response, with 1 μM concentration imposing maximum cytotoxicity and 0.22 ± 0.11% of viable cells in 3D samples versus 1.02 ± 0.28% viable cells in 2D cultures, after 5 days of culture. Moreover, a flow cytometric analysis study was conducted to study CMs proliferation in the presence of DOX and antioxidants. Our data support the use of a 3D bioprinted cardiac spheroidal droplet as a robust and high-throughput screening model for drug toxicity. In the future, this 3D spheroidal droplet model can be adopted as a human-derived tissue-engineered equivalent to address challenges in other various aspects of biomedical pre-clinical research.
Collapse
Affiliation(s)
- Raven El Khoury
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso El Paso TX 79968 USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso 500 W. University Avenue El Paso TX 79968 USA
| | - Salma P Ramirez
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso El Paso TX 79968 USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso 500 W. University Avenue El Paso TX 79968 USA
| | - Carla D Loyola
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso El Paso TX 79968 USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso 500 W. University Avenue El Paso TX 79968 USA
| | - Binata Joddar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso El Paso TX 79968 USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso 500 W. University Avenue El Paso TX 79968 USA
- Border Biomedical Research Center, The University of Texas at El Paso 500 W. University Avenue El Paso TX 79968 USA
| |
Collapse
|
8
|
Mohandas S, Gayatri V, Kumaran K, Gopinath V, Paulmurugan R, Ramkumar KM. New Frontiers in Three-Dimensional Culture Platforms to Improve Diabetes Research. Pharmaceutics 2023; 15:pharmaceutics15030725. [PMID: 36986591 PMCID: PMC10056755 DOI: 10.3390/pharmaceutics15030725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Diabetes mellitus is associated with defects in islet β-cell functioning and consequent hyperglycemia resulting in multi-organ damage. Physiologically relevant models that mimic human diabetic progression are urgently needed to identify new drug targets. Three-dimensional (3D) cell-culture systems are gaining a considerable interest in diabetic disease modelling and are being utilized as platforms for diabetic drug discovery and pancreatic tissue engineering. Three-dimensional models offer a marked advantage in obtaining physiologically relevant information and improve drug selectivity over conventional 2D (two-dimensional) cultures and rodent models. Indeed, recent evidence persuasively supports the adoption of appropriate 3D cell technology in β-cell cultivation. This review article provides a considerably updated view of the benefits of employing 3D models in the experimental workflow compared to conventional animal and 2D models. We compile the latest innovations in this field and discuss the various strategies used to generate 3D culture models in diabetic research. We also critically review the advantages and the limitations of each 3D technology, with particular attention to the maintenance of β-cell morphology, functionality, and intercellular crosstalk. Furthermore, we emphasize the scope of improvement needed in the 3D culture systems employed in diabetes research and the promises they hold as excellent research platforms in managing diabetes.
Collapse
Affiliation(s)
- Sundhar Mohandas
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Vijaya Gayatri
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Kriya Kumaran
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Vipin Gopinath
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Centre for Cancer Early Detection, Bio-X Program, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Molecular Oncology Division, Malabar Cancer Centre, Moozhikkara P.O, Thalassery 670103, Kerala, India
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Centre for Cancer Early Detection, Bio-X Program, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Correspondence: (R.P.); (K.M.R.)
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Centre for Cancer Early Detection, Bio-X Program, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Correspondence: (R.P.); (K.M.R.)
| |
Collapse
|
9
|
Tripathi S, Mandal SS, Bauri S, Maiti P. 3D bioprinting and its innovative approach for biomedical applications. MedComm (Beijing) 2023; 4:e194. [PMID: 36582305 PMCID: PMC9790048 DOI: 10.1002/mco2.194] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 12/26/2022] Open
Abstract
3D bioprinting or additive manufacturing is an emerging innovative technology revolutionizing the field of biomedical applications by combining engineering, manufacturing, art, education, and medicine. This process involved incorporating the cells with biocompatible materials to design the required tissue or organ model in situ for various in vivo applications. Conventional 3D printing is involved in constructing the model without incorporating any living components, thereby limiting its use in several recent biological applications. However, this uses additional biological complexities, including material choice, cell types, and their growth and differentiation factors. This state-of-the-art technology consciously summarizes different methods used in bioprinting and their importance and setbacks. It also elaborates on the concept of bioinks and their utility. Biomedical applications such as cancer therapy, tissue engineering, bone regeneration, and wound healing involving 3D printing have gained much attention in recent years. This article aims to provide a comprehensive review of all the aspects associated with 3D bioprinting, from material selection, technology, and fabrication to applications in the biomedical fields. Attempts have been made to highlight each element in detail, along with the associated available reports from recent literature. This review focuses on providing a single platform for cancer and tissue engineering applications associated with 3D bioprinting in the biomedical field.
Collapse
Affiliation(s)
- Swikriti Tripathi
- School of Material Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| | - Subham Shekhar Mandal
- School of Material Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| | - Sudepta Bauri
- School of Material Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| | - Pralay Maiti
- School of Material Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| |
Collapse
|
10
|
Pires IGS, Silva e Souza JA, de Melo Bisneto AV, Passos XS, Carneiro CC. Clinical efficacy of stem-cell therapy on diabetes mellitus: A systematic review and meta-analysis. Transpl Immunol 2022; 75:101740. [DOI: 10.1016/j.trim.2022.101740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 09/12/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022]
|
11
|
Labusca L. Adipose tissue in bone regeneration - stem cell source and beyond. World J Stem Cells 2022; 14:372-392. [PMID: 35949397 PMCID: PMC9244952 DOI: 10.4252/wjsc.v14.i6.372] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/30/2021] [Accepted: 05/27/2022] [Indexed: 02/06/2023] Open
Abstract
Adipose tissue (AT) is recognized as a complex organ involved in major home-ostatic body functions, such as food intake, energy balance, immunomodulation, development and growth, and functioning of the reproductive organs. The role of AT in tissue and organ homeostasis, repair and regeneration is increasingly recognized. Different AT compartments (white AT, brown AT and bone marrow AT) and their interrelation with bone metabolism will be presented. AT-derived stem cell populations - adipose-derived mesenchymal stem cells and pluripotent-like stem cells. Multilineage differentiating stress-enduring and dedifferentiated fat cells can be obtained in relatively high quantities compared to other sources. Their role in different strategies of bone and fracture healing tissue engineering and cell therapy will be described. The current use of AT- or AT-derived stem cell populations for fracture healing and bone regenerative strategies will be presented, as well as major challenges in furthering bone regenerative strategies to clinical settings.
Collapse
Affiliation(s)
- Luminita Labusca
- Magnetic Materials and Sensors, National Institute of Research and Development for Technical Physics, Iasi 700050, Romania
- Orthopedics and Traumatology, County Emergency Hospital Saint Spiridon Iasi, Iasi 700050, Romania.
| |
Collapse
|
12
|
Shinkar K, Rhode K. Could 3D extrusion bioprinting serve to be a real alternative to organ transplantation in the future? ANNALS OF 3D PRINTED MEDICINE 2022. [DOI: 10.1016/j.stlm.2022.100066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
13
|
El Khoury R, Nagiah N, Mudloff JA, Thakur V, Chattopadhyay M, Joddar B. 3D Bioprinted Spheroidal Droplets for Engineering the Heterocellular Coupling between Cardiomyocytes and Cardiac Fibroblasts. CYBORG AND BIONIC SYSTEMS 2021; 2021:9864212. [PMID: 35795473 PMCID: PMC9254634 DOI: 10.34133/2021/9864212] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
Since conventional human cardiac two-dimensional (2D) cell culture and multilayered three-dimensional (3D) models fail in recapitulating cellular complexity and possess inferior translational capacity, we designed and developed a high-throughput scalable 3D bioprinted cardiac spheroidal droplet-organoid model with cardiomyocytes and cardiac fibroblasts that can be used for drug screening or regenerative engineering applications. This study helped establish the parameters for bioprinting and cross-linking a gelatin-alginate-based bioink into 3D spheroidal droplets. A flattened disk-like structure developed in prior studies from our laboratory was used as a control. The microstructural and mechanical stability of the 3D spheroidal droplets was assessed and was found to be ideal for a cardiac scaffold. Adult human cardiac fibroblasts and AC16 cardiomyocytes were mixed in the bioink and bioprinted. Live-dead assay and flow cytometry analysis revealed robust biocompatibility of the 3D spheroidal droplets that supported the growth and proliferation of the cardiac cells in the long-term cultures. Moreover, the heterocellular gap junctional coupling between the cardiomyocytes and cardiac fibroblasts further validated the 3D cardiac spheroidal droplet model.
Collapse
Affiliation(s)
- Raven El Khoury
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX 79968, USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W. University Avenue, El Paso, TX 79968, USA
| | - Naveen Nagiah
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX 79968, USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W. University Avenue, El Paso, TX 79968, USA
| | - Joel A. Mudloff
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX 79968, USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W. University Avenue, El Paso, TX 79968, USA
| | - Vikram Thakur
- Department of Molecular and Translational Medicine, Center of Emphasis in Diabetes and Metabolism, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, TX 79905, USA
| | - Munmun Chattopadhyay
- Department of Molecular and Translational Medicine, Center of Emphasis in Diabetes and Metabolism, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, TX 79905, USA
| | - Binata Joddar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX 79968, USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W. University Avenue, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, 500 W. University Avenue, El Paso, TX 79968, USA
| |
Collapse
|
14
|
Employing Extracellular Matrix-Based Tissue Engineering Strategies for Age-Dependent Tissue Degenerations. Int J Mol Sci 2021; 22:ijms22179367. [PMID: 34502277 PMCID: PMC8431718 DOI: 10.3390/ijms22179367] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 01/10/2023] Open
Abstract
Tissues and organs are not composed of solely cellular components; instead, they converge with an extracellular matrix (ECM). The composition and function of the ECM differ depending on tissue types. The ECM provides a microenvironment that is essential for cellular functionality and regulation. However, during aging, the ECM undergoes significant changes along with the cellular components. The ECM constituents are over- or down-expressed, degraded, and deformed in senescence cells. ECM aging contributes to tissue dysfunction and failure of stem cell maintenance. Aging is the primary risk factor for prevalent diseases, and ECM aging is directly or indirectly correlated to it. Hence, rejuvenation strategies are necessitated to treat various age-associated symptoms. Recent rejuvenation strategies focus on the ECM as the basic biomaterial for regenerative therapies, such as tissue engineering. Modified and decellularized ECMs can be used to substitute aged ECMs and cell niches for culturing engineered tissues. Various tissue engineering approaches, including three-dimensional bioprinting, enable cell delivery and the fabrication of transplantable engineered tissues by employing ECM-based biomaterials.
Collapse
|
15
|
Generation of high yield insulin-producing cells (IPCs) from various sources of stem cells. VITAMINS AND HORMONES 2021; 116:235-268. [PMID: 33752820 DOI: 10.1016/bs.vh.2021.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Type 1 diabetes mellitus occurs when beta cell mass is reduced to less than 20% of the normal level due to immune system destruction of beta cell resulting in an inability to secrete enough insulin. The prevalence of diabetes is expanding according to the American Diabetes Association and the World Health Organization (WHO), foretold to exceed 350 million by 2030. The current treatment does not cure many of the serious complications associated with the disease such as neuropathy, nephropathy, dyslipidemia, retinopathy and cardiovascular disease. Whole pancreas or isolated pancreatic islet transplantation as an alternative therapy can prevent or reduce some of the complications of diabetes. However, the shortage of matched organ or islets cells donor and alloimmune responses limit this therapeutic strategy. Recently, several reports have raised extremely promising results to use different sources of stem cells to differentiate insulin-producing cells and focus on the expansion of these alternative sources. Stem cells, due to their potential for multiple differentiation and self-renewal can differentiate into all cell types, including insulin-producing cells (IPCs). Generation of new beta cells can be achieved from various stem cell sources, including embryonic stem cells (ESCs), adult stem cells, such as mesenchymal stem cells (MSCs) and induced pluripotent stem cells (iPSCs). Thus, this chapter discusses on the assistance of cellular reprogramming of various stem cells as candidates for the generation of IPCs using transcription factors/miRNA, cytokines/small molecules and tissue engineering.
Collapse
|
16
|
Li H, Zhu H, Ge T, Wang Z, Zhang C. Mesenchymal Stem Cell-Based Therapy for Diabetes Mellitus: Enhancement Strategies and Future Perspectives. Stem Cell Rev Rep 2021; 17:1552-1569. [PMID: 33675006 DOI: 10.1007/s12015-021-10139-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2021] [Indexed: 12/11/2022]
Abstract
Diabetes mellitus (DM), a chronic disorder of carbohydrate metabolism, is characterized by the unbridled hyperglycemia resulted from the impaired ability of the body to either produce or respond to insulin. As a cell-based regenerative therapy, mesenchymal stem cells (MSCs) hold immense potency for curing DM duo to their easy isolation, multi-differentiation potential, and immunomodulatory property. However, despite the promising efficacy in pre-clinical animal models, naive MSC administration fails to exhibit clinically satisfactory therapeutic outcomes, which varies greatly among individuals with DM. Recently, numbers of innovative strategies have been applied to improve MSC-based therapy. Preconditioning, genetic modification, combination therapy and exosome application are representative strategies to maximize the therapeutic benefits of MSCs. Therefore, in this review, we summarize recent advancements in mechanistic studies of MSCs-based treatment for DM, and mainly focus on the novel approaches aiming to improve the anti-diabetic potentials of naive MSCs. Additionally, the potential directions of MSCs-based therapy for DM are also proposed at a glance.
Collapse
Affiliation(s)
- Haisen Li
- Department of Plastic and Reconstructive Surgery, Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China.,Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.,Sinoneural Cell Engineering Group Holdings Co., Ltd., Shanghai 201100, China
| | - Hao Zhu
- Sinoneural Cell Engineering Group Holdings Co., Ltd., Shanghai 201100, China
| | - Ting Ge
- Xinxiang First People's Hospital, Xinxiang 453000, China
| | - Zhifeng Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China. .,Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China. .,Sinoneural Cell Engineering Group Holdings Co., Ltd., Shanghai 201100, China.
| | - Chao Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China. .,Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
17
|
Sokolowska P, Janikiewicz J, Jastrzebska E, Brzozka Z, Dobrzyn A. Combinations of regenerative medicine and Lab-on-a-chip systems: New hope to restoring the proper function of pancreatic islets in diabetes. Biosens Bioelectron 2020; 167:112451. [DOI: 10.1016/j.bios.2020.112451] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/18/2020] [Accepted: 07/13/2020] [Indexed: 12/27/2022]
|
18
|
Javaid M, Haleem A. 3D printing applications towards the required challenge of stem cells printing. CLINICAL EPIDEMIOLOGY AND GLOBAL HEALTH 2020. [DOI: 10.1016/j.cegh.2020.02.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
19
|
Doulgkeroglou MN, Di Nubila A, Niessing B, König N, Schmitt RH, Damen J, Szilvassy SJ, Chang W, Csontos L, Louis S, Kugelmeier P, Ronfard V, Bayon Y, Zeugolis DI. Automation, Monitoring, and Standardization of Cell Product Manufacturing. Front Bioeng Biotechnol 2020; 8:811. [PMID: 32766229 PMCID: PMC7381146 DOI: 10.3389/fbioe.2020.00811] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/23/2020] [Indexed: 12/18/2022] Open
Abstract
Although regenerative medicine products are at the forefront of scientific research, technological innovation, and clinical translation, their reproducibility and large-scale production are compromised by automation, monitoring, and standardization issues. To overcome these limitations, new technologies at software (e.g., algorithms and artificial intelligence models, combined with imaging software and machine learning techniques) and hardware (e.g., automated liquid handling, automated cell expansion bioreactor systems, automated colony-forming unit counting and characterization units, and scalable cell culture plates) level are under intense investigation. Automation, monitoring and standardization should be considered at the early stages of the developmental cycle of cell products to deliver more robust and effective therapies and treatment plans to the bedside, reducing healthcare expenditure and improving services and patient care.
Collapse
Affiliation(s)
- Meletios-Nikolaos Doulgkeroglou
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland.,Science Foundation Ireland, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Alessia Di Nubila
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland.,Science Foundation Ireland, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | | | - Niels König
- Fraunhofer Institute for Production Technology, Aachen, Germany
| | - Robert H Schmitt
- Production Engineering Cluster, RWTH Aachen University, Aachen, Germany
| | - Jackie Damen
- STEMCELL Technologies Inc., Vancouver, BC, Canada
| | | | - Wing Chang
- STEMCELL Technologies Ltd., Cambridge, United Kingdom
| | - Lynn Csontos
- STEMCELL Technologies Ltd., Cambridge, United Kingdom
| | - Sharon Louis
- STEMCELL Technologies Inc., Vancouver, BC, Canada
| | | | - Vincent Ronfard
- College System of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, United States.,Cutiss AG, Zurich, Switzerland.,HairClone, Manchester, United Kingdom
| | - Yves Bayon
- Medtronic - Sofradim Production, Trévoux, France
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland.,Science Foundation Ireland, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
20
|
Thakur G, Lee HJ, Jeon RH, Lee SL, Rho GJ. Small Molecule-Induced Pancreatic β-Like Cell Development: Mechanistic Approaches and Available Strategies. Int J Mol Sci 2020; 21:E2388. [PMID: 32235681 PMCID: PMC7178115 DOI: 10.3390/ijms21072388] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/26/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes is a metabolic disease which affects not only glucose metabolism but also lipid and protein metabolism. It encompasses two major types: type 1 and 2 diabetes. Despite the different etiologies of type 1 and 2 diabetes mellitus (T1DM and T2DM, respectively), the defining features of the two forms are insulin deficiency and resistance, respectively. Stem cell therapy is an efficient method for the treatment of diabetes, which can be achieved by differentiating pancreatic β-like cells. The consistent generation of glucose-responsive insulin releasing cells remains challenging. In this review article, we present basic concepts of pancreatic organogenesis, which intermittently provides a basis for engineering differentiation procedures, mainly based on the use of small molecules. Small molecules are more auspicious than any other growth factors, as they have unique, valuable properties like cell-permeability, as well as a nonimmunogenic nature; furthermore, they offer immense benefits in terms of generating efficient functional beta-like cells. We also summarize advances in the generation of stem cell-derived pancreatic cell lineages, especially endocrine β-like cells or islet organoids. The successful induction of stem cells depends on the quantity and quality of available stem cells and the efficient use of small molecules.
Collapse
Affiliation(s)
- Gitika Thakur
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| | - Hyeon-Jeong Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| | - Ryoung-Hoon Jeon
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Sung-Lim Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| | - Gyu-Jin Rho
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| |
Collapse
|
21
|
Alessandra G, Algerta M, Paola M, Carsten S, Cristina L, Paolo M, Elisa M, Gabriella T, Carla P. Shaping Pancreatic β-Cell Differentiation and Functioning: The Influence of Mechanotransduction. Cells 2020; 9:E413. [PMID: 32053947 PMCID: PMC7072458 DOI: 10.3390/cells9020413] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/29/2020] [Accepted: 02/07/2020] [Indexed: 02/08/2023] Open
Abstract
Embryonic and pluripotent stem cells hold great promise in generating β-cells for both replacing medicine and novel therapeutic discoveries in diabetes mellitus. However, their differentiation in vitro is still inefficient, and functional studies reveal that most of these β-like cells still fail to fully mirror the adult β-cell physiology. For their proper growth and functioning, β-cells require a very specific environment, the islet niche, which provides a myriad of chemical and physical signals. While the nature and effects of chemical stimuli have been widely characterized, less is known about the mechanical signals. We here review the current status of knowledge of biophysical cues provided by the niche where β-cells normally live and differentiate, and we underline the possible machinery designated for mechanotransduction in β-cells. Although the regulatory mechanisms remain poorly understood, the analysis reveals that β-cells are equipped with all mechanosensors and signaling proteins actively involved in mechanotransduction in other cell types, and they respond to mechanical cues by changing their behavior. By engineering microenvironments mirroring the biophysical niche properties it is possible to elucidate the β-cell mechanotransductive-regulatory mechanisms and to harness them for the promotion of β-cell differentiation capacity in vitro.
Collapse
Affiliation(s)
- Galli Alessandra
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Marku Algerta
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Marciani Paola
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Schulte Carsten
- CIMAINA, Department of Physics, Università degli Studi di Milano, 20133 Milan, Italy
| | - Lenardi Cristina
- CIMAINA, Department of Physics, Università degli Studi di Milano, 20133 Milan, Italy
| | - Milani Paolo
- CIMAINA, Department of Physics, Università degli Studi di Milano, 20133 Milan, Italy
| | - Maffioli Elisa
- Department of Veterinary Medicine, Università degli Studi di Milano, 20133 Milan, Italy
| | - Tedeschi Gabriella
- Department of Veterinary Medicine, Università degli Studi di Milano, 20133 Milan, Italy
| | - Perego Carla
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| |
Collapse
|
22
|
|