51
|
Moon SH, Park TY, Cha HJ, Yang YJ. Photo-/thermo-responsive bioink for improved printability in extrusion-based bioprinting. Mater Today Bio 2024; 25:100973. [PMID: 38322663 PMCID: PMC10844750 DOI: 10.1016/j.mtbio.2024.100973] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/08/2024] Open
Abstract
Extrusion-based bioprinting has demonstrated significant potential for manufacturing constructs, particularly for 3D cell culture. However, there is a greatly limited number of bioink candidates exploited with extrusion-based bioprinting, as they meet the opposing requirements for printability with indispensable rheological features and for biochemical functionality with desirable microenvironment. In this study, a blend of silk fibroin (SF) and iota-carrageenan (CG) was chosen as a cell-friendly printable material. The SF/CG ink exhibited suitable viscosity and shear-thinning properties, coupled with the rapid sol-gel transition of CG. By employing photo-crosslinking of SF, the printability with Pr value close to 1 and structural integrity of the 3D constructs were significantly improved within a matter of seconds. The printed constructs demonstrated a Young's modulus of approximately 250 kPa, making them suitable for keratinocyte and myoblast cell culture. Furthermore, the high cell adhesiveness and viability (maximum >98%) of the loaded cells underscored the considerable potential of this 3D culture scaffold applied for skin and muscle tissues, which can be easily manipulated using an extrusion-based bioprinter.
Collapse
Affiliation(s)
- Seo Hyung Moon
- Department of Biological Sciences and Bioengineering, Inha University, Incheon, 22212, Republic of Korea
| | - Tae Yoon Park
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Hyung Joon Cha
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
- Medical Science and Engineering, School of Convergence Science and Technology, Pohang University of Science, Pohang, 37673, Republic of Korea
| | - Yun Jung Yang
- Department of Biological Sciences and Bioengineering, Inha University, Incheon, 22212, Republic of Korea
- Inha University Hospital, Incheon, 22332, Republic of Korea
| |
Collapse
|
52
|
Xu R, Chen R, Tu C, Gong X, Liu Z, Mei L, Ren X, Li Z. 3D Models of Sarcomas: The Next-generation Tool for Personalized Medicine. PHENOMICS (CHAM, SWITZERLAND) 2024; 4:171-186. [PMID: 38884054 PMCID: PMC11169319 DOI: 10.1007/s43657-023-00111-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 06/18/2024]
Abstract
Sarcoma is a complex and heterogeneous cancer that has been difficult to study in vitro. While two-dimensional (2D) cell cultures and mouse models have been the dominant research tools, three-dimensional (3D) culture systems such as organoids have emerged as promising alternatives. In this review, we discuss recent developments in sarcoma organoid culture, with a focus on their potential as tools for drug screening and biobanking. We also highlight the ways in which sarcoma organoids have been used to investigate the mechanisms of gene regulation, drug resistance, metastasis, and immune interactions. Sarcoma organoids have shown to retain characteristics of in vivo biology within an in vitro system, making them a more representative model for sarcoma research. Our review suggests that sarcoma organoids offer a potential path forward for translational research in this field and may provide a platform for developing personalized therapies for sarcoma patients.
Collapse
Affiliation(s)
- Ruiling Xu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Changsha, 410011 Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, No. 139 Renmin Road, Changsha, 410011 Hunan China
| | - Ruiqi Chen
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Changsha, 410011 Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, No. 139 Renmin Road, Changsha, 410011 Hunan China
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Changsha, 410011 Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, No. 139 Renmin Road, Changsha, 410011 Hunan China
| | - Xiaofeng Gong
- College of Life Science, Fudan University, Shanghai, 200433 China
| | - Zhongyue Liu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Changsha, 410011 Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, No. 139 Renmin Road, Changsha, 410011 Hunan China
| | - Lin Mei
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Changsha, 410011 Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, No. 139 Renmin Road, Changsha, 410011 Hunan China
| | - Xiaolei Ren
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Changsha, 410011 Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, No. 139 Renmin Road, Changsha, 410011 Hunan China
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Changsha, 410011 Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, No. 139 Renmin Road, Changsha, 410011 Hunan China
| |
Collapse
|
53
|
Suryavanshi P, Bodas D. Knockout cancer by nano-delivered immunotherapy using perfusion-aided scaffold-based tumor-on-a-chip. Nanotheranostics 2024; 8:380-400. [PMID: 38751938 PMCID: PMC11093718 DOI: 10.7150/ntno.87818] [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: 12/01/2023] [Accepted: 01/20/2024] [Indexed: 05/18/2024] Open
Abstract
Cancer is a multifactorial disease produced by mutations in the oncogenes and tumor suppressor genes, which result in uncontrolled cell proliferation and resistance to cell death. Cancer progresses due to the escape of altered cells from immune monitoring, which is facilitated by the tumor's mutual interaction with its microenvironment. Understanding the mechanisms involved in immune surveillance evasion and the significance of the tumor microenvironment might thus aid in developing improved therapies. Although in vivo models are commonly utilized, they could be better for time, cost, and ethical concerns. As a result, it is critical to replicate an in vivo model and recreate the cellular and tissue-level functionalities. A 3D cell culture, which gives a 3D architecture similar to that found in vivo, is an appropriate model. Furthermore, numerous cell types can be cocultured, establishing cellular interactions between TME and tumor cells. Moreover, microfluidics perfusion can provide precision flow rates, thus simulating tissue/organ function. Immunotherapy can be used with the perfused 3D cell culture technique to help develop successful therapeutics. Immunotherapy employing nano delivery can target the spot and silence the responsible genes, ensuring treatment effectiveness while minimizing adverse effects. This study focuses on the importance of 3D cell culture in understanding the pathophysiology of 3D tumors and TME, the function of TME in drug resistance, tumor progression, and the development of advanced anticancer therapies for high-throughput drug screening.
Collapse
Affiliation(s)
- Pooja Suryavanshi
- Nanobioscience Group, Agharkar Research Institute, G.G. Agarkar Road, Pune 411 004 India
- Savitribai Phule Pune University, Ganeshkhind Road, Pune 411 007 India
| | - Dhananjay Bodas
- Nanobioscience Group, Agharkar Research Institute, G.G. Agarkar Road, Pune 411 004 India
- Savitribai Phule Pune University, Ganeshkhind Road, Pune 411 007 India
| |
Collapse
|
54
|
Wubshet NH, Cai G, Chen SJ, Sullivan M, Reeves M, Mays D, Harrison M, Varnado P, Yang B, Arreguin-Martinez E, Qu Y, Lin SS, Duran P, Aguilar C, Giza S, Clements T, Liu AP. Cellular mechanotransduction of human osteoblasts in microgravity. NPJ Microgravity 2024; 10:35. [PMID: 38514677 PMCID: PMC10957960 DOI: 10.1038/s41526-024-00386-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/13/2024] [Indexed: 03/23/2024] Open
Abstract
Astronauts experience significant and rapid bone loss as a result of an extended stay in space, making the International Space Station (ISS) the perfect laboratory for studying osteoporosis due to the accelerated nature of bone loss on the ISS. This prompts the question, how does the lack of load due to zero-gravity propagate to bone-forming cells, human fetal osteoblasts (hFOBs), altering their maturation to mineralization? Here, we aim to study the mechanotransduction mechanisms by which bone loss occurs in microgravity. Two automated experiments, microfluidic chips capable of measuring single-cell mechanics via aspiration and cell spheroids incubated in pressure-controlled chambers, were each integrated into a CubeLab deployed to the ISS National Laboratory. For the first experiment, we report protrusion measurements of aspirated cells after exposure to microgravity at the ISS and compare these results to ground control conducted inside the CubeLab. We found slightly elongated protrusions for space samples compared to ground samples indicating softening of hFOB cells in microgravity. In the second experiment, we encapsulated osteoblast spheroids in collagen gel and incubated the samples in pressure-controlled chambers. We found that microgravity significantly reduced filamentous actin levels in the hFOB spheroids. When subjected to pressure, the spheroids exhibited increased pSMAD1/5/9 expression, regardless of the microgravity condition. Moreover, microgravity reduced YAP expression, while pressure increased YAP levels, thus restoring YAP expression for spheroids in microgravity. Our study provides insights into the influence of microgravity on the mechanical properties of bone cells and the impact of compressive pressure on cell signaling in space.
Collapse
Affiliation(s)
- Nadab H Wubshet
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Grace Cai
- Applied Physics Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Samuel J Chen
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | | | | | | | | | - Benjamin Yang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Yunjia Qu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Shan-Shan Lin
- Applied Physics Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Pamela Duran
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Carlos Aguilar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | | | - Allen P Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Applied Physics Program, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biophysics, University of Michigan, Ann Arbor, MI, 48109, USA.
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
55
|
Wubshet NH, Cai G, Chen SJ, Sullivan M, Reeves M, Mays D, Harrison M, Varnado P, Yang B, Arreguin-Martinez E, Qu Y, Lin SS, Duran P, Aguilar C, Giza S, Clements T, Liu AP. Cellular mechanotransduction of human osteoblasts in microgravity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.03.583164. [PMID: 38464311 PMCID: PMC10925314 DOI: 10.1101/2024.03.03.583164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Astronauts experience significant and rapid bone loss as a result of an extended stay in space, making the International Space Station (ISS) the perfect laboratory for studying osteoporosis due to the accelerated nature of bone loss on the ISS. This prompts the question, how does the lack of load due to zero-gravity propagate to bone-forming cells, human fetal osteoblasts (hFOBs), altering their maturation to mineralization? Here, we aim to study the mechanotransduction mechanisms by which bone loss occurs in microgravity. Two automated experiments, 4 microfluidic chips capable of measuring single-cell mechanics of hFOBs via aspiration and cell spheroids incubated in pressure-controlled chambers, were each integrated into a CubeLab deployed to the ISS National Laboratory. For the first experiment, we report protrusion measurements of aspirated cells after exposure to microgravity at the ISS and compare these results to ground control conducted inside the CubeLab. Our analysis revealed slightly elongated protrusions for space samples compared to ground samples indicating softening of hFOB cells in microgravity. In the second experiment, we encapsulated osteoblast spheroids in collagen gel and incubated the samples in pressure-controlled chambers. We found that microgravity significantly reduced filamentous actin levels in the hFOB spheroids. When subjected to pressure, the spheroids exhibited increased pSMAD1/5/9 expression, regardless of the microgravity condition. Moreover, microgravity reduced YAP expression, while pressure increased YAP levels, thus restoring YAP expression for spheroids in microgravity. Our study provides insights into the influence of microgravity on the mechanical properties of bone cells and the impact of compressive pressure on cell behavior and signaling in space.
Collapse
Affiliation(s)
- Nadab H. Wubshet
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Grace Cai
- Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
| | - Samuel J. Chen
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | | | - Benjamin Yang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | | - Yunjia Qu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Shan-Shan Lin
- Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
| | - Pamela Duran
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Carlos Aguilar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Allen P. Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
- Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Biophysics, University of Michigan, Ann Arbor, MI, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
56
|
Hung HC, Mao TL, Fan MH, Huang GZ, Minhalina AP, Chen CL, Liu CL. Enhancement of Tumorigenicity, Spheroid Niche, and Drug Resistance of Pancreatic Cancer Cells in Three-Dimensional Culture System. J Cancer 2024; 15:2292-2305. [PMID: 38495500 PMCID: PMC10937281 DOI: 10.7150/jca.87494] [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: 06/26/2023] [Accepted: 01/27/2024] [Indexed: 03/19/2024] Open
Abstract
The three-dimensional (3D) cell culture technique has been applied comprehensively as a variable platform for medical research, biochemical signal pathway analysis, and evaluation of anti-tumor treatment response due to an excellent recapitulation of a tumor microenvironment (TME) in the in vitro cultured cancer cells. Pancreatic cancer (PaC) is one of the toughest malignancies with a complex TME and refractory treatment response. To comprehensively study the TME of PaC, there is an eager need to develop a 3D culture model to decompose the cellular components and their cross interactions. Herein, we establish a 3D PaC culture system with cancer stem cell (CSC) and scalability properties. To validate our model, we tested the individual PaC cell and the combined effects with cancer-associated fibroblasts (CAFs) on cancer tumorigenicity, the cellular interaction through the CXCR3/CXCL10 axis, and cellular responses reflection of anti-cancer treatments. With the help of our 3D technology, a simulated malignant spheroid with important stromal populations and TME physiochemical properties may be successfully recreated. It can be used in a wide range of preclinical research and helpful in advancing basic and translational cancer biology.
Collapse
Affiliation(s)
- Hao-Chien Hung
- Department of General Surgery, Chang-Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
| | - Tsui-Lien Mao
- Department of Pathology, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
- Department of Pathology, National Taiwan University Hospital, Taipei 10002, Taiwan
| | - Ming-Huei Fan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Guan-Zhi Huang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Ainani Priza Minhalina
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Chi-Long Chen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Pathology, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Chao-Lien Liu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- PhD Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
57
|
Zheng K, Ma Y, Chiu C, Xue M, Zhang C, Du D. Enhanced articular cartilage regeneration using costal chondrocyte-derived scaffold-free tissue engineered constructs with ascorbic acid treatment. J Orthop Translat 2024; 45:140-154. [PMID: 38559899 PMCID: PMC10979122 DOI: 10.1016/j.jot.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/29/2024] [Accepted: 02/20/2024] [Indexed: 04/04/2024] Open
Abstract
Background Cartilage tissue engineering faces challenges related to the use of scaffolds and limited seed cells. This study aims to propose a cost-effective and straightforward approach using costal chondrocytes (CCs) as an alternative cell source to overcome these challenges, eliminating the need for special culture equipment or scaffolds. Methods CCs were cultured at a high cell density with and without ascorbic acid treatment, serving as the experimental and control groups, respectively. Viability and tissue-engineered constructs (TEC) formation were evaluated until day 14. Slices of TEC samples were used for histological staining to evaluate the secretion of glycosaminoglycans and different types of collagen proteins within the extracellular matrix. mRNA sequencing and qPCR were performed to examine gene expression related to cartilage matrix secretion in the chondrocytes. In vivo experiments were conducted by implanting TECs from different groups into the defect site, followed by sample collection after 12 weeks for histological staining and scoring to evaluate the extent of cartilage regeneration. Hematoxylin-eosin (HE), Safranin-O-Fast Green, and Masson's trichrome stainings were used to examine the content of cartilage-related matrix components in the in vivo repair tissue. Immunohistochemical staining for type I and type II collagen, as well as aggrecan, was performed to assess the presence and distribution of these specific markers. Additionally, immunohistochemical staining for type X collagen was used to observe any hypertrophic changes in the repaired tissue. Results Viability of the chondrocytes remained high throughout the culture period, and the TECs displayed an enriched extracellular matrix suitable for surgical procedures. In vitro study revealed glycosaminoglycan and type II collagen production in both groups of TEC, while the TEC matrix treated with ascorbic acid displayed greater abundance. The results of mRNA sequencing and qPCR showed that genes related to cartilage matrix secretion such as Sox9, Col2, and Acan were upregulated by ascorbic acid in costal chondrocytes. Although the addition of Asc-2P led to an increase in COL10 expression according to qPCR and RNA-seq results, the immunofluorescence staining results of the two groups of TECs exhibited similar distribution and fluorescence intensity. In vivo experiments showed that both groups of TEC could adhere to the defect sites and kept hyaline cartilage morphology until 12 weeks. TEC treated with ascorbic acid showed superior cartilage regeneration as evidenced by significantly higher ICRS and O'Driscoll scores and stronger Safranin-O and collagen staining mimicking native cartilage when compared to other groups. In addition, the immunohistochemical staining results of Collgan X indicated that, after 12 weeks, the ascorbic acid-treated TEC did not exhibit further hypertrophy upon transplantation into the defect site, but maintained an expression profile similar to untreated TECs, while slightly higher than the sham-operated group. Conclusion These results suggest that CC-derived scaffold-free TEC presents a promising method for articular cartilage regeneration. Ascorbic acid treatment enhances outcomes by promoting cartilage matrix production. This study provides valuable insights and potential advancements in the field of cartilage tissue engineering. The translational potential of this article Cartilage tissue engineering is an area of research with immense clinical potential. The approach presented in this article offers a cost-effective and straightforward solution, which can minimize the complexity of cell culture and scaffold fabrication. This simplification could offer several translational advantages, such as ease of use, rapid scalability, lower costs, and the potential for patient-specific clinical translation. The use of costal chondrocytes, which are easily obtainable, and the scaffold-free approach, which does not require specialized equipment or membranes, could be particularly advantageous in clinical settings, allowing for in situ regeneration of cartilage.
Collapse
Affiliation(s)
- Kaiwen Zheng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiyang Ma
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng Chiu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengxin Xue
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changqing Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dajiang Du
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
58
|
Yun C, Kim SH, Kim KM, Yang MH, Byun MR, Kim JH, Kwon D, Pham HTM, Kim HS, Kim JH, Jung YS. Advantages of Using 3D Spheroid Culture Systems in Toxicological and Pharmacological Assessment for Osteogenesis Research. Int J Mol Sci 2024; 25:2512. [PMID: 38473760 DOI: 10.3390/ijms25052512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Bone differentiation is crucial for skeletal development and maintenance. Its dysfunction can cause various pathological conditions such as rickets, osteoporosis, osteogenesis imperfecta, or Paget's disease. Although traditional two-dimensional cell culture systems have contributed significantly to our understanding of bone biology, they fail to replicate the intricate biotic environment of bone tissue. Three-dimensional (3D) spheroid cell cultures have gained widespread popularity for addressing bone defects. This review highlights the advantages of employing 3D culture systems to investigate bone differentiation. It highlights their capacity to mimic the complex in vivo environment and crucial cellular interactions pivotal to bone homeostasis. The exploration of 3D culture models in bone research offers enhanced physiological relevance, improved predictive capabilities, and reduced reliance on animal models, which have contributed to the advancement of safer and more effective strategies for drug development. Studies have highlighted the transformative potential of 3D culture systems for expanding our understanding of bone biology and developing targeted therapeutic interventions for bone-related disorders. This review explores how 3D culture systems have demonstrated promise in unraveling the intricate mechanisms governing bone homeostasis and responses to pharmacological agents.
Collapse
Affiliation(s)
- Chawon Yun
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Sou Hyun Kim
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Kyung Mok Kim
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Min Hye Yang
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Mi Ran Byun
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Joung-Hee Kim
- Department of Medical Beauty Care, Dongguk University Wise, Gyeongju 38066, Republic of Korea
| | - Doyoung Kwon
- Jeju Research Institute of Pharmaceutical Sciences, College of Pharmacy, Jeju National University, Jeju 63243, Republic of Korea
| | - Huyen T M Pham
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Hyo-Sop Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Jae-Ho Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Young-Suk Jung
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
59
|
Ueno H, Yamamura S. Fabrication Method for Shape-Controlled 3D Tissue Using High-Porosity Porous Structure. Bioengineering (Basel) 2024; 11:160. [PMID: 38391646 PMCID: PMC10885993 DOI: 10.3390/bioengineering11020160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 01/30/2024] [Accepted: 02/04/2024] [Indexed: 02/24/2024] Open
Abstract
Shape-controlled 3D tissues resemble natural living tissues in human and animal bodies and are essential materials for developing and improving technologies in regenerative medicine, drug discovery, and biological robotics. In previous studies, shape-controlled 3D tissues were fabricated using scaffold structures or 3D bioprinting techniques. However, controlling the shape of 3D tissues without leaving non-natural materials inside the 3D tissue and efficiently fabricating them remains challenging. In this paper, we propose a novel method for fabricating shape-controlled 3D tissues free of non-natural materials using a flexible high-porosity porous structure (HPPS). The HPPS consisted of a micromesh with pore sizes of 14.87 ± 1.83 μm, lattice widths of 2.24 ± 0.10 μm, thicknesses of 9.96 ± 0.92 μm, porosity of 69.06 ± 3.30%, and an I-shaped microchamber of depth 555.26 ± 11.17 μm. U-87 human glioma cells were cultured in an I-shaped HPPS microchamber for 48 h. After cultivation, the 3D tissue was released within a few seconds while maintaining its I-shape. Specific chemicals, such as proteolytic enzymes, were not used. Moreover, the viability of the released cells composed of shape-controlled 3D tissues free of non-natural materials was above 90%. Therefore, the proposed fabrication method is recommended for shape-controlled 3D tissues free of non-natural materials without applying significant stresses to the cells.
Collapse
Affiliation(s)
- Hidetaka Ueno
- Center for Advanced Medical Engineering Research & Development (CAMED), Kobe University, 1-5-1 Minatojima-minamimachi, Chuo-ku, Kobe-city 650-0047, Hyogo, Japan
- Department of Medical Device Engineering, Graduate School of Medicine, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe-city 650-0017, Hyogo, Japan
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 2217-14 Hayashi-cho, Takamatsu-city 761-0395, Kagawa, Japan
| | - Shohei Yamamura
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 2217-14 Hayashi-cho, Takamatsu-city 761-0395, Kagawa, Japan
| |
Collapse
|
60
|
Simelane NWN, Abrahamse H. Zinc phthalocyanine loaded- antibody functionalized nanoparticles enhance photodynamic therapy in monolayer (2-D) and multicellular tumour spheroid (3-D) cell cultures. Front Mol Biosci 2024; 10:1340212. [PMID: 38259685 PMCID: PMC10801020 DOI: 10.3389/fmolb.2023.1340212] [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: 11/21/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
In conventional photodynamic therapy (PDT), effective delivery of photosensitizers (PS) to cancer cells can be challenging, prompting the exploration of active targeting as a promising strategy to enhance PS delivery. Typically, two-dimensional (2-D) monolayer cell culture models are used for investigating targeted photodynamic therapy. However, despite their ease of use, these cell culture models come with certain limitations due to their structural simplicity when compared to three-dimensional (3-D) cell culture models such as multicellular tumour spheroids (MCTSs). In this study, we prepared gold nanoparticles (AuNPs) that were functionalized with antibodies and loaded with tetra sulphonated zinc phthalocyanine (ZnPcS4). Characterization techniques including transmission electron microscopy (TEM) was used to determine the size and morphology of the prepared nanoconjugates. We also conducted a comparative investigation to assess the photodynamic effects of ZnPcS4 alone and/or conjugated onto the bioactively functionalized nanodelivery system in colorectal Caco-2 cells cultured in both in vitro 2-D monolayers and 3-D MCTSs. TEM micrographs revealed small, well distributed, and spherical shaped nanoparticles. Our results demonstrated that biofunctionalized nanoparticle mediated PDT significantly inhibited cell proliferation and induced apoptosis in Caco-2 cancer monolayers and, to a lesser extent, in Caco-2 MCTSs. Live/dead assays further elucidated the impact of actively targeted nanoparticle-photosensitizer nanoconstruct, revealing enhanced cytotoxicity in 2-D cultures, with a notable increase in dead cells post-PDT. In 3-D spheroids, however, while the presence of targeted nanoparticle-photosensitizer system facilitated improved therapeutic outcomes, the live/dead results showed a higher number of viable cells after PDT treatment compared to their 2-D monolayer counterparts suggesting that MCTSs showed more resistance to PS drug as compared to 2-D monolayers. These findings suggest a high therapeutic potential of the multifunctional nanoparticle as a targeted photosensitizer delivery system in PDT of colorectal cancer. Furthermore, the choice of cell culture model influenced the response of cancer cells to PDT treatment, highlighting the feasibility of using MCTSs for targeted PS delivery to colorectal cancer cells.
Collapse
Affiliation(s)
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
61
|
Morrocchi E, van Haren S, Palma P, Levy O. Modeling human immune responses to vaccination in vitro. Trends Immunol 2024; 45:32-47. [PMID: 38135599 PMCID: PMC11688643 DOI: 10.1016/j.it.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023]
Abstract
The human immune system is a complex network of coordinated components that are crucial for health and disease. Animal models, commonly used to study immunomodulatory agents, are limited by species-specific differences, low throughput, and ethical concerns. In contrast, in vitro modeling of human immune responses can enable species- and population-specific mechanistic studies and translational development within the same study participant. Translational accuracy of in vitro models is enhanced by accounting for genetic, epigenetic, and demographic features such as age, sex, and comorbidity. This review explores various human in vitro immune models, considers evidence that they may resemble human in vivo responses, and assesses their potential to accelerate and de-risk vaccine discovery and development.
Collapse
Affiliation(s)
- Elena Morrocchi
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, Rome, Italy; Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
| | - Simon van Haren
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Paolo Palma
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, Rome, Italy; Chair of Pediatrics, Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy.
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
62
|
Pasupuleti V, Vora L, Prasad R, Nandakumar DN, Khatri DK. Glioblastoma preclinical models: Strengths and weaknesses. Biochim Biophys Acta Rev Cancer 2024; 1879:189059. [PMID: 38109948 DOI: 10.1016/j.bbcan.2023.189059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 12/20/2023]
Abstract
Glioblastoma multiforme is a highly malignant brain tumor with significant intra- and intertumoral heterogeneity known for its aggressive nature and poor prognosis. The complex signaling cascade that regulates this heterogeneity makes targeted drug therapy ineffective. The development of an optimal preclinical model is crucial for the comprehension of molecular heterogeneity and enhancing therapeutic efficacy. The ideal model should establish a relationship between various oncogenes and their corresponding responses. This review presents an analysis of preclinical in vivo and in vitro models that have contributed to the advancement of knowledge in model development. The experimental designs utilized in vivo models consisting of both immunodeficient and immunocompetent mice induced with intracranial glioma. The transgenic model was generated using various techniques, like the viral vector delivery system, transposon system, Cre-LoxP model, and CRISPR-Cas9 approaches. The utilization of the patient-derived xenograft model in glioma research is valuable because it closely replicates the human glioma microenvironment, providing evidence of tumor heterogeneity. The utilization of in vitro techniques in the initial stages of research facilitated the comprehension of molecular interactions. However, these techniques are inadequate in reproducing the interactions between cells and extracellular matrix (ECM). As a result, bioengineered 3D-in vitro models, including spheroids, scaffolds, and brain organoids, were developed to cultivate glioma cells in a three-dimensional environment. These models have enabled researchers to understand the influence of ECM on the invasive nature of tumors. Collectively, these preclinical models effectively depict the molecular pathways and facilitate the evaluation of multiple molecules while tailoring drug therapy.
Collapse
Affiliation(s)
- Vasavi Pasupuleti
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Lalitkumar Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK.
| | - Renuka Prasad
- Department of Anatomy, Korea University College of Medicine, Moonsuk Medical Research Building, 516, 5th floor, 73 Inchon-ro, Seongbuk-gu, Seoul 12841, Republic of Korea
| | - D N Nandakumar
- Department of Neurochemistry National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore 560029, India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India.
| |
Collapse
|
63
|
Strauß S, Diemer M, Bucan V, Kuhbier JW, Asendorf T, Vogt PM, Schlottmann F. Spider silk enhanced tissue engineering of cartilage tissue: Approach of a novel bioreactor model using adipose derived stromal cells. J Appl Biomater Funct Mater 2024; 22:22808000241226656. [PMID: 38253568 DOI: 10.1177/22808000241226656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024] Open
Abstract
Human cartilage tissue remains a challenge for the development of therapeutic options due to its poor vascularization and reduced regenerative capacities. There are a variety of research approaches dealing with cartilage tissue engineering. In addition to different biomaterials, numerous cell populations have been investigated in bioreactor-supported experimental setups to improve cartilage tissue engineering. The concept of the present study was to investigate spider silk cocoons as scaffold seeded with adipose-derived stromal cells (ASC) in a custom-made bioreactor model using cyclic axial compression to engineer cartilage-like tissue. For chemical induction of differentiation, BMP-7 and TGF-β2 were added and changes in cell morphology and de-novo tissue formation were investigated using histological staining to verify chondrogenic differentiation. By seeding spider silk cocoons with ASC, a high colonization density and cell proliferation could be achieved. Mechanical induction of differentiation using a newly established bioreactor model led to a more roundish cell phenotype and new extracellular matrix formation, indicating a chondrogenic differentiation. The addition of BMP-7 and TGF-β2 enhanced the expression of cartilage specific markers in immunohistochemical staining. Overall, the present study can be seen as pilot study and valuable complementation to the published literature.
Collapse
Affiliation(s)
- Sarah Strauß
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Maximilian Diemer
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Vesna Bucan
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Jörn W Kuhbier
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
- Department of Plastic, Aesthetic and Hand Surgery, Helios Klinikum Hildesheim, Hildesheim, Germany
| | - Tomke Asendorf
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Peter M Vogt
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Frederik Schlottmann
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
64
|
Kim JH, Lee HJ, Song HJ, Park JB. Impact of 17β-Estradiol on the Shape, Survival, Osteogenic Transformation, and mRNA Expression of Gingiva-Derived Stem Cell Spheroids. MEDICINA (KAUNAS, LITHUANIA) 2023; 60:60. [PMID: 38256321 PMCID: PMC10817649 DOI: 10.3390/medicina60010060] [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: 11/11/2023] [Revised: 12/05/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024]
Abstract
Background and Objectives: Mesenchymal stem cells hold promise for tissue regeneration, given their robust growth and versatile differentiation capabilities. An analysis of bone marrow-sourced mesenchymal stem cell proliferation showed that 17β-estradiol could enhance their growth. This study aims to investigate the influence of 17β-estradiol on the shape, survival, osteogenic differentiation, and mineralization of human mesenchymal stem cells. Materials and Methods: Spheroids made from human gingiva-derived stem cells were cultivated with varying concentrations of 17β-estradiol: 0, 0.01, 0.1, 1, and 10 nM. Morphology was assessed on days 1, 3, and 5. The live/dead kit assay was employed on day 3 for qualitative cell viability, while cell counting kit-8 was used for quantitative viability assessments on days 1, 3, and 5. To evaluate the osteogenic differentiation of the spheroids, a real-time polymerase chain reaction assessed the expressions of RUNX2 and COL1A1 on day 7. Results: The stem cells formed cohesive spheroids, and the inclusion of 17β-estradiol did not noticeably alter their shape. The spheroid diameter remained consistent across concentrations of 0, 0.01, 0.1, 1, and 10 nM of 17β-estradiol. However, cellular viability was boosted with the addition of 1 and 10 nM of 17β-estradiol. The highest expression levels for RUNX2 and COL1A1 were observed with the introduction of 17β-estradiol at 0.1 nM. Conclusions: In conclusion, from the results obtained, it can be inferred that 17β-estradiol can be utilized for differentiating stem cell spheroids. Furthermore, the localized and controlled use, potentially through localized delivery systems or biomaterials, can be an area of active research. While 17β-estradiol holds promise for enhancing stem cell applications, any clinical use requires a thorough understanding of its mechanisms, careful control of its dosage and delivery, and extensive testing to ensure safety and efficacy.
Collapse
Affiliation(s)
- Ju-Hwan Kim
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.-H.K.); (H.-J.L.)
| | - Hyun-Jin Lee
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.-H.K.); (H.-J.L.)
| | - Hye-Jung Song
- Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Jun-Beom Park
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.-H.K.); (H.-J.L.)
- Dental Implantology, Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Medicine, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
65
|
Zhao H, Han R, Wang Z, Xian J, Bai X. Colorectal Cancer Stem Cells and Targeted Agents. Pharmaceutics 2023; 15:2763. [PMID: 38140103 PMCID: PMC10748092 DOI: 10.3390/pharmaceutics15122763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Since their discovery, cancer stem cells have become a hot topic in cancer therapy research. These cells possess stem cell-like self-renewal and differentiation capacities and are important factors that dominate cancer metastasis, therapy-resistance and recurrence. Worse, their inherent characteristics make them difficult to eliminate. Colorectal cancer is the third-most common cancer and the second leading cause of cancer death worldwide. Targeting colorectal cancer stem cells (CR-CSCs) can inhibit colorectal cancer metastasis, enhance therapeutic efficacy and reduce recurrence. Here, we introduced the origin, biomarker proteins, identification, cultivation and research techniques of CR-CSCs, and we summarized the signaling pathways that regulate the stemness of CR-CSCs, such as Wnt, JAK/STAT3, Notch and Hh signaling pathway. In addition to these, we also reviewed recent anti-CR-CSC drugs targeting signaling pathways, biomarkers and other regulators. These will help researchers gain insight into the current agents targeting to CR-CSCs, explore new cancer drugs and propose potential therapies.
Collapse
Affiliation(s)
- Haobin Zhao
- Department of General Practice, People’s Hospital of Longhua, 38 Jinglong Jianshe Road, Shenzhen 518109, China; (H.Z.); (J.X.)
- Endocrinology Department, People’s Hospital of Longhua, 38 Jinglong Jianshe Road, Shenzhen 518109, China
| | - Ruining Han
- Obstetric Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518033, China;
| | - Zhankun Wang
- Emergency Department, People’s Hospital of Longhua, 38 Jinglong Jianshe Road, Shenzhen 518109, China;
| | - Junfang Xian
- Department of General Practice, People’s Hospital of Longhua, 38 Jinglong Jianshe Road, Shenzhen 518109, China; (H.Z.); (J.X.)
| | - Xiaosu Bai
- Endocrinology Department, People’s Hospital of Longhua, 38 Jinglong Jianshe Road, Shenzhen 518109, China
| |
Collapse
|
66
|
Méndez Rodríguez KB, Jiménez Avalos JA, Fernández Macias JC, González Palomo AK. Microplastics: challenges of assessment in biological samples and their implication for in vitro and in vivo effects. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:119733-119749. [PMID: 37971585 DOI: 10.1007/s11356-023-30853-6] [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: 05/26/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
Microplastics (MPs) have attracted global interest because they have been recognized as emerging pollutants that require urgent attention. MPs are plastic particles with a size between 1 micron and 5 mm (1 µm-5mm); those measuring less than 1 µm are known as nanoplastics (NPs). MP is distributed in the environment in various physical forms that depend on the degradation process, the erosion factors to which it was subjected, or the original form in which it was intentionally manufactured. Humans may be exposed to these pollutants mainly by ingestion or inhalation, which could adversely affect human health with effects that are still unknown due to limitations that are often dependent on their analytical determination and lack of studies over time, as it is a relatively new topic. Therefore, this review focuses on the challenges currently faced by laboratories for determining MPs in different matrices. We highlight the application of methods and techniques to assess the precise levels of exposure to MPs in biological samples. In addition, exposure pathways, sources, and evidence of adverse effects reported in vitro and in vivo studies are described to generate knowledge about their potential threat to human health.
Collapse
Affiliation(s)
- Karen Beatriz Méndez Rodríguez
- Coordinación para la Innovación y Aplicación de la Ciencia y Tecnología (CIACYT), Universidad Autónoma de San Luis Potosí (UASLP), San Luis Potosí, San Luis Potosí, México
| | | | - Juan Carlos Fernández Macias
- Coordinación para la Innovación y Aplicación de la Ciencia y Tecnología (CIACYT), Universidad Autónoma de San Luis Potosí (UASLP), San Luis Potosí, San Luis Potosí, México
| | - Ana Karen González Palomo
- Coordinación para la Innovación y Aplicación de la Ciencia y Tecnología (CIACYT), Universidad Autónoma de San Luis Potosí (UASLP), San Luis Potosí, San Luis Potosí, México.
| |
Collapse
|
67
|
Wang E, Andrade MJ, Smith Q. Vascularized liver-on-a-chip model to investigate nicotine-induced dysfunction. BIOMICROFLUIDICS 2023; 17:064108. [PMID: 38155919 PMCID: PMC10754629 DOI: 10.1063/5.0172677] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023]
Abstract
The development of physiologically relevant in vitro systems for simulating disease onset and progression and predicting drug metabolism holds tremendous value in reducing drug discovery time and cost. However, many of these platforms lack accuracy in replicating the tissue architecture and multicellular interactions. By leveraging three-dimensional cell culture, biomimetic soft hydrogels, and engineered stimuli, in vitro models have continued to progress. Nonetheless, the incorporation of the microvasculature has been met with many challenges, specifically with the addition of parenchymal cell types. Here, a systematic approach to investigating the initial seeding density of endothelial cells and its effects on interconnected networks was taken and combined with hepatic spheroids to form a liver-on-a-chip model. Leveraging this system, nicotine's effects on microvasculature and hepatic function were investigated. The findings indicated that nicotine led to interrupted adherens junctions, decreased guanosine triphosphate cyclohydrolase 1 expression, impaired angiogenesis, and lowered barrier function, all key factors in endothelial dysfunction. With the combination of the optimized microvascular networks, a vascularized liver-on-a-chip was formed, providing functional xenobiotic metabolism and synthesis of both albumin and urea. This system provides insight into potential hepatotoxicity caused by various drugs and allows for assessing vascular dysfunction in a high throughput manner.
Collapse
Affiliation(s)
- Eric Wang
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California 92697, USA
| | - Melisa J. Andrade
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California 92697, USA
| | | |
Collapse
|
68
|
Freitas de Morais E, Siquara da Rocha LDO, de Souza Santos JL, Freitas RD, Souza BSDF, Coletta RD, Gurgel Rocha CA. Use of Three-Dimensional Cell Culture Models in Drug Assays for Anti-Cancer Agents in Oral Cancer: Protocol for a Scoping Review. J Pers Med 2023; 13:1618. [PMID: 38003933 PMCID: PMC10672016 DOI: 10.3390/jpm13111618] [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: 09/13/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
Advances in the development of pharmacological treatment in oral cancer require tumor models capable of simulating the complex biology of the tumor microenvironment. The spread of three-dimensional models has changed the scenery of in vitro cell culture techniques, contributing to translational oncology. Still, the full extent of their application in preclinical drug trials is yet to be understood. Therefore, the present scoping review protocol was established to screen the literature on using three-dimensional cell culture models in drug-testing assays in the context of oral cancer. This scoping review will be conducted based on the guidelines established by the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Extension for Scoping Review guidelines (PRISMA-ScR). We will search the PubMed/Medline, Web of Science, Scopus, and Embase databases, as well as the gray literature, including peer-reviewed research articles involving 3D models applied to drug-assessment assays in oral cancer published from 1 March 2013 until 1 March 2023. Data will be charted, and findings will be described according to the predetermined questions of interest. We will present these findings in a narrative manner.
Collapse
Affiliation(s)
- Everton Freitas de Morais
- Department of Oral Diagnosis, School of Dentistry, University of Campinas (UNICAMP), Piracicaba 13414-903, SP, Brazil; (E.F.d.M.); (R.D.C.)
| | - Leonardo de Oliveira Siquara da Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil; (L.d.O.S.d.R.); (J.L.d.S.S.); (B.S.d.F.S.)
- Department of Pathology and Forensic Medicine, School of Medicine, Federal University of Bahia, Salvador 40110-100, BA, Brazil
| | - John Lenon de Souza Santos
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil; (L.d.O.S.d.R.); (J.L.d.S.S.); (B.S.d.F.S.)
- Department of Pathology and Forensic Medicine, School of Medicine, Federal University of Bahia, Salvador 40110-100, BA, Brazil
| | - Raíza Dias Freitas
- Department of Social and Pediatric Dentistry, School of Dentistry, Federal University of Bahia, Salvador 40110-150, BA, Brazil;
| | - Bruno Solano de Freitas Souza
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil; (L.d.O.S.d.R.); (J.L.d.S.S.); (B.S.d.F.S.)
- D’Or Institute for Research and Education (IDOR), Salvador 41253-190, BA, Brazil
| | - Ricardo D. Coletta
- Department of Oral Diagnosis, School of Dentistry, University of Campinas (UNICAMP), Piracicaba 13414-903, SP, Brazil; (E.F.d.M.); (R.D.C.)
- Graduate Program in Oral Biology, School of Dentistry, University of Campinas, Piracicaba 13414-903, SP, Brazil
| | - Clarissa A. Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil; (L.d.O.S.d.R.); (J.L.d.S.S.); (B.S.d.F.S.)
- Department of Pathology and Forensic Medicine, School of Medicine, Federal University of Bahia, Salvador 40110-100, BA, Brazil
- D’Or Institute for Research and Education (IDOR), Salvador 41253-190, BA, Brazil
- Graduate Program in Oral Biology, School of Dentistry, University of Campinas, Piracicaba 13414-903, SP, Brazil
- Department of Propaedeutics, School of Dentistry, Federal University of Bahia, Salvador 40110-150, BA, Brazil
| |
Collapse
|
69
|
Wu J, Kang K, Liu S, Ma Y, Yu M, Zhao X. Recent Progress of In Vitro 3D Culture of Male Germ Stem Cells. J Funct Biomater 2023; 14:543. [PMID: 37998112 PMCID: PMC10672244 DOI: 10.3390/jfb14110543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/23/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023] Open
Abstract
Male germline stem cells (mGSCs), also known as spermatogonial stem cells (SSCs), are the fundamental seed cells of male animal reproductive physiology. However, environmental influences, drugs, and harmful substances often pose challenges to SSCs, such as population reduction and quality decline. With advancements in bioengineering technology and biomaterial technology, an increasing number of novel cell culture methods and techniques have been employed for studying the proliferation and differentiation of SSCs in vitro. This paper provides a review on recent progress in 3D culture techniques for SSCs in vitro; we summarize the microenvironment of SSCs and spermatocyte development, with a focus on scaffold-based culture methods and 3D printing cell culture techniques for SSCs. Additionally, decellularized testicular matrix (DTM) and other biological substrates are utilized through various combinations and approaches to construct an in vitro culture microenvironment suitable for SSC growth. Finally, we present some perspectives on current research trends and potential opportunities within three areas: the 3D printing niche environment, alternative options to DTM utilization, and advancement of the in vitro SSC culture technology system.
Collapse
Affiliation(s)
- Jiang Wu
- Coastal Agricultural College, Guangdong Ocean University, Zhanjiang 524000, China; (J.W.)
| | - Kai Kang
- Coastal Agricultural College, Guangdong Ocean University, Zhanjiang 524000, China; (J.W.)
| | - Siqi Liu
- Coastal Agricultural College, Guangdong Ocean University, Zhanjiang 524000, China; (J.W.)
| | - Yaodan Ma
- Coastal Agricultural College, Guangdong Ocean University, Zhanjiang 524000, China; (J.W.)
| | - Meng Yu
- State Key Laboratory for Mechanical Behavior of Materials, Xi’an Jiaotong University, Xi’an 710049, China
| | - Xin Zhao
- State Key Laboratory for Mechanical Behavior of Materials, Xi’an Jiaotong University, Xi’an 710049, China
| |
Collapse
|
70
|
Lv S, He E, Luo J, Liu Y, Liang W, Xu S, Zhang K, Yang Y, Wang M, Song Y, Wu Y, Cai X. Using Human-Induced Pluripotent Stem Cell Derived Neurons on Microelectrode Arrays to Model Neurological Disease: A Review. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301828. [PMID: 37863819 PMCID: PMC10667858 DOI: 10.1002/advs.202301828] [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: 03/22/2023] [Revised: 09/04/2023] [Indexed: 10/22/2023]
Abstract
In situ physiological signals of in vitro neural disease models are essential for studying pathogenesis and drug screening. Currently, an increasing number of in vitro neural disease models are established using human-induced pluripotent stem cell (hiPSC) derived neurons (hiPSC-DNs) to overcome interspecific gene expression differences. Microelectrode arrays (MEAs) can be readily interfaced with two-dimensional (2D), and more recently, three-dimensional (3D) neural stem cell-derived in vitro models of the human brain to monitor their physiological activity in real time. Therefore, MEAs are emerging and useful tools to model neurological disorders and disease in vitro using human iPSCs. This is enabling a real-time window into neuronal signaling at the network scale from patient derived. This paper provides a comprehensive review of MEA's role in analyzing neural disease models established by hiPSC-DNs. It covers the significance of MEA fabrication, surface structure and modification schemes for hiPSC-DNs culturing and signal detection. Additionally, this review discusses advances in the development and use of MEA technology to study in vitro neural disease models, including epilepsy, autism spectrum developmental disorder (ASD), and others established using hiPSC-DNs. The paper also highlights the application of MEAs combined with hiPSC-DNs in detecting in vitro neurotoxic substances. Finally, the future development and outlook of multifunctional and integrated devices for in vitro medical diagnostics and treatment are discussed.
Collapse
Affiliation(s)
- Shiya Lv
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Enhui He
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
- The State Key Lab of Brain‐Machine IntelligenceZhejiang UniversityHangzhou321100China
| | - Jinping Luo
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yaoyao Liu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wei Liang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Shihong Xu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Kui Zhang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yan Yang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Mixia Wang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yilin Song
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yirong Wu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Xinxia Cai
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
71
|
Farouk SM, Khafaga AF, Abdellatif AM. Bladder cancer: therapeutic challenges and role of 3D cell culture systems in the screening of novel cancer therapeutics. Cancer Cell Int 2023; 23:251. [PMID: 37880676 PMCID: PMC10601189 DOI: 10.1186/s12935-023-03069-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 09/17/2023] [Indexed: 10/27/2023] Open
Abstract
Bladder cancer (BC) is the sixth most common worldwide urologic malignancy associated with elevated morbidity and mortality rates if not well treated. The muscle-invasive form of BC develops in about 25% of patients. Moreover, according to estimates, 50% of patients with invasive BC experience fatal metastatic relapses. Currently, resistance to drug-based therapy is the major tumble to BC treatment. The three-dimensional (3D) cell cultures are clearly more relevant not only as a novel evolving gadget in drug screening but also as a bearable therapeutic for different diseases. In this review, various subtypes of BC and mechanisms of drug resistance to the commonly used anticancer therapies are discussed. We also summarize the key lineaments of the latest cell-based assays utilizing 3D cell culture systems and their impact on understanding the pathophysiology of BC. Such knowledge could ultimately help to address the most efficient BC treatment.
Collapse
Affiliation(s)
- Sameh M Farouk
- Department of Cytology & Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt.
| | - Asmaa F Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina, 22758, Egypt
| | - Ahmed M Abdellatif
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
72
|
Allcock B, Wei W, Goncalves K, Hoyle H, Robert A, Quelch-Cliffe R, Hayward A, Cooper J, Przyborski S. Impact of the Physical Cellular Microenvironment on the Structure and Function of a Model Hepatocyte Cell Line for Drug Toxicity Applications. Cells 2023; 12:2408. [PMID: 37830622 PMCID: PMC10572302 DOI: 10.3390/cells12192408] [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: 08/18/2023] [Revised: 09/26/2023] [Accepted: 10/05/2023] [Indexed: 10/14/2023] Open
Abstract
It is widely recognised that cells respond to their microenvironment, which has implications for cell culture practices. Growth cues provided by 2D cell culture substrates are far removed from native 3D tissue structure in vivo. Geometry is one of many factors that differs between in vitro culture and in vivo cellular environments. Cultured cells are far removed from their native counterparts and lose some of their predictive capability and reliability. In this study, we examine the cellular processes that occur when a cell is cultured on 2D or 3D surfaces for a short period of 8 days prior to its use in functional assays, which we term: "priming". We follow the process of mechanotransduction from cytoskeletal alterations, to changes to nuclear structure, leading to alterations in gene expression, protein expression and improved functional capabilities. In this study, we utilise HepG2 cells as a hepatocyte model cell line, due to their robustness for drug toxicity screening. Here, we demonstrate enhanced functionality and improved drug toxicity profiles that better reflect the in vivo clinical response. However, findings more broadly reflect in vitro cell culture practises across many areas of cell biology, demonstrating the fundamental impact of mechanotransduction in bioengineering and cell biology.
Collapse
Affiliation(s)
- Benjamin Allcock
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (B.A.); (W.W.); (K.G.)
| | - Wenbin Wei
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (B.A.); (W.W.); (K.G.)
| | - Kirsty Goncalves
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (B.A.); (W.W.); (K.G.)
| | - Henry Hoyle
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (B.A.); (W.W.); (K.G.)
| | - Alisha Robert
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (B.A.); (W.W.); (K.G.)
| | - Rebecca Quelch-Cliffe
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (B.A.); (W.W.); (K.G.)
| | - Adam Hayward
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (B.A.); (W.W.); (K.G.)
| | - Jim Cooper
- European Collection of Authenticated Cell Cultures, Salisbury SP4 0JG, UK
| | - Stefan Przyborski
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (B.A.); (W.W.); (K.G.)
- Reprocell Europe Ltd., Glasgow G20 0XA, UK
| |
Collapse
|
73
|
Tang P, Thongrom B, Arora S, Haag R. Polyglycerol-Based Biomedical Matrix for Immunomagnetic Circulating Tumor Cell Isolation and Their Expansion into Tumor Spheroids for Drug Screening. Adv Healthc Mater 2023; 12:e2300842. [PMID: 37402278 PMCID: PMC11469197 DOI: 10.1002/adhm.202300842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/08/2023] [Accepted: 06/29/2023] [Indexed: 07/06/2023]
Abstract
Circulating tumor cells (CTCs) are established as distinct cancer biomarkers for diagnosis, as preclinical models, and therapeutic targets. Their use as preclinical models is limited owing to low purity after isolation and the lack of effective techniques to create 3D cultures that accurately mimic in vivo conditions. Herein, a two-component system for detecting, isolating, and expanding CTCs to generate multicellular tumor spheroids that mimic the physiology and microenvironment of the diseased organ is proposed. First, an antifouling biointerface on magnetic beads is fabricated by adding a bioinert polymer layer and conjugation of biospecific ligands to isolate cancer cells, dramatically enhancing the selectivity and purity of the isolated cancer cells. Next, the isolated cells are encapsulated into self-degradable hydrogels synthesized using a thiol-click approach. The hydrogels are mechanochemically tuned to enable tumor spheroid growth to a size greater than 300 µm and to further release the grown spheroids while retaining their tumor-like characteristics. In addition, drug treatment highlights the need for 3D culture environments rather than conventional 2D culture. The designed biomedical matrix shows potential as a universal method to ensure mimicry of in vivo tumor characteristics in individual patients and to improve the predictability of preclinical screening of personalized therapeutics.
Collapse
Affiliation(s)
- Peng Tang
- Institute for Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Boonya Thongrom
- Institute for Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Smriti Arora
- Institute for Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Rainer Haag
- Institute for Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| |
Collapse
|
74
|
Hodge JG, Robinson JL, Mellott AJ. Mesenchymal Stem Cell Extracellular Vesicles from Tissue-Mimetic System Enhance Epidermal Regeneration via Formation of Migratory Cell Sheets. Tissue Eng Regen Med 2023; 20:993-1013. [PMID: 37515738 PMCID: PMC10519905 DOI: 10.1007/s13770-023-00565-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND The secretome of adipose-derived mesenchymal stem cells (ASCs) offers a unique approach to understanding and treating wounds, including the critical process of epidermal regeneration orchestrated by keratinocytes. However, 2D culture techniques drastically alter the secretory dynamics of ASCs, which has led to ambiguity in understanding which secreted compounds (e.g., growth factors, exosomes, reactive oxygen species) may be driving epithelialization. METHODS A novel tissue-mimetic 3D hydrogel system was utilized to enhance the retainment of a more regenerative ASC phenotype and highlight the functional secretome differences between 2D and 3D. Subsequently, the ASC-secretome was stratified by molecular weight and the presence/absence of extracellular vesicles (EVs). The ASC-secretome fractions were then evaluated to assess for the capacity to augment specific keratinocyte activities. RESULTS Culture of ASCs within the tissue-mimetic system enhanced protein secretion ~ 50%, exclusively coming from the > 100 kDa fraction. The ASC-secretome ability to modulate epithelialization functions, including migration, proliferation, differentiation, and morphology, resided within the "> 100 kDa" fraction, with the 3D ASC-secretome providing the greatest improvement. 3D ASC EV secretion was enhanced two-fold and exhibited dose-dependent effects on epidermal regeneration. Notably, ASC-EVs induced morphological changes in keratinocytes reminiscent of native regeneration, including formation of stratified cell sheets. However, only 3D-EVs promoted collective cell sheet migration and an epithelial-to-mesenchymal-like transition in keratinocytes, whereas 2D-EVs contained an anti-migratory stimulus. CONCLUSION This study demonstrates how critical the culture environment is on influencing ASC-secretome regenerative capabilities. Additionally, the critical role of EVs in modulating epidermal regeneration is revealed and their translatability for future clinical therapies is discussed.
Collapse
Affiliation(s)
- Jacob G Hodge
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA
- Department of Plastic Surgery, University of Kansas Medical Center, 3901 Rainbow Blvd., Mail Stop: 3051, Kansas City, KS, USA
| | - Jennifer L Robinson
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS, USA
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Adam J Mellott
- Department of Plastic Surgery, University of Kansas Medical Center, 3901 Rainbow Blvd., Mail Stop: 3051, Kansas City, KS, USA.
- Ronawk Inc., Olathe, KS, USA.
| |
Collapse
|
75
|
Alsobaie S, Alsobaie T, Alshammary AF, Abudawood M, Mantalaris A. Alginate Beads as a Promising Tool for Successful Production of Viable and Pluripotent Human-Induced Pluripotent Stem Cells in a 3D Culture System. Stem Cells Cloning 2023; 16:61-73. [PMID: 37790697 PMCID: PMC10544263 DOI: 10.2147/sccaa.s409139] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/13/2023] [Indexed: 10/05/2023] Open
Abstract
Purpose Two-dimensional (2D)-based cell culture systems, limited by their inherent heterogeneity and scalability, are a bottleneck in the production of high-quality cells for downstream biomedical applications. Finding the optimal conditions for large-scale stem cell culture while maintaining good cellular status is challenging. The aim of this study was to assess the effects of three-dimensional (3D) culture on the viability, proliferation, self-renewal, and differentiation of human induced pluripotent stem cells (IPSCs). Patients and Methods Various culture conditions were evaluated to determine the optimal conditions to maintain the viability and proliferation of human IPSCs in a 3D environment: static versus dynamic culture, type of adhesion protein added to alginate (Matrigel™ versus gelatin), and the addition of Y-27632t on long-term 3D culture. The proliferation ability of the cells was evaluated via the MTS proliferation assay; the expression levels of the pluripotency markers Nanog and Oct3/4, PAX6 as an ectoderm marker, and laminin-5 and fibronectin as markers of extracellular matrix synthesis were assessed; and HIF1α and HIF2α levels were measured using quantitative reverse transcription polymerase chain reaction. Results Using a high-aspect-ratio vessel bioreactor with a gentle, low-sheer, and low-turbulence environment with sufficient oxygenation and effective mass transfer of nutrients and waste, we verified its ability to promote cell proliferation and self-renewal. The findings showed that human IPSCs have the ability to maintain pluripotency in a feeder-free system and by inhibiting ROCK signaling and using hypoxia to improve single-cell viability in 3D culture. Furthermore, these cells demonstrated increased self-renewal and proliferation when inoculated as single cells in 3D alginate beads by adding RI during the culture period. Conclusion Dynamic 3D culture is desirable for the large-scale expansion of undifferentiated human IPSCs.
Collapse
Affiliation(s)
- Sarah Alsobaie
- Department of Clinical Laboratory Science, King Saud University, Riyadh, Saudi Arabia
| | - Tamador Alsobaie
- Biological Systems Engineering Laboratory, Department of Chemical Engineering, Imperial College London, London, UK
| | - Amal F Alshammary
- Department of Clinical Laboratory Science, King Saud University, Riyadh, Saudi Arabia
| | - Manal Abudawood
- Department of Clinical Laboratory Science, King Saud University, Riyadh, Saudi Arabia
| | - Athanasios Mantalaris
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
76
|
Choi J, Min J, Kim D, Kim J, Kim J, Yoon H, Lee J, Jeong Y, Kim CY, Ko SH. Hierarchical 3D Percolation Network of Ag-Au Core-Shell Nanowire-Hydrogel Composite for Efficient Biohybride Electrodes. ACS NANO 2023; 17:17966-17978. [PMID: 37668160 DOI: 10.1021/acsnano.3c04292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Metal nanomaterials are highly valued for their enhanced surface area and electrochemical properties, which are crucial for energy devices and bioelectronics. However, their practical applications are often limited by challenges, such as scalability and dimensional constraints. In this study, we developed a synthesis method for highly porous Ag-Au core-shell nanowire foam (AACNF) using a one-pot process based on a simultaneous nanowelding synthesis method. The unique characteristics of AACNF as metal-based electrodes show the lowest density among metal-based electrodes while demonstrating high electrical conductivity (99.33-753.04 S/m) and mechanical stability. The AACNF's excellent mass transport properties enable multiscale hierarchical incorporation with functional materials including polymeric precursors and living cells. The enhanced mechanical stability at the nanowelded junctions allows AACNF-hydrogel composites to exhibit large stretching (∼700%) and 10,000 times higher electrical conductivity than hydrogel-nanowire composites without the junction. Large particles in the 1-10 μm scale, including fibroblast cells and exoelectrogenic microbes, are also successfully incorporated with AACNF. AACNF-based microbial fuel cells show high power density (∼330.1 W/m3) within the optimal density range. AACNF's distinctive ability to form a hierarchical structure with substances in various scales showcases its potential for advanced energy devices and biohybrid electrodes in the future.
Collapse
Affiliation(s)
- Joonhwa Choi
- Applied Nano and Thermal Science Lab, Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - JinKi Min
- Applied Nano and Thermal Science Lab, Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Dohyung Kim
- Applied Nano and Thermal Science Lab, Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Jin Kim
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Jinsol Kim
- Applied Nano and Thermal Science Lab, Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Hyeokjun Yoon
- Applied Nano and Thermal Science Lab, Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Jinwoo Lee
- Applied Nano and Thermal Science Lab, Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Youngin Jeong
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Seung Hwan Ko
- Applied Nano and Thermal Science Lab, Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
- Institute of Engineering Research, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, 08826 Korea
| |
Collapse
|
77
|
Gangadaran P, Oh EJ, Rajendran RL, Oh JM, Kim HM, Kwak S, Chung HY, Lee J, Ahn BC, Hong CM. Three-dimensional culture conditioned bone marrow MSC secretome accelerates wound healing in a burn injury mouse model. Biochem Biophys Res Commun 2023; 673:87-95. [PMID: 37364390 DOI: 10.1016/j.bbrc.2023.05.088] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/09/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023]
Abstract
Mesenchymal stem cell (MSC)-based therapy has emerged as a promising regenerative therapeutic approach for wound healing. To determine the effects of cultured MSCs as a 2D monolayer (2D-MSCs) and 3D spheroids (3D-MSCs) on their secretomes, and to examine the effect of 3D-MSC secretomes on endothelial cells (ECs) and MSCs in a burn injury mouse model. MSCs were cultured as 2D monolayers (2D-MSCs) and 3D spheroids (3D-MSCs) and their cellular characteristics were evaluated by western blotting. 2D-MSC and 3D-MSC secretomes (condition medium: CM) were analyzed using an angiogenic array. The activation of ECs by 2D-MSC and 3D-MSC CMs was examined in cellular proliferation, migration, and tube formation assays. The wound healing effects of 2D-MSCs and 3D-MSCs were determined in vivo using a burn injury mouse model. 3D culture conditions altered the markers of components that regulate cell survival, cytoskeletal, adhesion, and proliferation. Interleukin-6 (IL-6), vascular endothelial growth factor A (VEGFA), IL-8, and chemokine (CXC motif) ligand 1 (CXCL1) were present at high levels in the CM of 3D-MSCs compared with 2D-MCs. 3D-MSC-CMs promoted the proliferation, migration, and tube formation of ECs. Furthermore, 3D-MSC treatment enhanced wound healing in a burn injury mouse model. 3D culture improves proangiogenic factors in the MSC secretome and 3D-MSCs represent a new cell-based treatment strategy for wound healing.
Collapse
Affiliation(s)
- Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea; Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea
| | - Eun Jung Oh
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, South Korea
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea
| | - Hyun Mi Kim
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, South Korea
| | - Suin Kwak
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea; Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, South Korea
| | - Ho Yun Chung
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea; Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, South Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, 41944, South Korea
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea; Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, 41944, South Korea.
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, 41944, South Korea.
| |
Collapse
|
78
|
Nkune NW, Abrahamse H. The Efficacy of Zinc Phthalocyanine Nanoconjugate on Melanoma Cells Grown as Three-Dimensional Multicellular Tumour Spheroids. Pharmaceutics 2023; 15:2264. [PMID: 37765232 PMCID: PMC10535874 DOI: 10.3390/pharmaceutics15092264] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Melanoma remains a major public health concern that is highly resistant to standard therapeutic approaches. Photodynamic therapy (PDT) is an underutilised cancer therapy with an increased potency and negligible side effects, and it is non-invasive compared to traditional treatment modalities. Three-dimensional multicellular tumour spheroids (MCTS) closely resemble in vivo avascular tumour features, allowing for the more efficient and precise screening of novel anticancer agents with various treatment combinations. In this study, we utilised A375 human melanoma spheroids to screen the phototoxic effect of zinc phthalocyanine tetrasulfonate (ZnPcS4) conjugated to gold nanoparticles (AuNP). The nanoconjugate was synthesised and characterised using ultraviolet-visible spectroscopy, a high-resolution transmission electron microscope (TEM), dynamic light scattering (DLS), and zeta potential (ZP). The phototoxicity of the nanoconjugate was tested on the A375 MCTS using PDT at a fluency of 10 J/cm2. After 24 h, the cellular responses were evaluated via microscopy, an MTT viability assay, an ATP luminescence assay, and cell death induction using annexin propidium iodide. The MTT viability assay demonstrated that the photoactivated ZnPcS4, at a concentration of 12.73 µM, caused an approximately 50% reduction in the cell viability of the spheroids. When conjugated to AuNPs, the latter significantly increased the cellular uptake and cytotoxicity in the melanoma spheroids via the induction of apoptosis. This novel Zinc Phthalocyanine Nanoconjugate shows promise as a more effective PDT treatment modality.
Collapse
Affiliation(s)
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, P.O. Box 17011, Johannesburg 2028, South Africa;
| |
Collapse
|
79
|
Xue W, Lee D, Kong Y, Kuss M, Huang Y, Kim T, Chung S, Dudley AT, Ro SH, Duan B. A Facile Strategy for the Fabrication of Cell-laden Porous Alginate Hydrogels Based on Two-phase Aqueous Emulsions. ADVANCED FUNCTIONAL MATERIALS 2023; 33:2214129. [PMID: 38131003 PMCID: PMC10732541 DOI: 10.1002/adfm.202214129] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Indexed: 12/23/2023]
Abstract
Porous alginate hydrogels possess many advantages as cell carriers. However, current pore generation methods require either complex or harsh fabrication processes, toxic components, or extra purification steps, limiting the feasibility and affecting the cellular survival and function. In this study, a simple and cell-friendly approach to generate highly porous cell-laden alginate hydrogels based on two-phase aqueous emulsions is reported. The pre-gel solutions, which contain two immiscible aqueous phases of alginate and caseinate, are crosslinked by calcium ions. The porous structure of the hydrogel construct is formed by subsequently removing the caseinate phase from the ion-crosslinked alginate hydrogel. Those porous alginate hydrogels possess heterogeneous pores around 100 μm and interconnected paths. Human white adipose progenitors (WAPs) encapsulated in these hydrogels self-organize into spheroids and show enhanced viability, proliferation, and adipogenic differentiation, compared to non-porous constructs. As a proof of concept, this porous alginate hydrogel platform is employed to prepare core-shell spheres for coculture of WAPs and colon cancer cells, with WAP clusters distributed around cancer cell aggregates, to investigate cellular crosstalk. This efficacious approach is believed to provide a robust and versatile platform for engineering porous-structured alginate hydrogels for applications as cell carriers and in disease modeling.
Collapse
Affiliation(s)
- Wen Xue
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA.; Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Donghee Lee
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA.; Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Yunfan Kong
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA.; Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA.; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Mitchell Kuss
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA.; Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Ying Huang
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Taesung Kim
- Department of Biochemistry and the Redox Biology Center, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, USA
| | - Soonkyu Chung
- Department of Nutrition, University of Massachusetts Amherst, Amherst, Massachusetts 01003, USA
| | - Andrew T Dudley
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA.; Department of Genetics, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Seung-Hyun Ro
- Department of Biochemistry and the Redox Biology Center, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, USA
| | - Bin Duan
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA.; Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA.; Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA.; Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, USA
| |
Collapse
|
80
|
Staedtke V, Topilko P, Le LQ, Grimes K, Largaespada DA, Cagan RL, Steensma MR, Stemmer-Rachamimov A, Blakeley JO, Rhodes SD, Ly I, Romo CG, Lee SY, Serra E. Existing and Developing Preclinical Models for Neurofibromatosis Type 1-Related Cutaneous Neurofibromas. J Invest Dermatol 2023; 143:1378-1387. [PMID: 37330719 PMCID: PMC11246562 DOI: 10.1016/j.jid.2023.01.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 06/19/2023]
Abstract
Neurofibromatosis type 1 (NF1) is caused by a nonfunctional copy of the NF1 tumor suppressor gene that predisposes patients to the development of cutaneous neurofibromas (cNFs), the skin tumor that is the hallmark of this condition. Innumerable benign cNFs, each appearing by an independent somatic inactivation of the remaining functional NF1 allele, form in nearly all patients with NF1. One of the limitations in developing a treatment for cNFs is an incomplete understanding of the underlying pathophysiology and limitations in experimental modeling. Recent advances in preclinical in vitro and in vivo modeling have substantially enhanced our understanding of cNF biology and created unprecedented opportunities for therapeutic discovery. We discuss the current state of cNF preclinical in vitro and in vivo model systems, including two- and three-dimensional cell cultures, organoids, genetically engineered mice, patient-derived xenografts, and porcine models. We highlight the models' relationship to human cNFs and how they can be used to gain insight into cNF development and therapeutic discovery.
Collapse
Affiliation(s)
- Verena Staedtke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Piotr Topilko
- Institut Mondor de Recherche Biomédicale (IMRB), Créteil, France
| | - Lu Q Le
- Department of Dermatology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Kevin Grimes
- SPARK Program in Translational Research, Stanford University School of Medicine, Stanford, California, USA; Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, USA
| | - David A Largaespada
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ross L Cagan
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Matthew R Steensma
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan, USA; Helen DeVos Children's Hospital, Spectrum Health System, Grand Rapids, Michigan, USA; Michigan State University College of Human Medicine, Grand Rapids, Michigan, USA
| | - Anat Stemmer-Rachamimov
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jaishri O Blakeley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Steven D Rhodes
- Division of Hematology-Oncology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ina Ly
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Carlos G Romo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sang Y Lee
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eduard Serra
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
81
|
Zhu Y, Zhang M, Sun Q, Wang X, Li X, Li Q. Advanced Mechanical Testing Technologies at the Cellular Level: The Mechanisms and Application in Tissue Engineering. Polymers (Basel) 2023; 15:3255. [PMID: 37571149 PMCID: PMC10422338 DOI: 10.3390/polym15153255] [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: 07/11/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Mechanics, as a key physical factor which affects cell function and tissue regeneration, is attracting the attention of researchers in the fields of biomaterials, biomechanics, and tissue engineering. The macroscopic mechanical properties of tissue engineering scaffolds have been studied and optimized based on different applications. However, the mechanical properties of the overall scaffold materials are not enough to reveal the mechanical mechanism of the cell-matrix interaction. Hence, the mechanical detection of cell mechanics and cellular-scale microenvironments has become crucial for unraveling the mechanisms which underly cell activities and which are affected by physical factors. This review mainly focuses on the advanced technologies and applications of cell-scale mechanical detection. It summarizes the techniques used in micromechanical performance analysis, including atomic force microscope (AFM), optical tweezer (OT), magnetic tweezer (MT), and traction force microscope (TFM), and analyzes their testing mechanisms. In addition, the application of mechanical testing techniques to cell mechanics and tissue engineering scaffolds, such as hydrogels and porous scaffolds, is summarized and discussed. Finally, it highlights the challenges and prospects of this field. This review is believed to provide valuable insights into micromechanics in tissue engineering.
Collapse
Affiliation(s)
- Yingxuan Zhu
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Mengqi Zhang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qingqing Sun
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaofeng Wang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaomeng Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qian Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
82
|
Banda Sánchez C, Cubo Mateo N, Saldaña L, Valdivieso A, Earl J, González Gómez I, Rodríguez-Lorenzo LM. Selection and Optimization of a Bioink Based on PANC-1- Plasma/Alginate/Methylcellulose for Pancreatic Tumour Modelling. Polymers (Basel) 2023; 15:3196. [PMID: 37571089 PMCID: PMC10421301 DOI: 10.3390/polym15153196] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
3D bioprinting involves using bioinks that combine biological and synthetic materials. The selection of the most appropriate cell-material combination for a specific application is complex, and there is a lack of consensus on the optimal conditions required. Plasma-loaded alginate and alginate/methylcellulose (Alg/MC) inks were chosen to study their viscoelastic behaviour, degree of recovery, gelation kinetics, and cell survival after printing. Selected inks showed a shear thinning behavior from shear rates as low as 0.2 s-1, and the ink composed of 3% w/v SA and 9% w/v MC was the only one showing a successful stacking and 96% recovery capacity. A 0.5 × 106 PANC-1 cell-laden bioink was extruded with an Inkredible 3D printer (Cellink) through a D = 410 μm tip conical nozzle into 6-well culture plates. Cylindrical constructs were printed and crosslinked with CaCl2. Bioinks suffered a 1.845 Pa maximum pressure at the tip that was not deleterious for cellular viability. Cell aggregates can be appreciated for the cut total length observed in confocal microscopy, indicating a good proliferation rate at different heights of the construct, and suggesting the viability of the selected bioink PANC-1/P-Alg3/MC9 for building up three-dimensional bioprinted pancreatic tumor constructs.
Collapse
Affiliation(s)
| | - Nieves Cubo Mateo
- Nebrija Research Group ARIES, Higher Polytechnic School, Antonio de Nebrija University, 28015 Madrid, Spain
- Institute for Physical and Information Technologies (ITEFI-CSIC), Sensors and Ultrasonic Systems, 28006 Madrid, Spain
| | - Laura Saldaña
- IdiPAZ, Hospital Universitario La Paz, 28046 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine, CIBER-BBN, 28029 Madrid, Spain
| | - Alba Valdivieso
- Institute for Physical and Information Technologies (ITEFI-CSIC), Sensors and Ultrasonic Systems, 28006 Madrid, Spain
| | - Julie Earl
- Ramón y Cajal Health Research Institute (IRYCIS), Molecular Epidemiology and Predictive Tumour Markers, 28034 Madrid, Spain
- Biomedical Research Network in Cancer (CIBERONC), 28034 Madrid, Spain
| | - Itziar González Gómez
- Institute for Physical and Information Technologies (ITEFI-CSIC), Sensors and Ultrasonic Systems, 28006 Madrid, Spain
| | | |
Collapse
|
83
|
Liu J, Liu S, Zeng L, Tsilioni I. Amyloid Beta Peptides Lead to Mast Cell Activation in a Novel 3D Hydrogel Model. Int J Mol Sci 2023; 24:12002. [PMID: 37569378 PMCID: PMC10419190 DOI: 10.3390/ijms241512002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disease and the world's primary cause of dementia among the elderly population. The aggregation of toxic amyloid-beta (Aβ) is one of the main pathological hallmarks of the AD brain. Recently, neuroinflammation has been recognized as one of the major features of AD, which involves a network of interactions between immune cells. The mast cell (MC) is an innate immune cell type known to serve as a first responder to pathological changes and crosstalk with microglia and neurons. Although an increased number of mast cells were found near the sites of Aβ deposition, how mast cells are activated in AD is not clear. We developed a 3D culture system to culture MCs and investigated the activation of MCs by Aβ peptides. Because collagen I is the major component of extracellular matrix (ECM) in the brain, we encapsulated human LADR MCs in gels formed by collagen I. We found that 3D-cultured MCs survived and proliferated at the same level as MCs in suspension. Additionally, they can be induced to secrete inflammatory cytokines as well as MC proteases tryptase and chymase by typical MC activators interleukin 33 (IL-33) and IgE/anti-IgE. Culturing with peptides Aβ1-42, Aβ1-40, and Aβ25-35 caused MCs to secrete inflammatory mediators, with Aβ1-42 inducing the maximum level of activation. These data indicate that MCs respond to amyloid deposition to elicit inflammatory responses and demonstrate the validity of collagen gel as a model system to investigate MCs in a 3D environment to understand neuroinflammation in AD.
Collapse
Affiliation(s)
- Jingshu Liu
- Department of Immunology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA; (J.L.)
| | - Sihan Liu
- Department of Immunology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA; (J.L.)
| | - Li Zeng
- Department of Immunology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA; (J.L.)
- Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University, 136 Harrison Avenue, Boston, MA 02111, USA
- Program in Pharmacology, Graduate School of Biomedical Sciences, Tufts University, 136 Harrison Avenue, Boston, MA 02111, USA
- Program in Immunology, Graduate School of Biomedical Sciences, Tufts University, 136 Harrison Avenue, Boston, MA 02111, USA
- Department of Orthopaedics, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Irene Tsilioni
- Department of Immunology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA; (J.L.)
| |
Collapse
|
84
|
Soares BX, Miranda CC, Fernandes TG. Systems bioengineering approaches for developmental toxicology. Comput Struct Biotechnol J 2023; 21:3272-3279. [PMID: 38213895 PMCID: PMC10781881 DOI: 10.1016/j.csbj.2023.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 01/13/2024] Open
Abstract
Developmental toxicology is the field of study that examines the effects of chemical and physical agents on developing organisms. By using principles of systems biology and bioengineering, a systems bioengineering approach could be applied to study the complex interactions between developing organisms, the environment, and toxic agents. This approach would result in a holistic understanding of the effects of toxic agents on organisms, by considering the interactions between different biological systems and the impacts of toxicants on those interactions. It would be useful in identifying key biological pathways and mechanisms affected by toxic agents, as well as in the development of predictive models to assess potential risks of exposure to toxicants during development. In this review, we discuss the relevance of systems bioengineering to the field of developmental toxicity and provide up-to-date examples that illustrate the use of engineering principles for this application.
Collapse
Affiliation(s)
- Beatriz Xavier Soares
- Department of Bioengineering and iBB – Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB – Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Cláudia C. Miranda
- Department of Bioengineering and iBB – Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB – Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- AccelBio, Collaborative Laboratory to Foster Translation and Drug Discovery, Cantanhede, Portugal
| | - Tiago G. Fernandes
- Department of Bioengineering and iBB – Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB – Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
85
|
Marques JROF, González-Alva P, Yu-Tong Lin R, Ferreira Fernandes B, Chaurasia A, Dubey N. Advances in tissue engineering of cancer microenvironment-from three-dimensional culture to three-dimensional printing. SLAS Technol 2023; 28:152-164. [PMID: 37019216 DOI: 10.1016/j.slast.2023.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/27/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023]
Abstract
Cancer treatment development is a complex process, with tumor heterogeneity and inter-patient variations limiting the success of therapeutic intervention. Traditional two-dimensional cell culture has been used to study cancer metabolism, but it fails to capture physiologically relevant cell-cell and cell-environment interactions required to mimic tumor-specific architecture. Over the past three decades, research efforts in the field of 3D cancer model fabrication using tissue engineering have addressed this unmet need. The self-organized and scaffold-based model has shown potential to study the cancer microenvironment and eventually bridge the gap between 2D cell culture and animal models. Recently, three-dimensional (3D) bioprinting has emerged as an exciting and novel biofabrication strategy aimed at developing a 3D compartmentalized hierarchical organization with the precise positioning of biomolecules, including living cells. In this review, we discuss the advancements in 3D culture techniques for the fabrication of cancer models, as well as their benefits and limitations. We also highlight future directions associated with technological advances, detailed applicative research, patient compliance, and regulatory challenges to achieve a successful bed-to-bench transition.
Collapse
Affiliation(s)
- Joana Rita Oliveira Faria Marques
- Oral Biology and Biochemistry Research Group (GIBBO), Unidade de Investigação em Ciências Orais e Biomédicas (UICOB), Faculdade de Medicina Dentária, Universidade de Lisboa, Lisboa, Portugal
| | - Patricia González-Alva
- Tissue Bioengineering Laboratory, Postgraduate Studies and Research Division, Faculty of Dentistry, National Autonomous University of Mexico (UNAM), 04510, Mexico, CDMX, Mexico
| | - Ruby Yu-Tong Lin
- Faculty of Dentistry, National University of Singapore, Singapore
| | - Beatriz Ferreira Fernandes
- Oral Biology and Biochemistry Research Group (GIBBO), Unidade de Investigação em Ciências Orais e Biomédicas (UICOB), Faculdade de Medicina Dentária, Universidade de Lisboa, Lisboa, Portugal
| | - Akhilanand Chaurasia
- Department of Oral Medicine, Faculty of Dental Sciences, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Nileshkumar Dubey
- Faculty of Dentistry, National University of Singapore, Singapore; ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore.
| |
Collapse
|
86
|
Babar Q, Saeed A, Tabish TA, Sarwar M, Thorat ND. Targeting the tumor microenvironment: Potential strategy for cancer therapeutics. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166746. [PMID: 37160171 DOI: 10.1016/j.bbadis.2023.166746] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/11/2023]
Abstract
Cellular and stromal components including tumor cells, immune cells, mesenchymal cells, cancer-linked fibroblasts, and extracellular matrix, constituent tumor microenvironment (TME). TME plays a crucial role in reprogramming tumor initiation, uncontrolled proliferation, invasion and metastasis as well as response to therapeutic modalities. In recent years targeting the TME has developed as a potential strategy for treatment of cancer because of its life-threatening functions in restricting tumor development and modulating responses to standard-of-care medicines. Cold atmospheric plasma, oncolytic viral therapy, bacterial therapy, nano-vaccine, and repurposed pharmaceuticals with combination therapy, antiangiogenic drugs, and immunotherapies are among the most effective therapies directed by TME that have either been clinically authorized or are currently being studied. This article discusses above-mentioned therapies in light of targeting TME. We also cover problems related to the TME-targeted therapies, as well as future insights and practical uses in this rapidly growing field.
Collapse
Affiliation(s)
- Quratulain Babar
- Department of Biochemistry Government College University, Faisalabad, Pakistan
| | - Ayesha Saeed
- Department of Biochemistry Government College University, Faisalabad, Pakistan
| | - Tanveer A Tabish
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Mohsin Sarwar
- Department of Biochemistry University of Management and Technology, Lahore, Pakistan
| | - Nanasaheb D Thorat
- Department of Physics, Bernal Institute, Castletroy, Limerick V94T9PX, Ireland; Nuffield Department of Women's and Reproductive Health, John Radcliffe Hospital, Medical Sciences Division, University of Oxford, Oxford OX3 9DU, United Kingdom; Limerick Digital Cancer Research Centre (LDCRC) University of Limerick, Castletroy, Limerick V94T9PX, Ireland.
| |
Collapse
|
87
|
Fu Z, Zhang Y, Geng X, Chi K, Liu C, Song C, Cai G, Chen X, Hong Q. Optimization strategies of mesenchymal stem cell-based therapy for acute kidney injury. Stem Cell Res Ther 2023; 14:116. [PMID: 37122024 PMCID: PMC10150535 DOI: 10.1186/s13287-023-03351-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 04/20/2023] [Indexed: 05/02/2023] Open
Abstract
Considering the high prevalence and the lack of targeted pharmacological management of acute kidney injury (AKI), the search for new therapeutic approaches for it is in urgent demand. Mesenchymal stem cells (MSCs) have been increasingly recognized as a promising candidate for the treatment of AKI. However, clinical translation of MSCs-based therapies is hindered due to the poor retention and survival rates as well as the impaired paracrine ability of MSCs post-delivery. To address these issues, a series of strategies including local administration, three-dimensional culture, and preconditioning have been applied. Owing to the emergence and development of these novel biotechnologies, the effectiveness of MSCs in experimental AKI models is greatly improved. Here, we summarize the different approaches suggested to optimize the efficacy of MSCs therapy, aiming at promoting the therapeutic effects of MSCs on AKI patients.
Collapse
Affiliation(s)
- Zhangning Fu
- Medical School of Chinese PLA, Beijing, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese PLA, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Yifan Zhang
- Medical School of Chinese PLA, Beijing, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese PLA, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Xiaodong Geng
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese PLA, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
- Beidaihe Rehabilitation and Recuperation Center, Chinese People's Liberation Army Joint Logistics Support Force, Qinhuangdao, China
| | - Kun Chi
- Medical School of Chinese PLA, Beijing, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese PLA, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Chao Liu
- Department of Critical Care Medicine, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chengcheng Song
- Department of Nephrology, Beijing Electric Power Hospital, Beijing, China
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese PLA, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese PLA, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Quan Hong
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese PLA, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China.
| |
Collapse
|
88
|
Bhoopathi P, Mannangatti P, Das SK, Fisher PB, Emdad L. Chemoresistance in pancreatic ductal adenocarcinoma: Overcoming resistance to therapy. Adv Cancer Res 2023; 159:285-341. [PMID: 37268399 DOI: 10.1016/bs.acr.2023.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a prominent cause of cancer deaths worldwide, is a highly aggressive cancer most frequently detected at an advanced stage that limits treatment options to systemic chemotherapy, which has provided only marginal positive clinical outcomes. More than 90% of patients with PDAC die within a year of being diagnosed. PDAC is increasing at a rate of 0.5-1.0% per year, and it is expected to be the second leading cause of cancer-related mortality by 2030. The resistance of tumor cells to chemotherapeutic drugs, which can be innate or acquired, is the primary factor contributing to the ineffectiveness of cancer treatments. Although many PDAC patients initially responds to standard of care (SOC) drugs they soon develop resistance caused partly by the substantial cellular heterogeneity seen in PDAC tissue and the tumor microenvironment (TME), which are considered key factors contributing to resistance to therapy. A deeper understanding of molecular mechanisms involved in PDAC progression and metastasis development, and the interplay of the TME in all these processes is essential to better comprehend the etiology and pathobiology of chemoresistance observed in PDAC. Recent research has recognized new therapeutic targets ushering in the development of innovative combinatorial therapies as well as enhancing our comprehension of several different cell death pathways. These approaches facilitate the lowering of the therapeutic threshold; however, the possibility of subsequent resistance development still remains a key issue and concern. Discoveries, that can target PDAC resistance, either alone or in combination, have the potential to serve as the foundation for future treatments that are effective without posing undue health risks. In this chapter, we discuss potential causes of PDAC chemoresistance and approaches for combating chemoresistance by targeting different pathways and different cellular functions associated with and mediating resistance.
Collapse
Affiliation(s)
- Praveen Bhoopathi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Padmanabhan Mannangatti
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
89
|
Do TD, Pham UT, Nguyen LP, Nguyen TM, Bui CN, Oliver S, Pham P, Tran TQ, Hoang BT, Pham MTH, Pham DTN, Nguyen DT. Fabrication of a Low-Cost Microfluidic Device for High-Throughput Drug Testing on Static and Dynamic Cancer Spheroid Culture Models. Diagnostics (Basel) 2023; 13:diagnostics13081394. [PMID: 37189495 DOI: 10.3390/diagnostics13081394] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/17/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Drug development is a complex and expensive process from new drug discovery to product approval. Most drug screening and testing rely on in vitro 2D cell culture models; however, they generally lack in vivo tissue microarchitecture and physiological functionality. Therefore, many researchers have used engineering methods, such as microfluidic devices, to culture 3D cells in dynamic conditions. In this study, a simple and low-cost microfluidic device was fabricated using Poly Methyl Methacrylate (PMMA), a widely available material, and the total cost of the completed device was USD 17.75. Dynamic and static cell culture examinations were applied to monitor the growth of 3D cells. α-MG-loaded GA liposomes were used as the drug to test cell viability in 3D cancer spheroids. Two cell culture conditions (i.e., static and dynamic) were also used in drug testing to simulate the effect of flow on drug cytotoxicity. Results from all assays showed that with the velocity of 0.005 mL/min, cell viability was significantly impaired to nearly 30% after 72 h in a dynamic culture. This device is expected to improve in vitro testing models, reduce and eliminate unsuitable compounds, and select more accurate combinations for in vivo testing.
Collapse
Affiliation(s)
- Tung Dinh Do
- Saint Paul General Hospital, No. 12, Chu Van An St., Ba Dinh Dist, Ha Noi 10000, Vietnam
| | - Uyen Thu Pham
- Institute for Tropical Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| | - Linh Phuong Nguyen
- School of Preventive Medicine and Public Health, Hanoi Medical University, 1 Ton That Tung St., Dong Da Dist., Hanoi 10000, Vietnam
| | - Trang Minh Nguyen
- Institute for Tropical Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| | - Cuong Nguyen Bui
- Hung Yen University of Technology and Education (UTEHY), 39A St., Khoai Chau Dist., Hung Yen 17000, Vietnam
| | - Susan Oliver
- Centre for Advanced Macromolecular Design and Australian Centre for NanoMedicine, School of Chemical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Phuong Pham
- Centre for Advanced Macromolecular Design and Australian Centre for NanoMedicine, School of Chemical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Toan Quoc Tran
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| | - Bich Thi Hoang
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| | - Minh Thi Hong Pham
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| | - Dung Thuy Nguyen Pham
- Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
- Faculty of Environmental and Food Engineering, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
| | - Duong Thanh Nguyen
- Institute for Tropical Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| |
Collapse
|
90
|
Mitrakas AG, Tsolou A, Didaskalou S, Karkaletsou L, Efstathiou C, Eftalitsidis E, Marmanis K, Koffa M. Applications and Advances of Multicellular Tumor Spheroids: Challenges in Their Development and Analysis. Int J Mol Sci 2023; 24:ijms24086949. [PMID: 37108113 PMCID: PMC10138394 DOI: 10.3390/ijms24086949] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/31/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Biomedical research requires both in vitro and in vivo studies in order to explore disease processes or drug interactions. Foundational investigations have been performed at the cellular level using two-dimensional cultures as the gold-standard method since the early 20th century. However, three-dimensional (3D) cultures have emerged as a new tool for tissue modeling over the last few years, bridging the gap between in vitro and animal model studies. Cancer has been a worldwide challenge for the biomedical community due to its high morbidity and mortality rates. Various methods have been developed to produce multicellular tumor spheroids (MCTSs), including scaffold-free and scaffold-based structures, which usually depend on the demands of the cells used and the related biological question. MCTSs are increasingly utilized in studies involving cancer cell metabolism and cell cycle defects. These studies produce massive amounts of data, which demand elaborate and complex tools for thorough analysis. In this review, we discuss the advantages and disadvantages of several up-to-date methods used to construct MCTSs. In addition, we also present advanced methods for analyzing MCTS features. As MCTSs more closely mimic the in vivo tumor environment, compared to 2D monolayers, they can evolve to be an appealing model for in vitro tumor biology studies.
Collapse
Affiliation(s)
- Achilleas G Mitrakas
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Avgi Tsolou
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Stylianos Didaskalou
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Lito Karkaletsou
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Christos Efstathiou
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Evgenios Eftalitsidis
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Konstantinos Marmanis
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Maria Koffa
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
91
|
Lyu X, Cui F, Zhou H, Cao B, Zhang X, Cai M, Yang S, Sun B, Li G. 3D co-culture of macrophages and fibroblasts in a sessile drop array for unveiling the role of macrophages in skin wound-healing. Biosens Bioelectron 2023; 225:115111. [PMID: 36731395 DOI: 10.1016/j.bios.2023.115111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/30/2023]
Abstract
Three-dimensional (3D) heterotypic multicellular spheroid models play important roles in researches of the proliferation and remodeling phases in wound healing. This study aimed to develop a sessile drop array to cultivate 3D spheroids and simulate wound healing stage in vitro using NIH-3T3 fibroblasts and M2-type macrophages. By the aid of the offset of surface tension and gravity, the sessile drop array is able to transfer cell suspensions to spheroids via the superhydrophobic surface of each microwell. Meanwhile, each microwell has a cylinder hole at its bottom that provides adequate oxygen to the spheroid. It demonstrated that the NIH-3T3 fibroblast spheroid and the 3T3 fibroblast/M2-type macrophage heterotypic multicellular spheroid can form and maintain physiological activities within nine days. In order to further investigate the structure without destroying the entire spheroid, we reconstructed its 3D morphology using transparent processing technology and the Z-stack function of confocal microscopy. Additionally, a nano antibody-based 3D immunostaining assay was used to analyze the proliferation and differentiation characteristics of these cells. It found that M2-type macrophages were capable of promoting the differentiation of 3T3 fibroblast spheroid. In this study, a novel, inexpensive platform was constructed for developing spheroids, as well as a 3D immunofluorescence method for investigating the macrophage-associated wound healing microenvironment.
Collapse
Affiliation(s)
- Xiaoyan Lyu
- Department of Dermatology, Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Feiyun Cui
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Hang Zhou
- The Ministry of Education Key Laboratory of Clinical Diagnostics, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Bo Cao
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Xiaolan Zhang
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Minghui Cai
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Shulong Yang
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Bangyong Sun
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education, Defense Key Disciplines Lab of Novel Micro-Nano Devices and System Technology, Chongqing University, Chongqing, 400044, China
| | - Gang Li
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education, Defense Key Disciplines Lab of Novel Micro-Nano Devices and System Technology, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
92
|
Susceptibility of Ovine Bone Marrow-Derived Mesenchymal Stem Cell Spheroids to Scrapie Prion Infection. Animals (Basel) 2023; 13:ani13061043. [PMID: 36978584 PMCID: PMC10044354 DOI: 10.3390/ani13061043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/16/2023] [Accepted: 03/09/2023] [Indexed: 03/15/2023] Open
Abstract
In neurodegenerative diseases, including prion diseases, cellular in vitro models appear as fundamental tools for the study of pathogenic mechanisms and potential therapeutic compounds. Two-dimensional (2D) monolayer cell culture systems are the most used cell-based assays, but these platforms are not able to reproduce the microenvironment of in vivo cells. This limitation can be surpassed using three-dimensional (3D) culture systems such as spheroids that more effectively mimic in vivo cell interactions. Herein, we evaluated the effect of scrapie prion infection in monolayer-cultured ovine bone marrow-derived mesenchymal stem cells (oBM-MSCs) and oBM-MSC-derived spheroids in growth and neurogenic conditions, analyzing their cell viability and their ability to maintain prion infection. An MTT assay was performed in oBM-MSCs and spheroids subjected to three conditions: inoculated with brain homogenate from scrapie-infected sheep, inoculated with brain homogenate from healthy sheep, and non-inoculated controls. The 3D conditions improved the cell viability in most cases, although in scrapie-infected spheroids in growth conditions, a decrease in cell viability was observed. The levels of pathological prion protein (PrPSc) in scrapie-infected oBM-MSCs and spheroids were measured by ELISA. In neurogenic conditions, monolayer cells and spheroids maintained the levels of PrPSc over time. In growth conditions, however, oBM-MSCs showed decreasing levels of PrPSc throughout time, whereas spheroids were able to maintain stable PrPSc levels. The presence of PrPSc in spheroids was also confirmed by immunocytochemistry. Altogether, these results show that a 3D culture microenvironment improves the permissiveness of oBM-MSCs to scrapie infection in growth conditions and maintains the infection ability in neurogenic conditions, making this model of potential use for prion studies.
Collapse
|
93
|
Abdolahzadeh H, Rad NK, Shpichka A, Golroo R, Rahi K, Timashev P, Hassan M, Vosough M. Progress and promise of cell sheet assisted cardiac tissue engineering in regenerative medicine. Biomed Mater 2023; 18. [PMID: 36758240 DOI: 10.1088/1748-605x/acbad4] [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: 09/26/2022] [Accepted: 02/09/2023] [Indexed: 02/11/2023]
Abstract
Cardiovascular diseases (CVDs) are the most common leading causes of premature deaths in all countries. To control the harmful side effects of CVDs on public health, it is necessary to understand the current and prospective strategies in prevention, management, and monitoring CVDs.In vitro,recapitulating of cardiac complex structure with its various cell types is a challenging topic in tissue engineering. Cardiac tissue engineering (CTE) is a multi-disciplinary strategy that has been considered as a novel alternative approach for cardiac regenerative medicine and replacement therapies. In this review, we overview various cell types and approaches in cardiac regenerative medicine. Then, the applications of cell-sheet-assisted CTE in cardiac diseases were discussed. Finally, we described how this technology can improve cardiac regeneration and function in preclinical and clinical models.
Collapse
Affiliation(s)
- Hadis Abdolahzadeh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Khoshdel Rad
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Anastasia Shpichka
- World-Class Research Center 'Digital Biodesign and Personalized Healthcare', Sechenov University, Moscow, Russia.,Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.,Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Reihaneh Golroo
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Kosar Rahi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Peter Timashev
- World-Class Research Center 'Digital Biodesign and Personalized Healthcare', Sechenov University, Moscow, Russia.,Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.,Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Chemistry Department, Lomonosov Moscow State University, Moscow, Russia.,Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
94
|
Prévôt ME, Ustunel S, Freychet G, Webb CR, Zhernenkov M, Pindak R, Clements RJ, Hegmann E. Physical Models from Physical Templates Using Biocompatible Liquid Crystal Elastomers as Morphologically Programmable Inks For 3D Printing. Macromol Biosci 2023; 23:e2200343. [PMID: 36415071 DOI: 10.1002/mabi.202200343] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/17/2022] [Indexed: 11/24/2022]
Abstract
Advanced manufacturing has received considerable attention as a tool for the fabrication of cell scaffolds however, finding ideal biocompatible and biodegradable materials that fit the correct parameters for 3D printing and guide cells to align remain a challenge. Herein, a photocrosslinkable smectic-A (Sm-A) liquid crystal elastomer (LCE) designed for 3D printing is presented, that promotes cell proliferation but most importantly induces cell anisotropy. The LCE-based bio-ink allows the 3D duplication of a highly complex brain structure generated from an animal model. Vascular tissue models are generated from fluorescently stained mouse tissue spatially imaged using confocal microscopy and subsequently processed to create a digital 3D model suitable for printing. The 3D structure is reproduced using a Digital Light Processing (DLP) stereolithography (SLA) desktop 3D printer. Synchrotron Small-Angle X-ray Diffraction (SAXD) data reveal a strong alignment of the LCE layering within the struts of the printed 3D scaffold. The resultant anisotropy of the LCE struts is then shown to direct cell growth. This study offers a simple approach to produce model tissues built within hours that promote cellular alignment.
Collapse
Affiliation(s)
- Marianne E Prévôt
- Advanced Materials and Liquid Crystal Institute, Kent State University, Kent, OH, 44242, USA
| | - Senay Ustunel
- Advanced Materials and Liquid Crystal Institute, Kent State University, Kent, OH, 44242, USA.,Materials Science Graduate Program, Kent State University, Kent, OH, 44242, USA
| | - Guillaume Freychet
- Brookhaven National Laboratory, National Synchrotron Light Source-II, Upton, NY, 11973, USA
| | - Caitlyn R Webb
- Advanced Materials and Liquid Crystal Institute, Kent State University, Kent, OH, 44242, USA.,Department of Biological Sciences, Kent State University, Kent, OH, 44242, USA
| | - Mikhail Zhernenkov
- Brookhaven National Laboratory, National Synchrotron Light Source-II, Upton, NY, 11973, USA
| | - Ron Pindak
- Brookhaven National Laboratory, National Synchrotron Light Source-II, Upton, NY, 11973, USA
| | - Robert J Clements
- Advanced Materials and Liquid Crystal Institute, Kent State University, Kent, OH, 44242, USA.,Department of Biological Sciences, Kent State University, Kent, OH, 44242, USA.,Biomedical Sciences Program, Kent State University, Kent, OH, 44242, USA
| | - Elda Hegmann
- Advanced Materials and Liquid Crystal Institute, Kent State University, Kent, OH, 44242, USA.,Materials Science Graduate Program, Kent State University, Kent, OH, 44242, USA.,Department of Biological Sciences, Kent State University, Kent, OH, 44242, USA.,Biomedical Sciences Program, Kent State University, Kent, OH, 44242, USA.,Brain Health Research Institute, Kent State University, Kent, OH, 44242, USA
| |
Collapse
|
95
|
Wang Y, Liu M, Zhang Y, Liu H, Han L. Recent methods of droplet microfluidics and their applications in spheroids and organoids. LAB ON A CHIP 2023; 23:1080-1096. [PMID: 36628972 DOI: 10.1039/d2lc00493c] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Droplet microfluidic techniques have long been known as a high-throughput approach for cell manipulation. The capacity to compartmentalize cells into picolitre droplets in microfluidic devices has opened up a range of new ways to extract information from cells. Spheroids and organoids are crucial in vitro three-dimensional cell culture models that physiologically mimic natural tissues and organs. With the aid of developments in cell biology and materials science, droplet microfluidics has been applied to construct spheroids and organoids in numerous formats. In this article, we divide droplet microfluidic approaches for managing spheroids and organoids into three categories based on the droplet module format: liquid droplet, microparticle, and microcapsule. We discuss current advances in the use of droplet microfluidics for the generation of tumour spheroids, stem cell spheroids, and organoids, as well as the downstream applications of these methods in high-throughput screening and tissue engineering.
Collapse
Affiliation(s)
- Yihe Wang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237 P. R. China.
| | - Mengqi Liu
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237 P. R. China.
| | - Yu Zhang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237 P. R. China.
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100 P. R. China.
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237 P. R. China.
- Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, 250100 P. R. China
| |
Collapse
|
96
|
Tahara S, de Faria FCC, Sarchet P, Calore F, Sharick J, Leight JL, Casadei L, Pollock RE. Three dimensional models of dedifferentiated liposarcoma cell lines: scaffold-based and scaffold-free approaches. Hum Cell 2023; 36:1081-1089. [PMID: 36763259 DOI: 10.1007/s13577-023-00865-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/17/2023] [Indexed: 02/11/2023]
Abstract
Sarcomas are rare malignancies, the number of reports is limited, and this rarity makes further research difficult even though liposarcoma is one of major sarcomas. 2D cell culture remains an important role in establishing basic tumor biology research, but its various shortcomings and limitations are still of concern, and it is now well-accepted that the behavior of 3D-cultured cells is more reflective of in vivo cellular responses compared to 2D models. This study aimed to establish 3D cell culture of liposarcomas using two different methods: scaffold-based (Matrigel extracellular matrix [ECM] scaffold method) and scaffold-free (Ultra-low attachment [ULA] plate). Lipo246, Lipo224 and Lipo863 cell lines were cultured, and distinctive differences in structures were observed in Matrigel 3D model: Lipo224 and Lipo863 formed spheroids, whereas Lipo246 grew radially without forming spheres. In ULA plate approaches, all cell lines formed spheroids, but Lipo224 and Lipo863 spheroids showed bigger size and looser aggregation than Lipo246. Formalin fixed, paraffin embedded (FFPE) blocks were obtained from all 3D models, confirming the spheroid structures. The expression of MDM2, Ki-67 positivity and MDM2 amplification were confirmed by IHC and DNAscope™, respectively. Protein and DNA were extracted from all samples and MDM2 upregulation was confirmed by western blot and qPCR analysis. After treatment with MDM2 inhibitor SAR405838, DDLPS spheroids demonstrated different sensitivity patterns from 2D models. Taken together, we believed that 3D models would have a possibility to provide us a new predictability of efficacy and toxicity, and considered as one important process in in vitro pre-clinical phase prior to moving forward to clinical trials.
Collapse
Affiliation(s)
- Sayumi Tahara
- Comprehensive Cancer Center, The Ohio State University, 460 West 10th Avenue. Suite D920E, Columbus, OH, 43210, USA
| | - Fernanda Costas C de Faria
- Comprehensive Cancer Center, The Ohio State University, 460 West 10th Avenue. Suite D920E, Columbus, OH, 43210, USA
| | - Patricia Sarchet
- Comprehensive Cancer Center, The Ohio State University, 460 West 10th Avenue. Suite D920E, Columbus, OH, 43210, USA
| | - Federica Calore
- Comprehensive Cancer Center, The Ohio State University, 460 West 10th Avenue. Suite D920E, Columbus, OH, 43210, USA
| | - Joe Sharick
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Jennifer L Leight
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Lucia Casadei
- Comprehensive Cancer Center, The Ohio State University, 460 West 10th Avenue. Suite D920E, Columbus, OH, 43210, USA
| | - Raphael E Pollock
- Comprehensive Cancer Center, The Ohio State University, 460 West 10th Avenue. Suite D920E, Columbus, OH, 43210, USA.
| |
Collapse
|
97
|
Nikolits I, Radwan S, Liebner F, Dietrich W, Egger D, Chariyev-Prinz F, Kasper C. Hydrogels from TEMPO-Oxidized Nanofibrillated Cellulose Support In Vitro Cultivation of Encapsulated Human Mesenchymal Stem Cells. ACS APPLIED BIO MATERIALS 2023; 6:543-551. [PMID: 36745634 PMCID: PMC9945099 DOI: 10.1021/acsabm.2c00854] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs) are the most prominent type of adult stem cells for clinical applications. Three-dimensional (3D) cultivation of MSCs in biomimetic hydrogels provides a more physiologically relevant cultivation microenvironment for in vitro testing and modeling, thus overcoming the limitations of traditional planar cultivation methods. Cellulose nanofibers are an excellent candidate biomaterial for synthesis of hydrogels for this application, due to their biocompatibility, tunable properties, availability, and low cost. Herein, we demonstrate the capacity of hydrogels prepared from 2,2,6,6-tetramethylpiperidine-1-oxyl -oxidized and subsequently individualized cellulose-nanofibrils to support physiologically relevant 3D in vitro cultivation of human MSCs at low solid contents (0.2-0.5 wt %). Our results show that MSCs can spread, proliferate, and migrate inside the cellulose hydrogels, while the metabolic activity and proliferative capacity of the cells as well as their morphological characteristics benefit more in the lower bulk cellulose concentration hydrogels.
Collapse
Affiliation(s)
- Ilias Nikolits
- Institute
of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences
BOKU Vienna, Muthgasse 18, 1190 Vienna, Austria
| | - Sara Radwan
- Department
of Life Science Engineering, University
of Applied Sciences Technikum Vienna, Höchstädtplatz 6, 1200 Vienna, Austria
| | - Falk Liebner
- Institute
of Chemistry of Renewable Resources, Department of Chemistry, University of Natural Resources and Life Sciences
BOKU Vienna, Konrad Lorenz Straße 24, 3430 Tulln, Austria
| | - Wolf Dietrich
- Department
of Gynecology and Obstetrics, Karl Landsteiner
University of Health Sciences, Alter Ziegelweg 10, 3430 Tulln, Austria
| | - Dominik Egger
- Institute
of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences
BOKU Vienna, Muthgasse 18, 1190 Vienna, Austria
| | - Farhad Chariyev-Prinz
- Institute
of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences
BOKU Vienna, Muthgasse 18, 1190 Vienna, Austria
| | - Cornelia Kasper
- Institute
of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences
BOKU Vienna, Muthgasse 18, 1190 Vienna, Austria,
| |
Collapse
|
98
|
Tundisi LL, Ataide JA, Costa JSR, Coêlho DDF, Liszbinski RB, Lopes AM, Oliveira-Nascimento L, de Jesus MB, Jozala AF, Ehrhardt C, Mazzola PG. Nanotechnology as a tool to overcome macromolecules delivery issues. Colloids Surf B Biointerfaces 2023; 222:113043. [PMID: 36455361 DOI: 10.1016/j.colsurfb.2022.113043] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/09/2022] [Accepted: 11/18/2022] [Indexed: 11/21/2022]
Abstract
Nanocarriers can deliver drugs to specific organs or cells, potentially bridging the gap between a drug's function and its interaction with biological systems such as human physiology. The untapped potential of nanotechnology stems from its ability to manipulate materials, allowing control over physical and chemical properties and overcoming drug-related problems, e.g., poor solubility or poor bioavailability. For example, most protein drugs are administered parenterally, each with challenges and peculiarities. Some problems faced by bioengineered macromolecule drugs leading to poor bioavailability are short biological half-life, large size and high molecular weight, low permeability through biological membranes, and structural instability. Nanotechnology emerges as a promising strategy to overcome these problems. Nevertheless, the delivery system should be carefully chosen considering loading efficiency, physicochemical properties, production conditions, toxicity, and regulations. Moving from the bench to the bedside is still one of the major bottlenecks in nanomedicine, and toxicological issues are the greatest challenges to overcome. This review provides an overview of biotech drug delivery approaches, associated nanotechnology novelty, toxicological issues, and regulations.
Collapse
Affiliation(s)
| | - Janaína Artem Ataide
- Faculty of Pharmaceutical Sciences, University of Campinas (Unicamp), Campinas, Brazil.
| | - Juliana Souza Ribeiro Costa
- Faculty of Pharmaceutical Sciences, University of Campinas (Unicamp), Campinas, Brazil; Laboratory of Pharmaceutical Technology (Latef), Faculty of Pharmaceutical Sciences, University of Campinas (Unicamp), Campinas, Brazil
| | | | - Raquel Bester Liszbinski
- Nano-Cell Interactions Lab., Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (Unicamp), Campinas, Brazil
| | - André Moreni Lopes
- Faculty of Pharmaceutical Sciences, University of Campinas (Unicamp), Campinas, Brazil
| | - Laura Oliveira-Nascimento
- Faculty of Pharmaceutical Sciences, University of Campinas (Unicamp), Campinas, Brazil; Laboratory of Pharmaceutical Technology (Latef), Faculty of Pharmaceutical Sciences, University of Campinas (Unicamp), Campinas, Brazil
| | - Marcelo Bispo de Jesus
- Nano-Cell Interactions Lab., Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (Unicamp), Campinas, Brazil
| | - Angela Faustino Jozala
- LAMINFE - Laboratory of Industrial Microbiology and Fermentation Process, University of Sorocaba, Sorocaba, Brazil
| | - Carsten Ehrhardt
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute Trinity College Dublin, Dublin, Ireland
| | - Priscila Gava Mazzola
- Faculty of Pharmaceutical Sciences, University of Campinas (Unicamp), Campinas, Brazil
| |
Collapse
|
99
|
Anti-Hypoxia Nanoplatforms for Enhanced Photosensitizer Uptake and Photodynamic Therapy Effects in Cancer Cells. Int J Mol Sci 2023; 24:ijms24032656. [PMID: 36768975 PMCID: PMC9916860 DOI: 10.3390/ijms24032656] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/18/2023] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
Photodynamic therapy (PDT) holds great promise in cancer eradication due to its target selectivity, non-invasiveness, and low systemic toxicity. However, due to the hypoxic nature of many native tumors, PDT is frequently limited in its therapeutic effect. Additionally, oxygen consumption during PDT may exacerbate the tumor's hypoxic condition, which stimulates tumor proliferation, metastasis, and invasion, resulting in poor treatment outcomes. Therefore, various strategies have been developed to combat hypoxia in PDT, such as oxygen carriers, reactive oxygen supplements, and the modulation of tumor microenvironments. However, most PDT-related studies are still conducted on two-dimensional (2D) cell cultures, which fail to accurately reflect tissue complexity. Thus, three-dimensional (3D) cell cultures are ideal models for drug screening, disease simulation and targeted cancer therapy, since they accurately replicate the tumor tissue architecture and microenvironment. This review summarizes recent advances in the development of strategies to overcome tumor hypoxia for enhanced PDT efficiency, with a particular focus on nanoparticle-based photosensitizer (PS) delivery systems, as well as the advantages of 3D cell cultures.
Collapse
|
100
|
Gong S, Shahriyari L. A Review of Stochastic and Deterministic Modeling of Stem Cell Dynamics. CURRENT STEM CELL REPORTS 2023. [DOI: 10.1007/s40778-023-00225-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|