1
|
Huang Y, Ye Q, Wang J, Zhu K, Yang H, Jiang X, Shen M. Recent progress in the identification and in vitro culture of skin organoids. Regen Ther 2025; 29:341-351. [PMID: 40242086 PMCID: PMC12000699 DOI: 10.1016/j.reth.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/13/2024] [Accepted: 01/04/2025] [Indexed: 04/18/2025] Open
Abstract
An organoid is a cell-based structure that shows organ-specific properties and shares a similar spatial organization as the corresponding organ. Organoids possess powerful capability to reproduce the key functions of the associated organ structures, and their similarity to the organs makes them physiologically relevant systems. The primary challenge associated with the development of skin organoids is the complexity of the human skin architecture, which encompasses the epidermis and the dermis as well as accessory structures, including hair follicles, sweat glands, and sebaceous glands, that perform various functions such as thermoregulation. The ultimate objectives of developing skin organoids are to regenerate the complete skin structure in vitro and reconstruct the skin in vivo. Consequently, safety, reliability, and the fidelity of the tissue interfaces are key considerations in this process. For this purpose, the present article reviews the most recent advances in this field, focusing on the cell sources, culture methods, culture conditions, and biomarkers for identifying the structure and function of skin organoids developed in vitro or in vivo. The subsequent sections summarize the recent applications of skin organoids in related disease diagnosis and treatments, and discuss the future prospects of these organoids in terms of clinical applications. This review of skin organoids can provide an important foundation for studies on human skin development, disease modeling, and reconstructive surgery, with broad utility for promising future opportunities in both biomedical research and clinical practice.
Collapse
Affiliation(s)
- Yanan Huang
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | - Qing Ye
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | | | - Kaimin Zhu
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | - Haojie Yang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, China
| | - Xiaoping Jiang
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | - Meihua Shen
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| |
Collapse
|
2
|
Xia X, Hu M, Zhou W, Jin Y, Yao X. Engineering cardiology with miniature hearts. Mater Today Bio 2025; 31:101505. [PMID: 39911371 PMCID: PMC11795835 DOI: 10.1016/j.mtbio.2025.101505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/28/2024] [Accepted: 01/18/2025] [Indexed: 02/07/2025] Open
Abstract
Cardiac organoids offer sophisticated 3D structures that emulate key aspects of human heart development and function. This review traces the evolution of cardiac organoid technology, from early stem cell differentiation protocols to advanced bioengineering approaches. We discuss the methodologies for creating cardiac organoids, including self-organization techniques, biomaterial-based scaffolds, 3D bioprinting, and organ-on-chip platforms, which have significantly enhanced the structural complexity and physiological relevance of in vitro cardiac models. We examine their applications in fundamental research and medical innovations, highlighting their potential to transform our understanding of cardiac biology and pathology. The integration of multiple cell types, vascularization strategies, and maturation protocols has led to more faithful representations of the adult human heart. However, challenges remain in achieving full functional maturity and scalability. We critically assess the current limitations and outline future directions for advancing cardiac organoid technology. By providing a comprehensive analysis of the field, this review aims to catalyze further innovation in cardiac tissue engineering and facilitate its translation to clinical applications.
Collapse
Affiliation(s)
- Xiaojun Xia
- Department of Cardiology, Center of Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Miner Hu
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310000, China
| | - Wenyan Zhou
- School of Medicine, Taizhou University, Taizhou, Zhejiang, 318000, China
| | - Yunpeng Jin
- Department of Cardiology, Center of Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xudong Yao
- Department of Cardiology, Center of Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| |
Collapse
|
3
|
Yang J, Li W, Zhang Z, Xu Z, Zhu W, Wang J, Wang W. Development and Applications of Organoids in Gynecological Diseases. Stem Cell Rev Rep 2025; 21:629-644. [PMID: 39666266 PMCID: PMC11965162 DOI: 10.1007/s12015-024-10833-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Organoids are rapidly self-organizing 3D in vitro cultures derived from pluripotent stem cells (PSCs) or adult stem cells (ASCs) that possess disease-like characteristics with high success rates. Due to their ability to retain tissue structure, biological phenotypes, and genetic information, they have been utilized as a novel in vitro model for disease research. In recent years, scientists have established self-organizing 3D organoids for human endometrium, fallopian tubes, ovaries, and cervix by culturing stem cells with cytokines in 3D scaffolds. The integration of organoids with animal models, organ-on-a-chip systems, and 3D printing technologies offers a novel preclinical model for exploring disease mechanisms and developing treatments. This review elaborate on the recent research progress of stem cells-formed organoids in the field of gynecology from the aspects of constructing gynecological disease organoids, drug screening and new drug development, simulation modeling, allogeneic transplantation, regenerative medicine and personalized treatment."
Collapse
Affiliation(s)
- Jian Yang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wenwen Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zihan Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhonglei Xu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wenjing Zhu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Anhui Women and Children's Medical Center, Hefei, Anhui, China
| | - Wenyan Wang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
4
|
Zhao KY, Du YX, Cao HM, Su LY, Su XL, Li X. The biological macromolecules constructed Matrigel for cultured organoids in biomedical and tissue engineering. Colloids Surf B Biointerfaces 2025; 247:114435. [PMID: 39647422 DOI: 10.1016/j.colsurfb.2024.114435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 12/10/2024]
Abstract
Matrigel is the most commonly used matrix for 3D organoid cultures. Research on the biomaterial basis of Matrigel for organoid cultures is a highly challenging field. Currently, many studies focus on Matrigel-based biological macromolecules or combinations to construct natural Matrigel and synthetic hydrogel scaffolds based on collagen, peptides, polysaccharides, microbial transglutaminase, DNA supramolecules, and polymers for organoid culture. In this review, we discuss the limitations of both natural and synthetic Matrigel, and describe alternative scaffolds that have been employed for organoid cultures. The patient-derived organoids were constructed in different cancer types and limitations of animal-derived organoids based on the hydrogel or Matrigel. The constructed techniques utilizing 3D bioprinting platforms, air-liquid interface (ALI) culture, microfluidic culture, and organ-on-a-chip platform are summarized. Given the potential of organoids for a wide range of therapeutic, tissue engineering and pharmaceutical applications, it is indeed imperative to develop defined and customized hydrogels in addition to Matrigel.
Collapse
Affiliation(s)
- Ke-Yu Zhao
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China; Key Laboratory of Medical Cell Biology in Inner Mongolia, Inner Mongolia Bioactive Peptide Engineering Laboratory, 1 North Tongdao Street, Hohhot, Inner Mongolia 010050, China
| | - Yi-Xiang Du
- Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China
| | - Hui-Min Cao
- Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China
| | - Li-Ya Su
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China
| | - Xiu-Lan Su
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China; Key Laboratory of Medical Cell Biology in Inner Mongolia, Inner Mongolia Bioactive Peptide Engineering Laboratory, 1 North Tongdao Street, Hohhot, Inner Mongolia 010050, China
| | - Xian Li
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China; Key Laboratory of Medical Cell Biology in Inner Mongolia, Inner Mongolia Bioactive Peptide Engineering Laboratory, 1 North Tongdao Street, Hohhot, Inner Mongolia 010050, China.
| |
Collapse
|
5
|
Chu X, Zhou Z, Qian X, Shen H, Cheng H, Zhang J. Functional regeneration strategies of hair follicles: advances and challenges. Stem Cell Res Ther 2025; 16:77. [PMID: 39985119 PMCID: PMC11846195 DOI: 10.1186/s13287-025-04210-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 01/29/2025] [Indexed: 02/24/2025] Open
Abstract
Hair follicles are essential appendages of human skin that function in protection, sensation, thermoregulation and social interactions. The multicellular components, particularly the dermal papilla, matrix and bulge housing stem cells, enable cyclic hair growth postnatally. However, miniaturization and loss of hair follicles can occur in the context of ageing, trauma and various alopecia-related diseases. Conventional treatments involve the redistribution of existing follicles, which may not be viable in patients lacking follicular resources. Recent progress in the comprehension of morphogenesis and the development of biomaterials has significantly advanced follicle reconstruction, incorporating organ germ assembling, stem cell induction and bioprinting techniques. Despite these advancements, fully restoring hair follicles remains challenging due to the complexities of replicating embryonic signals and sustaining growth cycles. Identifying suitable cell sources for clinical applications also presents a hurdle. Here, we retrospect the progress made in the field of hair follicle regeneration, aiming to offer an exhaustive analysis on the benefits and limitations of these methods, and to foster the development of innovative solutions.
Collapse
Affiliation(s)
- Xi Chu
- Department of Plastic and Cosmetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 261 Huansha Road, Hangzhou, 310000, Zhejiang, China
| | - Zhentao Zhou
- Department of Plastic and Cosmetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 261 Huansha Road, Hangzhou, 310000, Zhejiang, China
| | - Xifei Qian
- School of Medicine, Zhejiang Chinese Medical University, Hangzhou, 310000, Zhejiang, China
| | - Haiyan Shen
- Department of Plastic and Cosmetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 261 Huansha Road, Hangzhou, 310000, Zhejiang, China
| | - Hanxiao Cheng
- Department of Plastic and Cosmetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 261 Huansha Road, Hangzhou, 310000, Zhejiang, China
| | - Jufang Zhang
- Department of Plastic and Cosmetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 261 Huansha Road, Hangzhou, 310000, Zhejiang, China.
| |
Collapse
|
6
|
Feng QS, Shan XF, Yau V, Cai ZG, Xie S. Facilitation of Tumor Stroma-Targeted Therapy: Model Difficulty and Co-Culture Organoid Method. Pharmaceuticals (Basel) 2025; 18:62. [PMID: 39861125 PMCID: PMC11769033 DOI: 10.3390/ph18010062] [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: 12/10/2024] [Revised: 12/28/2024] [Accepted: 01/05/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Tumors, as intricate ecosystems, comprise oncocytes and the highly dynamic tumor stroma. Tumor stroma, representing the non-cancerous and non-cellular composition of the tumor microenvironment (TME), plays a crucial role in oncogenesis and progression, through its interactions with biological, chemical, and mechanical signals. This review aims to analyze the challenges of stroma mimicry models, and highlight advanced personalized co-culture approaches for recapitulating tumor stroma using patient-derived tumor organoids (PDTOs). Methods: This review synthesizes findings from recent studies on tumor stroma composition, stromal remodeling, and the spatiotemporal heterogeneities of the TME. It explores popular stroma-related models, co-culture systems integrating PDTOs with stromal elements, and advanced techniques to improve stroma mimicry. Results: Stroma remodeling, driven by stromal cells, highlights the dynamism and heterogeneity of the TME. PDTOs, derived from tumor tissues or cancer-specific stem cells, accurately mimic the tissue-specific and genetic features of primary tumors, making them valuable for drug screening. Co-culture models combining PDTOs with stromal elements effectively recreate the dynamic TME, showing promise in personalized anti-cancer therapy. Advanced co-culture techniques and flexible combinations enhance the precision of tumor-stroma recapitulation. Conclusions: PDTO-based co-culture systems offer a promising platform for stroma mimicry and personalized anti-cancer therapy development. This review underscores the importance of refining these models to advance precision medicine and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Qiu-Shi Feng
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22# Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (Q.-S.F.); (X.-F.S.)
| | - Xiao-Feng Shan
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22# Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (Q.-S.F.); (X.-F.S.)
| | - Vicky Yau
- Division of Oral and Maxillofacial Surgery, Columbia Irving Medical Center, New York City, NY 10027, USA;
| | - Zhi-Gang Cai
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22# Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (Q.-S.F.); (X.-F.S.)
| | - Shang Xie
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22# Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (Q.-S.F.); (X.-F.S.)
| |
Collapse
|
7
|
Wang Z, Zhao F, Lang H, Ren H, Zhang Q, Huang X, He C, Xu C, Tan C, Ma J, Duan S, Wang Z. Organoids in skin wound healing. BURNS & TRAUMA 2025; 13:tkae077. [PMID: 39759541 PMCID: PMC11697111 DOI: 10.1093/burnst/tkae077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/30/2024] [Accepted: 11/11/2024] [Indexed: 01/07/2025]
Abstract
Stem cells (SCs) can self-replicate and differentiate into multiple lineages. Organoids, 3D cultures derived from SCs, can replicate the spatial structure and physiological characteristics of organs in vitro. Skin organoids can effectively simulate the physiological structure and function of skin tissue, reliably restoring the natural skin ecology in various in vitro environments. Skin organoids have been employed extensively in skin development and pathology research, offering valuable insights for drug screening. Moreover, they play crucial roles in skin regeneration and tissue repair. This in-depth review explores the construction and applications of skin organoids in wound healing, with a focus on their construction process, including skin appendage integration, and significant advancements in wound-healing research.
Collapse
Affiliation(s)
- Zitong Wang
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Feng Zhao
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, No. 77 Puhe Road, Shenyang, Liaoning 110013, China
| | - Hongxin Lang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, No. 77 Puhe Road, Shenyang, Liaoning 110013, China
| | - Haiyue Ren
- Department of Pathology, Wuhan Hospital of Traditional Chinese and Western Medicine (Wuhan No. 1 Hospital), No. 215 Zhongshan Street, Wuhan, Hubei 430022, China
| | - Qiqi Zhang
- Department of Pathology, Chengdu Third People's Hospital, No. 82 Qinglong Street, Chengdu, Sichuan 610031, China
| | - Xing Huang
- Department of Anaesthesiology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yantaxi Road, Xi'an, Shanxi 710061, China
| | - Cai He
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Chengcheng Xu
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Chiyu Tan
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Jiajie Ma
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Shu Duan
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Zhe Wang
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| |
Collapse
|
8
|
Zhang T, Sheng S, Cai W, Yang H, Li J, Niu L, Chen W, Zhang X, Zhou Q, Gao C, Li Z, Zhang Y, Wang G, Shen H, Zhang H, Hu Y, Yin Z, Chen X, Liu Y, Cui J, Su J. 3-D bioprinted human-derived skin organoids accelerate full-thickness skin defects repair. Bioact Mater 2024; 42:257-269. [PMID: 39285913 PMCID: PMC11404058 DOI: 10.1016/j.bioactmat.2024.08.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
The healing of large skin defects remains a significant challenge in clinical settings. The lack of epidermal sources, such as autologous skin grafting, limits full-thickness skin defect repair and leads to excessive scar formation. Skin organoids have the potential to generate a complete skin layer, supporting in-situ skin regeneration in the defect area. In this study, skin organoid spheres, created with human keratinocytes, fibroblasts, and endothelial cells, showed a specific structure with a stromal core surrounded by surface keratinocytes. We selected an appropriate bioink and innovatively combined an extrusion-based bioprinting technique with dual-photo source cross-linking technology to ensure the overall mechanical properties of the 3D bioprinted skin organoid. Moreover, the 3D bioprinted skin organoid was customized to match the size and shape of the wound site, facilitating convenient implantation. When applied to full-thickness skin defects in immunodeficient mice, the 3D bioprinted human-derived skin organoid significantly accelerated wound healing through in-situ regeneration, epithelialization, vascularization, and inhibition of excessive inflammation. The combination of skin organoid and 3D bioprinting technology can overcome the limitations of current skin substitutes, offering a novel treatment strategy to address large-area skin defects.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200433, China
- Department of Orthopedics, First Affiliated Hospital, Naval Medical University, Shanghai, 200433, China
| | - Shihao Sheng
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200433, China
| | - Weihuang Cai
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, 200444, China
| | - Huijian Yang
- Department of Laboratory Medicine, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Jiameng Li
- Department of Orthopedics, First Affiliated Hospital, Naval Medical University, Shanghai, 200433, China
| | - Luyu Niu
- Department of Orthopedics, First Affiliated Hospital, Naval Medical University, Shanghai, 200433, China
| | - Wanzhuo Chen
- Department of Orthopedics, First Affiliated Hospital, Naval Medical University, Shanghai, 200433, China
| | - Xiuyuan Zhang
- Department of Orthopedics, First Affiliated Hospital, Naval Medical University, Shanghai, 200433, China
| | - Qirong Zhou
- Department of Orthopedics, First Affiliated Hospital, Naval Medical University, Shanghai, 200433, China
| | - Chuang Gao
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, 200444, China
| | - Zuhao Li
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200433, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yuanwei Zhang
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200433, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Guangchao Wang
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200433, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Hao Shen
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200433, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Hao Zhang
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200433, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yan Hu
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200433, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zhifeng Yin
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Xiao Chen
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200433, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yuanyuan Liu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, 200444, China
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, China
| | - Jin Cui
- Department of Orthopedics, First Affiliated Hospital, Naval Medical University, Shanghai, 200433, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200433, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Institute of Biomedicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), SHU Branch, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
9
|
Zhang YX, Zhou Y, Xiong YY, Li YM. Beyond skin deep: Revealing the essence of iPS cell-generated skin organoids in regeneration. Burns 2024; 50:107194. [PMID: 39317530 DOI: 10.1016/j.burns.2024.06.011] [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: 12/24/2023] [Revised: 03/13/2024] [Accepted: 06/23/2024] [Indexed: 09/26/2024]
Abstract
Various methods have been used for in vivo and in vitro skin regeneration, including stem cell therapy, tissue engineering, 3D printing, and platelet-rich plasma (PRP) injection therapy. However, these approaches are rooted in the existing knowledge of skin structures, which overlook the normal physiological processes of skin development and fall short of replicating the skin's regenerative processes outside the body. This comprehensive review primarily focuses on skin organoids derived from human pluripotent stem cells, which have the capacity to regenerate human skin tissue by restoring the embryonic skin structure, thus offering a novel avenue for producing in vitro skin substitutes. Furthermore, they contribute to the repair of damaged skin lesions in patients with systemic sclerosis or severe burns. Particular emphasis will be placed on the origins, generations, and applications of skin organoids, especially in dermatology, and the challenges that must be addressed before clinical implementation.
Collapse
Affiliation(s)
- Yu-Xuan Zhang
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Yuan Zhou
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Yu-Yun Xiong
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.
| | - Yu-Mei Li
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
10
|
Lv Y, Yang W, Kannan PR, Zhang H, Zhang R, Zhao R, Kong X. Materials-based hair follicle engineering: Basic components and recent advances. Mater Today Bio 2024; 29:101303. [PMID: 39498149 PMCID: PMC11532916 DOI: 10.1016/j.mtbio.2024.101303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
The hair follicle (HF) is a significant skin appendage whose primary function is to produce the hair shaft. HFs are a non-renewable resource; skin damage or follicle closure may lead to permanent hair loss. Advances in biomaterials and biomedical engineering enable the feasibility of manipulating the HF-associated cell function for follicle reconstruction via rational design. The regeneration of bioengineered HF addresses the issue of limited resources and contributes to advancements in research and applications in hair loss treatment, HF development, and drug screening. Based on these requirements, this review summarizes the basic and recent advances in hair follicle regulation, including four components: acquisition of stem cells, signaling pathways, materials, and engineering methods. Recent studies have focused on efficiently combining these components and reproducing functionality, which would boost fabrication in HF rebuilding ex vivo, thereby eliminating the obstacles of transplantation into animals to promote mature development.
Collapse
Affiliation(s)
- Yudie Lv
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Weili Yang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Perumal Ramesh Kannan
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Han Zhang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Rui Zhang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Ruibo Zhao
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xiangdong Kong
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| |
Collapse
|
11
|
Ge JY, Wang Y, Li QL, Liu FK, Lei QK, Zheng YW. Trends and challenges in organoid modeling and expansion with pluripotent stem cells and somatic tissue. PeerJ 2024; 12:e18422. [PMID: 39619184 PMCID: PMC11608026 DOI: 10.7717/peerj.18422] [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: 04/28/2024] [Accepted: 10/08/2024] [Indexed: 03/10/2025] Open
Abstract
The increasing demand for disease modeling, preclinical drug testing, and long waiting lists for alternative organ substitutes has posed significant challenges to current limitations in organoid technology. Consequently, organoid technology has emerged as a cutting-edge tool capable of accurately recapitulating the complexity of actual organs in physiology and functionality. To bridge the gaps between basic research and pharmaceutical as well as clinical applications, efforts have been made to develop organoids from tissue-derived stem cells or pluripotent stem cells. These developments include optimizing starting cells, refining culture systems, and introducing genetic modifications. With the rapid development of organoid technology, organoid composition has evolved from single-cell to multi-cell types, enhancing their level of biomimicry. Tissue structure has become more refined, and core challenges like vascularization are being addressed actively. These improvements are expected to pave the way for the construction of organoid atlases, automated large-scale cultivation, and universally compatible organoid biobanks. However, major obstacles remain to be overcome before urgently proof-of-concept organoids can be readily converted to practical applications. These obstacles include achieving structural and functional summarily to native tissue, remodeling the microenvironment, and scaling up production. This review aims to summarize the status of organoid development and applications, highlight recent progress, acknowledge existing limitations and challenges, and provide insights into future advancements. It is expected that this will contribute to the establishment of a reliable, scalable, and practical platform for organoid production and translation, further promoting their use in the pharmaceutical industry and regenerative medicine.
Collapse
Affiliation(s)
- Jian-Yun Ge
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong, China
- Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Chinese Academy of Medical Sciences, Tianjin, China
- Innovation and Transformation Center, University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Yun Wang
- Institute of Regenerative Medicine, and Department of Dermatology, Affilated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Dermatology, The First People’s Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Qi-Lin Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Fan-Kai Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Quan-Kai Lei
- Institute of Regenerative Medicine, and Department of Dermatology, Affilated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yun-Wen Zheng
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong, China
- Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Chinese Academy of Medical Sciences, Tianjin, China
- Institute of Regenerative Medicine, and Department of Dermatology, Affilated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
12
|
Ningsih SS, Jusman SWA, Syaidah R, Nauli R, Fadilah F. Efficient protocol for isolating human fibroblast from primary skin cell cultures: application to keloid, hypertrophic scar, and normal skin biopsies. Biol Methods Protoc 2024; 9:bpae082. [PMID: 39554256 PMCID: PMC11565194 DOI: 10.1093/biomethods/bpae082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/16/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024] Open
Abstract
This protocol introduces a streamlined and efficient method for isolating human fibroblast from skin primary cell culture with a specific focus on its application to keloid, hypertrophic scar, and normal skin biopsies. Additionally, the absence of suitable animal models for keloid and hypertrophic scar has led preclinical research to rely on in vitro studies using primary cell cultures. This approach addresses the challenges of existing protocols in terms of time, cost, equipment, and technical expertise required. The method involves derivation, culture, and characterization analysis including cell proliferation, migration, and fibroblastic marker (Vimentin, CD90, CD73, and CD105) expression. Our study yielded high amounts of fibroblast from tested skin explants while maintaining their in vivo-like characteristics and behaviour. Immunostaining assay confirmed that the cultivated fibroblast was positively expressed Vimentin. Flowcytometry results showed high expression of CD90 and CD73 while relatively showing lower expression of CD105. Fibroblast derived from keloid tissue showed the highest rate of proliferation and migration ability compared to the other samples. These findings suggest an efficient and reproducible technique to cultivate high qualified fibroblast from human skin in normal or pathological condition, particularly for keloid and hypertrophic scar. The application of this protocol provides a foundation for further studies to investigate the progression and potential intervention of aberrant fibrotic dermatological disorder, in vitro.
Collapse
Affiliation(s)
- Sri Suciati Ningsih
- Doctoral Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Faculty of Medicine, Universitas Muhammadiyah Prof Dr Hamka, Jakarta 12130, Indonesia
| | - Sri Widia A Jusman
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Center of Hypoxia and Oxidative Stress Studies, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Rahimi Syaidah
- Doctoral Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Department of Histology, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Raisa Nauli
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Fadilah Fadilah
- Doctoral Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Department of Medical Chemistry, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| |
Collapse
|
13
|
Yao Q, Cheng S, Pan Q, Yu J, Cao G, Li L, Cao H. Organoids: development and applications in disease models, drug discovery, precision medicine, and regenerative medicine. MedComm (Beijing) 2024; 5:e735. [PMID: 39309690 PMCID: PMC11416091 DOI: 10.1002/mco2.735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Organoids are miniature, highly accurate representations of organs that capture the structure and unique functions of specific organs. Although the field of organoids has experienced exponential growth, driven by advances in artificial intelligence, gene editing, and bioinstrumentation, a comprehensive and accurate overview of organoid applications remains necessary. This review offers a detailed exploration of the historical origins and characteristics of various organoid types, their applications-including disease modeling, drug toxicity and efficacy assessments, precision medicine, and regenerative medicine-as well as the current challenges and future directions of organoid research. Organoids have proven instrumental in elucidating genetic cell fate in hereditary diseases, infectious diseases, metabolic disorders, and malignancies, as well as in the study of processes such as embryonic development, molecular mechanisms, and host-microbe interactions. Furthermore, the integration of organoid technology with artificial intelligence and microfluidics has significantly advanced large-scale, rapid, and cost-effective drug toxicity and efficacy assessments, thereby propelling progress in precision medicine. Finally, with the advent of high-performance materials, three-dimensional printing technology, and gene editing, organoids are also gaining prominence in the field of regenerative medicine. Our insights and predictions aim to provide valuable guidance to current researchers and to support the continued advancement of this rapidly developing field.
Collapse
Affiliation(s)
- Qigu Yao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Sheng Cheng
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Qiaoling Pan
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jiong Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Guoqiang Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Lanjuan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Hongcui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic‐Chemical and Aging‐Related InjuriesHangzhouChina
| |
Collapse
|
14
|
Luca T, Pezzino S, Puleo S, Castorina S. Lesson on obesity and anatomy of adipose tissue: new models of study in the era of clinical and translational research. J Transl Med 2024; 22:764. [PMID: 39143643 PMCID: PMC11323604 DOI: 10.1186/s12967-024-05547-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 07/28/2024] [Indexed: 08/16/2024] Open
Abstract
Obesity is a serious global illness that is frequently associated with metabolic syndrome. Adipocytes are the typical cells of adipose organ, which is composed of at least two different tissues, white and brown adipose tissue. They functionally cooperate, interconverting each other under physiological conditions, but differ in their anatomy, physiology, and endocrine functions. Different cellular models have been proposed to study adipose tissue in vitro. They are also useful for elucidating the mechanisms that are responsible for a pathological condition, such as obesity, and for testing therapeutic strategies. Each cell model has its own characteristics, culture conditions, advantages and disadvantages. The choice of one model rather than another depends on the specific study the researcher is conducting. In recent decades, three-dimensional cultures, such as adipose spheroids, have become very attractive because they more closely resemble the phenotype of freshly isolated cells. The use of such models has developed in parallel with the evolution of translational research, an interdisciplinary branch of the biomedical field, which aims to learn a scientific translational approach to improve human health and longevity. The focus of the present review is on the growing body of data linking the use of new cell models and the spread of translational research. Also, we discuss the possibility, for the future, to employ new three-dimensional adipose tissue cell models to promote the transition from benchside to bedsite and vice versa, allowing translational research to become routine, with the final goal of obtaining clinical benefits in the prevention and treatment of obesity and related disorders.
Collapse
Affiliation(s)
- Tonia Luca
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, Catania, 95123, Italy.
| | | | - Stefano Puleo
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| | - Sergio Castorina
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, Catania, 95123, Italy
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| |
Collapse
|
15
|
Yang H, Niu S, Guo M, Xue Y. Applications of 3D organoids in toxicological studies: a comprehensive analysis based on bibliometrics and advances in toxicological mechanisms. Arch Toxicol 2024; 98:2309-2330. [PMID: 38806717 DOI: 10.1007/s00204-024-03777-4] [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: 01/22/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024]
Abstract
A mechanism exploration is an important part of toxicological studies. However, traditional cell and animal models can no longer meet the current needs for in-depth studies of toxicological mechanisms. The three-dimensional (3D) organoid derived from human embryonic stem cells (hESC) or induced pluripotent stem cells (hiPSC) is an ideal experimental model for the study of toxicological effects and mechanisms, which further recapitulates the human tissue microenvironment and provides a reliable method for studying complex cell-cell interactions. This article provides a comprehensive overview of the state of the 3D organoid technology in toxicological studies, including a bibliometric analysis of the existing literature and an exploration of the latest advances in toxicological mechanisms. The use of 3D organoids in toxicology research is growing rapidly, with applications in disease modeling, organ-on-chips, and drug toxicity screening being emphasized, but academic communications among countries/regions, institutions, and research scholars need to be further strengthened. Attempts to study the toxicological mechanisms of exogenous chemicals such as heavy metals, nanoparticles, drugs and organic pollutants are also increasing. It can be expected that 3D organoids can be better applied to the safety evaluation of exogenous chemicals by establishing a standardized methodology.
Collapse
Affiliation(s)
- Haitao Yang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Shuyan Niu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Menghao Guo
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yuying Xue
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
16
|
Mazari‐Arrighi E, Lépine M, Ayollo D, Faivre L, Larghero J, Chatelain F, Fuchs A. Self-Organization of Long-Lasting Human Endothelial Capillary-Like Networks Guided by DLP Bioprinting. Adv Healthc Mater 2024; 13:e2302830. [PMID: 38366136 PMCID: PMC11468676 DOI: 10.1002/adhm.202302830] [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: 08/25/2023] [Revised: 01/29/2024] [Indexed: 02/18/2024]
Abstract
Tissue engineering holds great promise for regenerative medicine, drug discovery, and as an alternative to animal models. However, as soon as the dimensions of engineered tissue exceed the diffusion limit of oxygen and nutriments, a necrotic core forms leading to irreversible damage. To overcome this constraint, the establishment of a functional perfusion network is essential. In this work, digital light processing bioprinting is used to encapsulate endothelial progenitor cells (EPCs) in 3D light-cured hydrogel scaffolds to guide them toward vascular network formation. In these scaffolds, EPCs proliferate and self-organize within a few days into branched tubular structures with predefined geometry, forming capillary-like vascular tubes or trees of diameters in the range of 10 to 100 µm. Presenting a confluent monolayer wall of cells strongly connect by tight junctions around a central lumen-like space, these structures can be microinjected with a fluorescent dye and are stable for several weeks in vitro. These endothelial structures can be recovered and manipulated in an alginate patch without altering their shape or viability. This approach opens new opportunities for future applications, such as stacking with other cell sheets or multicellular constructs to yield bioengineered tissue with higher complexity and functionality.
Collapse
Affiliation(s)
- Elsa Mazari‐Arrighi
- Université de ParisU976 HIPI, InsermParisF‐75006France
- AP‐HPHôpital Saint‐Louis1 avenue VellefauxParisF‐75010France
| | - Matthieu Lépine
- Université de ParisU976 HIPI, InsermParisF‐75006France
- AP‐HPHôpital Saint‐Louis1 avenue VellefauxParisF‐75010France
| | - Dmitry Ayollo
- Université de ParisU976 HIPI, InsermParisF‐75006France
- AP‐HPHôpital Saint‐Louis1 avenue VellefauxParisF‐75010France
| | - Lionel Faivre
- Université de ParisU976 HIPI, InsermParisF‐75006France
- AP‐HPHôpital Saint‐Louis1 avenue VellefauxParisF‐75010France
| | - Jérôme Larghero
- Université de ParisU976 HIPI, InsermParisF‐75006France
- AP‐HPHôpital Saint‐Louis1 avenue VellefauxParisF‐75010France
| | - François Chatelain
- Université de ParisU976 HIPI, InsermParisF‐75006France
- CEAIRIGGrenobleF‐38000France
| | - Alexandra Fuchs
- Université de ParisU976 HIPI, InsermParisF‐75006France
- CEAIRIGGrenobleF‐38000France
| |
Collapse
|
17
|
Piñón Hofbauer J, Guttmann-Gruber C, Wally V, Sharma A, Gratz IK, Koller U. Challenges and progress related to gene editing in rare skin diseases. Adv Drug Deliv Rev 2024; 208:115294. [PMID: 38527624 DOI: 10.1016/j.addr.2024.115294] [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: 12/15/2023] [Revised: 03/01/2024] [Accepted: 03/21/2024] [Indexed: 03/27/2024]
Abstract
Genodermatoses represent a large group of inherited skin disorders encompassing clinically-heterogeneous conditions that manifest in the skin and other organs. Depending on disease variant, associated clinical manifestations and secondary complications can severely impact patients' quality of life and currently available treatments are transient and not curative. Multiple emerging approaches using CRISPR-based technologies offer promising prospects for therapy. Here, we explore current advances and challenges related to gene editing in rare skin diseases, including different strategies tailored to mutation type and structural organization of the affected gene, considerations for in vivo and ex vivo applications, the critical issue of delivery into the skin, and immune aspects of therapy. Against the backdrop of a landmark FDA approval for the first re-dosable gene replacement therapy for a rare genetic skin disorder, gene editing approaches are inching closer to the clinics and the possibility of a local permanent cure for patients affected by these disorders.
Collapse
Affiliation(s)
- Josefina Piñón Hofbauer
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Christina Guttmann-Gruber
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Verena Wally
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Anshu Sharma
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria
| | - Iris K Gratz
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria; Center for Tumor Biology and Immunology, University of Salzburg, 5020 Salzburg, Austria
| | - Ulrich Koller
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria.
| |
Collapse
|
18
|
Choudhury S, Dhoke NR, Chawla S, Das A. Bioengineered MSC Cxcr2 transdifferentiated keratinocyte-like cell-derived organoid potentiates skin regeneration through ERK1/2 and STAT3 signaling in diabetic wound. Cell Mol Life Sci 2024; 81:172. [PMID: 38597972 PMCID: PMC11006766 DOI: 10.1007/s00018-023-05057-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 11/17/2023] [Accepted: 11/18/2023] [Indexed: 04/11/2024]
Abstract
Skin regeneration is severely compromised in diabetic foot ulcers. Allogeneic mesenchymal stem cell (MSC) transplantation is limited due to the poor engraftment, mitogenic, and differentiation potential in the harsh wound microenvironment. Thus, to improve the efficacy of cell therapy, the chemokine receptor Cxcr2 was overexpressed in MSCs (MSCCxcr2). CXCL2/CXCR2 axis induction led to the enhanced proliferation of MSCs through the activation of STAT3 and ERK1/2 signaling. Transcriptional upregulation of FGFR2IIIb (KGF Receptor) promoter by the activated STAT3 and ERK1/2 suggested trans-differentiation of MSCs into keratinocytes. These stable MSCCxcr2 in 2D and 3D (spheroid) cell cultures efficiently transdifferentiated into keratinocyte-like cells (KLCs). An in vivo therapeutic potential of MSCCxcr2 transplantation and its keratinocyte-specific cell fate was observed by accelerated skin tissue regeneration in an excisional splinting wound healing murine model of streptozotocin-induced type 1 diabetes. Finally, 3D skin organoids generated using MSCCxcr2-derived KLCs upon grafting in a relatively avascular and non-healing wounds of type 2 diabetic db/db transgenic old mice resulted in a significant enhancement in the rate of wound closure by increased epithelialization (epidermal layer) and endothelialization (dermal layer). Our findings emphasize the therapeutic role of the CXCL2/CXCR2 axis in inducing trans-differentiation of the MSCs toward KLCs through the activation of ERK1/2 and STAT3 signaling and enhanced skin regeneration potential of 3D organoids grafting in chronic diabetic wounds.
Collapse
Affiliation(s)
- Subholakshmi Choudhury
- Department of Applied Biology, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad, 500007, TS, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Neha R Dhoke
- Department of Applied Biology, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad, 500007, TS, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shilpa Chawla
- Department of Applied Biology, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad, 500007, TS, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Amitava Das
- Department of Applied Biology, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad, 500007, TS, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
19
|
Ying X, Yu C, Yang W, Ye L, Sun R, Gu T, Fan S, Yao S. The transformation of multifunctional bio-patch to hydrogel on skin wounds for efficient scarless wound healing. Mater Today Bio 2024; 24:100901. [PMID: 38188643 PMCID: PMC10770564 DOI: 10.1016/j.mtbio.2023.100901] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/19/2023] [Accepted: 12/03/2023] [Indexed: 01/09/2024] Open
Abstract
Hydrogels have been widely used in various biomedical applications, including skin regeneration and tissue repair. However, the capability of certain hydrogels to absorb exudate or blood from surrounding wounds, coupled with the challenge in their long-term storage to prevent bacterial growth, can pose limitations to their efficacy in biological applications. To address these challenges, the development of a multifunctional aloin-arginine-alginate (short for 3A) bio-patch capable of transforming into a hydrogel upon absorbing exudate or blood from neighboring wounds for cutaneous regeneration is proposed. The 3A bio-patch exhibits outstanding features, including an excellent porous structure, swelling properties, and biodegradability. These characteristics allow for the rapid absorption of wound exudates and subsequent transformation into a hydrogel that is suitable for treating skin wounds. Furthermore, the 3A bio-patch exhibits remarkable antibacterial and anti-inflammatory properties, leading to accelerated wound healing and scarless repair in vivo. This study presents a novel approach to the development of cutaneous wound dressing materials.
Collapse
Affiliation(s)
- Xiaozhang Ying
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
- Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, 310003, China
| | - Congcong Yu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Wentao Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Lin Ye
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Rongtai Sun
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Tianyuan Gu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Shasha Yao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| |
Collapse
|
20
|
Ma M, Ge JY, Nie YZ, Li YM, Zheng YW. Developing Humanized Animal Models with Transplantable Human iPSC-Derived Cells. FRONT BIOSCI-LANDMRK 2024; 29:34. [PMID: 38287837 DOI: 10.31083/j.fbl2901034] [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: 09/16/2023] [Revised: 12/02/2023] [Accepted: 12/22/2023] [Indexed: 01/31/2024]
Abstract
Establishing reliable and reproducible animal models for disease modelling, drug screening and the understanding of disease susceptibility and pathogenesis is critical. However, traditional animal models differ significantly from humans in terms of physiology, immune response, and pathogenesis. As a result, it is difficult to translate laboratory findings into biomedical applications. Although several animal models with human chimeric genes, organs or systems have been developed in the past, their limited engraftment rate and physiological functions are a major obstacle to realize convincing models of humans. The lack of human transplantation resources and insufficient immune tolerance of recipient animals are the main challenges that need to be overcome to generate fully humanized animals. Recent advances in gene editing and pluripotent stem cell-based xenotransplantation technologies offer opportunities to create more accessible human-like models for biomedical research. In this article, we have combined our laboratory expertise to summarize humanized animal models, with a focus on hematopoietic/immune system and liver. We discuss their generation strategies and the potential donor cell sources, with particular attention given to human pluripotent stem cells. In particular, we discuss the advantages, limitations and emerging trends in their clinical and pharmaceutical applications. By providing insights into the current state of humanized animal models and their potential for biomedical applications, this article aims to advance the development of more accurate and reliable animal models for disease modeling and drug screening.
Collapse
Affiliation(s)
- Min Ma
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, 212001 Zhenjiang, Jiangsu, China
| | - Jian-Yun Ge
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and South China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, 529020 Jiangmen, Guangdong, China
| | - Yun-Zhong Nie
- Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, 108-8639 Tokyo, Japan
| | - Yu-Mei Li
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, 212001 Zhenjiang, Jiangsu, China
| | - Yun-Wen Zheng
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, 212001 Zhenjiang, Jiangsu, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and South China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, 529020 Jiangmen, Guangdong, China
- Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, 108-8639 Tokyo, Japan
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 278-8510 Noda, Japan
| |
Collapse
|
21
|
Jing Jia, Ma B, Zhao X. Fetal endothelial colony-forming cells: Possible targets for prevention of the fetal origins of adult diseases. Placenta 2024; 145:80-88. [PMID: 38100962 DOI: 10.1016/j.placenta.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/20/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Endothelial colony-forming cells (ECFCs), a subset of circulating and resident endothelial progenitor cells, are capable of self-renewal and de novo vessel formation, and are known key regulators of vascular integrity and homeostasis. Numerous studies have found that exposure to hostile environment during the fetal development exerts a profound influence on the level and function of ECFCs, which may be the underlying factor linking endothelial dysfunction to cardiovascular disease of the offspring in later life. Herein, we focus on the latest findings regarding the effects of pregnancy-related disorders on the frequency and function of fetal ECFCs. Subsequently, we discuss about placental ECFCs and put forward some details that should be paid attention to in the process of ECFC isolation and culture. Overall, the information presented in this review highlight the potential of ECFCs as a future biomarker or even therapeutic targets for the pregnancy-related adverse maternal and fetal outcomes.
Collapse
Affiliation(s)
- Jing Jia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Baitao Ma
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xianlan Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
22
|
Novis T, Takiya CM. Skin Resident Stem Cells. RESIDENT STEM CELLS AND REGENERATIVE THERAPY 2024:205-249. [DOI: 10.1016/b978-0-443-15289-4.00005-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
23
|
Zhao H, Chen Z, Kang X, Yang B, Luo P, Li H, He Q. The frontline of alternatives to animal testing: novel in vitro skin model application in drug development and evaluation. Toxicol Sci 2023; 196:152-169. [PMID: 37702017 DOI: 10.1093/toxsci/kfad093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023] Open
Abstract
The FDA Modernization Act 2.0 has brought nonclinical drug evaluation into a new era. In vitro models are widely used and play an important role in modern drug development and evaluation, including early candidate drug screening and preclinical drug efficacy and toxicity assessment. Driven by regulatory steering and facilitated by well-defined physiology, novel in vitro skin models are emerging rapidly, becoming the most advanced area in alternative testing research. The revolutionary technologies bring us many in vitro skin models, either laboratory-developed or commercially available, which were all built to emulate the structure of the natural skin to recapitulate the skin's physiological function and particular skin pathology. During the model development, how to achieve balance among complexity, accessibility, capability, and cost-effectiveness remains the core challenge for researchers. This review attempts to introduce the existing in vitro skin models, align them on different dimensions, such as structural complexity, functional maturity, and screening throughput, and provide an update on their current application in various scenarios within the scope of chemical testing and drug development, including testing in genotoxicity, phototoxicity, skin sensitization, corrosion/irritation. Overall, the review will summarize a general strategy for in vitro skin model to enhance future model invention, application, and translation in drug development and evaluation.
Collapse
Affiliation(s)
- He Zhao
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhaozeng Chen
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| | - Xingchen Kang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| | - Hui Li
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| |
Collapse
|
24
|
Hong ZX, Zhu ST, Li H, Luo JZ, Yang Y, An Y, Wang X, Wang K. Bioengineered skin organoids: from development to applications. Mil Med Res 2023; 10:40. [PMID: 37605220 PMCID: PMC10463602 DOI: 10.1186/s40779-023-00475-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/01/2023] [Indexed: 08/23/2023] Open
Abstract
Significant advancements have been made in recent years in the development of highly sophisticated skin organoids. Serving as three-dimensional models that mimic human skin, these organoids have evolved into complex structures and are increasingly recognized as effective alternatives to traditional culture models and human skin due to their ability to overcome the limitations of two-dimensional systems and ethical concerns. The inherent plasticity of skin organoids allows for their construction into physiological and pathological models, enabling the study of skin development and dynamic changes. This review provides an overview of the pivotal work in the progression from 3D layered epidermis to cyst-like skin organoids with appendages. Furthermore, it highlights the latest advancements in organoid construction facilitated by state-of-the-art engineering techniques, such as 3D printing and microfluidic devices. The review also summarizes and discusses the diverse applications of skin organoids in developmental biology, disease modelling, regenerative medicine, and personalized medicine, while considering their prospects and limitations.
Collapse
Affiliation(s)
- Zi-Xuan Hong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Shun-Tian Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Hao Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Jing-Zhi Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Yu Yang
- Department of Hepatopancreatobiliary Surgery, the Third Affiliated Hospital of Soochow University, Changzhou, 213000, Jiangsu, China
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China.
| | - Xi Wang
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China.
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China.
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China.
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Beijing, 100037, China.
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
25
|
Walz S, Pollehne P, Vollmer P, Aicher WK, Stenzl A, Harland N, Amend B. Effects of Scaffolds on Urine- and Urothelial Carcinoma Tissue-Derived Organoids from Bladder Cancer Patients. Cells 2023; 12:2108. [PMID: 37626918 PMCID: PMC10453567 DOI: 10.3390/cells12162108] [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: 07/05/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Organoids are three-dimensional constructs generated by placing cells in scaffolds to facilitate the growth of cultures with cell-cell and cell-matrix interactions close to the in vivo situation. Organoids may contain different types of cells, including cancer cells, progenitor cells, or differentiated cells. As distinct culture conditions have significant effects on cell metabolism, we explored the expansion of cells and expression of marker genes in bladder cancer cells expanded in two different common scaffolds. The cells were seeded in basement membrane extract (BME; s.c., Matrigel®) or in a cellulose-derived hydrogel (GrowDex®, GD) and cultured. The size of organoids and expression of marker genes were studied. We discovered that BME facilitated the growth of significantly larger organoids of cancer cell line RT112 (p < 0.05), cells from a solid tumor (p < 0.001), and a voiding urine sample (p < 0.001). Expression of proliferation marker Ki76, transcription factor TP63, cytokeratin CK20, and cell surface marker CD24 clearly differed in these different tumor cells upon expansion in BME when compared to cells in GD. We conclude that the choice of scaffold utilized for the generation of organoids has an impact not only on cell growth and organoid size but also on protein expression. The disadvantages of batch-to-batch-variations of BME must be balanced with the phenotypic bias observed with GD scaffolds when standardizing organoid cultures for clinical diagnoses.
Collapse
Affiliation(s)
- Simon Walz
- Department of Urology, University of Tuebingen Hospital, 72076 Tübingen, Germany
| | - Paul Pollehne
- Center for Medical Research, University of Tuebingen, 72074 Tübingen, Germany
| | - Philipp Vollmer
- Center for Medical Research, University of Tuebingen, 72074 Tübingen, Germany
| | - Wilhelm K. Aicher
- Center for Medical Research, University of Tuebingen, 72074 Tübingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University of Tuebingen Hospital, 72076 Tübingen, Germany
| | - Niklas Harland
- Department of Urology, University of Tuebingen Hospital, 72076 Tübingen, Germany
| | - Bastian Amend
- Department of Urology, University of Tuebingen Hospital, 72076 Tübingen, Germany
| |
Collapse
|
26
|
Urzì O, Gasparro R, Costanzo E, De Luca A, Giavaresi G, Fontana S, Alessandro R. Three-Dimensional Cell Cultures: The Bridge between In Vitro and In Vivo Models. Int J Mol Sci 2023; 24:12046. [PMID: 37569426 PMCID: PMC10419178 DOI: 10.3390/ijms241512046] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Although historically, the traditional bidimensional in vitro cell system has been widely used in research, providing much fundamental information regarding cellular functions and signaling pathways as well as nuclear activities, the simplicity of this system does not fully reflect the heterogeneity and complexity of the in vivo systems. From this arises the need to use animals for experimental research and in vivo testing. Nevertheless, animal use in experimentation presents various aspects of complexity, such as ethical issues, which led Russell and Burch in 1959 to formulate the 3R (Replacement, Reduction, and Refinement) principle, underlying the urgent need to introduce non-animal-based methods in research. Considering this, three-dimensional (3D) models emerged in the scientific community as a bridge between in vitro and in vivo models, allowing for the achievement of cell differentiation and complexity while avoiding the use of animals in experimental research. The purpose of this review is to provide a general overview of the most common methods to establish 3D cell culture and to discuss their promising applications. Three-dimensional cell cultures have been employed as models to study both organ physiology and diseases; moreover, they represent a valuable tool for studying many aspects of cancer. Finally, the possibility of using 3D models for drug screening and regenerative medicine paves the way for the development of new therapeutic opportunities for many diseases.
Collapse
Affiliation(s)
- Ornella Urzì
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (O.U.); (R.G.); (E.C.); (R.A.)
| | - Roberta Gasparro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (O.U.); (R.G.); (E.C.); (R.A.)
| | - Elisa Costanzo
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (O.U.); (R.G.); (E.C.); (R.A.)
| | - Angela De Luca
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche, 40136 Bologna, Italy; (A.D.L.); (G.G.)
| | - Gianluca Giavaresi
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche, 40136 Bologna, Italy; (A.D.L.); (G.G.)
| | - Simona Fontana
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (O.U.); (R.G.); (E.C.); (R.A.)
| | - Riccardo Alessandro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (O.U.); (R.G.); (E.C.); (R.A.)
| |
Collapse
|
27
|
Jang HJ, Lee JB, Yoon JK. Advanced In Vitro Three-Dimensional Skin Models of Atopic Dermatitis. Tissue Eng Regen Med 2023; 20:539-552. [PMID: 36995643 PMCID: PMC10313606 DOI: 10.1007/s13770-023-00532-1] [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: 01/13/2023] [Revised: 02/11/2023] [Accepted: 02/19/2023] [Indexed: 03/31/2023] Open
Abstract
Atopic dermatitis (AD) is one of the most prevalent inflammatory skin diseases that is characterized by eczematous rashes, intense itching, dry skin, and sensitive skin. Although AD significantly impacts the quality of life and the number of patients keeps increasing, its pathological mechanism is still unknown because of its complexity. The importance of developing new in vitro three-dimensional (3D) models has been underlined in order to understand the mechanisms for the development of therapeutics since the limitations of 2D models or animal models have been repeatedly reported. Thus, the new in vitro AD models should not only be created in 3D structure, but also reflect the pathological characteristics of AD, which are known to be associated with Th2-mediated inflammatory responses, epidermal barrier disruption, increased dermal T-cell infiltration, filaggrin down-regulation, or microbial imbalance. In this review, we introduce various types of in vitro skin models including 3D culture methods, skin-on-a-chips, and skin organoids, as well as their applications to AD modeling for drug screening and mechanistic studies.
Collapse
Affiliation(s)
- Hye-Jeong Jang
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea
| | - Jung Bok Lee
- Department of Biological Sciences, Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea.
| |
Collapse
|
28
|
Liu ZJ, Wang MJ, Luo J, Tan YT, Hou M, Wang SC. A bibliometric analysis of hotpots and trends for the relationship between skin inflammation and regeneration. Front Surg 2023; 10:1180624. [PMID: 37151861 PMCID: PMC10160476 DOI: 10.3389/fsurg.2023.1180624] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 03/29/2023] [Indexed: 05/09/2023] Open
Abstract
BACKGROUND Skin regeneration is a challenging issue worldwide. Increasing research has highlighted the role of immune cells in healing and the underlying regulatory mechanism. The purpose of this study was to identify the hotspots and trends in skin regeneration and inflammation research through bibliometrics and to provide insights into the future development of fundamental research and disease treatment. METHODS Publications were collected from the Web of Science Core Collection on March 1, 2022. Articles and reviews published in English from January 1, 1999, to December 31, 2022, were selected, and statistical analyses of countries, institutions, authors, references, and keywords were performed using VOSviewer 1.6.18 and CiteSpace 5.8. RESULTS A total of 3,894 articles and reviews were selected. The number of publications on skin inflammation and regeneration showed an increasing trend over time. Additionally, authors and institutions in the United States, United Kingdom, Canada, and China appeared to be at the forefront of research in the field of skin inflammation and regeneration. Werner Sabine published some of the most cited papers. Wound Repair and Regeneration was the most productive journal, while Journal of Investigative Dermatology was the most cited journal. Angiogenesis, diamonds, collagen, cytokine, and keratinocytes were the five most commonly used keywords. CONCLUSION The number of publications on skin inflammation and regeneration show an increasing trend. Moreover, a series of advanced technologies and treatments for skin regeneration, such as exosomes, hydrogels, and wound dressings, are emerging, which will provide precise information for the treatment of skin wounds. This study can enhance our understanding of current hotspots and future trends in skin inflammation and regeneration research, as well as provide guidelines for fundamental research and clinical treatment.
Collapse
Affiliation(s)
- Zhen-jiang Liu
- Department of Cardiology, Cardiac Catheterization Lab, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Mei-juan Wang
- Medical Imaging Center, Qingdao West Coast New District People's Hospital, Qingdao, China
| | - Jia Luo
- Hunan key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China
| | - Ya-ting Tan
- Center for Medical Research, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Min Hou
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital of Central South University, Changsha, China
- Party Committee Office, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shu-chao Wang
- Center for Medical Research, The Second Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
29
|
Hochmann S, Ou K, Poupardin R, Mittermeir M, Textor M, Ali S, Wolf M, Ellinghaus A, Jacobi D, Elmiger JAJ, Donsante S, Riminucci M, Schäfer R, Kornak U, Klein O, Schallmoser K, Schmidt-Bleek K, Duda GN, Polansky JK, Geissler S, Strunk D. The enhancer landscape predetermines the skeletal regeneration capacity of stromal cells. Sci Transl Med 2023; 15:eabm7477. [PMID: 36947595 DOI: 10.1126/scitranslmed.abm7477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Multipotent stromal cells are considered attractive sources for cell therapy and tissue engineering. Despite numerous experimental and clinical studies, broad application of stromal cell therapeutics is not yet emerging. A major challenge is the functional diversity of available cell sources. Here, we investigated the regenerative potential of clinically relevant human stromal cells from bone marrow (BMSCs), white adipose tissue, and umbilical cord compared with mature chondrocytes and skin fibroblasts in vitro and in vivo. Although all stromal cell types could express transcription factors related to endochondral ossification, only BMSCs formed cartilage discs in vitro that fully regenerated critical-size femoral defects after transplantation into mice. We identified cell type-specific epigenetic landscapes as the underlying molecular mechanism controlling transcriptional stromal differentiation networks. Binding sites of commonly expressed transcription factors in the enhancer and promoter regions of ossification-related genes, including Runt and bZIP families, were accessible only in BMSCs but not in extraskeletal stromal cells. This suggests an epigenetically predetermined differentiation potential depending on cell origin that allows common transcription factors to trigger distinct organ-specific transcriptional programs, facilitating forward selection of regeneration-competent cell sources. Last, we demonstrate that viable human BMSCs initiated defect healing through the secretion of osteopontin and contributed to transient mineralized bone hard callus formation after transplantation into immunodeficient mice, which was eventually replaced by murine recipient bone during final tissue remodeling.
Collapse
Affiliation(s)
- Sarah Hochmann
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Kristy Ou
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), T Cell Epigenetics, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Rodolphe Poupardin
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Michaela Mittermeir
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Martin Textor
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Salaheddine Ali
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Martin Wolf
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Agnes Ellinghaus
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Dorit Jacobi
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Juri A J Elmiger
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Samantha Donsante
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Richard Schäfer
- Institute for Transfusion Medicine and Immunohematology, Goethe University Hospital, German Red Cross Blood Service Baden-Württemberg-Hessen gGmbH, 60323 Frankfurt am Main, Germany
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, 79106 Freiburg, Germany
| | - Uwe Kornak
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Oliver Klein
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
| | | | - Katharina Schmidt-Bleek
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Georg N Duda
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Julia K Polansky
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), T Cell Epigenetics, Augustenburger Platz 1, 13353 Berlin, Germany
- German Rheumatism Research Centre (DRFZ), 10117 Berlin, Germany
| | - Sven Geissler
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Center for Advanced Therapies (BECAT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Dirk Strunk
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| |
Collapse
|
30
|
Zwicklhuber J, Kocher T, Liemberger B, Hainzl S, Bischof J, Strunk D, Raninger AM, Gratz I, Wally V, Guttmann-Gruber C, Hofbauer JP, Bauer JW, Koller U. A Novel Fluorescence-Based Screen of Gene Editing Molecules for Junctional Epidermolysis Bullosa. Int J Mol Sci 2023; 24:ijms24065197. [PMID: 36982270 PMCID: PMC10049061 DOI: 10.3390/ijms24065197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
Junctional epidermolysis bullosa (JEB) is a severe blistering skin disease caused by mutations in genes encoding structural proteins essential for skin integrity. In this study, we developed a cell line suitable for gene expression studies of the JEB-associated COL17A1 encoding type XVII collagen (C17), a transmembrane protein involved in connecting basal keratinocytes to the underlying dermis of the skin. Using the CRISPR/Cas9 system of Streptococcus pyogenes we fused the coding sequence of GFP to COL17A1 leading to the constitutive expression of GFP-C17 fusion proteins under the control of the endogenous promoter in human wild-type and JEB keratinocytes. We confirmed the accurate full-length expression and localization of GFP-C17 to the plasma membrane via fluorescence microscopy and Western blot analysis. As expected, the expression of GFP-C17mut fusion proteins in JEB keratinocytes generated no specific GFP signal. However, the CRISPR/Cas9-mediated repair of a JEB-associated frameshift mutation in GFP-COL17A1mut-expressing JEB cells led to the restoration of GFP-C17, apparent in the full-length expression of the fusion protein, its accurate localization within the plasma membrane of keratinocyte monolayers as well as within the basement membrane zone of 3D-skin equivalents. Thus, this fluorescence-based JEB cell line provides the potential to serve as a platform to screen for personalized gene editing molecules and applications in vitro and in appropriate animal models in vivo.
Collapse
Affiliation(s)
- Janine Zwicklhuber
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Thomas Kocher
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Bernadette Liemberger
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Stefan Hainzl
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Johannes Bischof
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Dirk Strunk
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria
| | - Anna M. Raninger
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria
| | - Iris Gratz
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
- Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria
| | - Verena Wally
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Christina Guttmann-Gruber
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Josefina Piñón Hofbauer
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Johann W. Bauer
- Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Ulrich Koller
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
- Correspondence:
| |
Collapse
|
31
|
Otsuka H. Nanofabrication Technologies to Control Cell and Tissue Function in Three-Dimension. Gels 2023; 9:gels9030203. [PMID: 36975652 PMCID: PMC10048556 DOI: 10.3390/gels9030203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/14/2023] [Accepted: 02/24/2023] [Indexed: 03/29/2023] Open
Abstract
In the 2000s, advances in cellular micropatterning using microfabrication contributed to the development of cell-based biosensors for the functional evaluation of newly synthesized drugs, resulting in a revolutionary evolution in drug screening. To this end, it is essential to utilize cell patterning to control the morphology of adherent cells and to understand contact and paracrine-mediated interactions between heterogeneous cells. This suggests that the regulation of the cellular environment by means of microfabricated synthetic surfaces is not only a valuable endeavor for basic research in biology and histology, but is also highly useful to engineer artificial cell scaffolds for tissue regeneration. This review particularly focuses on surface engineering techniques for the cellular micropatterning of three-dimensional (3D) spheroids. To establish cell microarrays, composed of a cell adhesive region surrounded by a cell non-adherent surface, it is quite important to control a protein-repellent surface in the micro-scale. Thus, this review is focused on the surface chemistries of the biologically inspired micropatterning of two-dimensional non-fouling characters. As cells are formed into spheroids, their survival, functions, and engraftment in the transplanted site are significantly improved compared to single-cell transplantation. To improve the therapeutic effect of cell spheroids even further, various biomaterials (e.g., fibers and hydrogels) have been developed for spheroid engineering. These biomaterials not only can control the overall spheroid formation (e.g., size, shape, aggregation speed, and degree of compaction), but also can regulate cell-to-cell and cell-to-matrix interactions in spheroids. These important approaches to cell engineering result in their applications to tissue regeneration, where the cell-biomaterial composite is injected into diseased area. This approach allows the operating surgeon to implant the cell and polymer combinations with minimum invasiveness. The polymers utilized in hydrogels are structurally similar to components of the extracellular matrix in vivo, and are considered biocompatible. This review will provide an overview of the critical design to make hydrogels when used as cell scaffolds for tissue engineering. In addition, the new strategy of injectable hydrogel will be discussed as future directions.
Collapse
Affiliation(s)
- Hidenori Otsuka
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, 1-3 Kagurazaka, Shinjuku-ku, Tokyo 162-8601, Japan
| |
Collapse
|
32
|
Jiang Z, Xu Y, Fu M, Zhu D, Li N, Yang G. Genetically modified cell spheroids for tissue engineering and regenerative medicine. J Control Release 2023; 354:588-605. [PMID: 36657601 DOI: 10.1016/j.jconrel.2023.01.033] [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: 11/05/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/21/2023]
Abstract
Cell spheroids offer cell-to-cell interactions and show advantages in survival rate and paracrine effect to solve clinical and biomedical inquiries ranging from tissue engineering and regenerative medicine to disease pathophysiology. Therefore, cell spheroids are ideal vehicles for gene delivery. Genetically modified spheroids can enhance specific gene expression to promote tissue regeneration. Gene deliveries to cell spheroids are via viral vectors or non-viral vectors. Some new technologies like CRISPR/Cas9 also have been used in genetically modified methods to deliver exogenous gene to the host chromosome. It has been shown that genetically modified cell spheroids had the potential to differentiate into bone, cartilage, vascular, nerve, cardiomyocytes, skin, and skeletal muscle as well as organs like the liver to replace the diseased organ in the animal and pre-clinical trials. This article reviews the recent articles about genetically modified spheroid cells and explains the fabrication, applications, development timeline, limitations, and future directions of genetically modified cell spheroid.
Collapse
Affiliation(s)
- Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Yi Xu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Mengdie Fu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Danji Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Na Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| |
Collapse
|
33
|
Textor M, Hoburg A, Lehnigk R, Perka C, Duda GN, Reinke S, Blankenstein A, Hochmann S, Stockinger A, Resch H, Wolf M, Strunk D, Geissler S. Chondrocyte Isolation from Loose Bodies-An Option for Reducing Donor Site Morbidity for Autologous Chondrocyte Implantation. Int J Mol Sci 2023; 24:ijms24021484. [PMID: 36675010 PMCID: PMC9867247 DOI: 10.3390/ijms24021484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/21/2022] [Accepted: 12/31/2022] [Indexed: 01/15/2023] Open
Abstract
Loose bodies (LBs) from patients with osteochondritis dissecans (OCD) are usually removed and discarded during surgical treatment of the defect. In this study, we address the question of whether these LBs contain sufficient viable and functional chondrocytes that could serve as a source for autologous chondrocyte implantation (ACI) and how the required prolonged in vitro expansion affects their phenotype. Chondrocytes were isolated from LBs of 18 patients and compared with control chondrocyte from non-weight-bearing joint regions (n = 7) and bone marrow mesenchymal stromal cells (BMSCs, n = 6) obtained during primary arthroplasty. No significant differences in the initial cell yield per isolation and the expression of the chondrocyte progenitor cell markers CD44 + /CD146+ were found between chondrocyte populations from LBs (LB-CH) and control patients (Ctrl-CH). During long-term expansion, LB-CH exhibited comparable viability and proliferation rates to control cells and no ultimate cell cycle arrest was observed within 12 passages respectively 15.3 ± 1.1 mean cumulative populations doublings (CPD). The chondrogenic differentiation potential was comparable between LB-CH and Ctrl-CH, but both groups showed a significantly higher ability to form a hyaline cartilage matrix in vitro than BMSC. Our data suggest that LBs are a promising cell source for obtaining qualitatively and quantitatively suitable chondrocytes for therapeutic applications, thereby circumventing donor site morbidity as a consequence of the biopsies required for the current ACI procedure.
Collapse
Affiliation(s)
- Martin Textor
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Arnd Hoburg
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Centrum für Muskuloskelettale Chirugie (CBMSC), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Med Center 360 Degree Berlin, Kieler Straße 1, 12163 Berlin, Germany
| | - Rex Lehnigk
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Centrum für Muskuloskelettale Chirugie (CBMSC), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Carsten Perka
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Centrum für Muskuloskelettale Chirugie (CBMSC), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Georg N. Duda
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Centrum für Muskuloskelettale Chirugie (CBMSC), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02138, USA
| | - Simon Reinke
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Antje Blankenstein
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Sarah Hochmann
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | | | - Herbert Resch
- Department of Traumatology, Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Martin Wolf
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Dirk Strunk
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Sven Geissler
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Center for Advanced Therapies (BECAT), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany
- Correspondence:
| |
Collapse
|
34
|
Transplantation of Endothelial Progenitor Cells: Summary and prospect. Acta Histochem 2023; 125:151990. [PMID: 36587456 DOI: 10.1016/j.acthis.2022.151990] [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] [Received: 09/15/2022] [Revised: 12/17/2022] [Accepted: 12/18/2022] [Indexed: 12/31/2022]
Abstract
Endothelial Progenitor Cells (EPCs) are precursor cells of endothelial cells (ECs), which can differentiate into vascular ECs, protect from endothelial dysfunction and tissue ischemia, and reduce vascular hyperplasia. Due to these functions, EPCs are used as a candidate cell source for transplantation strategies. In recent years, a great progress was achieved in EPCs biology research, and EPCs transplantation has become a research hotspot. At present, transplanted EPCs have been used to treat ischemic diseases due to their powerful vasculogenesis and beneficial paracrine effects. Although EPCs transplantation has been proved to play an important role, the clinical application of EPCs still faces many challenges. This review briefly summarized the basic characteristics of EPCs, the process of EPCs transplantation promoting the healing of ischemic tissue, and the ways to improve the efficiency of EPCs transplantation. In addition, the application of EPCs in neurological improvement, cardiovascular and respiratory diseases and the challenges and problems in clinical application of EPCs were also discussed. In the end, the application of EPCs transplantation in regenerative medicine and tissue engineering was discussed.
Collapse
|
35
|
Oceguera-Yanez F, Avila-Robinson A, Woltjen K. Differentiation of pluripotent stem cells for modeling human skin development and potential applications. Front Cell Dev Biol 2022; 10:1030339. [PMID: 36506084 PMCID: PMC9728031 DOI: 10.3389/fcell.2022.1030339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
The skin of mammals is a multilayered and multicellular tissue that forms an environmental barrier with key functions in protection, regulation, and sensation. While animal models have long served to study the basic functions of the skin in vivo, new insights are expected from in vitro models of human skin development. Human pluripotent stem cells (PSCs) have proven to be invaluable tools for studying human development in vitro. To understand the mechanisms regulating human skin homeostasis and injury repair at the molecular level, recent efforts aim to differentiate PSCs towards skin epidermal keratinocytes, dermal fibroblasts, and skin appendages such as hair follicles and sebaceous glands. Here, we present an overview of the literature describing strategies for human PSC differentiation towards the components of skin, with a particular focus on keratinocytes. We highlight fundamental advances in the field employing patient-derived human induced PSCs (iPSCs) and skin organoid generation. Importantly, PSCs allow researchers to model inherited skin diseases in the search for potential treatments. Skin differentiation from human PSCs holds the potential to clarify human skin biology.
Collapse
Affiliation(s)
- Fabian Oceguera-Yanez
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan,*Correspondence: Fabian Oceguera-Yanez, ; Knut Woltjen,
| | | | - Knut Woltjen
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan,*Correspondence: Fabian Oceguera-Yanez, ; Knut Woltjen,
| |
Collapse
|
36
|
Organoids and Their Research Progress in Plastic and Reconstructive Surgery. Aesthetic Plast Surg 2022; 47:880-891. [PMID: 36401134 DOI: 10.1007/s00266-022-03129-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/25/2022] [Indexed: 11/19/2022]
Abstract
Organoids are 3D structures generated from stem cells. Their functions and physiological characteristics are similar to those of normal organs. They are used in disease mechanism research, new drug development, organ transplantation and other fields. In recent years, the application of 3D materials in plastic surgery for repairing injuries, filling, tissue reconstruction and regeneration has also been investigated. The PubMed/MEDLINE database was queried to search for animal and human studies published through July of 2022 with search terms related to Organoids, Plastic Surgery, Pluripotent Stem Cells, Bioscaffold, Skin Reconstruction, Bone and Cartilage Regeneration. This review presents stem cells, scaffold materials and methods for the construction of organoids for plastic surgery, and it summarizes their research progress in plastic surgery in recent years.Level of Evidence III This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
|
37
|
Extra-hematopoietic immunomodulatory role of the guanine-exchange factor DOCK2. Commun Biol 2022; 5:1246. [DOI: 10.1038/s42003-022-04078-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 10/06/2022] [Indexed: 11/16/2022] Open
Abstract
AbstractStromal cells interact with immune cells during initiation and resolution of immune responses, though the precise underlying mechanisms remain to be resolved. Lessons learned from stromal cell-based therapies indicate that environmental signals instruct their immunomodulatory action contributing to immune response control. Here, to the best of our knowledge, we show a novel function for the guanine-exchange factor DOCK2 in regulating immunosuppressive function in three human stromal cell models and by siRNA-mediated DOCK2 knockdown. To identify immune function-related stromal cell molecular signatures, we first reprogrammed mesenchymal stem/progenitor cells (MSPCs) into induced pluripotent stem cells (iPSCs) before differentiating these iPSCs in a back-loop into MSPCs. The iPSCs and immature iPS-MSPCs lacked immunosuppressive potential. Successive maturation facilitated immunomodulation, while maintaining clonogenicity, comparable to their parental MSPCs. Sequential transcriptomics and methylomics displayed time-dependent immune-related gene expression trajectories, including DOCK2, eventually resembling parental MSPCs. Severe combined immunodeficiency (SCID) patient-derived fibroblasts harboring bi-allelic DOCK2 mutations showed significantly reduced immunomodulatory capacity compared to non-mutated fibroblasts. Conditional DOCK2 siRNA knockdown in iPS-MSPCs and fibroblasts also immediately reduced immunomodulatory capacity. Conclusively, CRISPR/Cas9-mediated DOCK2 knockout in iPS-MSPCs also resulted in significantly reduced immunomodulation, reduced CDC42 Rho family GTPase activation and blunted filopodia formation. These data identify G protein signaling as key element devising stromal cell immunomodulation.
Collapse
|
38
|
Three-in-one customized bioink for islet organoid: GelMA/ECM/PRP orchestrate pro-angiogenic and immunoregulatory function. Colloids Surf B Biointerfaces 2022; 221:113017. [DOI: 10.1016/j.colsurfb.2022.113017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/30/2022] [Accepted: 11/10/2022] [Indexed: 11/15/2022]
|
39
|
Talbott HE, Mascharak S, Griffin M, Wan DC, Longaker MT. Wound healing, fibroblast heterogeneity, and fibrosis. Cell Stem Cell 2022; 29:1161-1180. [PMID: 35931028 PMCID: PMC9357250 DOI: 10.1016/j.stem.2022.07.006] [Citation(s) in RCA: 285] [Impact Index Per Article: 95.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Fibroblasts are highly dynamic cells that play a central role in tissue repair and fibrosis. However, the mechanisms by which they contribute to both physiologic and pathologic states of extracellular matrix deposition and remodeling are just starting to be understood. In this review article, we discuss the current state of knowledge in fibroblast biology and heterogeneity, with a primary focus on the role of fibroblasts in skin wound repair. We also consider emerging techniques in the field, which enable an increasingly nuanced and contextualized understanding of these complex systems, and evaluate limitations of existing methodologies and knowledge. Collectively, this review spotlights a diverse body of research examining an often-overlooked cell type-the fibroblast-and its critical functions in wound repair and beyond.
Collapse
Affiliation(s)
- Heather E Talbott
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shamik Mascharak
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michelle Griffin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Derrick C Wan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Michael T Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
40
|
Wu SD, Dai NT, Liao CY, Kang LY, Tseng YW, Hsu SH. Planar-/Curvilinear-Bioprinted Tri-Cell-Laden Hydrogel for Healing Irregular Chronic Wounds. Adv Healthc Mater 2022; 11:e2201021. [PMID: 35758924 DOI: 10.1002/adhm.202201021] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Indexed: 01/24/2023]
Abstract
Chronic cutaneous wounds from tissue trauma or extensive burns can impair skin barrier function and cause severe infection. Fabrication of a customizable tissue-engineered skin is a promising strategy for regeneration of uneven wounds. Herein, a planar-/curvilinear-bioprintable hydrogel is developed to produce tissue-engineered skin and evaluated in rat models of chronic and irregular wounds. The hydrogel is composed of biodegradable polyurethane (PU) and gelatin. The hydrogel laden with cells displays good 3D printability and structure stability. The circular wounds of normal and diabetes mellitus (DM) rats treated with planar-printed tri-cell-laden (fibroblasts, keratinocytes, and endothelial progenitor cells (EPCs)) hydrogel demonstrate full reepithelization and dermal repair as well as large amounts of neovascularization and collagen production after 28 days. Furthermore, the curvilinear module is fabricated based on the corresponding wound topography for curvilinear-bioprinting of the irregular tissue-engineered skin. The large and irregular rat skin wounds treated with curvilinear-printed tri-cell-laden hydrogel demonstrate full repair after 28 days. This planar-/curvilinear-bioprintable tri-cell-laden hydrogel shows great potential for customized biofabrication in skin tissue engineering.
Collapse
Affiliation(s)
- Shin-Da Wu
- Institute of Polymer Science and Engineering, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 10617, Taiwan
| | - Niann-Tzyy Dai
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chao-Yaug Liao
- Department of Mechanical Engineering, National Central University, Taoyuan, 32001, Taiwan
| | - Lan-Ya Kang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Wen Tseng
- Department of Mechanical Engineering, National Central University, Taoyuan, 32001, Taiwan
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 10617, Taiwan.,Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
41
|
Xie S, Chen L, Zhang M, Zhang C, Li H. Self-assembled complete hair follicle organoids by coculture of neonatal mouse epidermal cells and dermal cells in Matrigel. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:767. [PMID: 35965801 PMCID: PMC9372662 DOI: 10.21037/atm-22-3252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/08/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND 3D organoid cultures of hair follicles (HFs) are powerful models that mimic native HF for both in-depth study of HF disease and precision therapy. However, few studies have investigated the complete structure and properties of HF organoids. To investigate and characterize the complete HF organoids self-assembled by coculture of neonatal mouse epidermal cells (MECs) and dermal cells in Matrigel. METHODS Fresh epidermal and dermal cells from newborn mice (n=4) were isolated, and cocultured (1:1 ratio) in Matrigel using DMEM/F12 medium for 1 week. During the culture, an inverted microscope was used to observe the morphology of the 3D constructs. After 1 week, hematoxylin-eosin (HE) and immunofluorescence (IF) staining of HF-related markers (K5, K73, AE13, and K10), HF stem cell markers (K15, CD34, CD49f), skin-derived precursor-related marker (Nestin), and dermal papillae (DP)-specific markers (SOX2 and ALP) was performed in the harvested constructs to identify the HF organoids. RESULTS Epidermal and dermal cells self-assembled into HF organoids comprising an infundibular cyst-like structure, a lower segment-like structure, and a bulb-like structure from tail to root. The HF organoid had multiple, well-defined compartments similar to native anagen HF. Of the three segments, K73 was expressed in the inner root sheath-like layer, AE13 was localized in the hair shaft-like structure, K5, K15, CD34, and CD49f were present in the outer root sheath-like layer, Nestin labeled the connective tissue sheath-like layer, and SOX2 and ALP were expressed in the DP-like structure. Furthermore, K10 and K73 were expressed in the infundibular cyst-like structure. The expression of these molecular proteins was consistent with native anagen HF. CONCLUSIONS The complete HF organoid regenerated in Matrigel has specific compartments and is an excellent model to study HF disease and precision therapy.
Collapse
Affiliation(s)
- Sitian Xie
- Department of Plastic Surgery and Burn Center, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Liyun Chen
- Department of Plastic Surgery and Burn Center, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Mingjun Zhang
- Department of Plastic Surgery and Burn Center, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Cuiping Zhang
- Wound Healing and Cell Biology Laboratory, The First Affiliated Hospital, Chinese PLA General Hospital, Beijing, China
| | - Haihong Li
- Department of Plastic Surgery and Burn Center, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
- Department of Wound Repair, Institute of Wound Repair and Regeneration Medicine, Southern University of Science and Technology Hospital, Southern University of Science and Technology School of Medicine, Shenzhen, China
| |
Collapse
|
42
|
Wolf M, Poupardin RW, Ebner‐Peking P, Andrade AC, Blöchl C, Obermayer A, Gomes FG, Vari B, Maeding N, Eminger E, Binder H, Raninger AM, Hochmann S, Brachtl G, Spittler A, Heuser T, Ofir R, Huber CG, Aberman Z, Schallmoser K, Volk H, Strunk D. A functional corona around extracellular vesicles enhances angiogenesis, skin regeneration and immunomodulation. J Extracell Vesicles 2022; 11:e12207. [PMID: 35398993 PMCID: PMC8994701 DOI: 10.1002/jev2.12207] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/08/2022] [Accepted: 03/02/2022] [Indexed: 02/06/2023] Open
Abstract
Nanoparticles can acquire a plasma protein corona defining their biological identity. Corona functions were previously considered for cell-derived extracellular vesicles (EVs). Here we demonstrate that nano-sized EVs from therapy-grade human placental-expanded (PLX) stromal cells are surrounded by an imageable and functional protein corona when enriched with permissive technology. Scalable EV separation from cell-secreted soluble factors via tangential flow-filtration (TFF) and subtractive tandem mass-tag (TMT) proteomics revealed significant enrichment of predominantly immunomodulatory and proangiogenic proteins. Western blot, calcein-based flow cytometry, super-resolution and electron microscopy verified EV identity. PLX-EVs partly protected corona proteins from protease digestion. EVs significantly ameliorated human skin regeneration and angiogenesis in vivo, induced differential signalling in immune cells, and dose-dependently inhibited T cell proliferation in vitro. Corona removal by size-exclusion or ultracentrifugation abrogated angiogenesis. Re-establishing an artificial corona by cloaking EVs with fluorescent albumin as a model protein or defined proangiogenic factors was depicted by super-resolution microscopy, electron microscopy and zeta-potential shift, and served as a proof-of-concept. Understanding EV corona formation will improve rational EV-inspired nano-therapy design.
Collapse
Affiliation(s)
- Martin Wolf
- Cell Therapy InstituteSpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - Rodolphe W. Poupardin
- Cell Therapy InstituteSpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - Patricia Ebner‐Peking
- Cell Therapy InstituteSpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - André Cronemberger Andrade
- Cell Therapy InstituteSpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - Constantin Blöchl
- Department of BiosciencesParis Lodron University SalzburgSalzburgAustria
| | - Astrid Obermayer
- Department of BiosciencesParis Lodron University SalzburgSalzburgAustria
| | - Fausto Gueths Gomes
- Cell Therapy InstituteSpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
- Department of Transfusion Medicine and SCI‐TReCSPMUSalzburgAustria
| | - Balazs Vari
- Cell Therapy InstituteSpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - Nicole Maeding
- Cell Therapy InstituteSpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - Essi Eminger
- Cell Therapy InstituteSpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - Heide‐Marie Binder
- Cell Therapy InstituteSpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - Anna M. Raninger
- Cell Therapy InstituteSpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - Sarah Hochmann
- Cell Therapy InstituteSpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - Gabriele Brachtl
- Cell Therapy InstituteSpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - Andreas Spittler
- Core Facility Flow Cytometry and Department of SurgeryResearch LaboratoriesMedical University of ViennaViennaAustria
| | | | | | - Christian G. Huber
- Department of BiosciencesParis Lodron University SalzburgSalzburgAustria
| | | | | | - Hans‐Dieter Volk
- Berlin Institute of Health at Charité – UniversitätsmedizinBIH Centre for Regenerative Therapies (BCRT)BerlinGermany
| | - Dirk Strunk
- Cell Therapy InstituteSpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| |
Collapse
|
43
|
Gomes FG, Andrade AC, Wolf M, Hochmann S, Krisch L, Maeding N, Regl C, Poupardin R, Ebner-Peking P, Huber CG, Meisner-Kober N, Schallmoser K, Strunk D. Synergy of Human Platelet-Derived Extracellular Vesicles with Secretome Proteins Promotes Regenerative Functions. Biomedicines 2022; 10:238. [PMID: 35203448 PMCID: PMC8869293 DOI: 10.3390/biomedicines10020238] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 11/29/2022] Open
Abstract
Platelet-rich plasma is a promising regenerative therapeutic with controversial efficacy. We and others have previously demonstrated regenerative functions of human platelet lysate (HPL) as an alternative platelet-derived product. Here we separated extracellular vesicles (EVs) from soluble factors of HPL to understand the mode of action during skin-organoid formation and immune modulation as model systems for tissue regeneration. HPL-EVs were isolated by tangential-flow filtration (TFF) and further purified by size-exclusion chromatography (SEC) separating EVs from (lipo)protein-enriched soluble fractions. We characterized samples by tunable resistive pulse sensing, western blot, tandem mass-tag proteomics and super-resolution microscopy. We evaluated EV function during angiogenesis, wound healing, organoid formation and immune modulation. We characterized EV enrichment by TFF and SEC according to MISEV2018 guidelines. Proteomics showed three major clusters of protein composition separating TSEC-EVs from HPL clustering with TFF soluble fractions and TFF-EVs clustering with TSEC soluble fractions, respectively. HPL-derived TFF-EVs promoted skin-organoid formation and inhibited T-cell proliferation more efficiently than TSEC-EVs or TSEC-soluble fractions. Recombining TSEC-EVs with TSEC soluble fractions re-capitulated TFF-EV effects. Zeta potential and super-resolution imaging further evidenced protein corona formation on TFF-EVs. Corona depletion on SEC-EVs could be artificially reconstituted by TSEC late fraction add-back. In contrast to synthetic nanoparticles, which commonly experience reduced function after corona formation, the corona-bearing EVs displayed improved functionality. We conclude that permissive isolation technology, such as TFF, and better understanding of the mechanism of EV corona function are required to realize the complete potential of platelet-based regenerative therapies.
Collapse
Affiliation(s)
- Fausto Gueths Gomes
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (F.G.G.); (A.C.A.); (M.W.); (S.H.); (L.K.); (N.M.); (R.P.); (P.E.-P.)
- Department of Transfusion Medicine and SCI-TReCS, Paracelsus Medical University (PMU), 5020 Salzburg, Austria;
| | - André Cronemberger Andrade
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (F.G.G.); (A.C.A.); (M.W.); (S.H.); (L.K.); (N.M.); (R.P.); (P.E.-P.)
| | - Martin Wolf
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (F.G.G.); (A.C.A.); (M.W.); (S.H.); (L.K.); (N.M.); (R.P.); (P.E.-P.)
| | - Sarah Hochmann
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (F.G.G.); (A.C.A.); (M.W.); (S.H.); (L.K.); (N.M.); (R.P.); (P.E.-P.)
| | - Linda Krisch
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (F.G.G.); (A.C.A.); (M.W.); (S.H.); (L.K.); (N.M.); (R.P.); (P.E.-P.)
- Department of Transfusion Medicine and SCI-TReCS, Paracelsus Medical University (PMU), 5020 Salzburg, Austria;
| | - Nicole Maeding
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (F.G.G.); (A.C.A.); (M.W.); (S.H.); (L.K.); (N.M.); (R.P.); (P.E.-P.)
| | - Christof Regl
- Department for Biosciences and Medical Biology, Paris Lodron University, 5020 Salzburg, Austria; (C.R.); (C.G.H.); (N.M.-K.)
| | - Rodolphe Poupardin
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (F.G.G.); (A.C.A.); (M.W.); (S.H.); (L.K.); (N.M.); (R.P.); (P.E.-P.)
| | - Patricia Ebner-Peking
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (F.G.G.); (A.C.A.); (M.W.); (S.H.); (L.K.); (N.M.); (R.P.); (P.E.-P.)
| | - Christian G. Huber
- Department for Biosciences and Medical Biology, Paris Lodron University, 5020 Salzburg, Austria; (C.R.); (C.G.H.); (N.M.-K.)
| | - Nicole Meisner-Kober
- Department for Biosciences and Medical Biology, Paris Lodron University, 5020 Salzburg, Austria; (C.R.); (C.G.H.); (N.M.-K.)
| | - Katharina Schallmoser
- Department of Transfusion Medicine and SCI-TReCS, Paracelsus Medical University (PMU), 5020 Salzburg, Austria;
| | - Dirk Strunk
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (F.G.G.); (A.C.A.); (M.W.); (S.H.); (L.K.); (N.M.); (R.P.); (P.E.-P.)
| |
Collapse
|
44
|
Sun H, Zhang YX, Li YM. Generation of Skin Organoids: Potential Opportunities and Challenges. Front Cell Dev Biol 2021; 9:709824. [PMID: 34805138 PMCID: PMC8600117 DOI: 10.3389/fcell.2021.709824] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 10/21/2021] [Indexed: 12/03/2022] Open
Abstract
Although several types of human skin substitutes are currently available, they usually do not include important skin appendages such as hair follicles and sweat glands, or various skin-related cells, such as dermal adipocytes and sensory neurons. This highlights the need to improve the in vitro human skin generation model for use as a tool for investigating skin diseases and as a source of cells or tissues for skin regeneration. Skin organoids are generated from stem cells and are expected to possess the complexity and function of natural skin. Here, we summarize the current literatures relating to the "niches" of the local skin stem cell microenvironment and the formation of skin organoids, and then discuss the opportunities and challenges associated with multifunctional skin organoids.
Collapse
Affiliation(s)
- Hui Sun
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yi-Xuan Zhang
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yu-Mei Li
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
45
|
Krisch L, Brachtl G, Hochmann S, Andrade AC, Oeller M, Ebner-Peking P, Schallmoser K, Strunk D. Improving Human Induced Pluripotent Stem Cell-Derived Megakaryocyte Differentiation and Platelet Production. Int J Mol Sci 2021; 22:8224. [PMID: 34360992 PMCID: PMC8348107 DOI: 10.3390/ijms22158224] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
Several protocols exist for generating megakaryocytes (MKs) and platelets from human induced pluripotent stem cells (hiPSCs) with limited efficiency. We observed previously that mesoderm induction improved endothelial and stromal differentiation. We, therefore, hypothesized that a protocol modification prior to hemogenic endothelial cell (HEC) differentiation will improve MK progenitor (MKP) production and increase platelet output. We further asked if basic media composition affects MK maturation. In an iterative process, we first compared two HEC induction protocols. We found significantly more HECs using the modified protocol including activin A and CHIR99021, resulting in significantly increased MKs. MKs released comparable platelet amounts irrespective of media conditions. In a final validation phase, we obtained five-fold more platelets per hiPSC with the modified protocol (235 ± 84) compared to standard conditions (51 ± 15; p < 0.0001). The regenerative potency of hiPSC-derived platelets was compared to adult donor-derived platelets by profiling angiogenesis-related protein expression. Nineteen of 24 angiogenesis-related proteins were expressed equally, lower or higher in hiPSC-derived compared to adult platelets. The hiPSC-platelet's coagulation hyporeactivity compared to adult platelets was confirmed by thromboelastometry. Further stepwise improvement of hiPSC-platelet production will, thus, permit better identification of platelet-mediated regenerative mechanisms and facilitate manufacture of sufficient amounts of functional platelets for clinical application.
Collapse
Affiliation(s)
- Linda Krisch
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (L.K.); (G.B.); (S.H.); (A.C.A.); (P.E.-P.)
- Department of Transfusion Medicine and SCI-TReCS, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (M.O.); (K.S.)
| | - Gabriele Brachtl
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (L.K.); (G.B.); (S.H.); (A.C.A.); (P.E.-P.)
| | - Sarah Hochmann
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (L.K.); (G.B.); (S.H.); (A.C.A.); (P.E.-P.)
| | - André Cronemberger Andrade
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (L.K.); (G.B.); (S.H.); (A.C.A.); (P.E.-P.)
| | - Michaela Oeller
- Department of Transfusion Medicine and SCI-TReCS, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (M.O.); (K.S.)
| | - Patricia Ebner-Peking
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (L.K.); (G.B.); (S.H.); (A.C.A.); (P.E.-P.)
| | - Katharina Schallmoser
- Department of Transfusion Medicine and SCI-TReCS, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (M.O.); (K.S.)
| | - Dirk Strunk
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (L.K.); (G.B.); (S.H.); (A.C.A.); (P.E.-P.)
| |
Collapse
|