1
|
Kang MS, Kim JM, Jo HJ, Heo HJ, Kim YH, Park KM, Han DW. 3D bioprintable Mg 2+-incorporated hydrogels tailored for regeneration of volumetric muscle loss. Theranostics 2025; 15:2185-2200. [PMID: 39990217 PMCID: PMC11840723 DOI: 10.7150/thno.103677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/06/2025] [Indexed: 02/25/2025] Open
Abstract
Rationale: Current therapeutic approaches for volumetric muscle loss (VML) face challenges owing to limited graft availability and insufficient bioactivity. Three-dimensional (3D) bioprinting has become an alternative technology for fabricating native tissue-mimetic grafts, allowing for tailored structures and complex designs. Methods: We developed an Mg2+-incorporated bioink composed of thiolated gelatin (GtnSH) and maleimide-conjugated gelatin (GtnMI) decorated with magnesium peroxide (MgO2), referred to as a GtnSH/GtnMI/MgO2 bioink. We designed in situ crosslinking between GtnSH and GtnMI to prepare cytocompatible bioink for 3D bioprinting of muscle mimetics. Results: The incorporated MgO2 particles provided oxygen supplementation and myogenic cues. In vitro assays demonstrated that C2C12 myoblasts encapsulated in the GtnSH/GtnMI/MgO2 bioink exhibited high viability, intrinsic proliferation rate, and increased expression of key myogenic markers. In vivo transplantation of the 3D bioprinted GtnSH/GtnMI/MgO2 constructs facilitated muscle mass restoration and M2 macrophage polarization. Additionally, they downregulate the activities of CD4+ and CD8+ lymphocytes, inducing a transition from the initial inflammatory to the restoration phase. Conclusion: The GtnSH/GtnMI/MgO2 bioink is a potential therapeutic strategy for enhancing myogenesis and skeletal muscle tissue regeneration.
Collapse
Affiliation(s)
- Moon Sung Kang
- Department of Cogno-mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Jeong Min Kim
- Department of Bioengineering and Nano-bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Hyo Jung Jo
- Department of Cogno-mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Hye Jin Heo
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Yun Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Biomedical Informatics, Pusan National University, Yangsan 50612, Republic of Korea
- Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
- Periodontal Disease Signaling Network Research Center and Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Kyung Min Park
- Department of Bioengineering and Nano-bioengineering, Incheon National University, Incheon 22012, Republic of Korea
- Research Center for Bio Materials & Process Development, Incheon National University, Incheon 22012, Republic of Korea
| | - Dong-Wook Han
- Department of Cogno-mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
- Institute of Nano-Bio Convergence, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
2
|
Villani V, Frank CN, Cravedi P, Hou X, Bin S, Kamitakahara A, Barbati C, Buono R, Da Sacco S, Lemley KV, De Filippo RE, Lai S, Laviano A, Longo VD, Perin L. A kidney-specific fasting-mimicking diet induces podocyte reprogramming and restores renal function in glomerulopathy. Sci Transl Med 2024; 16:eadl5514. [PMID: 39475573 DOI: 10.1126/scitranslmed.adl5514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 06/24/2024] [Accepted: 09/11/2024] [Indexed: 11/04/2024]
Abstract
Cycles of a fasting-mimicking diet (FMD) promote regeneration and reduce damage in the pancreases, blood, guts, and nervous systems of mice, but their effect on kidney disease is unknown. In addition, a FMD has not been tested in rats. Here, we show that cycles of a newly developed low-salt FMD (LS-FMD) restored normal proteinuria and nephron structure and function in rats with puromycin-induced nephrosis compared with that in animals with renal damage that did not receive the dietary intervention. LS-FMD induced modulation of a nephrogenic gene program, resembling renal developmental processes in multiple kidney structures. LS-FMD also activated podocyte-lineage reprogramming pathways and promoted a quiescent state in mature podocytes in the rat kidney damage model. In a pilot clinical study in patients with chronic kidney disease, FMD cycles of 5 days each month for 3 months promoted renoprotection, including reduction of proteinuria and improved endothelial function, compared with that in patients who did not receive the FMD cycles. These results show that FMD cycles, which promote the reprogramming of multiple renal cell types and lead to glomerular damage reversal in rats, should be tested further for the treatment of progressive kidney diseases.
Collapse
Affiliation(s)
- Valentina Villani
- GOFARR Laboratory, Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, Los Angeles, CA 90027, USA
| | - Camille Nicolas Frank
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Paolo Cravedi
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029-5674, USA
| | - Xiaogang Hou
- GOFARR Laboratory, Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, Los Angeles, CA 90027, USA
| | - Sofia Bin
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029-5674, USA
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna 40138, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna 40126, Italy
| | - Anna Kamitakahara
- Division of Neurology, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Cristiani Barbati
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Italian National Institute of Health, Rome 00185, Italy
| | - Roberta Buono
- Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Stefano Da Sacco
- GOFARR Laboratory, Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, Los Angeles, CA 90027, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kevin V Lemley
- Department of Pediatrics, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA
| | - Roger E De Filippo
- GOFARR Laboratory, Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, Los Angeles, CA 90027, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Silvia Lai
- Department of Translational and Precision Medicine, Nephrology Unit, Sapienza University of Rome, Rome 00185, Italy
| | - Alessandro Laviano
- Department of Translational and Precision Medicine, Sapienza University, Rome 00185, Italy
| | - Valter D Longo
- Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Laura Perin
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
3
|
Shi Y, Peng J, Liu M, Qi X, Li S, Li Q, Jiang Q, Zheng L, Xu J, Zhao Y, Zhang Y. Nicotinamide mononucleotide enhances fracture healing by promoting skeletal stem cell proliferation. Theranostics 2024; 14:5999-6015. [PMID: 39346542 PMCID: PMC11426247 DOI: 10.7150/thno.98149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/04/2024] [Indexed: 10/01/2024] Open
Abstract
The process of skeletal regeneration initiated by stem cells following injury, especially in fractures, is significantly impaired by aging and adverse factors. Nicotinamide mononucleotide (NMN), a critical endogenous precursor of nicotinamide adenine dinucleotide (NAD), has garnered extensive attention for its multifaceted regulatory functions in living organisms and its wide-ranging therapeutic potential. However, whether NMN contributes to trauma-induced skeletal regeneration remains unclear. Methods: The transverse femoral shaft fracture model was employed to evaluate the potential advantages of NMN administration for overall repair during the initial fracture stages in male mice through micro-CT analysis, histochemistry, and biomechanical testing. The pro-proliferative function of NMN on skeletal stem cells (SSCs) was investigated through flow cytometry, qRT-PCR, NAD content measurement, and cell proliferation assay. Results: In this study, we observed that the administration of NMN during the initial phase of fracture in mice led to a larger callus and corresponding improvement in micro-CT parameters. NMN enhances the cartilaginous component of the callus by elevating the NAD content, consequently accelerating subsequent endochondral ossification and the fracture healing process. Subsequent analyses elucidated that NMN was beneficial in promoting the expansion of diverse stem cells in vivo and in vitro potentially via modulation of the Notch signaling pathway. Moreover, the depletion of macrophages profoundly obstructs the proliferation of SSCs. Conclusion: Our discoveries provide a potential strategy for enhancing fracture healing through stimulation of callus SSC proliferation at an early stage, shedding light on the translational value of NMN as an enhancer for skeletal regeneration and highlighting the pivotal role of macrophage-stem cell interactions in governing the regenerative influence of NMN on stem cells.
Collapse
Affiliation(s)
- Yitian Shi
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, PR China
| | - Jiayin Peng
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, PR China
| | - Mengfan Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, PR China
| | - Xiling Qi
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, PR China
| | - Siyu Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, PR China
| | - Qiangqiang Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu 210008, PR China
| | - Qing Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu 210008, PR China
| | - Liming Zheng
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, PR China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong 999077, PR China
| | - Yun Zhao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, PR China
| | - Yifeng Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, PR China
- Shanghai Clinical Research and Trial Center, Shanghai 200000, PR China
| |
Collapse
|
4
|
Kim JW, Bae JH, Go GY, Lee JR, Jeong Y, Kim JY, Kim TH, Kim YK, Han JW, Oh JE, Hahn MJ, Kang JS, Bae GU. Epsti1 Regulates the Inflammatory Stage of Early Muscle Regeneration through STAT1-VCP Interaction. Int J Biol Sci 2024; 20:3530-3543. [PMID: 38993551 PMCID: PMC11234217 DOI: 10.7150/ijbs.94675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
During muscle regeneration, interferon-gamma (IFN-γ) coordinates inflammatory responses critical for activation of quiescent muscle stem cells upon injury via the Janus kinase (JAK) - signal transducer and activator of transcription 1 (STAT1) pathway. Dysregulation of JAK-STAT1 signaling results in impaired muscle regeneration, leading to muscle dysfunction or muscle atrophy. Until now, the underlying molecular mechanism of how JAK-STAT1 signaling resolves during muscle regeneration remains largely elusive. Here, we demonstrate that epithelial-stromal interaction 1 (Epsti1), an interferon response gene, has a crucial role in regulating the IFN-γ-JAK-STAT1 signaling at early stage of muscle regeneration. Epsti1-deficient mice exhibit impaired muscle regeneration with elevated inflammation response. In addition, Epsti1-deficient myoblasts display aberrant interferon responses. Epsti1 interacts with valosin-containing protein (VCP) and mediates the proteasomal degradation of IFN-γ-activated STAT1, likely contributing to dampening STAT1-mediated inflammation. In line with the notion, mice lacking Epsti1 exhibit exacerbated muscle atrophy accompanied by increased inflammatory response in cancer cachexia model. Our study suggests a crucial function of Epsti1 in the resolution of IFN-γ-JAK-STAT1 signaling through interaction with VCP which provides insights into the unexplored mechanism of crosstalk between inflammatory response and muscle regeneration.
Collapse
Affiliation(s)
- Jee Won Kim
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, South Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, South Korea
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
| | - Ju-Hyeon Bae
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, South Korea
| | - Ga-Yeon Go
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, South Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, South Korea
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
| | - Jae-Rin Lee
- Cell and Gene Therapy Products Division, National Institute of Food and Drug Safety Evaluation, Cheongju 28159, South Korea
| | - Yideul Jeong
- Research Institute of Aging Related Disease, AniMusCure Inc., Suwon 16419, South Korea
| | - Jun-Young Kim
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
| | - Tae Hyun Kim
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, South Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, South Korea
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
| | - Yong Kee Kim
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, South Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, South Korea
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
| | - Jeung-Whan Han
- Research Center for Epigenome Regulation, School of Pharmacy, Department of Biochemistry and Molecular Biology, Sungkyunkwan University, Suwon 16419, South Korea
| | - Ji-Eun Oh
- Department of Biomedical Laboratory Science, Far East University, 76-32 Daehakgil, Gamgok-myeon, Eumseong-gun, Chungbuk-do, 27601, Korea
| | - Myong-Joon Hahn
- Research Center for Epigenome Regulation, School of Pharmacy, Department of Biochemistry and Molecular Biology, Sungkyunkwan University, Suwon 16419, South Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, South Korea
- Research Institute of Aging Related Disease, AniMusCure Inc., Suwon 16419, South Korea
| | - Gyu-Un Bae
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, South Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, South Korea
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
- Research Institute of Aging Related Disease, AniMusCure Inc., Suwon 16419, South Korea
| |
Collapse
|
5
|
Liu Q, Zhang S, Shi L, Shi J, Sun C, Wang J, Zhou W, Zhou H, Shan F, Wang H, Wang J, Ren N, Feng S, Liu H, Wang S. Osteogenic Induction and Anti-Inflammatory Effects of Calcium-Chlorogenic Acid Nanoparticles Remodel the Osteoimmunology Microenvironment for Accelerating Bone Repair. Adv Healthc Mater 2024:e2401114. [PMID: 38885954 DOI: 10.1002/adhm.202401114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/12/2024] [Indexed: 06/20/2024]
Abstract
Successful bone regeneration requires close cooperation between bone marrow mesenchymal stem cells (BMSCs) and macrophages, but the low osteogenic differentiation efficiency of stem cells and the excessive inflammatory response of immune cells hinder the development of bone repair. It is necessary to develop a strategy that simultaneously regulates the osteogenic differentiation of BMSCs and the anti-inflammatory polarization of macrophages for accelerating the bone regeneration. Herein, calcium-chlorogenic acid nanoparticles (Ca-CGA NPs) are synthesized by combining the small molecules of chlorogenic acid (CGA) with Ca2+. Ca-CGA NPs internalized by cells can be dissolved to release free CGA and Ca2+ under low pH conditions in lysosomes. In vitro results demonstrate that Ca-CGA NPs can not only enhance the osteogenic differentiation of BMSCs but also promote the phenotype transformation of macrophages from M1 to M2. Furthermore, in vivo experiments confirm that Ca-CGA NPs treatment facilitates the recovery of rat skull defect model through both osteoinduction and immunomodulation. This study develops a new Ca-CGA NPs-based strategy to induce the differentiation of BMSCs into osteoblasts and the polarization of macrophages into M2 phenotype, which is promising for accelerating bone repair.
Collapse
Affiliation(s)
- Qi Liu
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, 250022, P. R. China
| | - Shuo Zhang
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, 250022, P. R. China
| | - Lusen Shi
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - Jiapei Shi
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, 250022, P. R. China
| | - Chunhui Sun
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, 250022, P. R. China
| | - Jingang Wang
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, 250022, P. R. China
| | - Weijia Zhou
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, 250022, P. R. China
| | - Hengxing Zhou
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
| | - Fengjuan Shan
- Ji'nan Pantheum Biological Technology Limited Company, Jinan, 250100, P. R. China
| | - Hongli Wang
- Ji'nan Pantheum Biological Technology Limited Company, Jinan, 250100, P. R. China
| | - Jie Wang
- Ji'nan Pantheum Biological Technology Limited Company, Jinan, 250100, P. R. China
| | - Na Ren
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, 250022, P. R. China
| | - Shiqing Feng
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P. R. China
- The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250033, P. R. China
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, 300052, P. R. China
| | - Hong Liu
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, 250022, P. R. China
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| | - Shuping Wang
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, 250022, P. R. China
| |
Collapse
|
6
|
Guisado D, Talware S, Wang X, Davis A, Fozilov E, Etra A, Colombel JF, Schaniel C, Tastad C, Levine JE, Ferrara JLM, Chuang LS, Sabic K, Singh S, Marcellino BK, Hoffman R, Cho J, Cohen LJ. The reparative immunologic consequences of stem cell transplantation as a cellular therapy for refractory Crohn's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596699. [PMID: 38895305 PMCID: PMC11185544 DOI: 10.1101/2024.05.30.596699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Background Treatment strategies for Crohn's disease (CD) suppress diverse inflammatory pathways but many patients remain refractory to treatment. Autologous hematopoietic stem cell transplantation (SCT) has emerged as a therapy for medically refractory CD. SCT was developed to rescue cancer patients from myelosuppressive chemotherapy but its use for CD and other immune diseases necessitates reimagining SCT as a cellular therapy that restores appropriately responsive immune cell populations from hematopoietic progenitors in the stem cell autograft (i.e. immune "reset"). Here we present a paradigm to understand SCT as a cellular therapy for immune diseases and reveal how SCT re-establishes cellular immunity utilizing high-dimensional cellular phenotyping and functional studies of the stem cell grafts. Methods Immunophenotyping using CyTOF, single cell RNA sequencing (scRNA-seq) and T cell receptor (TCR) sequencing was performed on peripheral blood and intestinal tissue samples from refractory CD patients who underwent SCT. The stem cell graft from these patients was analyzed using flow cytometry and functionally interrogated using a murine model for engraftment. Results Our study revealed a remodeling of intestinal macrophages capable of supporting mucosal healing that was independently validated using multimodal studies of immune reconstitution events including CyTOF and scRNA-seq. Functional interrogation of hematopoietic stem cells (HSCs) using a xenograft model demonstrated that HSCs shape the timing of immune reconstitution, the selected reconstitution of specific cell lineages and potentially the clinical efficacy of SCT. Conclusions These studies indicate that SCT serves as a myeloid-directed cellular therapy re-establishing homeostatic intestinal macrophages that support intestinal healing and suggest refractory CD evolves from impairment of restorative functions in myeloid cells. Furthermore, we report heterogeneity among HSCs from CD patients which may drive SCT outcomes and suggests an unrecognized impact of CD pathophysiology on HSC in the marrow niche.
Collapse
|
7
|
Karthikeyan S, Asakura A. Imaging analysis for muscle stem cells and regeneration. Front Cell Dev Biol 2024; 12:1411401. [PMID: 38774645 PMCID: PMC11106391 DOI: 10.3389/fcell.2024.1411401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 05/24/2024] Open
Abstract
Composed of a diverse variety of cells, the skeletal muscle is one of the body's tissues with the remarkable ability to regenerate after injury. One of the key players in the regeneration process is the muscle satellite cell (MuSC), a stem cell population for skeletal muscle, as it is the source of new myofibers. Maintaining MuSC quiescence during homeostasis involves complex interactions between MuSCs and other cells in their corresponding niche in adult skeletal muscle. After the injury, MuSCs are activated to enter the cell cycle for cell proliferation and differentiate into myotubes, followed by mature myofibers to regenerate muscle. Despite decades of research, the exact mechanisms underlying MuSC maintenance and activation remain elusive. Traditional methods of analyzing MuSCs, including cell cultures, animal models, and gene expression analyses, provide some insight into MuSC biology but lack the ability to replicate the 3-dimensional (3-D) in vivo muscle environment and capture dynamic processes comprehensively. Recent advancements in imaging technology, including confocal, intra-vital, and multi-photon microscopies, provide promising avenues for dynamic MuSC morphology and behavior to be observed and characterized. This chapter aims to review 3-D and live-imaging methods that have contributed to uncovering insights into MuSC behavior, morphology changes, interactions within the muscle niche, and internal signaling pathways during the quiescence to activation (Q-A) transition. Integrating advanced imaging modalities and computational tools provides a new avenue for studying complex biological processes in skeletal muscle regeneration and muscle degenerative diseases such as sarcopenia and Duchenne muscular dystrophy (DMD).
Collapse
Affiliation(s)
- Smrithi Karthikeyan
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, United States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Atsushi Asakura
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, United States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
8
|
Selvakumar B, Sekar P, Samsudin AR. Intestinal macrophages in pathogenesis and treatment of gut leakage: current strategies and future perspectives. J Leukoc Biol 2024; 115:607-619. [PMID: 38198217 DOI: 10.1093/jleuko/qiad165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 11/13/2023] [Accepted: 12/08/2023] [Indexed: 01/12/2024] Open
Abstract
Macrophages play key roles in tissue homeostasis, defense, disease, and repair. Macrophages are highly plastic and exhibit distinct functional phenotypes based on micro-environmental stimuli. In spite of several advancements in understanding macrophage biology and their different functional phenotypes in various physiological and pathological conditions, currently available treatment strategies targeting macrophages are limited. Macrophages' high plasticity and diverse functional roles-including tissue injury and wound healing mechanisms-mark them as potential targets to mine for efficient therapeutics to treat diseases. Despite mounting evidence on association of gut leakage with several extraintestinal diseases, there is no targeted standard therapy to treat gut leakage. Therefore, there is an urgent need to develop therapeutic strategies to treat this condition. Macrophages are the cells that play the largest role in interacting with the gut microbiota in the intestinal compartment and exert their intended functions in injury and repair mechanisms. In this review, we have summarized the current knowledge on the origins and phenotypes of macrophages. The specific role of macrophages in intestinal barrier function, their role in tissue repair mechanisms, and their association with gut microbiota are discussed. In addition, currently available therapies and the putative tissue repair mediators of macrophages for treating microbiota dysbiosis induced gut leakage are also discussed. The overall aim of this review is to convey the intense need to screen for microbiota induced macrophage-released prorepair mediators, which could lead to the identification of potential candidates that could be developed for treating the leaky gut and associated diseases.
Collapse
Affiliation(s)
- Balachandar Selvakumar
- Department of Microbiota, Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - Priyadharshini Sekar
- Department of Microbiota, Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - A Rani Samsudin
- Department of Microbiota, Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| |
Collapse
|
9
|
Yang Q, Barbachano-Guerrero A, Fairchild LM, Rowland TJ, Dowell RD, Allen MA, Warren CJ, Sawyer SL. Macrophages derived from human induced pluripotent stem cells (iPSCs) serve as a high-fidelity cellular model for investigating HIV-1, dengue, and influenza viruses. J Virol 2024; 98:e0156323. [PMID: 38323811 PMCID: PMC10949493 DOI: 10.1128/jvi.01563-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
Macrophages are important target cells for diverse viruses and thus represent a valuable system for studying virus biology. Isolation of primary human macrophages is done by culture of dissociated tissues or from differentiated blood monocytes, but these methods are both time consuming and result in low numbers of recovered macrophages. Here, we explore whether macrophages derived from human induced pluripotent stem cells (iPSCs)-which proliferate indefinitely and potentially provide unlimited starting material-could serve as a faithful model system for studying virus biology. Human iPSC-derived monocytes were differentiated into macrophages and then infected with HIV-1, dengue virus, or influenza virus as model human viruses. We show that iPSC-derived macrophages support the replication of these viruses with kinetics and phenotypes similar to human blood monocyte-derived macrophages. These iPSC-derived macrophages were virtually indistinguishable from human blood monocyte-derived macrophages based on surface marker expression (flow cytometry), transcriptomics (RNA sequencing), and chromatin accessibility profiling. iPSC lines were additionally generated from non-human primate (chimpanzee) fibroblasts. When challenged with dengue virus, human and chimpanzee iPSC-derived macrophages show differential susceptibility to infection, thus providing a valuable resource for studying the species-tropism of viruses. We also show that blood- and iPSC-derived macrophages both restrict influenza virus at a late stage of the virus lifecycle. Collectively, our results substantiate iPSC-derived macrophages as an alternative to blood monocyte-derived macrophages for the study of virus biology. IMPORTANCE Macrophages have complex relationships with viruses: while macrophages aid in the removal of pathogenic viruses from the body, macrophages are also manipulated by some viruses to serve as vessels for viral replication, dissemination, and long-term persistence. Here, we show that iPSC-derived macrophages are an excellent model that can be exploited in virology.
Collapse
Affiliation(s)
- Qing Yang
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | | | - Laurence M. Fairchild
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Teisha J. Rowland
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Robin D. Dowell
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Linda Crnic Institute for Down Syndrome Boulder Branch, BioFrontiers Institute, Boulder, Colorado, USA
- Department of Computer Science, University of Colorado Boulder, Boulder, Colorado, USA
| | - Mary A. Allen
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Linda Crnic Institute for Down Syndrome Boulder Branch, BioFrontiers Institute, Boulder, Colorado, USA
| | - Cody J. Warren
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
- Center for Retrovirus Research, The Ohio State University, Columbus, Ohio, USA
- Center for RNA Biology, The Ohio State University, Columbus, Ohio, USA
- Viruses and Emerging Pathogens Program, Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
| | - Sara L. Sawyer
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
10
|
Yadav P, Singh SK, Rajput S, Allawadhi P, Khurana A, Weiskirchen R, Navik U. Therapeutic potential of stem cells in regeneration of liver in chronic liver diseases: Current perspectives and future challenges. Pharmacol Ther 2024; 253:108563. [PMID: 38013053 DOI: 10.1016/j.pharmthera.2023.108563] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/04/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
The deposition of extracellular matrix and hyperplasia of connective tissue characterizes chronic liver disease called hepatic fibrosis. Progression of hepatic fibrosis may lead to hepatocellular carcinoma. At this stage, only liver transplantation is a viable option. However, the number of possible liver donors is less than the number of patients needing transplantation. Consequently, alternative cell therapies based on non-stem cells (e.g., fibroblasts, chondrocytes, keratinocytes, and hepatocytes) therapy may be able to postpone hepatic disease, but they are often ineffective. Thus, novel stem cell-based therapeutics might be potentially important cutting-edge approaches for treating liver diseases and reducing patient' suffering. Several signaling pathways provide targets for stem cell interventions. These include pathways such as TGF-β, STAT3/BCL-2, NADPH oxidase, Raf/MEK/ERK, Notch, and Wnt/β-catenin. Moreover, mesenchymal stem cells (MSCs) stimulate interleukin (IL)-10, which inhibits T-cells and converts M1 macrophages into M2 macrophages, producing an anti-inflammatory environment. Furthermore, it inhibits the action of CD4+ and CD8+ T cells and reduces the activity of TNF-α and interferon cytokines by enhancing IL-4 synthesis. Consequently, the immunomodulatory and anti-inflammatory capabilities of MSCs make them an attractive therapeutic approach. Importantly, MSCs can inhibit the activation of hepatic stellate cells, causing their apoptosis and subsequent promotion of hepatocyte proliferation, thereby replacing dead hepatocytes and reducing liver fibrosis. This review discusses the multidimensional therapeutic role of stem cells as cell-based therapeutics in liver fibrosis.
Collapse
Affiliation(s)
- Poonam Yadav
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Sumeet Kumar Singh
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Sonu Rajput
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Prince Allawadhi
- Department of Pharmacy, Vaish Institute of Pharmaceutical Education and Research (VIPER), Pandit Bhagwat Dayal Sharma University of Health Sciences (Pt. B. D. S. UHS), Rohtak, Haryana 124001, India
| | - Amit Khurana
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India; Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelsstr. 30, D-52074 Aachen, Germany.
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelsstr. 30, D-52074 Aachen, Germany.
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India; Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelsstr. 30, D-52074 Aachen, Germany.
| |
Collapse
|
11
|
Krzysiek-Maczka G, Brzozowski T, Ptak-Belowska A. Helicobacter pylori-activated fibroblasts as a silent partner in gastric cancer development. Cancer Metastasis Rev 2023; 42:1219-1256. [PMID: 37460910 PMCID: PMC10713772 DOI: 10.1007/s10555-023-10122-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/20/2023] [Indexed: 12/18/2023]
Abstract
The discovery of Helicobacter pylori (Hp) infection of gastric mucosa leading to active chronic gastritis, gastroduodenal ulcers, and MALT lymphoma laid the groundwork for understanding of the general relationship between chronic infection, inflammation, and cancer. Nevertheless, this sequence of events is still far from full understanding with new players and mediators being constantly identified. Originally, the Hp virulence factors affecting mainly gastric epithelium were proposed to contribute considerably to gastric inflammation, ulceration, and cancer. Furthermore, it has been shown that Hp possesses the ability to penetrate the mucus layer and directly interact with stroma components including fibroblasts and myofibroblasts. These cells, which are the source of biophysical and biochemical signals providing the proper balance between cell proliferation and differentiation within gastric epithelial stem cell compartment, when exposed to Hp, can convert into cancer-associated fibroblast (CAF) phenotype. The crosstalk between fibroblasts and myofibroblasts with gastric epithelial cells including stem/progenitor cell niche involves several pathways mediated by non-coding RNAs, Wnt, BMP, TGF-β, and Notch signaling ligands. The current review concentrates on the consequences of Hp-induced increase in gastric fibroblast and myofibroblast number, and their activation towards CAFs with the emphasis to the altered communication between mesenchymal and epithelial cell compartment, which may lead to inflammation, epithelial stem cell overproliferation, disturbed differentiation, and gradual gastric cancer development. Thus, Hp-activated fibroblasts may constitute the target for anti-cancer treatment and, importantly, for the pharmacotherapies diminishing their activation particularly at the early stages of Hp infection.
Collapse
Affiliation(s)
- Gracjana Krzysiek-Maczka
- Department of Physiology, the Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531, Kraków, Poland.
| | - Tomasz Brzozowski
- Department of Physiology, the Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531, Kraków, Poland.
| | - Agata Ptak-Belowska
- Department of Physiology, the Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531, Kraków, Poland
| |
Collapse
|
12
|
Rashchupkin IM, Shevela EY, Maksimova AA, Tikhonova MA, Ostanin AA, Chernykh ER. Effect of Differently Polarized Human Macrophages on the SH-SY5Y Cells Damaged by Ischemia and Hypoxia In Vitro. J Immunol Res 2023; 2023:5595949. [PMID: 37692837 PMCID: PMC10484653 DOI: 10.1155/2023/5595949] [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: 05/25/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 09/12/2023] Open
Abstract
Macrophages are the major cells of innate immunity with a wide range of biological effects due to their great plasticity and heterogeneity. Macrophages play a key role in neuroregeneration following nervous tissue injury. However, the neuroregenerative potential of various macrophage phenotypes, including those polarized by efferocytosis, remains unexplored. The aim of this study was to compare the neuroregenerative and neuroprotective activity of soluble factors secreted by variously activated human macrophages on the functions of neural progenitors in an in vitro model of ischemia or ischemia/hypoxia. Macrophages were polarized by interferon-γ (M1), IL-4 (M2a), or interaction with apoptotic cells (M2(LS)). The effect of macrophages conditioned media on the proliferation, differentiation, and survival of SH-SY5Y cells damaged by serum deprivation alone (ischemic conditions) or in combination with CoCl2 (ischemic/hypoxic conditions) was assessed. All studied macrophages stimulated the proliferation and differentiation of SH-SY5Y cells. On day 3, the pro-proliferating effect of M1 and M2 was similar and did not depend on the severity of the damaging effect (ischemia or ischemia/hypoxia), while on day 7 and under ischemic/hypoxic conditions, the effects of M2(LS) exceeded those of M1 and M2a cells. The prodifferentiation effects of macrophages were manifested in both short- and long-term cultures, mainly under ischemic/hypoxic conditions, and were most characteristic of M2(LS) cells. Importantly, the ischemia/hypoxia model was accompanied by the pronounced death of SH-SY5Y cells. Only macrophages with the M2 phenotype demonstrated antiapoptotic activity, and the effect of M2(LS) was higher than that of M2a. The results obtained indicate that human macrophages have neuroprotective and neuroregenerative activity, which is mediated by soluble factors, is most characteristic for macrophages activated by efferocytosis (M2(LS)), and is most prominent under in vitro conditions simulating the combined effect of ischemia/hypoxia.
Collapse
Affiliation(s)
| | | | | | | | | | - Elena Removna Chernykh
- Scientific Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| |
Collapse
|
13
|
Giannotti L, Di Chiara Stanca B, Spedicato F, Nitti P, Damiano F, Demitri C, Calabriso N, Carluccio MA, Palermo A, Siculella L, Stanca E. Progress in Regenerative Medicine: Exploring Autologous Platelet Concentrates and Their Clinical Applications. Genes (Basel) 2023; 14:1669. [PMID: 37761809 PMCID: PMC10530962 DOI: 10.3390/genes14091669] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
The goal of regenerative medicine is to achieve tissue regeneration. In the past, commonly used techniques included autologous or allogeneic transplantation and stem cell therapy, which have limitations, such as a lack of donor sites in the case of autologous transplantation and the invasiveness of stem cell harvesting. In recent years, research has, therefore, focused on new and less invasive strategies to achieve tissue regeneration. A step forward in this direction has been made with the development of autologous platelet concentrates (APCs), which are derived from the patient's own blood. They can be classified into three generations: platelet-rich plasma (PRP), platelet-rich fibrin (PRF), and concentrated growth factors (CGFs). These APCs have different structural characteristics, depending on the distinctive preparation method, and contain platelets, leukocytes, and multiple growth factors, including those most involved in regenerative processes. The purpose of this review is to clarify the most used techniques in the field of regenerative medicine in recent years, comparing the different types of APCs and analyzing the preparation protocols, the composition of the growth factors, the level of characterization achieved, and their clinical applications to date.
Collapse
Affiliation(s)
- Laura Giannotti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.G.); (B.D.C.S.); (F.S.); (F.D.); (E.S.)
| | - Benedetta Di Chiara Stanca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.G.); (B.D.C.S.); (F.S.); (F.D.); (E.S.)
| | - Francesco Spedicato
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.G.); (B.D.C.S.); (F.S.); (F.D.); (E.S.)
| | - Paola Nitti
- Department of Engineering for Innovation, University of Salento, 73100 Lecce, Italy; (P.N.); (C.D.)
| | - Fabrizio Damiano
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.G.); (B.D.C.S.); (F.S.); (F.D.); (E.S.)
| | - Christian Demitri
- Department of Engineering for Innovation, University of Salento, 73100 Lecce, Italy; (P.N.); (C.D.)
| | - Nadia Calabriso
- National Research Council (CNR), Institute of Clinical Physiology (IFC), 73100 Lecce, Italy; (N.C.); (M.A.C.)
| | - Maria Annunziata Carluccio
- National Research Council (CNR), Institute of Clinical Physiology (IFC), 73100 Lecce, Italy; (N.C.); (M.A.C.)
| | - Andrea Palermo
- Implant Dentistry College of Medicine and Dentistry, Birmingham B4 6BN, UK;
| | - Luisa Siculella
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.G.); (B.D.C.S.); (F.S.); (F.D.); (E.S.)
| | - Eleonora Stanca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.G.); (B.D.C.S.); (F.S.); (F.D.); (E.S.)
| |
Collapse
|
14
|
Wang B, Pang M, Song Y, Wang H, Qi P, Bai S, Lei X, Wei S, Zong Z, Lin S, Zhang X, Cen X, Wang X, Yang Y, Li Y, Wang Y, Xu H, Huang L, Tortorella M, Cheng B, Lee Y, Qin D, Li G. Human fetal mesenchymal stem cells secretome promotes scarless diabetic wound healing through heat-shock protein family. Bioeng Transl Med 2023; 8:e10354. [PMID: 36684113 PMCID: PMC9842061 DOI: 10.1002/btm2.10354] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 01/25/2023] Open
Abstract
The high mortality rate of patients with diabetic foot ulcers is urging the appearance of an effective biomedical drug. Senescence is one of the major reasons of aging-induced decline in the diabetic wound. Our previous studies have demonstrated the anti-senescence effect of secretomes derived from human fetal mesenchymal stem cells (hfMSC). The present study tends to explore the potential role of hfMSC secretome (HFS) in wound healing through anti-aging. Meanwhile, we try to overcome several obstacles in the clinical application of stem cell secretome. A verticle bioreactor and microcarriers are employed to expand hfMSC and produce the HFS on a large scale. The HFS was then subjected to lyophilization (L-HFS). The PLGA (poly lactic-co-glycolic acid) particles were used to encapsulate and protect L-HFS from degradation in the streptozotocin (STZ)-induced diabetic rat model. Results showed that HFS-PLGA significantly enhanced wound healing by promoting vascularization and inhibiting inflammation in the skin wound bed. We further analyzed the contents of HFS. Isobaric tag for relative and absolute quantitation (ITRAQ) and label-free methods were used to identify peptides in the secretome. Bioinformatics analysis indicated that exosome production-related singling pathways and heat-shock protein family could be used as bio-functional markers and quality control for stem cell secretome production.
Collapse
Affiliation(s)
- Bin Wang
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education InstitutesThe Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- The Chinese University of Hong Kong (CUHK)‐Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GDL) Advanced Institute for Regenerative MedicineBioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| | - Mengru Pang
- Department of Burn and Plastic SurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
| | - Yancheng Song
- Department of orthopedicsThe Affiliated Hospital of Guangdong Pharmaceutical UniversityGuangzhouChina
| | - Haixing Wang
- Department of Orthopaedics and Traumatology, Stem Cells, and Regenerative Medicine LaboratoryLi Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales HospitalShatinHong Kong
| | - Pan Qi
- Department of Orthopaedics and Traumatology, Stem Cells, and Regenerative Medicine LaboratoryLi Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales HospitalShatinHong Kong
| | - Shanshan Bai
- Department of Orthopaedics and Traumatology, Stem Cells, and Regenerative Medicine LaboratoryLi Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales HospitalShatinHong Kong
| | - Xiaoxuan Lei
- Department of Oral and Maxillofacial Surgery/PathologyAmsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement ScienceAmsterdamThe Netherlands
| | - Shikun Wei
- Department of Plastic SurgeryGeneral Hospital of Southern Theater Command, PLAGuangzhouChina
| | - Zhixian Zong
- Department of Orthopaedics and Traumatology, Stem Cells, and Regenerative Medicine LaboratoryLi Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales HospitalShatinHong Kong
| | - Sien Lin
- Department of Orthopaedics and Traumatology, Stem Cells, and Regenerative Medicine LaboratoryLi Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales HospitalShatinHong Kong
| | - Xiaoting Zhang
- Department of Orthopaedics and Traumatology, Stem Cells, and Regenerative Medicine LaboratoryLi Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales HospitalShatinHong Kong
| | - Xiaotong Cen
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education InstitutesThe Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- The Chinese University of Hong Kong (CUHK)‐Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GDL) Advanced Institute for Regenerative MedicineBioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| | - Xia Wang
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education InstitutesThe Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- The Chinese University of Hong Kong (CUHK)‐Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GDL) Advanced Institute for Regenerative MedicineBioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| | - Yongkang Yang
- Department of Orthopaedics and Traumatology, Stem Cells, and Regenerative Medicine LaboratoryLi Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales HospitalShatinHong Kong
| | - Yuan Li
- Department of Orthopaedics and Traumatology, Stem Cells, and Regenerative Medicine LaboratoryLi Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales HospitalShatinHong Kong
| | - Yan Wang
- Department of Burn and Plastic SurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
| | - Hongjie Xu
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education InstitutesThe Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- The Chinese University of Hong Kong (CUHK)‐Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GDL) Advanced Institute for Regenerative MedicineBioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| | - Lin Huang
- Division of Plastic, Reconstructive and Aesthetic Surgery, Department of SurgeryThe Chinese University of Hong Kong, Prince of Wales HospitalShatinHong Kong
| | - Micky Tortorella
- Centre for Regenerative Medicine and HealthHong Kong Institute of Science and Innovation, Chinese Academy of SciencesHong KongChina
| | - Biao Cheng
- Department of Plastic SurgeryGeneral Hospital of Southern Theater Command, PLAGuangzhouChina
| | - Yukwai Lee
- Department of Orthopaedics and Traumatology, Stem Cells, and Regenerative Medicine LaboratoryLi Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales HospitalShatinHong Kong
| | - Dajiang Qin
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education InstitutesThe Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- The Chinese University of Hong Kong (CUHK)‐Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GDL) Advanced Institute for Regenerative MedicineBioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| | - Gang Li
- Department of Orthopaedics and Traumatology, Stem Cells, and Regenerative Medicine LaboratoryLi Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales HospitalShatinHong Kong
| |
Collapse
|
15
|
Skeletal Muscle Stem Cells in Aging: Asymmetric/Symmetric Division Switching. Symmetry (Basel) 2022. [DOI: 10.3390/sym14122676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In aged muscle, satellite cells’ symmetric and asymmetric divisions are impaired, and intrinsic and extrinsic complex mechanisms govern these processes. This review presents many updated aspects regarding muscle stem cells’ fate in normal and aging conditions. The balance between self-renewal and commitment divisions contributes to muscle regeneration, muscle homeostasis, aging, and disease. Stimulating muscle regeneration in aging could be a therapeutic target, but there is still a need to understand the many mechanisms that influence each other in satellite cells and their niche. We highlight here the general outlines regarding satellite cell divisions, the primary markers present in muscle stem cells, the aging aspects concerning signaling pathways involved in symmetric/asymmetric divisions, the regenerative capacity of satellite cells and their niche alteration in senescent muscle, genetics and epigenetics mechanisms implied in satellite cells aging and exercise effect on muscle regeneration in the elderly.
Collapse
|
16
|
Palermo A, Giannotti L, Di Chiara Stanca B, Ferrante F, Gnoni A, Nitti P, Calabriso N, Demitri C, Damiano F, Batani T, Lungherini M, Carluccio MA, Rapone B, Qorri E, Scarano A, Siculella L, Stanca E, Rochira A. Use of CGF in Oral and Implant Surgery: From Laboratory Evidence to Clinical Evaluation. Int J Mol Sci 2022; 23:15164. [PMID: 36499489 PMCID: PMC9736623 DOI: 10.3390/ijms232315164] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/03/2022] Open
Abstract
Edentulism is the condition of having lost natural teeth, and has serious social, psychological, and emotional consequences. The need for implant services in edentulous patients has dramatically increased during the last decades. In this study, the effects of concentrated growth factor (CGF), an autologous blood-derived biomaterial, in improving the process of osseointegration of dental implants have been evaluated. Here, permeation of dental implants with CGF has been obtained by using a Round up device. These CGF-coated dental implants retained a complex internal structure capable of releasing growth factors (VEGF, TGF-β1, and BMP-2) and matrix metalloproteinases (MMP-2 and MMP-9) over time. The CGF-permeated implants induced the osteogenic differentiation of human bone marrow stem cells (hBMSC) as confirmed by matrix mineralization and the expression of osteogenic differentiation markers. Moreover, CGF provided dental implants with a biocompatible and biologically active surface that significantly improved adhesion of endothelial cells on CGF-coated implants compared to control implants (without CGF). Finally, data obtained from surgical interventions with CGF-permeated dental implants presented better results in terms of optimal osseointegration and reduced post-surgical complications. These data, taken together, highlight new and interesting perspectives in the use of CGF in the dental implantology field to improve osseointegration and promote the healing process.
Collapse
Affiliation(s)
- Andrea Palermo
- College of Medicine and Dentistry Birmingham, University of Birmingham, Birmingham B4 6BN, UK
| | - Laura Giannotti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy
| | - Benedetta Di Chiara Stanca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy
| | | | - Antonio Gnoni
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Paola Nitti
- Department of Engineering for Innovation, Campus Ecotekne, University of Salento, Via per Monteroni, 73100 Lecce, Italy
| | - Nadia Calabriso
- National Research Council (CNR) Institute of Clinical Physiology (IFC), 73100 Lecce, Italy
| | - Christian Demitri
- Department of Engineering for Innovation, Campus Ecotekne, University of Salento, Via per Monteroni, 73100 Lecce, Italy
| | - Fabrizio Damiano
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy
| | | | | | | | - Biagio Rapone
- Interdisciplinary Department of Medicine, Aldo Moro University of Bari, 70121 Bari, Italy
| | - Erda Qorri
- Faculty of Medical Science, Albanian University, Bulevardi Zogu I, 1001 Tirana, Albania
| | - Antonio Scarano
- Department of Oral Science, Nano and Biotechnology and CeSi-Met, University of Chieti-Pescara, 66100 Chieti, Italy
| | - Luisa Siculella
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy
| | - Eleonora Stanca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy
| | - Alessio Rochira
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy
| |
Collapse
|
17
|
Li FC, Shahin-Shamsabadi A, Selvaganapathy PR, Kishen A. Engineering a novel stem cells from apical papilla - macrophages organoid for regenerative endodontics. J Endod 2022; 48:741-748. [PMID: 35245579 DOI: 10.1016/j.joen.2022.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 02/11/2022] [Accepted: 02/21/2022] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Three-dimension (3D) tissue-construct with a heterogeneous cell population is critical to understand the interactions between immune cells and stem cells from apical papilla (SCAP) in the periapical region for developing treatment strategies in regenerative endodontics. This study aims to develop and characterize a 3D tissue-construct with binary cell system for studying the interactions between SCAP and macrophages in presence of lipopolysaccharide (LPS - pro-inflammatory) and interleukin-4 (IL-4 - anti-inflammatory) environments. METHODS SCAP and macrophages were seeded in the 3D printed dumbbell-shaped molds to generate tissue-constructs with binary cell population. Two experimental (LPS and IL-4) and control (non-stimulation) conditions were applied to the tissue-constructs to determine the characteristics of the tissue-construct, volume of viable cells and their morphology using a confocal laser scanning microscopy from 0 to 7 days period. Experiments were conducted in triplicates and data were analyzed with trend analysis and two-way analysis of variance at the significance of p < 0.05. RESULTS The tissue-constructs revealed distinct SCAP-macrophage interaction in pro-/anti-inflammatory environments. SCAP displayed characteristic self-organization as a cap-shaped structure in the tissue-construct. The growth of cells and cell-to-cell as well as cell-to-matrix interactions resulted in 70% and 30% decreased dimension of the tissue graft on the SCAP side and macrophage side respectively at day 7 (p < 0.0001). The tissue environments influenced macrophages-SCAP interactions, resulting in altered viable cell volume (p < 0.05), morphology and structural organization. CONCLUSIONS This study developed and characterized an apical papilla organoid in a 3D collagen based tissue-construct for studying SCAP-macrophage crosstalk in tissue regeneration as well as repair.
Collapse
Affiliation(s)
- Fang-Chi Li
- The Kishen Lab, Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, Canada
| | | | - P Ravi Selvaganapathy
- School of Biomedical Engineering, McMaster University, Hamilton, Canada; Department of Mechanical Engineering, McMaster University, Hamilton, Canada
| | - Anil Kishen
- The Kishen Lab, Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, Canada; Department of Dentistry, Mount Sinai Health System, Toronto, Canada
| |
Collapse
|
18
|
Dang J, Yang J, Yu Z, Chen L, Zhang Z, Wang K, Tang J, Yi C. Bone marrow mesenchymal stem cells enhance angiogenesis and promote fat retention in fat grafting via polarized macrophages. Stem Cell Res Ther 2022; 13:52. [PMID: 35120568 PMCID: PMC8817529 DOI: 10.1186/s13287-022-02709-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/09/2022] [Indexed: 11/30/2022] Open
Abstract
Background Fat grafting is one of the most common soft tissue filling methods in plastic surgery. Bone marrow mesenchymal stem cell (BM-MSC) transplantation is an effective method for improving graft retention. However, the role of BM-MSCs in fat transplantation is not completely clear. Methods Human fat particles, together with BM-MSCs or PBS as a control, were subcutaneously transplanted into the backs of nude mice. Samples were taken on days 14, 30 and 90 post-grafting to calculate the fat graft retention rate, and tissue staining was evaluated. Furthermore, macrophages were treated with BM-MSC conditioned medium (BM-MSC-CM) to identify the beneficial component secreted by these stem cells. Results In this study, we found that BM-MSCs improved retention by enhancing angiogenesis in fat grafting. Further analysis revealed that BM-MSCs could significantly inhibit the expression of the proinflammatory M1 macrophage markers interleukin (IL)-1β, tumor necrosis factor-α (TNF-α) and IL-6 in the early stages of fat grafting and promote the expression of the anti-inflammatory M2 macrophage markers Arg1, IL-10 and VEGF. Furthermore, our results showed that IL-10 secreted by BM-MSCs induced M2 macrophage polarization in vitro. Conclusions BM-MSC transplantation can improve the fat retention rate and promote angiogenesis, which may be related to M2 macrophages. These results help elucidate the role of BM-MSCs in fat grafting.
Collapse
Affiliation(s)
- Juanli Dang
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jizhong Yang
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhou Yu
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lin Chen
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhaoxiang Zhang
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Kai Wang
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiezhang Tang
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chenggang Yi
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China. .,Department of Plastic Surgery, The Second Affiliated Hospital, Medical School, Zhejiang University, Hangzhou, China.
| |
Collapse
|
19
|
Analysis of CGF Biomolecules, Structure and Cell Population: Characterization of the Stemness Features of CGF Cells and Osteogenic Potential. Int J Mol Sci 2021; 22:ijms22168867. [PMID: 34445573 PMCID: PMC8396261 DOI: 10.3390/ijms22168867] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/12/2021] [Accepted: 08/15/2021] [Indexed: 01/08/2023] Open
Abstract
Concentrated Growth Factors (CGF) represent new autologous (blood-derived biomaterial), attracting growing interest in the field of regenerative medicine. In this study, the chemical, structural, and biological characterization of CGF was carried out. CGF molecular characterization was performed by GC/MS to quantify small metabolites and by ELISA to measure growth factors and matrix metalloproteinases (MMPs) release; structural CGF characterization was carried out by SEM analysis and immunohistochemistry; CGF has been cultured, and its primary cells were isolated for the identification of their surface markers by flow cytometry, Western blot, and real-time PCR; finally, the osteogenic differentiation of CGF primary cells was evaluated through matrix mineralization by alizarin red staining and through mRNA quantification of osteogenic differentiation markers by real-time PCR. We found that CGF has a complex inner structure capable of influencing the release of growth factors, metabolites, and cells. These cells, which could regulate the production and release of the CGF growth factors, show stem features and are able to differentiate into osteoblasts producing a mineralized matrix. These data, taken together, highlight interesting new perspectives for the use of CGF in regenerative medicine.
Collapse
|