1
|
Shi J, Li Y, Zhao H, Yan C, Cui R, Wen Y, Yu X, Ding W, Zhao Y, Fang Y. Single-Cell Transcriptomics Reveals Stem Cell-Derived Exosomes Attenuate Inflammatory Gene Expression in Pulmonary Oxygen Toxicity. Int J Mol Sci 2025; 26:4462. [PMID: 40362698 PMCID: PMC12072907 DOI: 10.3390/ijms26094462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
In recent years, the role played by exosomes in lung diseases has been investigated. Exosomes have been shown to contribute to reductions in lung inflammation and pulmonary fibrosis. However, the role played by exosomes in pulmonary oxygen toxicity and the mechanism involved have not yet been reported. In the present work, we aimed to investigate the mechanism by which stem cell exosomes protect lung tissue and the potential molecular regulatory network involved. In this study, we employed single-cell RNA sequencing techniques to elucidate the unique cellular and molecular mechanisms underlying the progression of exosome therapy for pulmonary oxygen toxicity. We found changes in cell populations after exosome treatment, characterized by the expression of different molecular markers. We also integrated single-cell RNA sequencing (scRNA-seq) and bulk analysis to identify the protective effects of mesenchymal stem cell exosomes (MSC-Exos) in a mouse pulmonary oxygen toxicity (POT) model. scRNA-seq revealed dynamic shifts in the lung cellular composition after exosome treatment, including a reduction in inflammatory lymphoid cells (NK, B cells, CD8+ T, CD4+ T) and restored alveolar epithelial populations (AT1/AT2). A comprehensive gene expression analysis showed that inflammatory pathways associated with oxidative stress were significantly upregulated. In addition, our analysis of the intercellular interaction network revealed that there was a significant reduction in intercellular signal transduction in the POT group compared to the exosome-treated group. These results not only shed light on the unique cellular heterogeneity and potential pathogenesis following exosome therapy, but they also deepen our understanding of molecular pathophysiology and provide new avenues for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Jing Shi
- Naval Medical Center, Naval Medical University, Shanghai 200433, China; (J.S.); (H.Z.); (Y.W.); (X.Y.); (W.D.)
- Translational Medical Research Center, Naval Medical University, Shanghai 200433, China;
| | - Yabin Li
- Department of Life Science and Technology, Tongji University, Shanghai 200092, China;
| | - Houyu Zhao
- Naval Medical Center, Naval Medical University, Shanghai 200433, China; (J.S.); (H.Z.); (Y.W.); (X.Y.); (W.D.)
| | - Chenyang Yan
- Translational Medical Research Center, Naval Medical University, Shanghai 200433, China;
| | - Ruxia Cui
- College of Biology and Environmental Science, Jishou University, Jishou 416000, China;
| | - Yukun Wen
- Naval Medical Center, Naval Medical University, Shanghai 200433, China; (J.S.); (H.Z.); (Y.W.); (X.Y.); (W.D.)
| | - Xuhua Yu
- Naval Medical Center, Naval Medical University, Shanghai 200433, China; (J.S.); (H.Z.); (Y.W.); (X.Y.); (W.D.)
| | - Wei Ding
- Naval Medical Center, Naval Medical University, Shanghai 200433, China; (J.S.); (H.Z.); (Y.W.); (X.Y.); (W.D.)
| | - Yunpeng Zhao
- Translational Medical Research Center, Naval Medical University, Shanghai 200433, China;
| | - Yiqun Fang
- Naval Medical Center, Naval Medical University, Shanghai 200433, China; (J.S.); (H.Z.); (Y.W.); (X.Y.); (W.D.)
| |
Collapse
|
2
|
Wang B, Lyu FJ, Deng Z, Zheng Q, Ma Y, Peng Y, Guo S, Lei G, Lai Y, Li Q. Therapeutic potential of stem cell-derived exosomes for bone tissue regeneration around prostheses. J Orthop Translat 2025; 52:85-96. [PMID: 40291635 PMCID: PMC12023751 DOI: 10.1016/j.jot.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/09/2025] [Accepted: 03/14/2025] [Indexed: 04/30/2025] Open
Abstract
Artificial joint replacement is a widely recognized treatment for arthritis and other severe joint conditions. However, one of the primary causes of failure in joint replacements is the loosening of the prosthesis. After implantation, wear particles between the implant and the adjacent bone tissue are the principal contributors to this loosening. Recently, exosomes have garnered significant interest due to their low immunogenicity and effective membrane binding. They have shown potential in promoting bone regeneration via the paracrine pathway. This review examines the role and mechanisms of exosomes derived from mesenchymal stem cells (MSCs) in bone regeneration, their impact on the integration of various implants into surrounding bone tissue and current challenges and future directions for the clinical application of exosomes. The Translational Potential of this Article: Emerging evidence suggests that mesenchymal stem cell-derived exosomes may offer a promising therapeutic strategy for aseptic prosthesis loosening, potentially mediated through mechanisms such as modulation of inflammatory responses, suppression of osteoclastogenesis, enhancement of osteogenic differentiation and facilitation of bone regeneration. Preclinical studies further indicate that the therapeutic potential of these extracellular vesicles could be optimized through bioengineering strategies, including surface modification and cargo-loading techniques, warranting further investigation to advance their clinical translation.
Collapse
Affiliation(s)
- Biwu Wang
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, China
| | - Feng-Juan Lyu
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510000, China
- South China University of Technology-The University of Western Australia Joint Center for Regenerative Medicine Research, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yuanchen Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yujie Peng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Shantou University Medical College, Xinling Road 22, Shantou, 515041, China
| | - Shujun Guo
- South China University of Technology-The University of Western Australia Joint Center for Regenerative Medicine Research, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Guihua Lei
- South China University of Technology-The University of Western Australia Joint Center for Regenerative Medicine Research, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Yonggang Lai
- South China University of Technology-The University of Western Australia Joint Center for Regenerative Medicine Research, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Qingtian Li
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| |
Collapse
|
3
|
Wu H, Li Y, Shi L, Liu Y, Shen J. New Advances in Periodontal Functional Materials Based on Antibacterial, Anti-Inflammatory, and Tissue Regeneration Strategies. Adv Healthc Mater 2025; 14:e2403206. [PMID: 39895157 DOI: 10.1002/adhm.202403206] [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: 08/26/2024] [Revised: 01/17/2025] [Indexed: 02/04/2025]
Abstract
With the global population aging, awareness of oral health is rising. Periodontitis, a widespread bacterial infectious disease, is gaining attention. Current novel biomaterials address key clinical issues like bacterial infection, gum inflammation, tooth loosening, and loss, focusing on antibacterial, anti-inflammatory, and tissue regeneration properties. However, strategies that integrate the advantages of these biomaterials to achieve synergistic therapeutic effects by clearing oral biofilms, inhibiting inflammation activation, and restoring periodontal soft and hard tissue functions remain very limited. Recent studies highlight the link between periodontitis and systemic diseases, underscoring the complexity of the periodontal disease. There is an urgent need to find comprehensive treatment plans that address clinical requirements. Whether by integrating new biomaterials to enhance existing periodontal treatments or by developing novel approaches to replace traditional therapies, these efforts will drive advancements in periodontitis treatment. Therefore, this review compares novel biomaterials with traditional treatments. It highlights the design concepts and mechanisms of these functional materials, focusing on their antibacterial, anti-inflammatory, and tissue regeneration properties, and discusses the importance of developing comprehensive treatment strategies. This review aims to provide guidance for emerging periodontitis research and to promote the development of precise and efficient treatment strategies.
Collapse
Affiliation(s)
- Haoyue Wu
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Yuanfeng Li
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yong Liu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Jing Shen
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
| |
Collapse
|
4
|
Li X, Hegarty K, Lin F, Chang JL, Abdalla A, Dhanabalan K, Solomevich SO, Song W, Roder K, Yao C, Lu W, Carmeliet P, Choudhary G, Dennery PA, Yao H. Endothelial Cpt1a Inhibits Neonatal Hyperoxia-Induced Pulmonary Vascular Remodeling by Repressing Endothelial-Mesenchymal Transition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415824. [PMID: 39799584 PMCID: PMC11923872 DOI: 10.1002/advs.202415824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/15/2024] [Indexed: 01/15/2025]
Abstract
Pulmonary hypertension (PH) increases the mortality of preterm infants with bronchopulmonary dysplasia (BPD). There are no curative therapies for this disease. Lung endothelial carnitine palmitoyltransferase 1a (Cpt1a), the rate-limiting enzyme of the carnitine shuttle system, is reduced in a rodent model of BPD. It is unknown whether endothelial Cpt1a reduction causes pulmonary vascular (PV) remodeling. The latter can be the result of endothelial-mesenchymal transition (EndoMT). Here, endothelial cell (EC)-specific Cpt1a KO and WT mice (<12 h old) are exposed to hyperoxia (70% O2) for 14 days and allow them to recover in normoxia until postnatal day 28. Hyperoxia causes PH, which is aggravated in EC-specific Cpt1a KO mice. Upregulating endothelial Cpt1a expression inhibits hyperoxia-induced PV remodeling. Hyperoxia causes lung EndoMT, detected by immunofluorescence, scRNA-sequencing, and EC lineage tracing, which is further increased in EC-specific Cpt1a KO mice. Blocking EndoMT inhibits hyperoxia-induced PV remodeling. Male mice under the same high oxygen conditions develop a higher degree of PH than females, which is associated with reduced endothelial Cpt1a expression. Conclusively, neonatal hyperoxia causes PH by decreasing endothelial Cpt1a expression and upregulating EndoMT. This provides a valuable strategy for developing targeted therapies by upregulating endothelial Cpt1a levels or inhibiting EndoMT to treat BPD-associated PH.
Collapse
Affiliation(s)
- Xiaoyun Li
- Department of Molecular BiologyCellular Biology, and BiochemistryBrown UniversityProvidenceRI02912USA
- Providence VA Medical CenterProvidenceRI02908USA
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
- College of PharmacyJinan UniversityGuangzhouGuangdong510632China
| | - Katy Hegarty
- Department of Molecular BiologyCellular Biology, and BiochemistryBrown UniversityProvidenceRI02912USA
| | - Fanjie Lin
- State Key Laboratory of Respiratory DiseaseGuangdong Key Laboratory of Vascular DiseaseNational Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdong510120China
| | - Jason L. Chang
- Department of Molecular BiologyCellular Biology, and BiochemistryBrown UniversityProvidenceRI02912USA
| | - Amro Abdalla
- Providence VA Medical CenterProvidenceRI02908USA
| | - Karthik Dhanabalan
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Sergey O. Solomevich
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Wenliang Song
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Karim Roder
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Chenrui Yao
- College of Arts & SciencesBoston UniversityBostonMA02215USA
| | - Wenju Lu
- State Key Laboratory of Respiratory DiseaseGuangdong Key Laboratory of Vascular DiseaseNational Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdong510120China
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular MetabolismDepartment of Oncology and Leuven Cancer InstituteKU LeuvenVIB Center for Cancer Biology, VIBLeuvenBrussels3000Belgium
- Center for BiotechnologyKhalifa UniversityAbu Dhabi127788UAE
| | - Gaurav Choudhary
- Providence VA Medical CenterProvidenceRI02908USA
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Phyllis A. Dennery
- Department of Molecular BiologyCellular Biology, and BiochemistryBrown UniversityProvidenceRI02912USA
- Department of PediatricsWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Hongwei Yao
- Department of Molecular BiologyCellular Biology, and BiochemistryBrown UniversityProvidenceRI02912USA
- Providence VA Medical CenterProvidenceRI02908USA
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| |
Collapse
|
5
|
Wang Y, Ding H, Bai R, Li Q, Ren B, Lin P, Li C, Chen M, Xu X. Exosomes from adipose-derived stem cells accelerate wound healing by increasing the release of IL-33 from macrophages. Stem Cell Res Ther 2025; 16:80. [PMID: 39984984 PMCID: PMC11846291 DOI: 10.1186/s13287-025-04203-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC) -derived exosomes, especially adipose-derived mesenchymal stem cell exosomes (ADSC-Exos), have emerged as a promising alternative for skin damage repair with anti-inflammatory, angiogenic and cell proliferation effects while overcoming some of the limitations of MSC. However, the mechanism by which ADSC-Exos regulates inflammatory cells during wound healing remains unclear. This study investigated how ADSC-Exos regulate macrophages to promote wound healing. METHODS ADSC-Exos were isolated using ultracentrifugation, with subsequent quantification of exosomes particle number. To investigate their role in wound healing, the effects of ADSC-Exos on inflammation, angiogenesis, collagen deposition and macrophage polarization were evaluated through immunohistochemical staining, immunofluorescence and western blotting. Changes in gene expression associated with ADSC-Exos-induced macrophage polarization were analyzed using qPCR. RNA sequencing was performed to identify differentially expressed genes affected by ADSC-Exos. The critical role of IL-33 in the wound healing process was further confirmed using Il33-/- mice. Additionally, co-culture experiments were conducted to explore the effects of IL-33 on keratinocyte proliferation, collagen deposition and epithelialization. RESULTS ADSC-Exos inhibited the expression of TNF-α and IL-6, induced M2 macrophage polarization, promoted collagen deposition and angiogenesis, and accelerated wound healing. RNA sequencing identified IL-33 as a key mediator in this process. In Il33-/- mice, impaired wound healing and decreased M2 macrophage polarization were observed. The co-culture experiments showed that IL-33 enhanced keratinocyte function through activation of the Wnt/β-catenin signaling pathway. These findings highlight the therapeutic potential of ADSC-Exos in wound healing by modulating IL-33. CONCLUSIONS ADSC-Exos promote wound healing by regulating macrophage polarization and enhancing IL-33 release which drives keratinocyte proliferation, collagen deposition and epithelialization via the Wnt/β-catenin signaling pathway. These findings provide a mechanistic basis for the therapeutic potential of ADSC-Exos in tissue repair and regeneration.
Collapse
Affiliation(s)
- Yichen Wang
- Senior Department of Burns and Plastic Surgery, the Fourth Medical Center of Chinese PLA General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100048, China
- Chinese PLA Medical School , Beijing, 100853, China
| | - Hongfan Ding
- Senior Department of Burns and Plastic Surgery, the Fourth Medical Center of Chinese PLA General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Ruiqi Bai
- Senior Department of Burns and Plastic Surgery, the Fourth Medical Center of Chinese PLA General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Qiang Li
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Boyuan Ren
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Pianpian Lin
- Senior Department of Burns and Plastic Surgery, the Fourth Medical Center of Chinese PLA General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Chengfei Li
- Senior Department of Burns and Plastic Surgery, the Fourth Medical Center of Chinese PLA General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Minliang Chen
- Senior Department of Burns and Plastic Surgery, the Fourth Medical Center of Chinese PLA General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100048, China.
| | - Xiao Xu
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, No. 69 Yongding Road, Haidian District, Beijing, 100039, People's Republic of China.
| |
Collapse
|
6
|
Cerveró-Varona A, Prencipe G, Peserico A, Canciello A, House AH, Santos HA, Perugini M, Sulcanese L, Takano C, Miki T, Iannetta A, Russo V, Mattioli M, Barboni B. Amniotic epithelial Cell microvesicles uptake inhibits PBMCs and Jurkat cells activation by inducing mitochondria-dependent apoptosis. iScience 2025; 28:111830. [PMID: 39967871 PMCID: PMC11834128 DOI: 10.1016/j.isci.2025.111830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/15/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Amniotic epithelial cells (AECs) exhibit significant immunomodulatory and pro-regenerative properties, largely due to their intrinsic paracrine functions that are currently harnessed through the collection of their secretomes. While there is increasing evidence of the role of bioactive components freely secreted or carried by exosomes, the bioactive cargo of AEC microvesicles (MVs) and their crosstalk with the immune cells remains to be fully explored. We showed that under intrinsic conditions or in response to LPS, AEC-derived MV carries components such as lipid-mediated signaling molecules, ER, and mitochondria. They foster the intra/interspecific mitochondrial transfer into immune cells (PBMCs and Jurkat cells) in vitro and in vivo on the zebrafish larvae model of injury. The internalization of MV cargoes through macropinocytosis induces hyperpolarization of PBMC mitochondrial membranes and triggers MV-mediated apoptosis. This powerful immune suppressive mechanism triggered by AEC-MV cargo delivery paves the way for controlled and targeted cell-free therapeutic approaches.
Collapse
Affiliation(s)
- Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Alessia Peserico
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Angelo Canciello
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Andrew H. House
- Helsinki University Lipidomics Unit, Helsinki Institute for Life Science (HiLIFE), Biocenter 3, Viikinkaari 1, 00790 Helsinki, Finland
| | - Hélder A. Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, 9713 AV Groningen, the Netherlands
| | - Monia Perugini
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Ludovica Sulcanese
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Chika Takano
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Toshio Miki
- Department of Physiology, Nihon University School of Medicine, Tokyo, Japan
| | - Annamaria Iannetta
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Valentina Russo
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Mauro Mattioli
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
7
|
Chen W, Kongsomros S, Thorman A, Esfandiari L, Morrow AL, Chutipongtanate S, Newburg DS. Extracellular vesicles and preterm infant diseases. Front Pediatr 2025; 13:1550115. [PMID: 40034714 PMCID: PMC11873092 DOI: 10.3389/fped.2025.1550115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/06/2025] [Indexed: 03/05/2025] Open
Abstract
With the continuous improvement in perinatal care, the number of viable preterm infants is gradually increasing, along with the rise in preterm-related diseases such as necrotizing enterocolitis, bronchopulmonary dysplasia, perinatal brain injury, retinopathy of prematurity, and sepsis. Due to the unique pathophysiology of preterm infants, diagnosing and treating these diseases has become particularly challenging, significantly affecting their survival rate and long-term quality of life. Extracellular vesicles (EVs), as key mediators of intercellular communication, play an important regulatory role in the pathophysiology of these diseases. Because of their biological characteristics, EVs could serve as biomarkers and potential therapeutic agents for preterm-related diseases. This review summarizes the biological properties of EVs, their relationship with preterm-related diseases, and their prospects for diagnosis and treatment. EVs face unique challenges and opportunities for clinical applications.
Collapse
Affiliation(s)
- Wenqain Chen
- Department of Neonatology, Fujian Maternity and Child Health Hospital; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Supasek Kongsomros
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Alexander Thorman
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Leyla Esfandiari
- Department of Biomedical Engineering, University of Cincinnati College of Engineering, Cincinnati, OH, United States
- Extracellular Vesicle Working Group, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Ardythe L. Morrow
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Extracellular Vesicle Working Group, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Department of Infectious Disease, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Somchai Chutipongtanate
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Extracellular Vesicle Working Group, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - David S. Newburg
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
8
|
Meng Y, Yao Z, Ke X, Hu M, Ren H, Gao S, Zhang H. Extracellular vesicles-based vaccines: Emerging immunotherapies against cancer. J Control Release 2025; 378:438-459. [PMID: 39667569 DOI: 10.1016/j.jconrel.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
Cancer vaccines are promising therapeutic approaches to enhance specific T-cell immunity against most solid tumors. By stimulating anti-tumor immunity, clearing minimal residual disease, and minimizing adverse effects, these vaccines target tumor cells and are effective when combined with immune checkpoint blockade or other immunotherapies. However, the development of tumor cell-based vaccines faces quality issues due to poor immunogenicity, tumor heterogeneity, a suppressive tumor immune microenvironment, and ineffective delivery methods. In contrast, extracellular vesicles (EVs), naturally released by cells, are considered the ideal drug carriers and vaccine platforms. EVs offer highly organ-specific targeting, induce broader and more effective immune responses, and demonstrate superior tissue delivery ability. The development of EV vaccines is crucial for advancing cancer immunotherapy. Compared to cell-based vaccines, EV vaccines produced under Good Manufacturing Practices (GMP) offer advantages such as high safety, ease of preservation and transport, and a wide range of sources. This review summarizes the latest research findings on EV vaccine and potential applications in this field. It also highlights novel neoantigens for the development of EV vaccines against cancer.
Collapse
Affiliation(s)
- Yuhua Meng
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Zhimeng Yao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China; Department of Urology Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xiurong Ke
- Department of Surgery, Laboratory for Translational Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mengyuan Hu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Hongzheng Ren
- Gongli Hospital of Shanghai Pudong New Area, Department of Pathology, Shanghai, China
| | - Shegan Gao
- College of Clinical Medicine, The First Affiliated Hospital of Henan University of Science and Technology, Henan Key Laboratory of Cancer Epigenetics, Luoyang, Henan, China.
| | - Hao Zhang
- Gongli Hospital of Shanghai Pudong New Area, Department of Pathology, Shanghai, China; Department of Pathology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, Guangdong, China; Department of Thoracic Surgery and General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
9
|
Saneh H, Wanczyk H, Walker J, Finck C. Stem cell-derived extracellular vesicles: a potential intervention for Bronchopulmonary Dysplasia. Pediatr Res 2025; 97:497-509. [PMID: 39251881 PMCID: PMC12014501 DOI: 10.1038/s41390-024-03471-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/06/2024] [Accepted: 07/16/2024] [Indexed: 09/11/2024]
Abstract
Despite advances in neonatal care, the incidence of Bronchopulmonary Dysplasia (BPD) remains high among extreme preterm infants. The pathogenesis of BPD is multifactorial, with inflammation playing a central role. There is strong evidence that stem cell therapy reduces inflammatory changes and restores normal lung morphology in animal models of hyperoxia-induced lung injury. These therapeutic effects occur without significant engraftment of the stem cells in the host lung, suggesting more of a paracrine mechanism mediated by their secretome. In addition, there are multiple concerns with stem cell therapy which may be alleviated by administering only the effective vesicles instead of the cells themselves. Extracellular vesicles (EVs) are cell-derived components secreted by most eukaryotic cells. They can deliver their bioactive cargo (mRNAs, microRNAs, proteins, growth factors) to recipient cells, which makes them a potential therapeutic vehicle in many diseases, including BPD. The following review will highlight recent studies that investigate the effectiveness of EVs derived from stem cells in preventing or repairing injury in the preterm lung, and the potential mechanisms of action that have been proposed. Current limitations will also be discussed as well as suggestions for advancing the field and easing the transition towards clinical translation in evolving or established BPD. IMPACT: Extracellular vesicles (EVs) derived from stem cells are a potential intervention for neonatal lung diseases. Their use might alleviate the safety concerns associated with stem cell therapy. This review highlights recent studies that investigate the effectiveness of stem cell-derived EVs in preclinical models of bronchopulmonary dysplasia. It adds to the existing literature by elaborating on the challenges associated with EV research. It also provides suggestions to advance the field and ease the transition towards clinical applications. Optimizing EV research could ultimately improve the quality of life of extreme preterm infants born at vulnerable stages of lung development.
Collapse
Affiliation(s)
- Hala Saneh
- Department of Neonatal Medicine, Connecticut Children's Medical Center, Hartford, CT, USA.
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA.
| | - Heather Wanczyk
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA
| | - Joanne Walker
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA
| | - Christine Finck
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA
- Department of Pediatric Surgery, Connecticut Children's Medical Center, Hartford, CT, USA
| |
Collapse
|
10
|
Wang H, Li L, Zhou G, Wang L, Wu Z. RPL39 Was Associated With Sex Differences in Pulmonary Arterial Hypertension. Can Respir J 2025; 2025:7139235. [PMID: 39957991 PMCID: PMC11824382 DOI: 10.1155/carj/7139235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 01/09/2025] [Indexed: 02/18/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is a malignant cardiovascular disease with a complex etiology, in which several types of cells play important roles. Sex differences in disease susceptibility and survival have been observed in PAH patients, but few studies have analyzed the effect of changes in cell type and number on sex differences in PAH at the single-cell level. In this study, we performed a series of analyses on GSE169471 and GSE228644 datasets and found significant changes in the ratio of several types of cells in male PAH lung tissues. Surprisingly, we found that the ratio of macrophages in male PAH samples was 7 times higher than that in females. Consistently, the ratio of M1 macrophages was also significantly increased in male PAH samples. The different expression genes (DEGs) in macrophages were mainly involved in the ribosome pathway, which is closely related to cell proliferation. Inhibition of ribosomal protein L39 (RPL39), a core gene in the ribosome pathway, can inhibit macrophage proliferation and attenuate the sex differences in PAH. In conclusion, our study suggests that ribosome pathway-associated cell proliferation of macrophages might be associated with sex differences in PAH.
Collapse
Affiliation(s)
- Haixia Wang
- National Health Commission Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (Co-Construction), Department of Scientific Research, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, China
- Department of Preventive Medicine, Shihezi University Medical School Shihezi, Xinjiang, China
| | - Ling Li
- Department of Preventive Medicine, Shihezi University Medical School Shihezi, Xinjiang, China
| | - Guangyuan Zhou
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lu Wang
- Department of Respiratory and Critical Care Medicine, Miyun Teaching Hospital of Capital Medical University, Beijing, China
| | - Zeang Wu
- National Health Commission Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (Co-Construction), Department of Scientific Research, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
11
|
Liao S, Guo F, Xiao Z, Xiao H, Pan QR, Guo Y, Chen J, Wang X, Wang S, Huang H, Yang L, Liu HF, Pan Q. Autophagy activation within inflammatory microenvironment improved the therapeutic effect of MSC-Derived extracellular Vesicle in SLE. J Adv Res 2025:S2090-1232(25)00063-3. [PMID: 39880074 DOI: 10.1016/j.jare.2025.01.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/06/2025] [Accepted: 01/25/2025] [Indexed: 01/31/2025] Open
Abstract
INTRODUCTION Developing strategies to improve the therapeutic efficacy of mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) in autoimmune diseases have garnered increased attention. OBJECTIVES To evaluate whether rapamycin-induced autophagy within the systemic lupus erythematosus (SLE) inflammatory microenvironment (Rapa-SLE) augments the therapeutic effects of MSC-derived EVs in SLE. METHODS The therapeutic potential of the resulting EVs (Rapa-SLE-EV) was assessed in MRL/lpr mice. Rapa-SLE-EVs were compared with EVs derived from MSCs from MSCs cultured with EV-depleted fetal bovine serum (FBS-EV), EVs from MSCs cultured with rapamycin-treated FBS (Rapa-FBS-EV), and EVs exposed to SLE serum without rapamycin (SLE-EV). The autoimmune response, renal function, and pathological damage were assessed among the mouse groups. Additionally, mechanistic investigations into the role of the anti-inflammatory protein IDO1 within the EVs. RESULTS Interaction with the SLE inflammatory microenvironment triggered autophagy in MSCs, which was further enhanced by rapamycin treatment. Rapa-SLE-EV administration significantly ameliorated the autoimmune response and renal damage in MRL/lpr mice, outperforming other MSC-EV groups. This treatment mitigated key manifestations of SLE, including reduced autoantibody levels, as well as splenomegaly, and lymphadenopathy. Furthermore, Rapa-SLE-EV demonstrated superior suppression of plasma inflammatory cytokines, preserved renal function, mitigated pathological damage, and reduced glomerular immune complex deposition. Mechanistically, Rapa-SLE-EV exhibits exceptional inhibitory effects on SLE-B cell function, benefited by the high expression of the anti-inflammatory protein IDO1, which was confirmed to enter SLE-B cells through EVs. CONCLUSIONS We developed a novel strategy to improve the therapeutic efficacy of MSC-EVs in SLE and confirmed that the immunomodulatory function of the MSC-EVs is enhanced through autophagic activation and interaction with the SLE serum microenvironment, a process likely benefited by the high expression of IDO1.
Collapse
Affiliation(s)
- Shuzhen Liao
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; Yue Bei People's Hospital Postdoctoral Innovation Practice Base, Southern Medical University, Guangzhou 510515, China
| | - Fengbiao Guo
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Zengzhi Xiao
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Quan-Ren Pan
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Yugan Guo
- Department of Radiation Oncology, Yuebei People's Hospital Affiliated to Shantou University School of Medicine, Shaoguan 512000, China
| | - Jiaxuan Chen
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Xi Wang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Shuting Wang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Haimin Huang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Lawei Yang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Hua-Feng Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China.
| | - Qingjun Pan
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; Department of Clinical Laboratory, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University. Guangzhou 510120, China.
| |
Collapse
|
12
|
Xu J, Li L, Zhou Y, Abudureheman Z, Xue L, Wu C, Zou X. Immunopathological characteristics and therapeutic effects of UC-MSCs in a pigeon breeder's lung mouse model. Acta Biochim Biophys Sin (Shanghai) 2025; 57:473-485. [PMID: 39844643 PMCID: PMC11986440 DOI: 10.3724/abbs.2025010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025] Open
Abstract
Hypersensitivity pneumonitis (HP), including pigeon breeder's lung (PBL), often progresses from acute inflammation to fibrosis, impairing lung function and limiting targeted therapeutic strategies. Mechanistic studies on PBL progression are limited by the lack of preclinical animal models and a predominant focus on patient data. This study explores the immunopathological characteristics of all stages of PBL in mice and evaluates the therapeutic potential of human umbilical cord-derived mesenchymal stem cells (UC-MSCs) during the non-fibrotic stage. PBL models are created in A/J mice through tracheal instillation of pigeon dropping extract (PDE) protein powder. Different doses (0.4 × 10 6, 0.8 × 10 6, and 1.6 × 10 6 cells per animal) and frequencies (1-2 times) are administered to the model. The immunopathological characteristics of PBL and the therapeutic effects of UC-MSCs are assessed using micro-CT, pulmonary function, histopathology, cell counts in BALF, HYP levels, inflammatory factor levels, immunohistochemistry, and fibrosis marker expression in lung tissues. The results show that PDE exposure consistently impairs pulmonary function and increases the levels of inflammation and fibrosis markers as the disease progresses. Model mice experience non-fibrotic stages (acute inflammation) from days 0-36, mild fibrosis from days 37-77, and severe fibrosis from day 78 onwards. UC-MSCs, particularly at the highest dose (1.6 × 10 6 cells), effectively treat non-fibrotic PBL by improving pulmonary function (lung ventilation area recovers) and reducing inflammation and fibrosis. This study successfully establishes PBL mouse models reflecting both the acute (inflammatory) and chronic (fibrotic) stages, and UC-MSCs have the potential to delay fibrosis, providing new therapeutic options for PBL and other inflammation-induced lung fibrotic diseases.
Collapse
Affiliation(s)
- Jingran Xu
- The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi830000China
| | - Li Li
- Department of Respiratory and Critical Care MedicineFirst People′s Hospital of KashiKashi844000China
| | - Yaping Zhou
- Department of Respiratory and Critical Care MedicineFirst People′s Hospital of KashiKashi844000China
| | - Zulipikaer Abudureheman
- Clinical Research Center of Infectious Diseases (Pulmonary Tuberculosis)First People’s Hospital of KashiKashi844000China
| | - Lexin Xue
- Department of Respiratory and Critical Care MedicineFirst People′s Hospital of KashiKashi844000China
| | - Chao Wu
- Department of Respiratory and Critical Care Medicinethe First Affiliated Hospital of Shihezi UniversityShihezi832061China
| | - Xiaoguang Zou
- Department of Respiratory and Critical Care MedicineFirst People′s Hospital of KashiKashi844000China
| |
Collapse
|
13
|
Doktor F, Antounians L, Figueira RL, Khalaj K, Duci M, Zani A. Amniotic fluid stem cell extracellular vesicles as a novel fetal therapy for pulmonary hypoplasia: a review on mechanisms and translational potential. Stem Cells Transl Med 2025; 14:szae095. [PMID: 39823257 PMCID: PMC11740888 DOI: 10.1093/stcltm/szae095] [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: 07/24/2024] [Accepted: 11/30/2024] [Indexed: 01/30/2025] Open
Abstract
Disruption of developmental processes affecting the fetal lung leads to pulmonary hypoplasia. Pulmonary hypoplasia results from several conditions including congenital diaphragmatic hernia (CDH) and oligohydramnios. Both entities have high morbidity and mortality, and no effective therapy that fully restores normal lung development. Hypoplastic lungs have impaired growth (arrested branching morphogenesis), maturation (decreased epithelial/mesenchymal differentiation), and vascularization (endothelial dysfunction and vascular remodeling leading to postnatal pulmonary hypertension). Herein, we discuss the pathogenesis of pulmonary hypoplasia and the role of microRNAs (miRNAs) during normal and pathological lung development. Since multiple cells and pathways are altered, the ideal strategy for hypoplastic lungs is to deliver a therapy that addresses all aspects of abnormal lung development. In this review, we report on a novel regenerative approach based on the administration of extracellular vesicles derived from amniotic fluid stem cells (AFSC-EVs). Specifically, we describe the effects of AFSC-EVs in rodent and human models of pulmonary hypoplasia, their mechanism of action via release of their cargo, including miRNAs, and their anti-inflammatory properties. We also compare cargo contents and regenerative effects of EVs from AFSCs and mesenchymal stromal cells (MSCs). Overall, there is compelling evidence that antenatal administration of AFSC-EVs rescues multiple features of fetal lung development in experimental models of pulmonary hypoplasia. Lastly, we discuss the steps that need to be taken to translate this promising EV-based therapy from the bench to the bedside. These include strategies to overcome barriers commonly associated with EV therapeutics and specific challenges related to stem cell-based therapies in fetal medicine.
Collapse
Affiliation(s)
- Fabian Doktor
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada M5G 0A4
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, Canada M5G 1X8
- Department of Pediatric Surgery, Leipzig University, Leipzig 04109, Germany
| | - Lina Antounians
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada M5G 0A4
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, Canada M5G 1X8
| | - Rebeca Lopes Figueira
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada M5G 0A4
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, Canada M5G 1X8
| | - Kasra Khalaj
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada M5G 0A4
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, Canada M5G 1X8
| | - Miriam Duci
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada M5G 0A4
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, Canada M5G 1X8
| | - Augusto Zani
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada M5G 0A4
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, Canada M5G 1X8
- Department of Surgery, University of Toronto, Toronto, ON, Canada M5T 1P5
| |
Collapse
|
14
|
Kim JE, Lee JW, Cha GD, Yoon JK. The Potential of Mesenchymal Stem Cell-Derived Exosomes to Treat Diabetes Mellitus. Biomimetics (Basel) 2025; 10:49. [PMID: 39851765 PMCID: PMC11760843 DOI: 10.3390/biomimetics10010049] [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: 12/12/2024] [Revised: 12/27/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Diabetes mellitus (DM) is a fatal metabolic disease characterized by persistent hyperglycemia. In recent studies, mesenchymal stem cell (MSC)-derived exosomes, which are being investigated clinically as a cell-free therapy for various diseases, have gained attention due to their biomimetic properties that closely resemble natural cellular communication systems. These MSC-derived exosomes inherit the regenerative and protective effects from MSCs, inducing pancreatic β-cell proliferation and inhibiting apoptosis, as well as ameliorating insulin resistance by suppressing the release of various inflammatory cytokines. Consequently, MSC-derived exosomes have attracted attention as a novel treatment for DM as an alternative to stem cell therapy. In this review, we will introduce the potential of MSC-derived exosomes for the treatment of DM by discussing the studies that have used MSC-derived exosomes to treat DM, which have shown therapeutic effects in both type 1 and type 2 DM.
Collapse
Affiliation(s)
| | | | | | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si 17546, Gyeonggi-do, Republic of Korea (G.D.C.)
| |
Collapse
|
15
|
Doktor F, Figueira RL, Fortuna V, Biouss G, Stasiewicz K, Obed M, Khalaj K, Antounians L, Zani A. Amniotic fluid stem cell extracellular vesicles promote lung development via TGF-beta modulation in a fetal rat model of oligohydramnios. J Control Release 2025; 377:427-441. [PMID: 39577465 DOI: 10.1016/j.jconrel.2024.11.043] [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: 06/08/2024] [Revised: 10/17/2024] [Accepted: 11/16/2024] [Indexed: 11/24/2024]
Abstract
Oligohydramnios (decreased amniotic fluid volume for gestational age) is a severe condition associated with high morbidity and mortality mainly due to fetal pulmonary hypoplasia. Currently, there are limited treatment options to promote fetal lung development. Administration of stem cells and their derivates have shown promising regenerative properties for several fetal and neonatal diseases related to arrested lung development. Herein, we first characterized pulmonary hypoplasia secondary to oligohydramnios in a surgical rat model. Experimental induction of oligohydramnios led to impaired lung growth, branching morphogenesis (fewer airspaces with decreased Fgf10, Nrp1, Ctnnb1 expression), proximal/distal progenitor cell patterning (decreased Sox2 and Sox9 expression), and TGF-β signaling. We then tested antenatal administration of extracellular vesicles derived from amniotic fluid stem cells (AFSC-EVs). In oligohydramnios lungs, AFSC-EV administration improved lung branching morphogenesis and airway progenitor cell patterning at least in part through the release of miR-93-5p. Our experiments suggest that AFSC-EV miR-93-5p blocked SMAD 7, resulting in upregulation of pSMAD2/3 and restoration of TGF-β signaling. Conversely, oligohydramnios lungs treated with antagomir 93-5p transfected AFSC-EVs had decreased branching morphogenesis and TGF-β signaling. This is the first study reporting that antenatal administration of stem cell derivatives could be a potential therapy to rescue lung development in fetuses with oligohydramnios.
Collapse
Affiliation(s)
- Fabian Doktor
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada; Department of Pediatric Surgery, Leipzig University, Leipzig 04109, Germany
| | - Rebeca Lopes Figueira
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Victoria Fortuna
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - George Biouss
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Kaya Stasiewicz
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Mikal Obed
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Kasra Khalaj
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Lina Antounians
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Augusto Zani
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada; Department of Surgery, University of Toronto, Toronto M5T 1P5, Canada.
| |
Collapse
|
16
|
Lou S, Hu W, Wei P, He D, Fu P, Ding K, Chen Z, Dong Z, Zheng J, Wang K. Artificial Nanovesicles Derived from Cells: A Promising Alternative to Extracellular Vesicles. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22-41. [PMID: 39692623 DOI: 10.1021/acsami.4c12567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
As naturally secreted vesicles by cells, extracellular vesicles (EVs) play essential roles in modulating cell-cell communication and have significant potential in tissue regeneration, immune regulation, and drug delivery. However, the low yield and uncontrollable heterogeneity of EVs have been obstacles to their widespread translation into clinical practice. Recently, it has been discovered that artificial nanovesicles (NVs) produced by cell processing can inherit the components and functions of the parent cells and possess similar structures and functions to EVs, with significantly higher yields and more flexible functionalization, making them a powerful complement to natural EVs. This review focuses on recent advances in the research of artificial NVs as replacements for natural EVs. We provide an overview comparing natural EVs and artificial NVs and summarize the top-down preparation strategies of NVs. The applications of NVs prepared from stem cells, differentiated cells, and engineered cells are presented, as well as the latest advances in NV engineering. Finally, the main challenges of artificial NVs are discussed.
Collapse
Affiliation(s)
- Saiyun Lou
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Wei Hu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Pengyao Wei
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo 315300, China
| | - Dongdong He
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo 315300, China
| | - Pan Fu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo 315300, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kejian Ding
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo,Zhejiang 315211, China
| | - Zhenyi Chen
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo,Zhejiang 315211, China
| | - Zhaoxing Dong
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Jianping Zheng
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo 315300, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kaizhe Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo 315300, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
17
|
Zhao S, Chen Z, Liu H, Wang X, Zhang X, Shi H. Maternal nutrition and offspring lung health: sex-specific pathway modulation in fibrosis, metabolism, and immunity. Food Nutr Res 2025; 69:11035. [PMID: 39790857 PMCID: PMC11708518 DOI: 10.29219/fnr.v69.11035] [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: 09/08/2024] [Revised: 10/26/2024] [Accepted: 11/26/2024] [Indexed: 01/12/2025] Open
Abstract
Background Maternal nutrition profoundly influences offspring health, impacting both prenatal and early postnatal development. Previous studies have demonstrated that maternal dietary habits can affect key developmental pathways in the offsprings, including those related to lung function and disease susceptibility. However, the sex-specific impact of a maternal high-salt diet (HSD) on offspring lung injury remains poorly understood. Objective This study aimed to investigate the sex-specific effects of maternal HSD on lung injury in mouse offsprings, focusing on pathways related to fibrosis, metabolism, immunity, and apoptosis. Design Pregnant C57BL/6J mice were subjected to either normal or HSD conditions during gestation. Lung tissues from the male and female offsprings were analyzed using high-throughput RNA sequencing and bioinformatics tools to examine transcriptomic changes. Wet-lab validation, including Masson trichrome staining, immunofluorescence for α-SMA, and qRT-PCR for fibrotic markers (α-SMA, collagen I, Fn1, and TGF-β), was conducted to confirm fibrosis and other injury markers. Lung structure and weight were also evaluated to assess physical alterations due to maternal diet. Results Maternal HSD significantly altered lung transcriptomes in a sex-specific manner. Male offsprings showed increased susceptibility to fibrosis, as confirmed by histological and molecular analyses, including elevated expression of α-SMA, collagen I, Fn1, and TGF-β. In contrast, female offsprings exhibited distinct changes in metabolic and immune pathways. These findings highlight the differential regulation of pulmonary injury mechanisms between male and female offsprings exposed to HSD. Conclusions Maternal HSD induces sex-specific lung injury in offsprings by disrupting critical pathways involved in fibrosis, metabolism, immunity, and apoptosis. The combination of transcriptomic and orthogonal data underscores the need for balanced maternal nutrition during pregnancy to promote long-term respiratory health in offsprings. These results provide new insights into the sex-specific vulnerabilities to lung disease arising from maternal diet.
Collapse
Affiliation(s)
- Shuangyi Zhao
- Department of Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhimin Chen
- Department of Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huina Liu
- Department of Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinyan Wang
- Department of Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiuru Zhang
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Huirong Shi
- Department of Gynaecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
18
|
Forbes LM, Bauer N, Bhadra A, Bogaard HJ, Choudhary G, Goss KN, Gräf S, Heresi GA, Hopper RK, Jose A, Kim Y, Klouda T, Lahm T, Lawrie A, Leary PJ, Leopold JA, Oliveira SD, Prisco SZ, Rafikov R, Rhodes CJ, Stewart DJ, Vanderpool RR, Yuan K, Zimmer A, Hemnes AR, de Jesus Perez VA, Wilkins MR. Precision Medicine for Pulmonary Vascular Disease: The Future Is Now (2023 Grover Conference Series). Pulm Circ 2025; 15:e70027. [PMID: 39749110 PMCID: PMC11693987 DOI: 10.1002/pul2.70027] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Pulmonary vascular disease is not a single condition; rather it can accompany a variety of pathologies that impact the pulmonary vasculature. Applying precision medicine strategies to better phenotype, diagnose, monitor, and treat pulmonary vascular disease is increasingly possible with the growing accessibility of powerful clinical and research tools. Nevertheless, challenges exist in implementing these tools to optimal effect. The 2023 Grover Conference Series reviewed the research landscape to summarize the current state of the art and provide a better understanding of the application of precision medicine to managing pulmonary vascular disease. In particular, the following aspects were discussed: (1) Clinical phenotypes, (2) genetics, (3) epigenetics, (4) biomarker discovery, (5) application of precision biology to clinical trials, (6) the right ventricle (RV), and (7) integrating precision medicine to clinical care. The present review summarizes the content of these discussions and the prospects for the future.
Collapse
Affiliation(s)
- Lindsay M. Forbes
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of ColoradoAuroraColoradoUSA
| | - Natalie Bauer
- Department of PharmacologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileAlabamaUSA
| | - Aritra Bhadra
- Department of PharmacologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
- Center for Lung BiologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
| | - Harm J. Bogaard
- Department of Pulmonary MedicineAmsterdam UMCAmsterdamNetherlands
| | - Gaurav Choudhary
- Division of CardiologyWarren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
- Lifespan Cardiovascular InstituteRhode Island and Miriam HospitalsProvidenceRhode IslandUSA
- Department of CardiologyProvidence VA Medical CenterProvidenceRhode IslandUSA
| | - Kara N. Goss
- Department of Medicine and PediatricsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Stefan Gräf
- Division of Computational Genomics and Genomic Medicine, Department of MedicineUniversity of Cambridge, Victor Phillip Dahdaleh Heart & Lung Research InstituteCambridgeUK
| | | | - Rachel K. Hopper
- Department of PediatricsStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Arun Jose
- Division of Pulmonary, Critical Care, and Sleep MedicineUniversity of CincinnatiCincinnatiOhioUSA
| | - Yunhye Kim
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Timothy Klouda
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Tim Lahm
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of ColoradoAuroraColoradoUSA
- Division of Pulmonary, Critical Care, and Sleep MedicineNational Jewish HealthDenverColoradoUSA
- Pulmonary and Critical Care SectionRocky Mountain Regional VA Medical CenterDenverColoradoUSA
| | - Allan Lawrie
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Peter J. Leary
- Departments of Medicine and EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Jane A. Leopold
- Division of Cardiovascular MedicineBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Suellen D. Oliveira
- Department of Anesthesiology, Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Sasha Z. Prisco
- Division of CardiovascularLillehei Heart Institute, University of MinnesotaMinneapolisMinnesotaUSA
| | - Ruslan Rafikov
- Department of MedicineIndiana UniversityIndianapolisIndianaUSA
| | | | - Duncan J. Stewart
- Ottawa Hospital Research InstituteFaculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | | | - Ke Yuan
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Alexsandra Zimmer
- Department of MedicineBrown UniversityProvidenceRhode IslandUSA
- Lifespan Cardiovascular InstituteRhode Island HospitalProvidenceRhode IslandUSA
| | - Anna R. Hemnes
- Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary and Critical Care MedicineStanford University Medical CenterStanfordCaliforniaUSA
| | | |
Collapse
|
19
|
Pierro M, Thébaud B. Cell-based strategies for the treatment of injury to the developing lung. THE LUNG 2025:403-426. [DOI: 10.1016/b978-0-323-91824-4.00020-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
20
|
Wang N, Ma F, Song H, He N, Zhang H, Li J, Liu Q, Xu C. Mesenchymal Stem Cell-Derived Extracellular Vesicles for Regenerative Applications and Radiotherapy. Cell Transplant 2025; 34:9636897241311019. [PMID: 39780320 PMCID: PMC11713979 DOI: 10.1177/09636897241311019] [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: 09/26/2024] [Revised: 12/12/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Tissue repair is an extremely crucial part of clinical treatment. During the course of disease treatment, surgery, chemotherapy, and radiotherapy cause tissue damage. On the other hand, Normal tissue from accidental or therapeutic exposure to high-dose radiation can cause severe tissue damage. There is an urgent need for developing medical countermeasures against radiation injury for tissue repair. Tissue repair involves the regeneration, proliferation, differentiation, and migration of tissue cells; imbalance of local tissue homeostasis, progressive chronic inflammation; decreased cell activity and stem cell function; and wound healing. Although many clinical treatments are currently available for tissue repair, they are expensive. The long recovery time and some unavoidable complications such as cell damage and the inflammatory reaction caused by radiotherapy have led to unsatisfactory results. Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) have similar tissue repair functions as MSCs. In tissue damage, EVs can be used as an alternative to stem cell therapy, thereby avoiding related complications such as immunological rejection. EVs play a major role in regulating tissue damage, anti-inflammation, pro-proliferation, and immune response, thus providing a diversified and efficient solution for the repair of disease- and radiotherapy-induced tissue damage. This article reviews the research progress of mesenchymal stem cell-derived EVs in promoting the repair of tissue including heart, lung, liver, intestine, skin, blood system, central nervous system, and tissue damage caused by radiotherapy, thereby aiming to offer new directions and ideas for the radiotherapy and regenerative applications.
Collapse
Affiliation(s)
- Ning Wang
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
| | - Feifei Ma
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
| | - Huijuan Song
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Ningning He
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Huanteng Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Jianguo Li
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
| | - Qiang Liu
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Chang Xu
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| |
Collapse
|
21
|
Lu Y, Huangfu S, Ma C, Ding Y, Zhang Y, Zhou C, Liao L, Li M, You J, Chen Y, Wang D, Chen A, Jiang B. Exosomes derived from umbilical cord mesenchymal stem cells promote healing of complex perianal fistulas in rats. Stem Cell Res Ther 2024; 15:414. [PMID: 39732731 DOI: 10.1186/s13287-024-04028-0] [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] [Received: 07/23/2024] [Accepted: 10/28/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Complex perianal fistulas, challenging to treat and prone to recurrence, often require surgical intervention that may cause fecal incontinence and lower quality of life due to large surgical wounds and potential sphincter damage. Human umbilical cord-derived MSCs (hUC-MSCs) and their exosomes (hUCMSCs-Exo) may promote wound healing. METHODS This study assessed the efficacy, mechanisms, and safety of these exosomes in treating complex perianal fistulas in SD rats. We established a rat model, divided rats with fistulas into the control and the exosome groups. We assessed treatment efficacy through ultrasound, clinical observations, and histopathological analysis. We also evaluated the activation of the HIF-1α/TGF-β/Smad signaling pathway via PCR and Western blot and assessed serological markers for HIF-1α and inflammatory indices through ELISA. We analyzed gut microbiota and the systemic metabolic environment via untargeted metabolomics. RESULTS The hUCMSCs-Exo effectively promoted healing of wound, regulated the immune balance enhanced collagen synthesis and angiogenesis in the perianal fistulas model of rats, and regulated the gut microbiota and metabolomic profiles. Results of PCR and Western blot analyses indicated that the exosomes activated HIF-1α/TGF-β/Smad signaling pathways. To the dosages tested, the 10ug/100ul concentration (medium dose) was found to be the most effective to the treatment of complex perianal fistulas. CONCLUSIONS The hUCMSCs-Exo significantly promoted the healing of wound in perianal fistulas of rats and demonstrated higher safety. The underlying mechanism facilitating the healing process was likely associated with the activation of the HIF-1α/TGF-β/Smad signaling pathway.
Collapse
Affiliation(s)
- Yafei Lu
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Shaohua Huangfu
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Chuanxue Ma
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Yan Ding
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Yajie Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
- Department of Biobank, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Chungen Zhou
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Lianming Liao
- Center of Laboratory Medicine, Union Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, People's Republic of China
| | - Ming Li
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui, People's Republic of China
| | - Jia You
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Yuting Chen
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Dawei Wang
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Ao Chen
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Bin Jiang
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China.
| |
Collapse
|
22
|
Thiruvenkataramani RP, Abdul-Hafez A, Kesaraju T, Mohamed H, Ibrahim SA, Othman A, Arif H, Zarea AA, Abdulmageed M, Arellano MG, Mohamed T, Kanada M, Madhukar BV, Omar SA. Small Extracellular Vesicles Derived from Cord Blood Plasma and Placental Mesenchymal Stem Cells Attenuate Acute Lung Injury Induced by Lipopolysaccharide (LPS). Int J Mol Sci 2024; 26:75. [PMID: 39795932 PMCID: PMC11719573 DOI: 10.3390/ijms26010075] [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: 11/18/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
Sepsis is a risk factor associated with increasing neonatal morbidity and mortality, acute lung injury, and chronic lung disease. While stem cell therapy has shown promise in alleviating acute lung injury, its effects are primarily exerted through paracrine mechanisms rather than local engraftment. Accumulating evidence suggests that these paracrine effects are mediated by mesenchymal stem cell (MSC)-derived small extracellular vesicles (sEVs), which play a critical role in immune system modulation and tissue regeneration. sEVs contain a diverse cargo of mRNA, miRNA, and proteins, contributing to their therapeutic potential. We hypothesize that sEVs derived from three distinct sources, cord blood plasma (CBP), Wharton jelly (WJ), and placental (PL) MSCs, may prevent the cytotoxicity induced by E. coli lipopolysaccharide (LPS) in lung alveolar epithelial cells. Objective: To determine the effects of CBP-, WJ-, and PL-MSCs-derived sEVs on cell viability, apoptosis, and proinflammatory cytokine production in alveolar epithelial cells and monocytes following LPS treatment. sEVs were collected from conditioned media of PL-MSCs, WJ-MSCs, and CBP using 50 nm membrane filters. sEVs were characterized based on nanoparticle tracking analysis (NTA), transmission electron microscopy (TEM), and Western blotting techniques. The protein concentration of isolated sEVs was used to standardize treatment doses. A549 cells and monocyte THP-1 cells were cultured and exposed to LPS in the presence or absence of sEVs for 72 h. Cell viability was measured using CellTiter-Glo 2.0 chemiluminescence-based assay. For cytokine analysis, A549 and THP-1 cells were pre-incubated for 24 h with or without PL- and CBP-sEVs, followed by exposure to LPS or control conditions for an additional 24 h. The conditioned media were collected, and interleukin-6 (IL-6) and interleukin-8 (IL-8) levels were quantified using ELISA. LPS treatment significantly reduced the viability of both A549 and THP-1 cells. The presence of CB- or WJ-sEVs significantly increased cell viability compared to controls. Cells treated with PL-sEVs showed increased cell viability but did not reach statistical significance. LPS-treated cells showed a significant increase in apoptosis and elevated levels of pro-inflammatory cytokines IL-6 and IL-8. All three sEVs types (CBP-, WJ-, and PL-sEVs) significantly reduced LPS-induced apoptosis and IL-6 release. Interestingly, while WJ-sEVs decreased IL-8, both CBP- and PL-sEVs led to an increase in IL-8 compared to their respective controls. CBP-, PL-, and WJ-derived sEVs demonstrated protective effects against LPS-induced injury in alveolar epithelial cells and monocytes, as evidenced by increased cell viability and modulation of pro-inflammatory cytokine release. These findings suggest that placenta-derived sEVs have the potential to modulate the immune response, mitigate inflammation, and prevent end-organ damage in neonatal sepsis.
Collapse
Affiliation(s)
- Ranga P. Thiruvenkataramani
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (T.K.); (H.M.); (S.A.I.); (A.O.); (H.A.); (M.A.); (M.G.A.); (T.M.); (B.V.M.); (S.A.O.)
- Regional Neonatal Intensive Care Unit, University of Michigan Health-Sparrow Hospital, Lansing, MI 48912, USA
| | - Amal Abdul-Hafez
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (T.K.); (H.M.); (S.A.I.); (A.O.); (H.A.); (M.A.); (M.G.A.); (T.M.); (B.V.M.); (S.A.O.)
| | - Tulasi Kesaraju
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (T.K.); (H.M.); (S.A.I.); (A.O.); (H.A.); (M.A.); (M.G.A.); (T.M.); (B.V.M.); (S.A.O.)
- Regional Neonatal Intensive Care Unit, University of Michigan Health-Sparrow Hospital, Lansing, MI 48912, USA
| | - Hend Mohamed
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (T.K.); (H.M.); (S.A.I.); (A.O.); (H.A.); (M.A.); (M.G.A.); (T.M.); (B.V.M.); (S.A.O.)
- Regional Neonatal Intensive Care Unit, University of Michigan Health-Sparrow Hospital, Lansing, MI 48912, USA
| | - Sherif Abdelfattah Ibrahim
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (T.K.); (H.M.); (S.A.I.); (A.O.); (H.A.); (M.A.); (M.G.A.); (T.M.); (B.V.M.); (S.A.O.)
| | - Amira Othman
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (T.K.); (H.M.); (S.A.I.); (A.O.); (H.A.); (M.A.); (M.G.A.); (T.M.); (B.V.M.); (S.A.O.)
- Regional Neonatal Intensive Care Unit, University of Michigan Health-Sparrow Hospital, Lansing, MI 48912, USA
| | - Hattan Arif
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (T.K.); (H.M.); (S.A.I.); (A.O.); (H.A.); (M.A.); (M.G.A.); (T.M.); (B.V.M.); (S.A.O.)
| | - Ahmed A. Zarea
- The Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824, USA; (A.A.Z.); (M.K.)
| | - Mohammed Abdulmageed
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (T.K.); (H.M.); (S.A.I.); (A.O.); (H.A.); (M.A.); (M.G.A.); (T.M.); (B.V.M.); (S.A.O.)
- Regional Neonatal Intensive Care Unit, University of Michigan Health-Sparrow Hospital, Lansing, MI 48912, USA
| | - Myrna Gonzalez Arellano
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (T.K.); (H.M.); (S.A.I.); (A.O.); (H.A.); (M.A.); (M.G.A.); (T.M.); (B.V.M.); (S.A.O.)
- Regional Neonatal Intensive Care Unit, University of Michigan Health-Sparrow Hospital, Lansing, MI 48912, USA
| | - Tarek Mohamed
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (T.K.); (H.M.); (S.A.I.); (A.O.); (H.A.); (M.A.); (M.G.A.); (T.M.); (B.V.M.); (S.A.O.)
- Regional Neonatal Intensive Care Unit, University of Michigan Health-Sparrow Hospital, Lansing, MI 48912, USA
| | - Masamitsu Kanada
- The Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824, USA; (A.A.Z.); (M.K.)
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Burra V. Madhukar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (T.K.); (H.M.); (S.A.I.); (A.O.); (H.A.); (M.A.); (M.G.A.); (T.M.); (B.V.M.); (S.A.O.)
| | - Said A. Omar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (T.K.); (H.M.); (S.A.I.); (A.O.); (H.A.); (M.A.); (M.G.A.); (T.M.); (B.V.M.); (S.A.O.)
- Regional Neonatal Intensive Care Unit, University of Michigan Health-Sparrow Hospital, Lansing, MI 48912, USA
| |
Collapse
|
23
|
Hu C, Wang L. Advances in the treatment of liver injury based on mesenchymal stem cell-derived exosomes. Stem Cell Res Ther 2024; 15:474. [PMID: 39696473 DOI: 10.1186/s13287-024-04087-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have shown a great potential role in treating liver injury. MSCs can promote liver regeneration by differentiating into hepatocytes, and can also secrete exosomes to participate in the repair of liver injury. Increasing evidence has shown that mesenchymal stem cell-derived exosomes (MSC-EXOs) play an important role in treating liver injury. In this review, the biogenesis and function of exosomes and the characteristics of MSC-EXOs were analyzed based on recent research results. MSC-EXOs are significant in liver injuries such as liver fibrosis, liver failure, hepatocellular carcinoma, oxidative stress, and lipid steatosis, and participate in the process of liver regeneration.
Collapse
Affiliation(s)
- Changlong Hu
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, 710000, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, 710000, China.
| |
Collapse
|
24
|
Chen T, Sun W, Xu ZJ. The immune mechanisms of acute exacerbations of idiopathic pulmonary fibrosis. Front Immunol 2024; 15:1450688. [PMID: 39737178 PMCID: PMC11682984 DOI: 10.3389/fimmu.2024.1450688] [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: 06/18/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
Acute exacerbations of idiopathic pulmonary fibrosis (AE-IPF) are the leading cause of mortality among patients with IPF. There is still a lack of effective treatments for AE-IPF, resulting in a hospitalization mortality rate as high as 70%-80%. To reveal the complicated mechanism of AE-IPF, more attention has been paid to its disturbed immune environment, as patients with IPF exhibit deficiencies in pathogen defense due to local immune dysregulation. During the development of AE-IPF, the classical stimulatory signals in adaptive immunity are inhibited, while the nonclassical immune reactions (Th17) are activated, attracting numerous neutrophils and monocytes to lung tissues. However, there is limited information about the specific changes in the immune response of AE-IPF. We summarized the immune mechanisms of AE-IPF in this review.
Collapse
Affiliation(s)
- Tao Chen
- Department of Respiratory and Critical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Sun
- Department of Respiratory and Critical Medicine, The second hospital of Tianjin Medical University, Tianjin, China
| | - Zuo-jun Xu
- Department of Respiratory and Critical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
25
|
Bao T, Zhu H, Ma M, Sun T, Hu J, Li J, Cao L, Cheng H, Tian Z. Implication of m6A Methylation Regulators in the Immune Microenvironment of Bronchopulmonary Dysplasia. Biochem Genet 2024; 62:5129-5143. [PMID: 38393623 DOI: 10.1007/s10528-024-10664-1] [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: 09/09/2022] [Accepted: 01/01/2024] [Indexed: 02/25/2024]
Abstract
N6-methyladenosine (m6A) regulates gene expression and governs many important biological processes. However, the function of m6A in the development of bronchopulmonary dysplasia (BPD) remains poorly characterized. Thus, the purpose of this investigation was to evaluate the effects of m6A RNA methylation regulators on the development of BPD. BPD-related transcriptome data were downloaded from the GEO database. Differentially expressed m6A methylation regulators between BPD and control group were identified. Consensus clustering was conducted for the classification of BPD and association between clusters and BPD phenotypes were explored. Analysis of differentially expressed genes (DEGs) and immune-related DEGs was performed. The GSEA, GO and KEGG analyses were used to interpret the functional enrichments. The composition of immune cell subtypes in BPD subsets was predicted by CIBERSORT analysis. Compared with the control group, expression of most m6A regulators showed significant alteration, especially for IGF2BP1/2/3. BPD was classified into 2 subsets, and cluster 1 was correlated with severe BPD. Furthermore, the results of functional enrichment analyses showed a disturbed immune-related signaling pathway. Based on CIBERSORT analysis, we found that the proportion of immune cell subsets changed between cluster 1 and cluster 2. Our study revealed the implication of m6A methylation regulators in the development of BPD, which might provide a novel insight for the diagnosis and treatment of BPD.
Collapse
Affiliation(s)
- Tianping Bao
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Haiyan Zhu
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Mengmeng Ma
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Tingting Sun
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Jingjing Hu
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - JingYan Li
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Linxia Cao
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Huaiping Cheng
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Zhaofang Tian
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China.
| |
Collapse
|
26
|
Jin T, Liu X, Li G, Sun S, Xie L. Intravenous injection of BMSCs modulate tsRNA expression and ameliorate lung remodeling in COPD mice. Stem Cell Res Ther 2024; 15:450. [PMID: 39587604 PMCID: PMC11590572 DOI: 10.1186/s13287-024-04066-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is characterized by lung remodeling induced by chronic inflammation, presenting challenges for effective treatment. Mesenchymal stem cells (MSCs) and their extracellular vesicles (EVs) have shown promise in mitigating inflammation and tissue repairing in various diseases, including COPD. However, the optimal therapeutic pathways for different stages of COPD remain unclear. Transfer RNA-derived small RNAs (tsRNAs) are emerging as key regulators of cellular processes. However, their role in COPD and MSC therapy remains poorly understood. METHODS This study explored the optimal administration routes and efficacy of bone marrow mesenchymal stem cells (BMSCs) and their extracellular vesicles (BMSC-EVs) in treating inflammatory or emphysematous COPD stages in mouse models. Male C57BL/6 mice were exposed to cigarette smoke daily for 6 or 16 weeks, with intraperitoneal CSE injections every 10 days, to model different stages of COPD. Mice were then treated with tracheal or intravenous injections of BMSCs or BMSC-EVs. PKH26 fluorescent dye labeled BMSCs and BMSC-EVs for pulmonary distribution observation. Lung tissue inflammation, apoptosis, EMT, and collagen deposition were assessed using HE staining, TUNEL assay, immunohistochemistry, and Sirius Red staining. Gene and tsRNA expression in lung tissues were analyzed by high-throughput sequencing. Differentially expressed tsRNAs (DE-tsRNAs) were validated by RT-qPCR. Statistical analysis was performed using GraphPad Prism 9.0. Data are presented as mean ± standard deviation (SD). RESULTS In 16-week COPD mice characterized by emphysema, tracheal administration of BMSC-EVs showed more significant lung distribution and inhibition of emphysematous pathology. In 6-week COPD mice characterized by inflammation, intravenous injection of BMSCs led to significant pulmonary homing, significantly reduced lung inflammation, apoptosis, EMT, and collagen deposition (P < 0.05). High-throughput sequencing indicated BMSC treatment downregulated genes related to these processes while upregulating mitochondrial function genes. Co-expression networks of DE-tsRNAs and target genes suggested potential roles in COPD. RT-qPCR confirmed significant differential expression of two DE-tsRNAs during COPD progression and BMSC treatment (P < 0.05). CONCLUSIONS Our study provides insights into selecting MSC and MSC-EV administration routes for different COPD stages. High-throughput sequencing supports BMSCs' inhibitory effects on lung remodeling and identifies the first tsRNA expression profile in a COPD model, warranting further investigation.
Collapse
Affiliation(s)
- Ting Jin
- Department of Respiratory and Critical Care Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xianyang Liu
- Department of Respiratory and Critical Care Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Guoan Li
- Department of Respiratory and Critical Care Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shenghua Sun
- Department of Respiratory and Critical Care Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Lihua Xie
- Department of Respiratory and Critical Care Medicine, The Third Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
27
|
Hao X, Li P, Wang Y, Zhang Q, Yang F. Mesenchymal Stem Cell-Exosomal miR-99a Attenuate Silica-Induced Lung Fibrosis by Inhibiting Pulmonary Fibroblast Transdifferentiation. Int J Mol Sci 2024; 25:12626. [PMID: 39684337 DOI: 10.3390/ijms252312626] [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: 10/02/2024] [Revised: 11/15/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Silicosis is one of the most prevalent and fatal occupational diseases worldwide, with unsatisfactory clinical outcomes. This study aimed to investigate the therapeutic effect and related molecular mechanisms of how mesenchymal stem cell (MSC)-secreted exosomes alleviate SiO2-induced pulmonary fibrosis. miR-99a-5p was significantly downregulated in silicosis models via high-throughput miRNA screening, and was overlapped with miRNAs in exosomes from MSCs. miR-99a-5p was significantly downregulated in the lung of a mice silicosis model and in TGFβ1-induced NIH-3T3 cells. In contrast, fibroblast growth factor receptor 3 (FGFR3), a direct target gene of miR-99a-5p, was upregulated in vitro and in vivo. Furthermore, we demonstrated that MSC-derived exosomes deliver enriched miR-99a-5p to target cells and inhibit TGF-β1-induced fibroblast transdifferentiation to reduce collagen protein production. Similarly, in a silicosis mouse model, MSC-derived exosome treatment through the tail veins of the mice counteracted the upregulation of fibrosis-related proteins and collagen deposition in the lung of the mice. By constructing exosomal therapeutic cell models with different miR-99a expressions, we further demonstrated that miR-99a-5p might attenuate pulmonary fibrosis by regulating target protein FGFR3 and downstream mitogen-activated protein kinase (MAPK) signalling pathways. Our study demonstrated that MSC-derived exosomes ameliorate SiO2-induced pulmonary fibrosis by inhibiting fibroblast transdifferentiation and represent an attractive method of pulmonary fibrosis treatment.
Collapse
Affiliation(s)
- Xiaohui Hao
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan 063210, China
- Hebei Coordinated Innovation Center of Occupational Health and Safety, North China University of Science and Technology, Tangshan 063210, China
| | - Peiyuan Li
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Yudi Wang
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Qinxin Zhang
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Fang Yang
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan 063210, China
| |
Collapse
|
28
|
Cheng T, Mao M, Liu Y, Xie L, Shi F, Liu H, Li X. The potential therapeutic effect of human umbilical cord mesenchymal stem cell-derived exosomes in bronchopulmonary dysplasia. Life Sci 2024; 357:123047. [PMID: 39260518 DOI: 10.1016/j.lfs.2024.123047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/25/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease of preterm infants, with its incidence rising due to improved survival rates of these infants. BPD results from a combination of prenatal and postnatal factors, such as mechanical ventilation, oxygen toxicity, and infections, all of which significantly impact the prognosis and growth of affected infants. Current treatment options for BPD are largely supportive and do not address the underlying pathology. Exosomes are cell-derived bilayer-enclosed membrane structures enclosing proteins, lipids, RNAs, growth factors, cytokines and metabolites. They have become recognized as crucial regulators of intercellular communication in various physiological and pathological processes. Previous studies have revealed the therapeutic potential of human umbilical cord mesenchymal stem cells-derived exosomes (HUCMSCs-Exos) in promoting tissue repair and regeneration. Therefore, HUCMSCs-Exos maybe a promising and effective therapeutic modality for BPD. In this review, we firstly provide a comprehensive overview of BPD, including its etiology and the mechanisms of lung injury. Then we detail the isolation, characterization, and contents of HUCMSCs-Exos, and discuss their potential mechanisms of HUCMSCs-Exos in BPD treatment. Additionally, we summarize current clinical trials and discuss the challenges in translating these findings from bench to bedside. This review aims to lay the groundwork for future clinical applications of HUCMSCs-Exos in treating BPD.
Collapse
Affiliation(s)
- Tianyu Cheng
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Min Mao
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yang Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Liang Xie
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Fang Shi
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hanmin Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Xin Li
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
29
|
Doktor F, Lo E, Fortuna V, Khalaj K, Garcia M, Figueira RL, Lacher M, Antounians L, Zani A. Sex-specific differences in the severity of pulmonary hypoplasia in experimental congenital diaphragmatic hernia and implications for extracellular vesicle-based therapy. Pediatr Surg Int 2024; 40:278. [PMID: 39467854 DOI: 10.1007/s00383-024-05856-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 10/30/2024]
Abstract
PURPOSE Amniotic fluid stem cell extracellular vesicles (AFSC-EVs) hold regenerative potential to treat hypoplastic lungs secondary to congenital diaphragmatic hernia (CDH). This study aims to investigate sex-specific differences in pulmonary hypoplasia severity and responses to AFSC-EV administration in an experimental CDH mouse model. METHODS C57BL/6J dams were fed with nitrofen + bisdiamine (left-sided CDH) or olive oil only (control) at embryonic day (E) 8.5. Lungs were dissected (E18.5), grown ex vivo and treated with medium ± AFSC-EVs that were collected via ultracentrifugation and characterized (nanoparticle tracking analysis, electron microscopy, Western blotting). Pulmonary hypoplasia was assessed via mean linear intercept (MLI). Gene and protein expression changes (Cd31, Enos, Il1b, TNFa) were measured via RT-qPCR and immunofluorescence. Pups were genotyped for Sry. RESULTS Experimental CDH showed a male predominance without sex differences for pulmonary hypoplasia severity, fetal lung vascularization, and inflammation. AFSC-EV administration led to improved lung growth (decreased MLI), improved fetal lung vascularization (increased Cd31 and Enos), and decreased fetal lung inflammation (Il1b, TNFa). There was no sex-specific response to AFSC-EV administration. CONCLUSION This study shows sex-independent impaired lung growth, vascularization and fetal lung inflammation in a CDH mouse model. Antenatal administration of AFSC-EVs reverses aspects of pulmonary hypoplasia secondary to CDH independent of the biological sex.
Collapse
Affiliation(s)
- Fabian Doktor
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
- Department of Pediatric Surgery, University of Leipzig, 04109, Leipzig, Germany
| | - Emily Lo
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
- Division of Neonatology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, M5G 1X8, Canada
| | - Victoria Fortuna
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Kasra Khalaj
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Miguel Garcia
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Rebeca Lopes Figueira
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Martin Lacher
- Department of Pediatric Surgery, University of Leipzig, 04109, Leipzig, Germany
| | - Lina Antounians
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Augusto Zani
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, M5G 0A4, Canada.
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.
- Department of Surgery, University of Toronto, Toronto, M5T 1P5, Canada.
| |
Collapse
|
30
|
Rao T, Zhou Y, Chen C, Chen J, Zhang J, Lin W, Jia D. Recent progress in neonatal hyperoxic lung injury. Pediatr Pulmonol 2024; 59:2414-2427. [PMID: 38742254 DOI: 10.1002/ppul.27062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/28/2024] [Accepted: 05/04/2024] [Indexed: 05/16/2024]
Abstract
With the progress in neonatal intensive care, there has been an increase in the survival rates of premature infants. However, this has also led to an increased incidence of neonatal hyperoxia lung injury and bronchopulmonary dysplasia (BPD), whose pathogenesis is believed to be influenced by various prenatal and postnatal factors, although the exact mechanisms remain unclear. Recent studies suggest that multiple mechanisms might be involved in neonatal hyperoxic lung injury and BPD, with sex also possibly playing an important role, and numerous drugs have been proposed and shown promise for improving the treatment outcomes of hyperoxic lung injury. Therefore, this paper aims to analyze and summarize sex differences in neonatal hyperoxic lung injury, potential pathogenesis and treatment progress to provide new ideas for basic and clinical research in this field.
Collapse
Affiliation(s)
- Tian Rao
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiyang Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chizhang Chen
- Department of Clinical Medicine, Chinese Medicine Hospital of Pingyang, Wenzhou, Zhejiang, China
| | - Jiayi Chen
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jie Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Lin
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Danyun Jia
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
31
|
Yu H, Li D, Zhao X, Fu J. Fetal origin of bronchopulmonary dysplasia: contribution of intrauterine inflammation. Mol Med 2024; 30:135. [PMID: 39227783 PMCID: PMC11373297 DOI: 10.1186/s10020-024-00909-5] [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: 08/16/2023] [Accepted: 08/23/2024] [Indexed: 09/05/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common chronic lung disease in infants and the most frequent adverse outcome of premature birth, despite major efforts to minimize injury. It is thought to result from aberrant repair response triggered by either prenatal or recurrent postnatal injury to the lungs during development. Intrauterine inflammation is an important risk factor for prenatal lung injury, which is also increasingly linked to BPD. However, the specific mechanisms remain unclear. This review summarizes clinical and animal research linking intrauterine inflammation to BPD. We assess how intrauterine inflammation affects lung alveolarization and vascular development. In addition, we discuss prenatal therapeutic strategies targeting intrauterine inflammation to prevent or treat BPD.
Collapse
Affiliation(s)
- Haoting Yu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Danni Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Xinyi Zhao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China.
| |
Collapse
|
32
|
Zhang S, Wang J, Xin Z, Sun C, Ju Z, Xue X, Jiang W, Xin Q, Wang J, Zhang Z, Luan Y. Effect of Oridonin on Experimental Animal Model of Bronchopulmonary Dysplasia. J Cell Biochem 2024; 125:e30632. [PMID: 39014931 DOI: 10.1002/jcb.30632] [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: 05/13/2024] [Revised: 06/24/2024] [Accepted: 07/04/2024] [Indexed: 07/18/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is a serious disease that occurs in premature and low-birth-weight infants. In recent years, the incidence of BPD has not decreased, and there is no effective treatment for it. Oridonin (Ori) is a traditional Chinese medicine with a wide range of biological activities, especially pharmacological and anti-inflammatory. It is well known that inflammation plays a key role in BPD. However, the therapeutic effect of Ori on BPD has not been studied. Therefore, in the present study, we will observe the anti-inflammatory activity of Ori in an experimental animal model of BPD. Here, we showed that Ori could significantly decrease hyperoxia-induced alveolar injury, inhibit neutrophil recruitment, myeloperoxidase concentrations, and release inflammatory factors in BPD neonatal rats. Taken together, the experimental results suggested that Ori can significantly improve BPD in neonatal rats by inhibiting inflammatory response.
Collapse
Affiliation(s)
- Shanshan Zhang
- Department of Emergency, The Second Hospital of Shandong University, Jinan, China
| | - Junfu Wang
- College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhihong Xin
- Department of Obstetrics, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Chao Sun
- Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, China
| | - Zhiye Ju
- Department of Ultrasound, Shandong Provincial Public Health Clinical Center, Jinan, China
| | - Xia Xue
- Department of Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wen Jiang
- Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, China
| | - Qian Xin
- Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, China
| | - Jue Wang
- Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, China
| | - Zhaohua Zhang
- Department of Obstetrics, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Yun Luan
- Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, China
- Renal Multidisciplinary Innovation Medical Center, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
33
|
Kumar N, Bidkhori HR, Yawno T, Lim R, Inocencio IM. Therapeutic potential of extracellular vesicles derived from human amniotic epithelial cells for perinatal cerebral and pulmonary injury. Stem Cells Transl Med 2024; 13:711-723. [PMID: 38895873 PMCID: PMC11328935 DOI: 10.1093/stcltm/szae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 04/19/2024] [Indexed: 06/21/2024] Open
Abstract
Lung and brain injury that occurs during the perinatal period leads to lifelong disability and is often driven and/or exacerbated by inflammation. Human amniotic epithelial cells (hAEC), which demonstrate immunomodulatory, anti-fibrotic, and regenerative capabilities, are being explored as a therapeutic candidate for perinatal injury. However, limitations regarding scalable manufacturing, storage, transport, and dose-related toxicity have impeded clinical translation. Isolated therapeutic extracellular vesicles (EVs) from stem and stem-like cells are thought to be key paracrine mediators of therapeutic efficacy. The unique characteristics of EVs suggest that they potentially circumvent the limitations of traditional cell-based therapies. However, given the novelty of EVs as a therapeutic, recommendations around ideal methods of production, isolation, storage, and delivery have not yet been created by regulatory agencies. In this concise review, we discuss the pertinence and limitations of cell-based therapeutics in perinatal medicine. We also review the preclinical evidence supporting the use of therapeutic EVs for perinatal therapy. Further, we summarize the arising considerations regarding adequate cell source, biodistribution, isolation and storage methods, and regulatory roadblocks for the development of therapeutic EVs.
Collapse
Affiliation(s)
- Naveen Kumar
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| | - Hamid Reza Bidkhori
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| | - Tamara Yawno
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
- Department of Paediatrics, Monash University, Clayton 3168, Victoria, Australia
| | - Rebecca Lim
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| | - Ishmael Miguel Inocencio
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| |
Collapse
|
34
|
Wang L, Xiao J, Zhang B, Hou A. Epigenetic modifications in the development of bronchopulmonary dysplasia: a review. Pediatr Res 2024; 96:632-642. [PMID: 38570557 DOI: 10.1038/s41390-024-03167-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 02/25/2024] [Accepted: 03/07/2024] [Indexed: 04/05/2024]
Abstract
While perinatal medicine advancements have bolstered survival outcomes for premature infants, bronchopulmonary dysplasia (BPD) continues to threaten their long-term health. Gene-environment interactions, mediated by epigenetic modifications such as DNA methylation, histone modification, and non-coding RNA regulation, take center stage in BPD pathogenesis. Recent discoveries link methylation variations across biological pathways with BPD. Also, the potential reversibility of histone modifications fuels new treatment avenues. The review also highlights the promise of utilizing mesenchymal stem cells and their exosomes as BPD therapies, given their ability to modulate non-coding RNA, opening novel research and intervention possibilities. IMPACT: The complexity and universality of epigenetic modifications in the occurrence and development of bronchopulmonary dysplasia were thoroughly discussed. Both molecular and cellular mechanisms contribute to the diverse nature of epigenetic changes, suggesting the need for deeper biochemical techniques to explore these molecular alterations. The utilization of innovative cell-specific drug delivery methods like exosomes and extracellular vesicles holds promise in achieving precise epigenetic regulation.
Collapse
Affiliation(s)
- Lichuan Wang
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China
| | - Jun Xiao
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China
| | - Bohan Zhang
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China
| | - Ana Hou
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
35
|
Din MAU, Wan A, Chu Y, Zhou J, Yan Y, Xu Z. Therapeutic role of extracellular vesicles from human umbilical cord mesenchymal stem cells and their wide therapeutic implications in inflammatory bowel disease and other inflammatory disorder. Front Med (Lausanne) 2024; 11:1406547. [PMID: 39139783 PMCID: PMC11319305 DOI: 10.3389/fmed.2024.1406547] [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: 03/25/2024] [Accepted: 07/18/2024] [Indexed: 08/15/2024] Open
Abstract
The chronic immune-mediated inflammatory condition known as inflammatory bowel disease (IBD) significantly affects the gastrointestinal system. While the precise etiology of IBD remains elusive, extensive research suggests that a range of pathophysiological pathways and immunopathological mechanisms may significantly contribute as potential factors. Mesenchymal stem cells (MSCs) have shown significant potential in the development of novel therapeutic approaches for various medical conditions. However, some MSCs have been found to exhibit tumorigenic characteristics, which limit their potential for medical treatments. The extracellular vesicles (EVs), paracrine factors play a crucial role in the therapeutic benefits conferred by MSCs. The EVs consist of proteins, microRNAs, and lipids, and are instrumental in facilitating intercellular communication. Due to the ease of maintenance, and decreased immunogenicity, tumorigenicity the EVs have become a new and exciting option for whole cell treatment. This review comprehensively assesses recent preclinical research on human umbilical cord mesenchymal stem cell (hUC-MSC)-derived EVs as a potential IBD therapy. It comprehensively addresses key aspects of various conditions, including diabetes, cancer, dermal injuries, neurological disorders, cardiovascular issues, liver and kidney diseases, and bone-related afflictions.
Collapse
Affiliation(s)
- Muhammad Azhar Ud Din
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine Jiangsu University, Zhenjiang, China
| | | | - Ying Chu
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
| | - Jing Zhou
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
| | - Yongmin Yan
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
| | - Zhiliang Xu
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
| |
Collapse
|
36
|
Antounians L, Figueira RL, Kukreja B, Litvack ML, Zani-Ruttenstock E, Khalaj K, Montalva L, Doktor F, Obed M, Blundell M, Wu T, Chan C, Wagner R, Lacher M, Wilson MD, Post M, Kalish BT, Zani A. Fetal hypoplastic lungs have multilineage inflammation that is reversed by amniotic fluid stem cell extracellular vesicle treatment. SCIENCE ADVANCES 2024; 10:eadn5405. [PMID: 39058789 PMCID: PMC11277482 DOI: 10.1126/sciadv.adn5405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 06/21/2024] [Indexed: 07/28/2024]
Abstract
Antenatal administration of extracellular vesicles from amniotic fluid stem cells (AFSC-EVs) reverses features of pulmonary hypoplasia in models of congenital diaphragmatic hernia (CDH). However, it remains unknown which lung cellular compartments and biological pathways are affected by AFSC-EV therapy. Herein, we conducted single-nucleus RNA sequencing (snRNA-seq) on rat fetal CDH lungs treated with vehicle or AFSC-EVs. We identified that intra-amniotically injected AFSC-EVs reach the fetal lung in rats with CDH, where they promote lung branching morphogenesis and epithelial cell differentiation. Moreover, snRNA-seq revealed that rat fetal CDH lungs have a multilineage inflammatory signature with macrophage enrichment, which is reversed by AFSC-EV treatment. Macrophage enrichment in CDH fetal rat lungs was confirmed by immunofluorescence, flow cytometry, and inhibition studies with GW2580. Moreover, we validated macrophage enrichment in human fetal CDH lung autopsy samples. Together, this study advances knowledge on the pathogenesis of pulmonary hypoplasia and further evidence on the value of an EV-based therapy for CDH fetuses.
Collapse
Affiliation(s)
- Lina Antounians
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Rebeca Lopes Figueira
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Bharti Kukreja
- Neurosciences and Mental Health Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
| | - Michael L. Litvack
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
| | - Elke Zani-Ruttenstock
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Kasra Khalaj
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Louise Montalva
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Fabian Doktor
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Mikal Obed
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Matisse Blundell
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Taiyi Wu
- Neurosciences and Mental Health Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
| | - Cadia Chan
- Genetics and Genome Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, Canada
| | - Richard Wagner
- Department of Pediatric Surgery, Leipzig University, Leipzig 04109, Germany
| | - Martin Lacher
- Department of Pediatric Surgery, Leipzig University, Leipzig 04109, Germany
| | - Michael D. Wilson
- Genetics and Genome Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, Canada
| | - Martin Post
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5T 1P5, Canada
| | - Brian T. Kalish
- Neurosciences and Mental Health Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, Canada
- Division of Neonatology, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Augusto Zani
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
- Department of Surgery, University of Toronto, Toronto M5T 1P5, Canada
| |
Collapse
|
37
|
Guo Y, Pan JJ, Zhu W, Wang MZ, Liu TY, Wang XX, Wu QQ, Cheng YX, Qian YS, Zhou XG, Yang Y. Hsa_circ_0001359 in Serum Exosomes: A Promising Marker to Predict Bronchopulmonary Dysplasia in Premature Infants. J Inflamm Res 2024; 17:5025-5037. [PMID: 39081873 PMCID: PMC11287472 DOI: 10.2147/jir.s463330] [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: 04/18/2024] [Accepted: 07/16/2024] [Indexed: 08/02/2024] Open
Abstract
Objective This prospective study is to explore the role of specific circRNAs in predicting the development of bronchopulmonary dysplasia (BPD). Methods From July 1, 2021 to December 1, 2021, peripheral blood samples were collected from 62 premature infants with gestational age (GA) ≤32 weeks on the 7th, 14th, and 28th day after birth. Then, on the 28th day, the included infants were divided into the BPD group and the non-BPD group according to the definition of BPD. Serum exosomal circRNAs from peripheral blood were identified, sequenced, and compared between the BPD and non-BPD groups at different time points. Specific differentially expressed circRNAs were further verified from another 42 enrolled premature infants (GA ≤32 weeks). The classical lung biological markers in serum were also measured simultaneously. Results Hsa_circ_0001359 in serum exosomes showed continuous differential expression between the BPD group and the non-BPD group on the 7th, 14th, and 28th day. Compared with that, classical lung biological markers like IL-6, IL-33, KL-6, and ET-1 did not exhibit continuous differences. Moreover, the expression of hsa_circ_0001359 on day 7 had a higher predictive value in predicting BPD (area under curve:0.853, 95% CI:0.738-0.968; adjusted odds ratio:6.033, 95% CI:2.373-13.326). The calibration curve further showed the mean absolute error = 0.033, mean squared error = 0.00231, and quantile of absolute error = 0.058. Conclusion Hsa_circ_0001359 in serum exosomes is a promising marker for predicting BPD in preterm infants with gestational age ≤32 weeks.
Collapse
Affiliation(s)
- Yan Guo
- Department of Neonatology, Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Jing-Jing Pan
- Department of Neonatology, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Wen Zhu
- Department of Neonatology, Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Mu-Zi Wang
- Department of Neonatology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, People’s Republic of China
| | - Tian-Yu Liu
- Department of Neonatology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, People’s Republic of China
| | - Xiao-Xin Wang
- Department of Pediatrics, Shandong Tumor Hospital, Jinan, People’s Republic of China
| | - Qian-Qian Wu
- Department of Neonatology, Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Yi-Xin Cheng
- Department of Neonatology, Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Yi-Sen Qian
- Department of Neonatology, Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Xiao-Guang Zhou
- Department of Neonatology, Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Yang Yang
- Department of Neonatology, Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
38
|
Rostami Z, Saharkhiz M, Khorashadizadeh M, Ayadilord M, Naseri M. The immunoregulatory property of mesenchymal stem cells in Crocin treatment by expression modulation of microRNA-155, microRNA-21, microRNA-23b, microRNA-126a, and their target inflammatory genes. Gene 2024; 916:148446. [PMID: 38583816 DOI: 10.1016/j.gene.2024.148446] [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: 02/10/2024] [Revised: 03/14/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Mesenchymal stem cells (MSCs) have high priority in clinical applications for treatment of immune disorders because of their immunomodulatory function. A lot of researches have currently been undertaken to enhance the stemness capacities of the cells and pick an excellent type of MSCs for clinical approaches. This study aims to assess the immunomodulatory related MicroRNAs (miRNAs) expression as well as their target genes in both adipose derived stem cells (Ad-SCs) and dental pulp derived stem cell (DP-SCs) in the presence or lack of Crocin (saffron plant's bioactive compound). For this purpose, first MSCs were extracted from adipose and dental pulp tissues, and then their mesenchymal nature was confirmed using flow cytometry and differentiation tests. Following the cell treatment with an optimal-non-toxic dose of Crocin (Obtained by MTT test), the expression of 4 selected immunomodulatory-related micro-RNAs (Mir-126, -21, -23, and-155) and their target genes (PI3K/ Akt 1 and 2/ NFKB and RELA) were assessed by RT-PCR. Our findings revealed that miRNA-23 and miRNA-126 were up-regulated in both types of cells treated with Crocin, while in the other side, miRNA-21 and miRNA-155 were down-regulated in DP-SCs and were up-regulated in Ad-SCs under treatment. Moreover, the real-time PCR results indicated that Crocin could significantly down regulate the expression of PI3K/ Akt1/ Akt2/ NFKB/ RELA genes in DP-SCs and PI3K/Akt2 genes in Ad-SCs and up regulate the expression of Akt1/ NFKB/ RELA genes in recent cells. Based on the analysis of the obtained data, the immunoregulatory effects of Crocin were higher in DP-SCs than in Ad-SCs. In conclusion, Crocin could control essential signaling pathways related to the inflammation by regulating the expression of related- miRNAs genes that play a key function in the immune regulation pathways in MSCs. Our findings can give an understanding of the mechanisms by which Crocin enhances the immunomodulatory feature of MSCs. According to the research findings, DP-SCs are probably a better immunomodulator in Crocin treatment than Ad-SCs and it may be helpful for MSCs selection in clinical applications for modulation or treatment of autoimmune disorders.
Collapse
Affiliation(s)
- Zeinab Rostami
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran; Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mansoore Saharkhiz
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran; Department of Immunology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohsen Khorashadizadeh
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran; Department of Medical Biotechnology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Malaksima Ayadilord
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Naseri
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran; Department of Immunology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
39
|
Hou W, Yu B, Li Y, Yan X, Su Q, Fang X, Zhou X, Yu Z. PC (16:0/14:0) ameliorates hyperoxia-induced bronchopulmonary dysplasia by upregulating claudin-1 and promoting alveolar type II cell repair. Int J Biochem Cell Biol 2024; 172:106587. [PMID: 38740281 DOI: 10.1016/j.biocel.2024.106587] [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: 12/22/2023] [Revised: 04/25/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
Bronchopulmonary dysplasia (BPD) remains a significant challenge in neonatal care, the pathogenesis of which potentially involves altered lipid metabolism. Given the critical role of lipids in lung development and the injury response, we hypothesized that specific lipid species could serve as therapeutic agents in BPD. This study aimed to investigate the role of the lipid Phosphatidylcholine (PC) (16:0/14:0) in modulating BPD pathology and to elucidate its underlying mechanisms of action. Our approach integrated in vitro and in vivo methodologies to assess the effects of PC (16:0/14:0) on the histopathology, cellular proliferation, apoptosis, and molecular markers in lung tissue. In a hyperoxia-induced BPD rat model, we observed a reduction in alveolar number and an enlargement in alveolar size, which were ameliorated by PC (16:0/14:0) treatment. Correspondingly, in BPD cell models, PC (16:0/14:0) intervention led to increased cell viability, enhanced proliferation, reduced apoptosis, and elevated surfactant protein C (SPC) expression. RNA sequencing revealed significant gene expression differences between BPD and PC (16:0/14:0) treated groups, with a particular focus on Cldn1 (encoding claudin 1), which was significantly enriched in our analysis. Our findings suggest that PC (16:0/14:0) might protect against hyperoxia-induced alveolar type II cell damage by upregulating CLDN1 expression, potentially serving as a novel therapeutic target for BPD. This study not only advances our understanding of the role of lipids in BPD pathogenesis, but also highlights the significance of PC (16:0/14:0) in the prevention and treatment of BPD, offering new avenues for future research and therapeutic development.
Collapse
Affiliation(s)
- Weiwei Hou
- Department of Neonatology, Nanjing Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, Jiangsu 210008, China; Division of Neonatology, Department of Pediatrics, Northern Jiangsu People's Hospital afiliated to Yangzhou University, 98 West Nantong Road, Yangzhou, Jiangsu 225001, China
| | - Boshi Yu
- Division of Neonatology, Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, 1017 North Dongmen Road, Shenzhen, Guangdong 518020, China.
| | - Yubai Li
- Division of Neonatology, Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, 1017 North Dongmen Road, Shenzhen, Guangdong 518020, China
| | - Xudong Yan
- Division of Neonatology, Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, 1017 North Dongmen Road, Shenzhen, Guangdong 518020, China
| | - Qian Su
- Division of Neonatology, Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, 1017 North Dongmen Road, Shenzhen, Guangdong 518020, China
| | - Xiaoyan Fang
- Division of Neonatology, Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, 1017 North Dongmen Road, Shenzhen, Guangdong 518020, China
| | - Xiaoguang Zhou
- Department of Neonatology, Nanjing Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, Jiangsu 210008, China.
| | - Zhangbin Yu
- Division of Neonatology, Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, 1017 North Dongmen Road, Shenzhen, Guangdong 518020, China.
| |
Collapse
|
40
|
Mohammed AN, Kohram F, Lan YW, Li E, Kolesnichenko OA, Kalin TV, Kalinichenko VV. Transplantation of alveolar macrophages improves the efficacy of endothelial progenitor cell therapy in mouse model of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2024; 327:L114-L125. [PMID: 38772902 PMCID: PMC11380942 DOI: 10.1152/ajplung.00274.2023] [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: 08/28/2023] [Revised: 04/12/2024] [Accepted: 05/07/2024] [Indexed: 05/23/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a severe complication of preterm births, which develops due to exposure to supplemental oxygen and mechanical ventilation. Published studies demonstrated that the number of endothelial progenitor cells (EPC) is decreased in mouse and human BPD lungs and that adoptive transfer of EPC is an effective approach in reversing the hyperoxia-induced lung damage in mouse model of BPD. Recent advancements in macrophage biology identified the specific subtypes of circulating and resident macrophages mediating the developmental and regenerative functions in the lungs. Several studies reported the successful application of macrophage therapy in accelerating the regenerative capacity of damaged tissues and enhancing the therapeutic efficacy of other transplantable progenitor cells. In the present study, we explored the efficacy of combined cell therapy with EPC and resident alveolar macrophages (rAM) in hyperoxia-induced BPD mouse model. rAM and EPC were purified from neonatal mouse lungs and were used for adoptive transfer to the recipient neonatal mice exposed to hyperoxia. Adoptive transfer of rAM alone did not result in engraftment of donor rAM into the lung tissue but increased the mRNA level and protein concentration of proangiogenic CXCL12 chemokine in recipient mouse lungs. Depletion of rAM by chlodronate-liposomes decreased the retention of donor EPC after their transplantation into hyperoxia-injured lungs. Adoptive transfer of rAM in combination with EPC enhanced the therapeutic efficacy of EPC as evidenced by increased retention of EPC, increased capillary density, improved arterial oxygenation, and alveolarization in hyperoxia-injured lungs. Dual therapy with EPC and rAM has promise in human BPD.NEW & NOTEWORTHY Recent studies demonstrated that transplantation of lung-resident endothelial progenitor cells (EPC) is an effective therapy in mouse model of bronchopulmonary dysplasia (BPD). However, key factors regulating the efficacy of EPC are unknown. Herein, we demonstrate that transplantation of tissue-resident alveolar macrophages (rAM) increases CXCL12 expression in neonatal mouse lungs. rAM are required for retention of donor EPC in hyperoxia-injured lungs. Co-transplantation of rAM and EPC improves the efficacy of EPC therapy in mouse BPD model.
Collapse
Affiliation(s)
- Afzaal Nadeem Mohammed
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States
| | - Fatemeh Kohram
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States
| | - Ying-Wei Lan
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States
| | - Enhong Li
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States
| | - Olena A Kolesnichenko
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Research Foundation, Cincinnati, Ohio, United States
| | - Tanya V Kalin
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States
| | - Vladimir V Kalinichenko
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States
- Division of Neonatology, Phoenix Children's Hospital, Phoenix, Arizona, United States
| |
Collapse
|
41
|
Lin KC, Fang WF, Yeh JN, Chiang JY, Chiang HJ, Shao PL, Sung PH, Yip HK. Outcomes of combined mitochondria and mesenchymal stem cells-derived exosome therapy in rat acute respiratory distress syndrome and sepsis. World J Stem Cells 2024; 16:690-707. [PMID: 38948095 PMCID: PMC11212548 DOI: 10.4252/wjsc.v16.i6.690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/08/2024] [Accepted: 05/09/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND The treatment of acute respiratory distress syndrome (ARDS) complicated by sepsis syndrome (SS) remains challenging. AIM To investigate whether combined adipose-derived mesenchymal-stem-cells (ADMSCs)-derived exosome (EXAD) and exogenous mitochondria (mitoEx) protect the lung from ARDS complicated by SS. METHODS In vitro study, including L2 cells treated with lipopolysaccharide (LPS) and in vivo study including male-adult-SD rats categorized into groups 1 (sham-operated-control), 2 (ARDS-SS), 3 (ARDS-SS + EXAD), 4 (ARDS-SS + mitoEx), and 5 (ARDS-SS + EXAD + mitoEx), were included in the present study. RESULTS In vitro study showed an abundance of mitoEx found in recipient-L2 cells, resulting in significantly higher mitochondrial-cytochrome-C, adenosine triphosphate and relative mitochondrial DNA levels (P < 0.001). The protein levels of inflammation [interleukin (IL)-1β/tumor necrosis factor (TNF)-α/nuclear factor-κB/toll-like receptor (TLR)-4/matrix-metalloproteinase (MMP)-9/oxidative-stress (NOX-1/NOX-2)/apoptosis (cleaved-caspase3/cleaved-poly (ADP-ribose) polymerase)] were significantly attenuated in lipopolysaccharide (LPS)-treated L2 cells with EXAD treatment than without EXAD treatment, whereas the protein expressions of cellular junctions [occluding/β-catenin/zonula occludens (ZO)-1/E-cadherin] exhibited an opposite pattern of inflammation (all P < 0.001). Animals were euthanized by 72 h post-48 h-ARDS induction, and lung tissues were harvested. By 72 h, flow cytometric analysis of bronchoalveolar lavage fluid demonstrated that the levels of inflammatory cells (Ly6G+/CD14+/CD68+/CD11b/c+/myeloperoxidase+) and albumin were lowest in group 1, highest in group 2, and significantly higher in groups 3 and 4 than in group 5 (all P < 0.0001), whereas arterial oxygen-saturation (SaO2%) displayed an opposite pattern of albumin among the groups. Histopathological findings of lung injury/fibrosis area and inflammatory/DNA-damaged markers (CD68+/γ-H2AX) displayed an identical pattern of SaO2% among the groups (all P < 0.0001). The protein expressions of inflammatory (TLR-4/MMP-9/IL-1β/TNF-α)/oxidative stress (NOX-1/NOX-2/p22phox/oxidized protein)/mitochondrial-damaged (cytosolic-cytochrome-C/dynamin-related protein 1)/autophagic (beclin-1/Atg-5/ratio of LC3B-II/LC3B-I) biomarkers exhibited a similar manner, whereas antioxidants [nuclear respiratory factor (Nrf)-1/Nrf-2]/cellular junctions (ZO-1/E-cadherin)/mitochondrial electron transport chain (complex I-V) exhibited an opposite manner of albumin among the groups (all P < 0.0001). CONCLUSION Combined EXAD-mitoEx therapy was better than merely one for protecting the lung against ARDS-SS induced injury.
Collapse
Affiliation(s)
- Kun-Chen Lin
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Wen-Feng Fang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Jui-Ning Yeh
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - John Y Chiang
- Department of Computer Science and Engineering, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
- Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hsin-Ju Chiang
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Pei-Lin Shao
- Department of Nursing, Asia University, Taichung 41354, Taiwan
| | - Pei-Hsun Sung
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Hon-Kan Yip
- Department of Nursing, Asia University, Taichung 41354, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| |
Collapse
|
42
|
Lin KC, Fang WF, Yeh JN, Chiang JY, Chiang HJ, Shao PL, Sung PH, Yip HK. Outcomes of combined mitochondria and mesenchymal stem cells-derived exosome therapy in rat acute respiratory distress syndrome and sepsis. World J Stem Cells 2024; 16:689-706. [DOI: 10.4252/wjsc.v16.i6.689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/08/2024] [Accepted: 05/09/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND The treatment of acute respiratory distress syndrome (ARDS) complicated by sepsis syndrome (SS) remains challenging.
AIM To investigate whether combined adipose-derived mesenchymal-stem-cells (ADMSCs)-derived exosome (EXAD) and exogenous mitochondria (mitoEx) protect the lung from ARDS complicated by SS.
METHODS In vitro study, including L2 cells treated with lipopolysaccharide (LPS) and in vivo study including male-adult-SD rats categorized into groups 1 (sham-operated-control), 2 (ARDS-SS), 3 (ARDS-SS + EXAD), 4 (ARDS-SS + mitoEx), and 5 (ARDS-SS + EXAD + mitoEx), were included in the present study.
RESULTS In vitro study showed an abundance of mitoEx found in recipient-L2 cells, resulting in significantly higher mitochondrial-cytochrome-C, adenosine triphosphate and relative mitochondrial DNA levels (P < 0.001). The protein levels of inflammation [interleukin (IL)-1β/tumor necrosis factor (TNF)-α/nuclear factor-κB/toll-like receptor (TLR)-4/matrix-metalloproteinase (MMP)-9/oxidative-stress (NOX-1/NOX-2)/apoptosis (cleaved-caspase3/cleaved-poly (ADP-ribose) polymerase)] were significantly attenuated in lipopolysaccharide (LPS)-treated L2 cells with EXAD treatment than without EXAD treatment, whereas the protein expressions of cellular junctions [occluding/β-catenin/zonula occludens (ZO)-1/E-cadherin] exhibited an opposite pattern of inflammation (all P < 0.001). Animals were euthanized by 72 h post-48 h-ARDS induction, and lung tissues were harvested. By 72 h, flow cytometric analysis of bronchoalveolar lavage fluid demonstrated that the levels of inflammatory cells (Ly6G+/CD14+/CD68+/CD11b/c+/myeloperoxidase+) and albumin were lowest in group 1, highest in group 2, and significantly higher in groups 3 and 4 than in group 5 (all P < 0.0001), whereas arterial oxygen-saturation (SaO2%) displayed an opposite pattern of albumin among the groups. Histopathological findings of lung injury/fibrosis area and inflammatory/DNA-damaged markers (CD68+/γ-H2AX) displayed an identical pattern of SaO2% among the groups (all P < 0.0001). The protein expressions of inflammatory (TLR-4/MMP-9/IL-1β/TNF-α)/oxidative stress (NOX-1/NOX-2/p22phox/oxidized protein)/mitochondrial-damaged (cytosolic-cytochrome-C/dynamin-related protein 1)/autophagic (beclin-1/Atg-5/ratio of LC3B-II/LC3B-I) biomarkers exhibited a similar manner, whereas antioxidants [nuclear respiratory factor (Nrf)-1/Nrf-2]/cellular junctions (ZO-1/E-cadherin)/mitochondrial electron transport chain (complex I-V) exhibited an opposite manner of albumin among the groups (all P < 0.0001).
CONCLUSION Combined EXAD-mitoEx therapy was better than merely one for protecting the lung against ARDS-SS induced injury.
Collapse
Affiliation(s)
- Kun-Chen Lin
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Wen-Feng Fang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Jui-Ning Yeh
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - John Y Chiang
- Department of Computer Science and Engineering, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
- Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hsin-Ju Chiang
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Pei-Lin Shao
- Department of Nursing, Asia University, Taichung 41354, Taiwan
| | - Pei-Hsun Sung
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Hon-Kan Yip
- Department of Nursing, Asia University, Taichung 41354, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| |
Collapse
|
43
|
Taherian M, Bayati P, Mojtabavi N. Stem cell-based therapy for fibrotic diseases: mechanisms and pathways. Stem Cell Res Ther 2024; 15:170. [PMID: 38886859 PMCID: PMC11184790 DOI: 10.1186/s13287-024-03782-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Fibrosis is a pathological process, that could result in permanent scarring and impairment of the physiological function of the affected organ; this condition which is categorized under the term organ failure could affect various organs in different situations. The involvement of the major organs, such as the lungs, liver, kidney, heart, and skin, is associated with a high rate of morbidity and mortality across the world. Fibrotic disorders encompass a broad range of complications and could be traced to various illnesses and impairments; these could range from simple skin scars with beauty issues to severe rheumatologic or inflammatory disorders such as systemic sclerosis as well as idiopathic pulmonary fibrosis. Besides, the overactivation of immune responses during any inflammatory condition causing tissue damage could contribute to the pathogenic fibrotic events accompanying the healing response; for instance, the inflammation resulting from tissue engraftment could cause the formation of fibrotic scars in the grafted tissue, even in cases where the immune system deals with hard to clear infections, fibrotic scars could follow and cause severe adverse effects. A good example of such a complication is post-Covid19 lung fibrosis which could impair the life of the affected individuals with extensive lung involvement. However, effective therapies that halt or slow down the progression of fibrosis are missing in the current clinical settings. Considering the immunomodulatory and regenerative potential of distinct stem cell types, their application as an anti-fibrotic agent, capable of attenuating tissue fibrosis has been investigated by many researchers. Although the majority of the studies addressing the anti-fibrotic effects of stem cells indicated their potent capabilities, the underlying mechanisms, and pathways by which these cells could impact fibrotic processes remain poorly understood. Here, we first, review the properties of various stem cell types utilized so far as anti-fibrotic treatments and discuss the challenges and limitations associated with their applications in clinical settings; then, we will summarize the general and organ-specific mechanisms and pathways contributing to tissue fibrosis; finally, we will describe the mechanisms and pathways considered to be employed by distinct stem cell types for exerting anti-fibrotic events.
Collapse
Affiliation(s)
- Marjan Taherian
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Paria Bayati
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Nazanin Mojtabavi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
44
|
Guo S, Wang D. Novel insights into the potential applications of stem cells in pulmonary hypertension therapy. Respir Res 2024; 25:237. [PMID: 38849894 PMCID: PMC11162078 DOI: 10.1186/s12931-024-02865-4] [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: 10/07/2023] [Accepted: 06/04/2024] [Indexed: 06/09/2024] Open
Abstract
Pulmonary hypertension (PH) refers to a group of deadly lung diseases characterized by vascular lesions in the microvasculature and a progressive increase in pulmonary vascular resistance. The prevalence of PH has increased over time. Currently, the treatment options available for PH patients have limited efficacy, and none of them can fundamentally reverse pulmonary vascular remodeling. Stem cells represent an ideal seed with proven efficacy in clinical studies focusing on liver, cardiovascular, and nerve diseases. Since the potential therapeutic effect of mesenchymal stem cells (MSCs) on PH was first reported in 2006, many studies have demonstrated the efficacy of stem cells in PH animal models and suggested that stem cells can help slow the deterioration of lung tissue. Existing PH treatment studies basically focus on the paracrine action of stem cells, including protein regulation, exosome pathway, and cell signaling; however, the specific mechanisms have not yet been clarified. Apoptotic and afunctional pulmonary microvascular endothelial cells (PMVECs) and alveolar epithelial cells (AECs) are two fundamental promoters of PH although they have not been extensively studied by researchers. This review mainly focuses on the supportive communication and interaction between PMVECs and AECs as well as the potential restorative effect of stem cells on their injury. In the future, more studies are needed to prove these effects and explore more radical cures for PH.
Collapse
Affiliation(s)
- Sijia Guo
- Stem Cell Laboratory, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China.
| | - Dachun Wang
- Stem Cell Laboratory, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- The Brown Foundation Institute of Molecular Medicine for the prevention of Human Diseases, University of Texas Medical School at Houston, Houston, TX, USA
| |
Collapse
|
45
|
Cerro Marín MJD, Ormazábal IG, Gimeno-Navarro A, Álvarez-Fuente M, López-Ortego P, Avila-Alvarez A, Arruza Gómez L, González-Menchen C, Labrandero de Lera C, Lozano Balseiro M, Moreno Gutiérrez L, Melen Frajilich G, Ramírez Orellana M, Saldaña García N, Pavón Delgado A, Vento Torres M. Repeated intravenous doses of human umbilical cord-derived mesenchymal stromal cells for bronchopulmonary dysplasia: results of a phase 1 clinical trial with 2-year follow-up. Cytotherapy 2024; 26:632-640. [PMID: 38556960 DOI: 10.1016/j.jcyt.2024.02.028] [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: 07/27/2023] [Revised: 12/29/2023] [Accepted: 02/29/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Currently, there is a lack of effective treatments or preventive strategies for bronchopulmonary dysplasia (BPD). Pre-clinical studies with mesenchymal stromal cells (MSCs) have yielded encouraging results. The safety of administering repeated intravenous doses of umbilical cord tissue-derived mesenchymal stromal cells (UC-MSCs) has not yet been tested in extremely-low-gestational-age newborns (ELGANs). AIMS to test the safety and feasibility of administering three sequential intravenous doses of UC-MSCs every 7 days to ELGANs at risk of developing BPD. METHODS In this phase 1 clinical trial, we recruited ELGANs (birth weight ≤1250 g and ≤28 weeks in gestational age [GA]) who were on invasive mechanical ventilation (IMV) with FiO2 ≥ 0.3 at postnatal days 7-14. Three doses of 5 × 106/kg of UC-MSCs were intravenously administered at weekly intervals. Adverse effects and prematurity-related morbidities were recorded. RESULTS From April 2019 to July 2020, 10 patients were recruited with a mean GA of 25.2 ± 0.8 weeks and a mean birth weight of 659.8 ± 153.8 g. All patients received three intravenous UC-MSC doses. The first dose was administered at a mean of 16.6 ± 2.9 postnatal days. All patients were diagnosed with BPD. All patients were discharged from the hospital. No deaths or any serious adverse events related to the infusion of UC-MSCs were observed during administration, hospital stays or at 2-year follow-up. CONCLUSIONS The administration of repeated intravenous infusion of UC-MSCs in ELGANs at a high risk of developing BPD was feasible and safe in the short- and mid-term follow-up.
Collapse
Affiliation(s)
- Maria Jesús Del Cerro Marín
- Pediatric Cardiology, Hospital Universitario Ramón y Cajal, Madrid and Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), Madrid, Spain.
| | - Itziar Garcia Ormazábal
- Pediatric Cardiology, Hospital Universitario Ramón y Cajal, Madrid and Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), Madrid, Spain
| | - Ana Gimeno-Navarro
- Division of Neonatology, Hospital Universitari i Politècnic La Fe (HULAFE) and Health Research Institute La Fe (IISLAFE), Valencia, Spain
| | - María Álvarez-Fuente
- Pediatric Cardiology, Hospital Universitario Ramón y Cajal, Madrid and Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), Madrid, Spain
| | | | - Alejandro Avila-Alvarez
- Neonatology Department, Complexo Hospitalario Universitario A Coruña (CHUAC), A Coruña, Spain
| | - Luis Arruza Gómez
- Department of Neonatology, Instituto del Niño y del Adolescente, Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Cristina González-Menchen
- Department of Neonatology, Instituto del Niño y del Adolescente, Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | | | - María Lozano Balseiro
- Neonatology Department, Complexo Hospitalario Universitario A Coruña (CHUAC), A Coruña, Spain
| | | | | | | | - Natalia Saldaña García
- Neonatology Department, Hospital Regional Universitario de Málaga and Biomedical Research Institute of Málaga, Málaga, Spain
| | | | - Máximo Vento Torres
- Division of Neonatology, Hospital Universitari i Politècnic La Fe (HULAFE) and Health Research Institute La Fe (IISLAFE), Valencia, Spain
| |
Collapse
|
46
|
Albertine KH, Rebentisch A, Dawson E, Van Boerum J, Major E, Štipka J, Foreman H, Headden D, Vordos Z, Beck E, Wang Z, Yang H, Yu B, Dahl MJ, Null DM, Bizzotto D, Veneroni C, Lavizzari A, Dellacà RL, Delavogia E, Mitsialis SA, Kourembanas S. Mesenchymal stromal cell extracellular vesicles improve lung development in mechanically ventilated preterm lambs. Am J Physiol Lung Cell Mol Physiol 2024; 326:L770-L785. [PMID: 38563994 PMCID: PMC11380989 DOI: 10.1152/ajplung.00349.2023] [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: 11/10/2023] [Revised: 03/18/2024] [Accepted: 03/26/2024] [Indexed: 04/04/2024] Open
Abstract
Novel therapies are needed for bronchopulmonary dysplasia (BPD) because no effective treatment exists. Mesenchymal stromal cell extracellular vesicles (MSC-sEVs) have therapeutic efficacy in a mouse pup neonatal hyperoxia BPD model. We tested the hypothesis that MSC-sEVs will improve lung functional and structural development in mechanically ventilated preterm lambs. Preterm lambs (∼129 days; equivalent to human lung development at ∼28 wk gestation) were exposed to antenatal steroids, surfactant, caffeine, and supported by mechanical ventilation for 6-7 days. Lambs were randomized to blinded treatment with either MSC-sEVs (human bone marrow MSC-derived; 2 × 1011 particles iv; n = 8; 4 F/4 M) or vehicle control (saline iv; 4 F/4 M) at 6 and 78 h post delivery. Physiological targets were pulse oximetry O2 saturation 90-94% ([Formula: see text] 60-90 mmHg), [Formula: see text] 45-60 mmHg (pH 7.25-7.35), and tidal volume 5-7 mL/kg. MSC-sEVs-treated preterm lambs tolerated enteral feedings compared with vehicle control preterm lambs. Differences in weight patterns were statistically significant. Respiratory severity score, oxygenation index, A-a gradient, distal airspace wall thickness, and smooth muscle thickness around terminal bronchioles and pulmonary arterioles were significantly lower for the MSC-sEVs group. S/F ratio, radial alveolar count, secondary septal volume density, alveolar capillary surface density, and protein abundance of VEGF-R2 were significantly higher for the MSC-sEVs group. MSC-sEVs improved respiratory system physiology and alveolar formation in mechanically ventilated preterm lambs. MSC-sEVs may be an effective and safe therapy for appropriate functional and structural development of the lung in preterm infants who require mechanical ventilation and are at risk of developing BPD.NEW & NOTEWORTHY This study focused on potential treatment of preterm infants at risk of developing bronchopulmonary dysplasia (BPD), for which no effective treatment exists. We tested treatment of mechanically ventilated preterm lambs with human mesenchymal stromal cell extracellular vesicles (MSC-sEVs). The results show improved respiratory gas exchange and parenchymal growth of capillaries and epithelium that are necessary for alveolar formation. Our study provides new mechanistic insight into potential efficacy of MSC-sEVs for preterm infants at risk of developing BPD.
Collapse
Affiliation(s)
- Kurt H Albertine
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Andrew Rebentisch
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Elaine Dawson
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Jakob Van Boerum
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Emily Major
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Juraj Štipka
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Hannah Foreman
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - David Headden
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Zoë Vordos
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Emily Beck
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Zhengming Wang
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Haixia Yang
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Baifeng Yu
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Mar Janna Dahl
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Donald M Null
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Davide Bizzotto
- TechRes Lab, Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano University, Milan, Italy
| | - Chiara Veneroni
- TechRes Lab, Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano University, Milan, Italy
| | - Anna Lavizzari
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Raffaele L Dellacà
- TechRes Lab, Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano University, Milan, Italy
| | - Eleni Delavogia
- Division of Neonatology, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States
| | - S Alex Mitsialis
- Division of Neonatology, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States
| | - Stella Kourembanas
- Division of Neonatology, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
47
|
Lin Y, Wang Z, Liu S, Liu J, Zhang Z, Ouyang Y, Su Z, Chen D, Guo L, Luo T. Roles of extracellular vesicles on macrophages in inflammatory bone diseases. Mol Cell Biochem 2024; 479:1401-1414. [PMID: 37436653 DOI: 10.1007/s11010-023-04809-w] [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: 04/02/2023] [Accepted: 07/02/2023] [Indexed: 07/13/2023]
Abstract
Inflammatory bone disease is a general term for a series of diseases caused by chronic inflammation, which leads to the destruction of bone homeostasis, that is, the osteolytic activity of osteoclasts increases, and the osteogenic activity of osteoblasts decreases, leading to osteolysis. Macrophages are innate immune cell with plasticity, and their polarization is related to inflammatory bone diseases. The dynamic balance of macrophages between the M1 phenotype and the M2 phenotype affects the occurrence and development of diseases. In recent years, an increasing number of studies have shown that extracellular vesicles existing in the extracellular environment can act on macrophages, affecting the progress of inflammatory diseases. This process is realized by influencing the physiological activity or functional activity of macrophages, inducing macrophages to secrete cytokines, and playing an anti-inflammatory or pro-inflammatory role. In addition, by modifying and editing extracellular vesicles, the potential of targeting macrophages can be used to provide new ideas for developing new drug carriers for inflammatory bone diseases.
Collapse
Affiliation(s)
- Yifan Lin
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ziyan Wang
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shirong Liu
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiaohong Liu
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhiyi Zhang
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuanting Ouyang
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhikang Su
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ding Chen
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lvhua Guo
- Guangzhou Medical University, Guangzhou, Guangdong, China.
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Tao Luo
- Guangzhou Medical University, Guangzhou, Guangdong, China.
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
48
|
Krishnan I, Chan AML, Law JX, Ng MH, Jayapalan JJ, Lokanathan Y. Proteomic Analysis of Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles: A Systematic Review. Int J Mol Sci 2024; 25:5340. [PMID: 38791378 PMCID: PMC11121203 DOI: 10.3390/ijms25105340] [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: 03/26/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Numerous challenges remain within conventional cell-based therapy despite the growing trend of stem cells used to treat various life-debilitating diseases. These limitations include batch-to-batch heterogeneity, induced alloreactivity, cell survival and integration, poor scalability, and high cost of treatment, thus hindering successful translation from lab to bedside. However, recent pioneering technology has enabled the isolation and enrichment of small extracellular vesicles (EVs), canonically known as exosomes. EVs are described as a membrane-enclosed cargo of functional biomolecules not limited to lipids, nucleic acid, and proteins. Interestingly, studies have correlated the biological role of MSC-EVs to the paracrine activity of MSCs. This key evidence has led to rigorous studies on MSC-EVs as an acellular alternative. Using EVs as a therapy was proposed as a model leading to improvements through increased safety; enhanced bioavailability due to size and permeability; reduced heterogeneity by selective and quantifiable properties; and prolonged shelf-life via long-term freezing or lyophilization. Yet, the identity and potency of EVs are still relatively unknown due to various methods of preparation and to qualify the final product. This is reflected by the absence of regulatory strategies overseeing manufacturing, quality control, clinical implementation, and product registration. In this review, the authors review the various production processes and the proteomic profile of MSC-EVs.
Collapse
Affiliation(s)
- Illayaraja Krishnan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (A.M.L.C.); (J.X.L.); (M.H.N.)
| | - Alvin Man Lung Chan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (A.M.L.C.); (J.X.L.); (M.H.N.)
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (A.M.L.C.); (J.X.L.); (M.H.N.)
| | - Min Hwei Ng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (A.M.L.C.); (J.X.L.); (M.H.N.)
| | | | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (A.M.L.C.); (J.X.L.); (M.H.N.)
| |
Collapse
|
49
|
Ransom MA, Blatt AM, Pua HH, Sucre JMS. The emerging role of extracellular vesicles in bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2024; 326:L517-L523. [PMID: 38469633 PMCID: PMC11380955 DOI: 10.1152/ajplung.00244.2023] [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: 08/02/2023] [Revised: 03/01/2024] [Accepted: 03/02/2024] [Indexed: 03/13/2024] Open
Abstract
Extracellular vesicle (EV) biology in neonatal lung development and disease is a rapidly growing area of investigation. Although EV research in the neonatal population lags behind EV research in adult lung diseases, recent discoveries demonstrate promise in furthering our understanding of the pathophysiology of bronchopulmonary dysplasia and the potential use of EVs in the clinical setting, as both biomarkers and therapeutic agents. This review article explores some of the recent advances in this field and our evolving knowledge of the role of EVs in bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Meaghan A Ransom
- Department of Pediatrics, Vanderbilt University Medical Center; Nashville, Tennessee, United States
| | - Alexander M Blatt
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Heather H Pua
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Vanderbilt Center for Immunobiology and Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Jennifer M S Sucre
- Department of Pediatrics, Vanderbilt University Medical Center; Nashville, Tennessee, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
50
|
Chen F, Chen Z, Wu HT, Chen XX, Zhan P, Wei ZY, Ouyang Z, Jiang X, Shen A, Luo MH, Liu Q, Zhou YP, Qin A. Mesenchymal Stem Cell-Derived Exosomes Attenuate Murine Cytomegalovirus-Infected Pneumonia via NF-κB/NLRP3 Signaling Pathway. Viruses 2024; 16:619. [PMID: 38675960 PMCID: PMC11054941 DOI: 10.3390/v16040619] [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: 02/18/2024] [Revised: 04/08/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Reactivation and infection with cytomegalovirus (CMV) are frequently observed in recipients of solid organ transplants, bone marrow transplants, and individuals with HIV infection. This presents an increasing risk of allograft rejection, opportunistic infection, graft failure, and patient mortality. Among immunocompromised hosts, interstitial pneumonia is the most critical clinical manifestation of CMV infection. Recent studies have demonstrated the potential therapeutic benefits of exosomes derived from mesenchymal stem cells (MSC-exos) in preclinical models of acute lung injury, including pneumonia, ARDS, and sepsis. However, the role of MSC-exos in the pathogenesis of infectious viral diseases, such as CMV pneumonia, remains unclear. In a mouse model of murine CMV-induced pneumonia, we observed that intravenous administration of mouse MSC (mMSC)-exos reduced lung damage, decreased the hyperinflammatory response, and shifted macrophage polarization from the M1 to the M2 phenotype. Treatment with mMSC-exos also significantly reduced the infiltration of inflammatory cells and pulmonary fibrosis. Furthermore, in vitro studies revealed that mMSC-exos reversed the hyperinflammatory phenotype of bone marrow-derived macrophages infected with murine CMV. Mechanistically, mMSC-exos treatment decreased activation of the NF-κB/NLRP3 signaling pathway both in vivo and in vitro. In summary, our findings indicate that mMSC-exo treatment is effective in severe CMV pneumonia by reducing lung inflammation and fibrosis through the NF-κB/NLRP3 signaling pathway, thus providing promising therapeutic potential for clinical CMV infection.
Collapse
Affiliation(s)
- Fei Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Sixth Affiliated Hospital, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China; (F.C.); (Z.C.); (H.-T.W.); (X.-X.C.); (P.Z.); (Z.-Y.W.); (X.J.); (A.S.)
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Center for Cancer Research and Translational Medicine, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhida Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Sixth Affiliated Hospital, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China; (F.C.); (Z.C.); (H.-T.W.); (X.-X.C.); (P.Z.); (Z.-Y.W.); (X.J.); (A.S.)
| | - Hui-Ting Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Sixth Affiliated Hospital, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China; (F.C.); (Z.C.); (H.-T.W.); (X.-X.C.); (P.Z.); (Z.-Y.W.); (X.J.); (A.S.)
| | - Xin-Xiang Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Sixth Affiliated Hospital, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China; (F.C.); (Z.C.); (H.-T.W.); (X.-X.C.); (P.Z.); (Z.-Y.W.); (X.J.); (A.S.)
| | - Peiqi Zhan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Sixth Affiliated Hospital, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China; (F.C.); (Z.C.); (H.-T.W.); (X.-X.C.); (P.Z.); (Z.-Y.W.); (X.J.); (A.S.)
| | - Zheng-Yi Wei
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Sixth Affiliated Hospital, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China; (F.C.); (Z.C.); (H.-T.W.); (X.-X.C.); (P.Z.); (Z.-Y.W.); (X.J.); (A.S.)
| | - Zizhang Ouyang
- Department of Pharmaceutical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan 511518, China;
| | - Xueyan Jiang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Sixth Affiliated Hospital, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China; (F.C.); (Z.C.); (H.-T.W.); (X.-X.C.); (P.Z.); (Z.-Y.W.); (X.J.); (A.S.)
| | - Ao Shen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Sixth Affiliated Hospital, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China; (F.C.); (Z.C.); (H.-T.W.); (X.-X.C.); (P.Z.); (Z.-Y.W.); (X.J.); (A.S.)
| | - Min-Hua Luo
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China;
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yue-Peng Zhou
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China
| | - Aiping Qin
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Sixth Affiliated Hospital, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China; (F.C.); (Z.C.); (H.-T.W.); (X.-X.C.); (P.Z.); (Z.-Y.W.); (X.J.); (A.S.)
| |
Collapse
|