1
|
Golmohammadi M, Sheikhha MH, Ganji F, Shirani A, Barati M, Kalantar SM, Haramshahi SMA, Karkuki Osguei N, Samadikuchaksaraei A. Human fetal lung mesenchymal stem cells ameliorate lung injury in an animal model. Sci Rep 2025; 15:6433. [PMID: 39984612 PMCID: PMC11845704 DOI: 10.1038/s41598-025-91406-0] [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: 08/31/2024] [Accepted: 02/20/2025] [Indexed: 02/23/2025] Open
Abstract
Acute lung injury (ALI) is a critical condition with limited treatment options. This study evaluates the therapeutic potential of human fetal lung-derived mesenchymal stem cells (hFL-MSCs) in an experimental model of ALI. Our proof-of-concept findings suggest a paradigm shift in the approach to cell sourcing for lung diseases, proposing that fetal lung cells may be potential targets for stem cell differentiation studies when the derived cells are intended to be used for lung cell therapy. After characterizing hFL-MSCs, 18-week fetal lung cells were intratracheally instilled into rats with bleomycin-induced ALI. All the animals were evaluated on days 3 - 28 post-injury for cell count and the cytokines in bronchoalveolar lavage fluid (BALF), lung wet/dry weight ratio, lung tissue histological staining and expression of an extracellular matrix component, inflammatory and fibrotic genes. The findings confirm mesenchymal stem cell identity of the isolated cells and stability in their cell cycle distribution. Analysis of BALF showed that immune cell response to acute inflammation and adaptive immunity was significantly ameliorated by cell therapy with hFL-MSCs. Same results were confirmed by the levels of IL-6, TNF-α, IL-10 and NO in BALF, the lung wet/dry weight ratio and histopathological analysis of lung tissues after H&E and Masson's trichrome staining. Effective modulation of key pro-inflammatory (Il6, Tnf, Il1b), pro-fibrotic (Tgfb1) and Col1a1 genes were also confirmed after therapy with hFL-MSCs. Our findings suggest that fetal lung tissue-specific stem cells are viable options for lung cell therapy and could be considered as targets for engineering of regenerative cells for lung diseases.
Collapse
Affiliation(s)
- Mahtab Golmohammadi
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Fatemeh Ganji
- Latner Research Laboratories, Division of Thoracic Surgery, University Health Network, Toronto, ON, Canada
| | - Ali Shirani
- The Persian Gulf Biomedical Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mahmood Barati
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Seyed Mehdi Kalantar
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Mohammad Amin Haramshahi
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Ali Samadikuchaksaraei
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran.
| |
Collapse
|
2
|
Wang F, Zhang S, Xu Y, He W, Wang X, He Z, Shang J, Zhenyu Z. Mapping the landscape: A bibliometric perspective on autophagy in spinal cord injury. Medicine (Baltimore) 2024; 103:e38954. [PMID: 39029042 PMCID: PMC11398829 DOI: 10.1097/md.0000000000038954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/26/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is a severe condition that often leads to persistent damage of nerve cells and motor dysfunction. Autophagy is an intracellular system that regulates the recycling and degradation of proteins and lipids, primarily through lysosomal-dependent organelle degradation. Numerous publications have highlighted the involvement of autophagy in the secondary injury of SCI. Therefore, gaining a comprehensive understanding of autophagy research is crucial for designing effective therapies for SCI. METHODS Dates were obtained from Web of Science, including articles and article reviews published from its inception to October 2023. VOSviewer, Citespace, and SCImago were used to visualized analysis. Bibliometric analysis was conducted using the Web of Science data, focusing on various categories such as publications, authors, journals, countries, organizations, and keywords. This analysis was aimed to summarize the knowledge map of autophagy and SCI. RESULTS From 2009 to 2023, the number of annual publications in this field exhibited wave-like growth, with the highest number of publications recorded in 2020 (44 publications). Our analysis identified Mei Xifan as the most prolific author, while Kanno H emerged as the most influential author based on co-citations. Neuroscience Letters was found to have published the largest number of papers in this field. China was the most productive country, contributing 232 publications, and Wenzhou Medical University was the most active organization, publishing 39 papers. CONCLUSION We demonstrated a comprehensive overview of the relationship between autophagy and SCI utilizing bibliometric tools. This article could help to enhance the understanding of the field about autophagy and SCI, foster collaboration among researchers and organizations, and identify potential therapeutic targets for treatment.
Collapse
Affiliation(s)
- Fei Wang
- Department of Orthopedic Surgery, Shaoxing People’s Hospital, Zhejiang University, School of Medicine, Shaoxing, Zhejiang Province, China
| | - Songou Zhang
- Ningbo University, School of Medicine, Ningbo, Zhejiang Province, China
| | - Yangjun Xu
- School of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province, China
| | - Wei He
- Department of Orthopedic Surgery, Shaoxing People’s Hospital, Zhejiang University, School of Medicine, Shaoxing, Zhejiang Province, China
| | - Xiang Wang
- Department of Thoracic Surgery, Shaoxing People’s Hospital, Shaoxing, Zhejiang Province, China
| | - Zhongwei He
- School of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province, China
| | - Jinxiang Shang
- Department of Orthopedic, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang Province, China
| | - Zhang Zhenyu
- School of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province, China
| |
Collapse
|
3
|
Aslan E, Maytalman E, Nemutlu Samur D, Köle E, Günizi ÖC. An in vitro pilot study investigating placenta-derived mesenchymal stem cell coating on polypropylene mesh materials. Int Urogynecol J 2024; 35:553-559. [PMID: 38206335 DOI: 10.1007/s00192-023-05687-y] [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: 08/14/2023] [Accepted: 10/31/2023] [Indexed: 01/12/2024]
Abstract
INTRODUCTION AND HYPOTHESIS Polypropylene meshes (PM) used in pelvic organ prolapse surgery are being withdrawn from the market. Although concerns about the usage of PMs in stress incontinence surgery have been raised, it is still one of the best methods of curing stress urinary incontinence. With advancements in stem cell-based therapies, especially mesenchymal stem cells (MSCs), it is believed that coating the synthetic meshes with MSCs may minimize excessive tissue reactions ultimately leading to clinical problems such as pain, erosion or extrusion of the implanted material. In our study we tried to show the possibility of coating the PM with placenta-derived MSCs. METHODS Mesenchymal stem cells obtained from six placentas were isolated, cultured, and identified. MSCs were then soaked in either fibronectin or collagen prior to co-culturing with strips of PMs. One group is used as a control, and hence was not pretreated before co-culturing. Specimens were fixed and stained with both Gram and hematoxylin and eosin and marked with Vybran Dil and DAPI. All preparations were examined under a light microscope. The IMAGEJ program was utilized to determine the surface area of meshes coated with MSCs. RESULTS We clearly showed that PMs can be coated successfully with placenta-derived MSCs. The percentage of the coated area is significantly increased when meshes were pretreated with fibronectin or collagen (p<0.0001). CONCLUSIONS Placenta-derived MSCs can successfully coat PMs. The immunomodulatory properties of MSCs, which may be of great advantage in preventing the side effects of meshes, should be tested by in vivo and hopefully human studies before clinical applications.
Collapse
Affiliation(s)
- Erdogan Aslan
- Faculty of Medicine, Department of Obstetrics and Gynecology, Alanya Alaaddin Keykubat University, Payallar Mh. Atatürk Cd. Yasemin Villaları, No:111/L-4, Alanya, Antalya, Türkiye.
| | - Erkan Maytalman
- Faculty of Medicine, Department of Pharmacology, Alanya Alaaddin Keykubat University, Alanya, Antalya, Türkiye
| | - Dilara Nemutlu Samur
- Faculty of Medicine, Department of Pharmacology, Alanya Alaaddin Keykubat University, Alanya, Antalya, Türkiye
| | - Emre Köle
- Faculty of Medicine, Department of Obstetrics and Gynecology, Alanya Alaaddin Keykubat University, Payallar Mh. Atatürk Cd. Yasemin Villaları, No:111/L-4, Alanya, Antalya, Türkiye
| | - Özlem Ceren Günizi
- Faculty of Medicine, Department of Pharmacology, Alanya Alaaddin Keykubat University, Alanya, Antalya, Türkiye
| |
Collapse
|
4
|
Russo E, Corrao S, Di Gaudio F, Alberti G, Caprnda M, Kubatka P, Kruzliak P, Miceli V, Conaldi PG, Borlongan CV, La Rocca G. Facing the Challenges in the COVID-19 Pandemic Era: From Standard Treatments to the Umbilical Cord-Derived Mesenchymal Stromal Cells as a New Therapeutic Strategy. Cells 2023; 12:1664. [PMID: 37371134 PMCID: PMC10297457 DOI: 10.3390/cells12121664] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/10/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), the pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which counts more than 650 million cases and more than 6.6 million of deaths worldwide, affects the respiratory system with typical symptoms such as fever, cough, sore throat, acute respiratory distress syndrome (ARDS), and fatigue. Other nonpulmonary manifestations are related with abnormal inflammatory response, the "cytokine storm", that could lead to a multiorgan disease and to death. Evolution of effective vaccines against SARS-CoV-2 provided multiple options to prevent the infection, but the treatment of the severe forms remains difficult to manage. The cytokine storm is usually counteracted with standard medical care and anti-inflammatory drugs, but researchers moved forward their studies on new strategies based on cell therapy approaches. The perinatal tissues, such as placental membranes, amniotic fluid, and umbilical cord derivatives, are enriched in mesenchymal stromal cells (MSCs) that exert a well-known anti-inflammatory role, immune response modulation, and tissue repair. In this review, we focused on umbilical-cord-derived MSCs (UC-MSCs) used in in vitro and in vivo studies in order to evaluate the weakening of the severe symptoms, and on recent clinical trials from different databases, supporting the favorable potential of UC-MSCs as therapeutic strategy.
Collapse
Affiliation(s)
- Eleonora Russo
- Section of Histology and Embryology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (E.R.); (G.A.)
| | - Simona Corrao
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per per i Trapianti e Terapie Ad Alta Specializzazione), 90127 Palermo, Italy; (S.C.); (V.M.); (P.G.C.)
| | | | - Giusi Alberti
- Section of Histology and Embryology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (E.R.); (G.A.)
| | - Martin Caprnda
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University, University Hospital Bratislava, 81499 Bratislava, Slovakia;
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03649 Martin, Slovakia;
| | - Peter Kruzliak
- Research and Development Services, Pradlacka 18, 61300 Brno, Czech Republic;
| | - Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per per i Trapianti e Terapie Ad Alta Specializzazione), 90127 Palermo, Italy; (S.C.); (V.M.); (P.G.C.)
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per per i Trapianti e Terapie Ad Alta Specializzazione), 90127 Palermo, Italy; (S.C.); (V.M.); (P.G.C.)
| | - Cesario Venturina Borlongan
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Giampiero La Rocca
- Section of Histology and Embryology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (E.R.); (G.A.)
| |
Collapse
|
5
|
Mižíková I, Thébaud B. Perinatal origins of bronchopulmonary dysplasia-deciphering normal and impaired lung development cell by cell. Mol Cell Pediatr 2023; 10:4. [PMID: 37072570 PMCID: PMC10113423 DOI: 10.1186/s40348-023-00158-2] [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: 06/30/2022] [Accepted: 03/26/2023] [Indexed: 04/20/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a multifactorial disease occurring as a consequence of premature birth, as well as antenatal and postnatal injury to the developing lung. BPD morbidity and severity depend on a complex interplay between prenatal and postnatal inflammation, mechanical ventilation, and oxygen therapy as well as associated prematurity-related complications. These initial hits result in ill-explored aberrant immune and reparative response, activation of pro-fibrotic and anti-angiogenic factors, which further perpetuate the injury. Histologically, the disease presents primarily by impaired lung development and an arrest in lung microvascular maturation. Consequently, BPD leads to respiratory complications beyond the neonatal period and may result in premature aging of the lung. While the numerous prenatal and postnatal stimuli contributing to BPD pathogenesis are relatively well known, the specific cell populations driving the injury, as well as underlying mechanisms are still not well understood. Recently, an effort to gain a more detailed insight into the cellular composition of the developing lung and its progenitor populations has unfold. Here, we provide an overview of the current knowledge regarding perinatal origin of BPD and discuss underlying mechanisms, as well as novel approaches to study the perturbed lung development.
Collapse
Affiliation(s)
- I Mižíková
- Experimental Pulmonology, Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
| | - B Thébaud
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Pediatrics, Children's Hospital of Eastern Ontario (CHEO), CHEO Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
6
|
Liu D, Xu C, Jiang L, Zhu X. Pulmonary endogenous progenitor stem cell subpopulation: Physiology, pathogenesis, and progress. JOURNAL OF INTENSIVE MEDICINE 2023; 3:38-51. [PMID: 36789358 PMCID: PMC9924023 DOI: 10.1016/j.jointm.2022.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/09/2022] [Accepted: 08/13/2022] [Indexed: 06/18/2023]
Abstract
Lungs are structurally and functionally complex organs consisting of diverse cell types from the proximal to distal axis. They have direct contact with the external environment and are constantly at risk of various injuries. Capable to proliferate and differentiate, pulmonary endogenous progenitor stem cells contribute to the maintenance of lung structure and function both under homeostasis and following injuries. Discovering candidate pulmonary endogenous progenitor stem cell types and underlying regenerative mechanisms provide insights into therapeutic strategy development for lung diseases. In this review, we reveal their compositions, roles in lung disease pathogenesis and injury repair, and the underlying mechanisms. We further underline the advanced progress in research approach and potential therapy for lung regeneration. We also demonstrate the feasibility and prospects of pulmonary endogenous stem cell transplantation for lung disease treatment.
Collapse
Affiliation(s)
- Di Liu
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Chufan Xu
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Xiaoyan Zhu
- Department of Physiology, Navy Medical University, 800 Xiangyin Road, Shanghai 200433, China
| |
Collapse
|
7
|
Kruk D, Yeung ACY, Faiz A, ten Hacken NHT, Timens W, van Kuppevelt TH, Daamen W, Hof D, Harmsen MC, Rojas M, Heijink IH. Gene expression profiles in mesenchymal stromal cells from bone marrow, adipose tissue and lung tissue of COPD patients and controls. Respir Res 2023; 24:22. [PMID: 36681830 PMCID: PMC9863276 DOI: 10.1186/s12931-023-02314-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/03/2023] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is characterized by irreversible lung tissue damage. Novel regenerative strategies are urgently awaited. Cultured mesenchymal stem/stromal cells (MSCs) have shown promising results in experimental models of COPD, but differences between sources may impact on their potential use in therapeutic strategies in patients. AIM To assess the transcriptome of lung-derived MSCs (LMSCs), bone marrow-derived MSCs (BM-MSC) and adipose-derived MSCs (AD-MSCs) from COPD patients and non-COPD controls. METHODS We studied differences in gene expression profiles between the MSC-subtypes, as well as between COPD and control using RNA sequencing (RNA-seq). RESULTS We show that besides heterogeneity between donors, MSCs from different sources have strongly divergent gene signatures. The growth factors FGF10 and HGF were predominantly expressed in LMSCs. MSCs from all sources displayed altered expression profiles in COPD, with most pronounced significantly up- and downregulated genes in MSCs from adipose tissue. Pathway analysis revealed that the most differentially expressed genes in COPD-derived AD-MSCs are involved in extracellular matrix (ECM) binding and expression. In LMSCs, the gene that differed most strongly between COPD and control was CSGALNACT1, an ECM modulating gene. CONCLUSION Autologous MSCs from COPD patients display abnormalities with respect to their transcriptome, which were surprisingly most profound in MSCs from extrapulmonary sources. LMSCs may be optimally equipped for lung tissue repair because of the expression of specific growth factor genes.
Collapse
Affiliation(s)
- Dennis Kruk
- grid.4494.d0000 0000 9558 4598Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Groningen The Netherlands ,grid.4494.d0000 0000 9558 4598Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anna C. Y. Yeung
- grid.117476.20000 0004 1936 7611Respiratory Bioinformatics and Molecular Biology (RBMB) Group, The University of Technology Sydney, Ultimo, NSW Australia ,grid.1013.30000 0004 1936 834XWoolcock Institute of Medical Research, The University of Sydney, Glebe, NSW Australia
| | - Alen Faiz
- grid.117476.20000 0004 1936 7611Respiratory Bioinformatics and Molecular Biology (RBMB) Group, The University of Technology Sydney, Ultimo, NSW Australia
| | - Nick H. T. ten Hacken
- grid.4494.d0000 0000 9558 4598Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands ,grid.4494.d0000 0000 9558 4598Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Wim Timens
- grid.4494.d0000 0000 9558 4598Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Groningen The Netherlands ,grid.4494.d0000 0000 9558 4598Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Toin H. van Kuppevelt
- grid.5590.90000000122931605Department of Biochemistry, University of Nijmegen, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Willeke Daamen
- grid.5590.90000000122931605Department of Biochemistry, University of Nijmegen, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Danique Hof
- grid.5590.90000000122931605Department of Biochemistry, University of Nijmegen, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martin C. Harmsen
- grid.4494.d0000 0000 9558 4598Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Groningen The Netherlands
| | - Mauricio Rojas
- grid.261331.40000 0001 2285 7943Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, Columbus, OH USA
| | - Irene H. Heijink
- grid.4494.d0000 0000 9558 4598Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Groningen The Netherlands ,grid.4494.d0000 0000 9558 4598Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands ,grid.4494.d0000 0000 9558 4598Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
8
|
Liu C, Xiao K, Xie L. Advances in mesenchymal stromal cell therapy for acute lung injury/acute respiratory distress syndrome. Front Cell Dev Biol 2022; 10:951764. [PMID: 36036014 PMCID: PMC9399751 DOI: 10.3389/fcell.2022.951764] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/19/2022] [Indexed: 11/29/2022] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) develops rapidly and has high mortality. ALI/ARDS is mainly manifested as acute or progressive hypoxic respiratory failure. At present, there is no effective clinical intervention for the treatment of ALI/ARDS. Mesenchymal stromal cells (MSCs) show promise for ALI/ARDS treatment due to their biological characteristics, easy cultivation, low immunogenicity, and abundant sources. The therapeutic mechanisms of MSCs in diseases are related to their homing capability, multidirectional differentiation, anti-inflammatory effect, paracrine signaling, macrophage polarization, the polarization of the MSCs themselves, and MSCs-derived exosomes. In this review, we discuss the pathogenesis of ALI/ARDS along with the biological characteristics and mechanisms of MSCs in the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Chang Liu
- School of Medicine, Nankai University, Tianjin, China
- Center of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
| | - Kun Xiao
- Center of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- *Correspondence: Kun Xiao, ; Lixin Xie,
| | - Lixin Xie
- School of Medicine, Nankai University, Tianjin, China
- Center of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- *Correspondence: Kun Xiao, ; Lixin Xie,
| |
Collapse
|
9
|
Shao F, Liu R, Tan X, Zhang Q, Ye L, Yan B, Zhuang Y, Xu J. MSC Transplantation Attenuates Inflammation, Prevents Endothelial Damage and Enhances the Angiogenic Potency of Endogenous MSCs in a Model of Pulmonary Arterial Hypertension. J Inflamm Res 2022; 15:2087-2101. [PMID: 35386223 PMCID: PMC8977867 DOI: 10.2147/jir.s355479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/18/2022] [Indexed: 12/14/2022] Open
Abstract
Purpose Pulmonary arterial hypertension (PAH) is a progressive and fatal pulmonary vascular disease initiated by endothelial dysfunction. Mesenchymal stromal cells (MSCs) have been shown to ameliorate PAH in various rodent models; however, these models do not recapitulate all the histopathological alterations observed in human PAH. Broiler chickens (Gallus gallus) can develop PAH spontaneously with neointimal and plexogenic arteriopathy strikingly similar to that in human patients. Herein, we examined the protective effects of MSC transplantation on the development of PAH in this avian model. Methods Mixed-sex broilers at 15 d of age were received 2×106 MSCs or PBS intravenously. One day later, birds were exposed to cool temperature with excessive salt in their drinking water to induce PAH. Cumulative morbidity from PAH and right-to-left ventricle ratio were recorded. Lung histologic features were evaluated for the presence of endothelial damage, endothelial proliferation and plexiform lesions. Expression of proinflammatory mediators and angiogenic factors in the lung was detected. Matrigel tube formation assay was performed to determine the angiogenic potential of endogenous MSCs. Results MSC administration reduced cumulative PAH morbidity and attenuated endothelial damage, plexiform lesions and production of inflammatory mediators in the lungs. No significant difference in the expression of paracrine angiogenic factors including VEGF-A and TGF-β was determined between groups, suggesting that they are not essential for the beneficial effect of MSC transplantation. Interestingly, the endogenous MSCs from birds receiving MSC transplantation demonstrated endothelial differentiatial capacity in vitro whereas those from the mock birds did not. Conclusion Our results support the therapeutic use of MSC transplantation for PAH treatment and suggest that exogenous MSCs produce beneficial effects through modulating inflammation and endogenous MSC-mediated vascular repair.
Collapse
Affiliation(s)
- Fengjin Shao
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.,Veterinary Medical Center, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Preventive Veterinary Sciences, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Rui Liu
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Xun Tan
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.,Veterinary Medical Center, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Preventive Veterinary Sciences, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.,Hainan Institute of Zhejiang University, Sanya, Hainan Province, People's Republic of China
| | - Qiaoyan Zhang
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.,Veterinary Medical Center, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Preventive Veterinary Sciences, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Lujie Ye
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.,Veterinary Medical Center, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Preventive Veterinary Sciences, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Bingxuan Yan
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.,Veterinary Medical Center, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Preventive Veterinary Sciences, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Ying Zhuang
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.,Veterinary Medical Center, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.,Hainan Institute of Zhejiang University, Sanya, Hainan Province, People's Republic of China
| | - Jiaxue Xu
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.,Veterinary Medical Center, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.,Hainan Institute of Zhejiang University, Sanya, Hainan Province, People's Republic of China
| |
Collapse
|
10
|
Mižíková I, Lesage F, Cyr-Depauw C, Cook DP, Hurskainen M, Hänninen SM, Vadivel A, Bardin P, Zhong S, Carpén O, Vanderhyden BC, Thébaud B. Single-Cell RNA Sequencing-Based Characterization of Resident Lung Mesenchymal Stromal Cells in Bronchopulmonary Dysplasia. Stem Cells 2022; 40:479-492. [PMID: 35445270 PMCID: PMC9199848 DOI: 10.1093/stmcls/sxab023] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/09/2021] [Indexed: 01/26/2023]
Abstract
Late lung development is a period of alveolar and microvascular formation, which is pivotal in ensuring sufficient and effective gas exchange. Defects in late lung development manifest in premature infants as a chronic lung disease named bronchopulmonary dysplasia (BPD). Numerous studies demonstrated the therapeutic properties of exogenous bone marrow and umbilical cord-derived mesenchymal stromal cells (MSCs) in experimental BPD. However, very little is known regarding the regenerative capacity of resident lung MSCs (L-MSCs) during normal development and in BPD. In this study we aimed to characterize the L-MSC population in homeostasis and upon injury. We used single-cell RNA sequencing (scRNA-seq) to profile in situ Ly6a+ L-MSCs in the lungs of normal and O2-exposed neonatal mice (a well-established model to mimic BPD) at 3 developmental timepoints (postnatal days 3, 7, and 14). Hyperoxia exposure increased the number and altered the expression profile of L-MSCs, particularly by increasing the expression of multiple pro-inflammatory, pro-fibrotic, and anti-angiogenic genes. In order to identify potential changes induced in the L-MSCs transcriptome by storage and culture, we profiled 15 000 Ly6a+ L-MSCs after in vitro culture. We observed great differences in expression profiles of in situ and cultured L-MSCs, particularly those derived from healthy lungs. Additionally, we have identified the location of Ly6a+/Col14a1+ L-MSCs in the developing lung and propose Serpinf1 as a novel, culture-stable marker of L-MSCs. Finally, cell communication analysis suggests inflammatory signals from immune and endothelial cells as main drivers of hyperoxia-induced changes in L-MSCs transcriptome.
Collapse
Affiliation(s)
- Ivana Mižíková
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Flore Lesage
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Chanele Cyr-Depauw
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - David P Cook
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
| | - Maria Hurskainen
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada,Division of Pediatric Cardiology, New Children’s Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland,Pediatric Research Center, New Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Satu M Hänninen
- Precision Cancer Pathology, Department of Pathology and Research Program in Systems Oncology, University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Arul Vadivel
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Pauline Bardin
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Shumei Zhong
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Olli Carpén
- Precision Cancer Pathology, Department of Pathology and Research Program in Systems Oncology, University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Barbara C Vanderhyden
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Obstetrics and Gynecology, University of Ottawa/The Ottawa Hospital, Ottawa, ON, Canada
| | - Bernard Thébaud
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada,Department of Pediatrics, Children’s Hospital of Eastern Ontario (CHEO) and CHEO Research Institute, University of Ottawa, Ottawa, ON, Canada,Corresponding author: Bernard Thébaud, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6.
| |
Collapse
|
11
|
Feyaerts D, Urbschat C, Gaudillière B, Stelzer IA. Establishment of tissue-resident immune populations in the fetus. Semin Immunopathol 2022; 44:747-766. [PMID: 35508672 PMCID: PMC9067556 DOI: 10.1007/s00281-022-00931-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/17/2022] [Indexed: 12/15/2022]
Abstract
The immune system establishes during the prenatal period from distinct waves of stem and progenitor cells and continuously adapts to the needs and challenges of early postnatal and adult life. Fetal immune development not only lays the foundation for postnatal immunity but establishes functional populations of tissue-resident immune cells that are instrumental for fetal immune responses amidst organ growth and maturation. This review aims to discuss current knowledge about the development and function of tissue-resident immune populations during fetal life, focusing on the brain, lung, and gastrointestinal tract as sites with distinct developmental trajectories. While recent progress using system-level approaches has shed light on the fetal immune landscape, further work is required to describe precise roles of prenatal immune populations and their migration and adaptation to respective organ environments. Defining points of prenatal susceptibility to environmental challenges will support the search for potential therapeutic targets to positively impact postnatal health.
Collapse
Affiliation(s)
- Dorien Feyaerts
- grid.168010.e0000000419368956Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA USA
| | - Christopher Urbschat
- grid.13648.380000 0001 2180 3484Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg, Hamburg, Germany
| | - Brice Gaudillière
- grid.168010.e0000000419368956Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA USA ,grid.168010.e0000000419368956Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA USA
| | - Ina A. Stelzer
- grid.168010.e0000000419368956Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA USA
| |
Collapse
|
12
|
Paracrine Regulation of Alveolar Epithelial Damage and Repair Responses by Human Lung-Resident Mesenchymal Stromal Cells. Cells 2021; 10:cells10112860. [PMID: 34831082 PMCID: PMC8616441 DOI: 10.3390/cells10112860] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/17/2021] [Accepted: 10/21/2021] [Indexed: 11/17/2022] Open
Abstract
COPD is characterized by irreversible lung tissue damage. We hypothesized that lung-derived mesenchymal stromal cells (LMSCs) reduce alveolar epithelial damage via paracrine processes, and may thus be suitable for cell-based strategies in COPD. We aimed to assess whether COPD-derived LMSCs display abnormalities. LMSCs were isolated from lung tissue of severe COPD patients and non-COPD controls. Effects of LMSC conditioned-medium (CM) on H2O2-induced, electric field- and scratch-injury were studied in A549 and NCI-H441 epithelial cells. In organoid models, LMSCs were co-cultured with NCI-H441 or primary lung cells. Organoid number, size and expression of alveolar type II markers were assessed. Pre-treatment with LMSC-CM significantly attenuated oxidative stress-induced necrosis and accelerated wound repair in A549. Co-culture with LMSCs supported organoid formation in NCI-H441 and primary epithelial cells, resulting in significantly larger organoids with lower type II-marker positivity in the presence of COPD-derived versus control LMSCs. Similar abnormalities developed in organoids from COPD compared to control-derived lung cells, with significantly larger organoids. Collectively, this indicates that LMSCs' secretome attenuates alveolar epithelial injury and supports epithelial repair. Additionally, LMSCs promote generation of alveolar organoids, with abnormalities in the supportive effects of COPD-derived LMCS, reflective of impaired regenerative responses of COPD distal lung cells.
Collapse
|
13
|
Sentek H, Klein D. Lung-Resident Mesenchymal Stem Cell Fates within Lung Cancer. Cancers (Basel) 2021; 13:cancers13184637. [PMID: 34572864 PMCID: PMC8472774 DOI: 10.3390/cancers13184637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Lung cancer remains the leading cause of cancer-related deaths worldwide. Herein, the heterogeneous tumor stroma decisively impacts on tumor progression, therapy resistance, and, thus, poor clinical outcome. Among the numerous non-epithelial cells constructing the complex environment of lung carcinomas, mesenchymal stem cells (MSC) gained attraction being stromal precursor cells that could be recruited and ‘educated’ by lung cancer cells to adopt a tumor-associated MSC phenotype, serve as source for activated fibroblasts and presumably for vascular mural cells finally reinforcing tumor progression. Lung-resident MSCs should be considered as ‘local MSCs in stand by’ ready to be arranged within the cancer stroma. Abstract Lung-resident mesenchymal stem cells (LR-MSCs) are non-hematopoietic multipotent stromal cells that predominately reside adventitial within lung blood vessels. Based on their self-renewal and differentiation properties, LR-MSCs turned out to be important regulators of normal lung homeostasis. LR-MSCs exert beneficial effects mainly by local secretion of various growth factors and cytokines that in turn foster pulmonary regeneration including suppression of inflammation. At the same time, MSCs derived from various tissues of origins represent the first choice of cells for cell-based therapeutic applications in clinical medicine. Particularly for various acute as well as chronic lung diseases, the therapeutic applications of exogenous MSCs were shown to mediate beneficial effects, hereby improving lung function and survival. In contrast, endogenous MSCs of normal lungs seem not to be sufficient for lung tissue protection or repair following a pathological trigger; LR-MSCs could even contribute to initiation and/or progression of lung diseases, particularly lung cancer because of their inherent tropism to migrate towards primary tumors and metastatic sites. However, the role of endogenous LR-MSCs to be multipotent tumor-associated (stromal) precursors remains to be unraveled. Here, we summarize the recent knowledge how ‘cancer-educated’ LR-MSCs impact on lung cancer with a focus on mesenchymal stem cell fates.
Collapse
Affiliation(s)
| | - Diana Klein
- Correspondence: ; Tel.: +49-(0)-201-7238-3342
| |
Collapse
|
14
|
Rangasamy T, Ghimire L, Jin L, Le J, Periasamy S, Paudel S, Cai S, Jeyaseelan S. Host Defense against Klebsiella pneumoniae Pneumonia Is Augmented by Lung-Derived Mesenchymal Stem Cells. THE JOURNAL OF IMMUNOLOGY 2021; 207:1112-1127. [PMID: 34341173 DOI: 10.4049/jimmunol.2000688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 06/16/2021] [Indexed: 11/19/2022]
Abstract
Klebsiella pneumoniae is a common cause of Gram-negative pneumonia. The spread of antibiotic-resistant and hypervirulent strains has made treatment more challenging. This study sought to determine the immunomodulatory, antibacterial, and therapeutic potential of purified murine stem cell Ag-1+ (Sca-1+) lung mesenchymal stem cells (LMSCs) using in vitro cell culture and an in vivo mouse model of pneumonia caused by K pneumoniae. Sca-1+ LMSCs are plastic adherent, possess colony-forming capacity, express mesenchymal stem cell markers, differentiate into osteogenic and adipogenic lineages in vitro, and exhibit a high proliferative capacity. Further, these Sca-1+ LMSCs are morphologically similar to fibroblasts but differ ultrastructurally. Moreover, Sca-1+ LMSCs have the capacity to inhibit LPS-induced secretion of inflammatory cytokines by bone marrow-derived macrophages and neutrophils in vitro. Sca-1+ LMSCs inhibit the growth of K pneumoniae more potently than do neutrophils. Sca-1+ LMSCs also possess the intrinsic ability to phagocytize and kill K. pneumoniae intracellularly. Whereas the induction of autophagy promotes bacterial replication, inhibition of autophagy enhances the intracellular clearance of K. pneumoniae in Sca-1+ LMSCs during the early time of infection. Adoptive transfer of Sca-1+ LMSCs in K. pneumoniae-infected mice improved survival, reduced inflammatory cells in bronchoalveolar lavage fluid, reduced inflammatory cytokine levels and pathological lesions in the lung, and enhanced bacterial clearance in the lung and in extrapulmonary organs. To our knowledge, these results together illustrate for the first time the protective role of LMSCs in bacterial pneumonia.
Collapse
Affiliation(s)
- Tirumalai Rangasamy
- Center for Lung Biology and Disease, Louisiana State University, Baton Rouge, LA; .,Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Laxman Ghimire
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Liliang Jin
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - John Le
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Sivakumar Periasamy
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Sagar Paudel
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Shanshan Cai
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Samithamby Jeyaseelan
- Center for Lung Biology and Disease, Louisiana State University, Baton Rouge, LA; .,Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and.,Division of Pulmonary and Critical Care, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA
| |
Collapse
|
15
|
Samarelli AV, Tonelli R, Heijink I, Martin Medina A, Marchioni A, Bruzzi G, Castaniere I, Andrisani D, Gozzi F, Manicardi L, Moretti A, Cerri S, Fantini R, Tabbì L, Nani C, Mastrolia I, Weiss DJ, Dominici M, Clini E. Dissecting the Role of Mesenchymal Stem Cells in Idiopathic Pulmonary Fibrosis: Cause or Solution. Front Pharmacol 2021; 12:692551. [PMID: 34290610 PMCID: PMC8287856 DOI: 10.3389/fphar.2021.692551] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is one of the most aggressive forms of idiopathic interstitial pneumonias, characterized by chronic and progressive fibrosis subverting the lung's architecture, pulmonary functional decline, progressive respiratory failure, and high mortality (median survival 3 years after diagnosis). Among the mechanisms associated with disease onset and progression, it has been hypothesized that IPF lungs might be affected either by a regenerative deficit of the alveolar epithelium or by a dysregulation of repair mechanisms in response to alveolar and vascular damage. This latter might be related to the progressive dysfunction and exhaustion of the resident stem cells together with a process of cellular and tissue senescence. The role of endogenous mesenchymal stromal/stem cells (MSCs) resident in the lung in the homeostasis of these mechanisms is still a matter of debate. Although endogenous MSCs may play a critical role in lung repair, they are also involved in cellular senescence and tissue ageing processes with loss of lung regenerative potential. In addition, MSCs have immunomodulatory properties and can secrete anti-fibrotic factors. Thus, MSCs obtained from other sources administered systemically or directly into the lung have been investigated for lung epithelial repair and have been explored as a potential therapy for the treatment of lung diseases including IPF. Given these multiple potential roles of MSCs, this review aims both at elucidating the role of resident lung MSCs in IPF pathogenesis and the role of administered MSCs from other sources for potential IPF therapies.
Collapse
Affiliation(s)
- Anna Valeria Samarelli
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children and Adults University Hospital of Modena and Reggio Emilia, Modena, Italy
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, Modena, Italy
| | - Roberto Tonelli
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children and Adults University Hospital of Modena and Reggio Emilia, Modena, Italy
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, Modena, Italy
| | - Irene Heijink
- University of Groningen, Departments of Pathology & Medical Biology and Pulmonology, GRIAC Research Institute, University Medical Center Groningen, Groningen, Netherlands
| | - Aina Martin Medina
- IdISBa (Institut d’Investigacio Sanitaria Illes Balears), Palma de Mallorca, Spain
| | - Alessandro Marchioni
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children and Adults University Hospital of Modena and Reggio Emilia, Modena, Italy
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, Modena, Italy
| | - Giulia Bruzzi
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children and Adults University Hospital of Modena and Reggio Emilia, Modena, Italy
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, Modena, Italy
| | - Ivana Castaniere
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children and Adults University Hospital of Modena and Reggio Emilia, Modena, Italy
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, Modena, Italy
| | - Dario Andrisani
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children and Adults University Hospital of Modena and Reggio Emilia, Modena, Italy
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, Modena, Italy
| | - Filippo Gozzi
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children and Adults University Hospital of Modena and Reggio Emilia, Modena, Italy
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, Modena, Italy
| | - Linda Manicardi
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children and Adults University Hospital of Modena and Reggio Emilia, Modena, Italy
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, Modena, Italy
| | - Antonio Moretti
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children and Adults University Hospital of Modena and Reggio Emilia, Modena, Italy
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, Modena, Italy
| | - Stefania Cerri
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children and Adults University Hospital of Modena and Reggio Emilia, Modena, Italy
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, Modena, Italy
| | - Riccardo Fantini
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, Modena, Italy
| | - Luca Tabbì
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, Modena, Italy
| | - Chiara Nani
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, Modena, Italy
| | - Ilenia Mastrolia
- Laboratory of Cellular Therapy, Program of Cell Therapy and Immuno-Oncology, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Daniel J. Weiss
- Department of Medicine, University of Vermont, Burlington, VT, United States
| | - Massimo Dominici
- Oncology Unit, University Hospital of Modena, University of Modena and Reggio Emilia, Modena, Italy
| | - Enrico Clini
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children and Adults University Hospital of Modena and Reggio Emilia, Modena, Italy
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, Modena, Italy
| |
Collapse
|
16
|
Brügger M, Démoulins T, Barut GT, Zumkehr B, Oliveira Esteves BI, Mehinagic K, Haas Q, Schögler A, Rameix-Welti MA, Eléouët JF, Moehrlen U, Marti TM, Schmid RA, Summerfield A, Posthaus H, Ruggli N, Hall SRR, Alves MP. Pulmonary mesenchymal stem cells are engaged in distinct steps of host response to respiratory syncytial virus infection. PLoS Pathog 2021; 17:e1009789. [PMID: 34320038 PMCID: PMC8351988 DOI: 10.1371/journal.ppat.1009789] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/09/2021] [Accepted: 07/08/2021] [Indexed: 02/06/2023] Open
Abstract
Lung-resident (LR) mesenchymal stem and stromal cells (MSCs) are key elements of the alveolar niche and fundamental regulators of homeostasis and regeneration. We interrogated their function during virus-induced lung injury using the highly prevalent respiratory syncytial virus (RSV) which causes severe outcomes in infants. We applied complementary approaches with primary pediatric LR-MSCs and a state-of-the-art model of human RSV infection in lamb. Remarkably, RSV-infection of pediatric LR-MSCs led to a robust activation, characterized by a strong antiviral and pro-inflammatory phenotype combined with mediators related to T cell function. In line with this, following in vivo infection, RSV invades and activates LR-MSCs, resulting in the expansion of the pulmonary MSC pool. Moreover, the global transcriptional response of LR-MSCs appears to follow RSV disease, switching from an early antiviral signature to repair mechanisms including differentiation, tissue remodeling, and angiogenesis. These findings demonstrate the involvement of LR-MSCs during virus-mediated acute lung injury and may have therapeutic implications.
Collapse
Affiliation(s)
- Melanie Brügger
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Thomas Démoulins
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - G. Tuba Barut
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Beatrice Zumkehr
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Blandina I. Oliveira Esteves
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Kemal Mehinagic
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Quentin Haas
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Aline Schögler
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Marie-Anne Rameix-Welti
- Université Paris-Saclay, INSERM, Université de Versailles St. Quentin, UMR 1173 (2I), Versailles, France
| | | | - Ueli Moehrlen
- Pediatric Surgery, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Thomas M. Marti
- Department of Biomedical Research, University of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ralph A. Schmid
- Department of Biomedical Research, University of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Artur Summerfield
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Horst Posthaus
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Nicolas Ruggli
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Sean R. R. Hall
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | - Marco P. Alves
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
17
|
Kwon DH, Park JB, Lee JS, Kim SJ, Choi B, Lee KY. Human delta like 1-expressing human mesenchymal stromal cells promote human T cell development and antigen-specific response in humanized NOD/SCID/IL-2R[Formula: see text] null (NSG) mice. Sci Rep 2021; 11:10603. [PMID: 34011992 PMCID: PMC8134586 DOI: 10.1038/s41598-021-90110-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/30/2021] [Indexed: 01/01/2023] Open
Abstract
Human delta-like 1 (hDlk1) is known to be able to regulate cell fate decisions during hematopoiesis. Mesenchymal stromal cells (MSCs) are known to exhibit potent immunomodulatory roles in a variety of diseases. Herein, we investigated in vivo functions of hDlk1-hMSCs and hDlk1+hMSCs in T cell development and T cell response to viral infection in humanized NOD/SCID/IL-2Rγnull (NSG) mice. Co-injection of hDlk1-hMSC with hCD34+ cord blood (CB) cells into the liver of NSG mice markedly suppressed the development of human T cells. In contrast, co-injection of hDlk1+hMSC with hCD34+ CB cells into the liver of NSG dramatically promoted the development of human T cells. Human T cells developed in humanized NSG mice represent markedly diverse, functionally active, TCR V[Formula: see text] usages, and the restriction to human MHC molecules. Upon challenge with Epstein-Barr virus (EBV), EBV-specific hCD8+ T cells in humanized NSG mice were effectively mounted with phenotypically activated T cells presented as hCD45+hCD3+hCD8+hCD45RO+hHLA-DR+ T cells, suggesting that antigen-specific T cell response was induced in the humanized NSG mice. Taken together, our data suggest that the hDlk1-expressing MSCs can effectively promote the development of human T cells and immune response to exogenous antigen in humanized NSG mice. Thus, the humanized NSG model might have potential advantages for the development of therapeutics targeting infectious diseases in the future.
Collapse
Affiliation(s)
- Do Hee Kwon
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, 16419 Gyeonggi-do Korea
| | - Jae Berm Park
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Joo Sang Lee
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon-Si, 440-746 Kyonggi-Do Korea
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419 Republic of Korea
| | - Sung Joo Kim
- GenNBio, Inc., Seoul, Republic of Korea
- Department of Medicine, Sungkyunkwan University School of Medicine, Suwon-Si, Kyonggi-Do, 440-746 Korea
| | - Bongkum Choi
- GenNBio, Inc., Seoul, Republic of Korea
- Department of Medicine, Sungkyunkwan University School of Medicine, Suwon-Si, Kyonggi-Do, 440-746 Korea
| | - Ki-Young Lee
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, 16419 Gyeonggi-do Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419 Republic of Korea
| |
Collapse
|
18
|
Rolandsson Enes S, Krasnodembskaya AD, English K, Dos Santos CC, Weiss DJ. Research Progress on Strategies that can Enhance the Therapeutic Benefits of Mesenchymal Stromal Cells in Respiratory Diseases With a Specific Focus on Acute Respiratory Distress Syndrome and Other Inflammatory Lung Diseases. Front Pharmacol 2021; 12:647652. [PMID: 33953680 PMCID: PMC8089479 DOI: 10.3389/fphar.2021.647652] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/29/2021] [Indexed: 01/16/2023] Open
Abstract
Recent advances in cell based therapies for lung diseases and critical illnesses offer significant promise. Despite encouraging preclinical results, the translation of efficacy to the clinical settings have not been successful. One of the possible reasons for this is the lack of understanding of the complex interaction between mesenchymal stromal cells (MSCs) and the host environment. Other challenges for MSC cell therapies include cell sources, dosing, disease target, donor variability, and cell product manufacturing. Here we provide an overview on advances and current issues with a focus on MSC-based cell therapies for inflammatory acute respiratory distress syndrome varieties and other inflammatory lung diseases.
Collapse
Affiliation(s)
- Sara Rolandsson Enes
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
| | - Anna D Krasnodembskaya
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queens University, Belfast, United Kingdom
| | - Karen English
- Cellular Immunology Laboratory, Biology Department, Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Claudia C Dos Santos
- Interdepartmental Division of Critical Care, Department of Medicine and the Keenan Center for Biomedical Research, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Daniel J Weiss
- Department of Medicine, 226 Health Science Research Facility, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| |
Collapse
|
19
|
In Vitro Generation of Vascular Wall-Typical Mesenchymal Stem Cells (VW-MSC) from Murine Induced Pluripotent Stem Cells Through VW-MSC-Specific Gene Transfer. Methods Mol Biol 2021. [PMID: 32474869 DOI: 10.1007/978-1-0716-0655-1_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Among the adult stem cells, multipotent mesenchymal stem cells (MSCs) turned out to be a promising option for cell-based therapies for the treatment of various diseases including autoimmune and cardiovascular disorders. MSCs bear a high proliferation and differentiation capability and exert immunomodulatory functions while being still clinically safe. As tissue-resident stem cells, MSCs can be isolated from various tissue including peripheral or umbilical cord blood, placenta, blood, fetal liver, lung, adipose tissue, and blood vessels, although the most commonly used source for MSCs is the bone marrow. However, the proportion of MSCs in primary isolates from adult tissue biopsies is rather low, and therefore MSCs must be intensively expanded in vitro before the MSCs find particular use in therapies that may require extensive and repetitive cell replacement. Therefore, more easily accessible sources of MSCs are needed. Here, we present a detailed protocol to generate tissue-typical MSCs by direct linage conversion using transcription factors defining target MSC identity from murine induced pluripotent stem cells (iPSCs).
Collapse
|
20
|
Tynecka M, Moniuszko M, Eljaszewicz A. Old Friends with Unexploited Perspectives: Current Advances in Mesenchymal Stem Cell-Based Therapies in Asthma. Stem Cell Rev Rep 2021; 17:1323-1342. [PMID: 33649900 PMCID: PMC7919631 DOI: 10.1007/s12015-021-10137-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs) have a great regenerative and immunomodulatory potential that was successfully tested in numerous pre-clinical and clinical studies of various degenerative, hematological and inflammatory disorders. Over the last few decades, substantial immunoregulatory effects of MSC treatment were widely observed in different experimental models of asthma. Therefore, it is tempting to speculate that stem cell-based treatment could become an attractive means to better suppress asthmatic airway inflammation, especially in subjects resistant to currently available anti-inflammatory therapies. In this review, we discuss mechanisms accounting for potent immunosuppressive properties of MSCs and the rationale for their use in asthma. We describe in detail an intriguing interplay between MSCs and other crucial players in the immune system as well as lung microenvironment. Finally, we reveal the potential of MSCs in maintaining airway epithelial integrity and alleviating lung remodeling.
Collapse
Affiliation(s)
- Marlena Tynecka
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269, Białystok, Poland
| | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269, Białystok, Poland.
- Department of Allergology and Internal Medicine, Medical University of Bialystok, ul. M. Skłodowskiej-Curie 24A, Białystok, 15-276, Poland.
| | - Andrzej Eljaszewicz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269, Białystok, Poland.
| |
Collapse
|
21
|
Nwabo Kamdje AH, Seke Etet PF, Simo Tagne R, Vecchio L, Lukong KE, Krampera M. Tumor Microenvironment Uses a Reversible Reprogramming of Mesenchymal Stromal Cells to Mediate Pro-tumorigenic Effects. Front Cell Dev Biol 2020; 8:545126. [PMID: 33330442 PMCID: PMC7710932 DOI: 10.3389/fcell.2020.545126] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 10/28/2020] [Indexed: 12/13/2022] Open
Abstract
The role of mesenchymal stromal cells (MSCs) in the tumor microenvironment is well described. Available data support that MSCs display anticancer activities, and that their reprogramming by cancer cells in the tumor microenvironment induces their switch toward pro-tumorigenic activities. Here we discuss the recent evidence of pro-tumorigenic effects of stromal cells, in particular (i) MSC support to cancer cells through the metabolic reprogramming necessary to maintain their malignant behavior and stemness, and (ii) MSC role in cancer cell immunosenescence and in the establishment and maintenance of immunosuppression in the tumor microenvironment. We also discuss the mechanisms of tumor microenvironment mediated reprogramming of MSCs, including the effects of hypoxia, tumor stiffness, cancer-promoting cells, and tumor extracellular matrix. Finally, we summarize the emerging strategies for reprogramming tumor MSCs to reactivate anticancer functions of these stromal cells.
Collapse
Affiliation(s)
- Armel H. Nwabo Kamdje
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences (FMBS), University of Ngaoundéré, Ngaoundéré, Cameroon
| | - Paul F. Seke Etet
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences (FMBS), University of Ngaoundéré, Ngaoundéré, Cameroon
- Center for Sustainable Health and Development, Garoua, Cameroon
| | - Richard Simo Tagne
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences (FMBS), University of Ngaoundéré, Ngaoundéré, Cameroon
| | - Lorella Vecchio
- Center for Sustainable Health and Development, Garoua, Cameroon
| | - Kiven Erique Lukong
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Mauro Krampera
- Section of Hematology, Stem Cell Research Laboratory, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
22
|
Nwabo Kamdje AH, Seke Etet PF, Simo RT, Vecchio L, Lukong KE, Krampera M. Emerging data supporting stromal cell therapeutic potential in cancer: reprogramming stromal cells of the tumor microenvironment for anti-cancer effects. Cancer Biol Med 2020; 17:828-841. [PMID: 33299638 PMCID: PMC7721102 DOI: 10.20892/j.issn.2095-3941.2020.0133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/20/2020] [Indexed: 02/03/2023] Open
Abstract
After more than a decade of controversy on the role of stromal cells in the tumor microenvironment, the emerging data shed light on pro-tumorigenic and potential anti-cancer factors, as well as on the roots of the discrepancies. We discuss the pro-tumorigenic effects of stromal cells, considering the effects of tumor drivers like hypoxia and tumor stiffness on these cells, as well as stromal cell-mediated adiposity and immunosuppression in the tumor microenvironment, and cancer initiating cells' cellular senescence and adaptive metabolism. We summarize the emerging data supporting stromal cell therapeutic potential in cancer, discuss the possibility to reprogram stromal cells of the tumor microenvironment for anti-cancer effects, and explore some causes of discrepancies on the roles of stromal cells in cancer in the available literature.
Collapse
Affiliation(s)
- Armel Hervé Nwabo Kamdje
- Department of Biomedical Sciences, University of Ngaoundere, Faculty of Science, Ngaoundere 454, Cameroon
| | - Paul Faustin Seke Etet
- Department of Physiological Sciences and Biochemistry, University of Ngaoundéré, Garoua 454, Cameroon
- Center for Sustainable Health and Development, Garoua 454, Cameroon
| | - Richard Tagne Simo
- Department of Biomedical Sciences, University of Ngaoundere, Faculty of Science, Ngaoundere 454, Cameroon
| | - Lorella Vecchio
- Center for Sustainable Health and Development, Garoua 454, Cameroon
| | - Kiven Erique Lukong
- Department of Biochemistry, Microbiology & Immunology, University of Saskatchewan, College of Medicine, Saskatoon SK S7N 5E5, Canada
| | - Mauro Krampera
- Department of Medicine, University of Verona, Section of Hematology, Stem Cell Research Laboratory, Verona 37134, Italy
| |
Collapse
|
23
|
Zhang YM, Zhang LY, Zhou H, Li YY, Wei KX, Li CH, Zhou T, Wang JF, Wei WJ, Hua JR, He Y, Hong T, Liu YQ. Astragalus polysaccharide inhibits radiation-induced bystander effects by regulating apoptosis in Bone Mesenchymal Stem Cells (BMSCs). Cell Cycle 2020; 19:3195-3207. [PMID: 33121344 DOI: 10.1080/15384101.2020.1838793] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
The purpose of this study was to investigate the effects of astragalus polysaccharides (APS) on the proliferation and apoptosis of bone marrow mesenchymal stem cells (BMSCs) induced by X-ray radiation-induced A549 cells bystander effect (RIBE), and to explore their mechanisms. In this study, APS increased the reduced cell proliferation rate induced by RIBE and inhibiting the apoptosis of bystander cells. In terms of mechanism, APS up-regulates the proteins Bcl-2, Bcl-xl, and down-regulates the proteins Bax and Bak, which induces a decrease in mitochondrial membrane potential, which induces the release of Cyt-c and AIF, which leads to caspase-dependent and caspase-independent pathway to cause apoptosis. In addition, we believe that ROS may be the main cause of these protein changes. APS can inhibit the generation of ROS in bystander cells and thus inhibit the activation of the mitochondrial pathway, further preventing cellular damage caused by RIBE.
Collapse
Affiliation(s)
- Yi-Ming Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Li-Ying Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Heng Zhou
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences , Lanzhou, China
| | - Yang-Yang Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Kong-Xi Wei
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Cheng-Hao Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Ting Zhou
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Ju-Fang Wang
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences , Lanzhou, China
| | - Wen-Jun Wei
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences , Lanzhou, China
| | - Jun-Rui Hua
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences , Lanzhou, China
| | - Yun He
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Tao Hong
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Yong-Qi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| |
Collapse
|
24
|
Steens J, Klar L, Hansel C, Slama A, Hager T, Jendrossek V, Aigner C, Klein D. The vascular nature of lung-resident mesenchymal stem cells. Stem Cells Transl Med 2020; 10:128-143. [PMID: 32830458 PMCID: PMC7780817 DOI: 10.1002/sctm.20-0191] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/13/2020] [Accepted: 07/27/2020] [Indexed: 12/14/2022] Open
Abstract
Human lungs bear their own reservoir of endogenous mesenchymal stem cells (MSCs). Although described as located perivascular, the cellular identity of primary lung MSCs remains elusive. Here we investigated the vascular nature of lung‐resident MSCs (LR‐MSCs) using healthy human lung tissue. LR‐MSCs predominately reside within the vascular stem cell niche, the so‐called vasculogenic zone of adult lung arteries. Primary LR‐MSCs isolated from normal human lung tissue showed typical MSC characteristics in vitro and were phenotypically and functionally indistinguishable from MSCs derived from the vascular wall of adult human blood vessels (VW‐MSCs). Moreover, LR‐MSCs expressed the VW‐MSC‐specific HOX code a characteristic to discriminate VW‐MSCs from phenotypical similar cells. Thus, LR‐MSC should be considered as VW‐MSCs. Immunofluorescent analyses of non‐small lung cancer (NSCLC) specimen further confirmed the vascular adventitia as stem cell niche for LR‐MSCs, and revealed their mobilization and activation in NSCLC progression. These findings have implications for understanding the role of MSC in normal lung physiology and pulmonary diseases, as well as for the rational design of additional therapeutic approaches.
Collapse
Affiliation(s)
- Jennifer Steens
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Essen, Germany
| | - Lea Klar
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Essen, Germany
| | - Christine Hansel
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Essen, Germany
| | - Alexis Slama
- Department of Thoracic Surgery and Surgical Endoscopy, Ruhrlandklinik-University Clinic Essen, Essen, Germany
| | - Thomas Hager
- Institute of Pathology, University Clinic Essen, University of Duisburg-Essen, Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Essen, Germany
| | - Clemens Aigner
- Department of Thoracic Surgery and Surgical Endoscopy, Ruhrlandklinik-University Clinic Essen, Essen, Germany
| | - Diana Klein
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Essen, Germany
| |
Collapse
|
25
|
Beisang DJ, Smith K, Yang L, Benyumov A, Gilbertsen A, Herrera J, Lock E, Racila E, Forster C, Sandri BJ, Henke CA, Bitterman PB. Single-cell RNA sequencing reveals that lung mesenchymal progenitor cells in IPF exhibit pathological features early in their differentiation trajectory. Sci Rep 2020; 10:11162. [PMID: 32636398 PMCID: PMC7341888 DOI: 10.1038/s41598-020-66630-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 05/06/2020] [Indexed: 12/12/2022] Open
Abstract
In Idiopathic Pulmonary Fibrosis (IPF), there is unrelenting scarring of the lung mediated by pathological mesenchymal progenitor cells (MPCs) that manifest autonomous fibrogenicity in xenograft models. To determine where along their differentiation trajectory IPF MPCs acquire fibrogenic properties, we analyzed the transcriptome of 335 MPCs isolated from the lungs of 3 control and 3 IPF patients at the single-cell level. Using transcriptional entropy as a metric for differentiated state, we found that the least differentiated IPF MPCs displayed the largest differences in their transcriptional profile compared to control MPCs. To validate entropy as a surrogate for differentiated state functionally, we identified increased CD44 as a characteristic of the most entropic IPF MPCs. Using FACS to stratify IPF MPCs based on CD44 expression, we determined that CD44hi IPF MPCs manifested an increased capacity for anchorage-independent colony formation compared to CD44lo IPF MPCs. To validate our analysis morphologically, we used two differentially expressed genes distinguishing IPF MPCs from control (CD44, cell surface; and MARCKS, intracellular). In IPF lung tissue, pathological MPCs resided in the highly cellular perimeter region of the fibroblastic focus. Our data support the concept that IPF fibroblasts acquire a cell-autonomous pathological phenotype early in their differentiation trajectory.
Collapse
Affiliation(s)
- Daniel J Beisang
- University of Minnesota, Department of Pediatrics, Division of Pediatric Pulmonology, Minneapolis, USA
| | - Karen Smith
- University of Minnesota, Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Minneapolis, USA
| | - Libang Yang
- University of Minnesota, Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Minneapolis, USA
| | - Alexey Benyumov
- University of Minnesota, Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Minneapolis, USA
| | - Adam Gilbertsen
- University of Minnesota, Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Minneapolis, USA
| | - Jeremy Herrera
- University of Manchester, School of Biological Sciences, Division of Cell Matrix Biology & Regenerative Medicine, Manchester, United Kingdom
| | - Eric Lock
- University of Minnesota, School of Public Health, Division of Biostatistics, Minneapolis, USA
| | - Emilian Racila
- University of Minnesota, Department of Laboratory Medicine and Pathology, Minneapolis, USA
| | - Colleen Forster
- University of Minnesota, Clinical and Translational Science Institute, Minneapolis, USA
| | - Brian J Sandri
- University of Minnesota, Department of Pediatrics, Division of Neonatology, Minneapolis, USA
| | - Craig A Henke
- University of Minnesota, Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Minneapolis, USA
| | - Peter B Bitterman
- University of Minnesota, Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Minneapolis, USA.
| |
Collapse
|
26
|
Abstract
Abstract
Purpose of Review
Mesenchymal stromal cell (MSC)–based therapies provide a platform for new therapeutic strategies in lung diseases. This review provides an overview of the current status of the field, along with some of the challenges ahead including better understanding of MSC actions in different lung diseases, personalized approaches to select patients most likely to benefit, and the growing problem of stem cell tourism.
Recent Findings
A newly evolving concept suggests that MSCs shape their immunomodulatory actions depending on the environment they encounter. Furthermore, in some models, it appears that dying or dead cells may contribute to the therapeutic efficacy by activating the host response.
Summary
Despite many pre-clinical studies demonstrating that MSCs can be used to treat lung disorders, clinical trials have failed to show improved outcome. Understanding the complex interaction between MSCs and the host microenvironment is likely to be an important area for enhancing the efficacy of MSC-based cell therapies.
Collapse
|
27
|
Sveiven SN, Nordgren TM. Lung-resident mesenchymal stromal cells are tissue-specific regulators of lung homeostasis. Am J Physiol Lung Cell Mol Physiol 2020; 319:L197-L210. [PMID: 32401672 DOI: 10.1152/ajplung.00049.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Until recently, data supporting the tissue-resident status of mesenchymal stromal cells (MSC) has been ambiguous since their discovery in the 1950-60s. These progenitor cells were first discovered as bone marrow-derived adult multipotent cells and believed to migrate to sites of injury, opposing the notion that they are residents of all tissue types. In recent years, however, it has been demonstrated that MSC can be found in all tissues and MSC from different tissues represent distinct populations with differential protein expression unique to each tissue type. Importantly, these cells are efficient mediators of tissue repair, regeneration, and prove to be targets for therapeutics, demonstrated by clinical trials (phase 1-4) for MSC-derived therapies for diseases like graft-versus-host-disease, multiple sclerosis, rheumatoid arthritis, and Crohn's disease. The tissue-resident status of MSC found in the lung is a key feature of their importance in the context of disease and injuries of the respiratory system, since these cells could be instrumental to providing more specific and targeted therapies. Currently, bone marrow-derived MSC have been established in the treatment of disease, including diseases of the lung. However, with lung-resident MSC representing a unique population with a different phenotypic and gene expression pattern than MSC derived from other tissues, their role in remediating lung inflammation and injury could provide enhanced efficacy over bone marrow-derived MSC methods. Through this review, lung-resident MSC will be characterized, using previously published data, by surface markers, gene expression patterns, and compared with bone-marrow MSC to highlight similarities and, importantly, differences in these cell types.
Collapse
Affiliation(s)
- Stefanie Noel Sveiven
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, California
| | - Tara M Nordgren
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, California
| |
Collapse
|
28
|
Florez-Sampedro L, Soto-Gamez A, Poelarends GJ, Melgert BN. The role of MIF in chronic lung diseases: looking beyond inflammation. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1183-L1197. [PMID: 32208924 DOI: 10.1152/ajplung.00521.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine that has been associated with many diseases. Most studies found in literature describe MIF as a proinflammatory cytokine involved in chronic inflammatory conditions, but evidence from last years suggests that many of its key effects are not directly related to inflammation. In fact, MIF is constitutively expressed in most human tissues and in some cases in high levels, which does not reflect the pattern of expression of a classic proinflammatory cytokine. Moreover, MIF is highly expressed during embryonic development and decreases during adulthood, which point toward a more likely role as growth factor. Accordingly, MIF knockout mice develop age-related spontaneous emphysema, suggesting that MIF presence (e.g., in younger individuals and wild-type animals) is part of a healthy lung. In view of this new line of evidence, we aimed to review data on the role of MIF in the pathogenesis of chronic lung diseases.
Collapse
Affiliation(s)
- Laura Florez-Sampedro
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands.,Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Abel Soto-Gamez
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands.,European Institute for the Biology of Aging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerrit J Poelarends
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Barbro N Melgert
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
29
|
Möbius MA, Freund D, Vadivel A, Koss S, McConaghy S, Ohls RK, Rüdiger M, Thébaud B. Oxygen Disrupts Human Fetal Lung Mesenchymal Cells. Implications for Bronchopulmonary Dysplasia. Am J Respir Cell Mol Biol 2019; 60:592-600. [PMID: 30562051 DOI: 10.1165/rcmb.2018-0358oc] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Exogenous mesenchymal stromal cells (MSCs) ameliorate experimental bronchopulmonary dysplasia. Moreover, data from term-born animal models and human tracheal aspirate-derived cells suggest altered mesenchymal signaling in the pathophysiology of neonatal lung disease. We hypothesized that hyperoxia, a factor contributing to the development of bronchopulmonary dysplasia, perturbs human lung-resident MSC function. Mesenchymal cells were isolated from human fetal lung tissue (16-18 wk of gestation), characterized and cultured in conditions resembling either intrauterine (5% O2) or extrauterine (21% and 60% O2) atmospheres. Secretome data were compared with MSCs obtained from term umbilical cord tissues. The human fetal lung mesenchyme almost exclusively contains CD146pos. MSCs expressing SOX-2 and OCT-4, which secrete elastin, fibroblast growth factors 7 and 10, vascular endothelial growth factor, angiogenin, and other lung cell-protecting/-maturing proteins. Exposure to extrauterine atmospheres in vitro leads to excessive proliferation, reduced colony-forming ability, alterations in the cell's surface marker profile, decreased elastin deposition, and impaired secretion of factors important for lung growth. Conversely, umbilical cord-derived MSCs abundantly secreted factors that impaired lung MSCs are unable to produce. Oxygen-impaired human fetal lung MSC function may contribute to disrupted repair capacity and arrested lung growth. Exogenous MSCs may act by triggering the signaling pathways lost by impaired endogenous lung mesenchymal cells.
Collapse
Affiliation(s)
- Marius A Möbius
- 1 Fachbereich Neonatologie und Pädiatrische Intensivmedizin, Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum und Medizinische Fakultät Carl Gustav Carus der Technischen Universität Dresden, Dresden, Saxony, Germany.,2 Deutsche Forschungsgemeinschaft Research Center and Cluster of Excellence for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Saxony, Germany.,3 Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Daniel Freund
- 1 Fachbereich Neonatologie und Pädiatrische Intensivmedizin, Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum und Medizinische Fakultät Carl Gustav Carus der Technischen Universität Dresden, Dresden, Saxony, Germany.,2 Deutsche Forschungsgemeinschaft Research Center and Cluster of Excellence for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Saxony, Germany
| | - Arul Vadivel
- 3 Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Sarah Koss
- 1 Fachbereich Neonatologie und Pädiatrische Intensivmedizin, Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum und Medizinische Fakultät Carl Gustav Carus der Technischen Universität Dresden, Dresden, Saxony, Germany
| | - Suzanne McConaghy
- 4 Division of Neonatology, Department of Pediatrics, University of New Mexico, Albuquerque, New Mexico; and
| | - Robin K Ohls
- 4 Division of Neonatology, Department of Pediatrics, University of New Mexico, Albuquerque, New Mexico; and
| | - Mario Rüdiger
- 1 Fachbereich Neonatologie und Pädiatrische Intensivmedizin, Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum und Medizinische Fakultät Carl Gustav Carus der Technischen Universität Dresden, Dresden, Saxony, Germany.,2 Deutsche Forschungsgemeinschaft Research Center and Cluster of Excellence for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Saxony, Germany
| | - Bernard Thébaud
- 3 Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,5 Division of Neonatology, Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| |
Collapse
|
30
|
Cruz T, López-Giraldo A, Noell G, Guirao A, Casas-Recasens S, Garcia T, Saco A, Sellares J, Agustí A, Faner R. Smoking Impairs the Immunomodulatory Capacity of Lung-Resident Mesenchymal Stem Cells in Chronic Obstructive Pulmonary Disease. Am J Respir Cell Mol Biol 2019; 61:575-583. [DOI: 10.1165/rcmb.2018-0351oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Tamara Cruz
- Centro Investigación Biomédica en Red Enfermedades Respiratorias, Barcelona, Spain
| | - Alejandra López-Giraldo
- Centro Investigación Biomédica en Red Enfermedades Respiratorias, Barcelona, Spain
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Guillaume Noell
- Centro Investigación Biomédica en Red Enfermedades Respiratorias, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; and
| | - Angela Guirao
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | | | - Tamara Garcia
- Centro Investigación Biomédica en Red Enfermedades Respiratorias, Barcelona, Spain
| | - Adela Saco
- Department of Pathology, Hospital Clinic, Barcelona, Spain
| | - Jacobo Sellares
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; and
| | - Alvar Agustí
- Centro Investigación Biomédica en Red Enfermedades Respiratorias, Barcelona, Spain
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; and
| | - Rosa Faner
- Centro Investigación Biomédica en Red Enfermedades Respiratorias, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; and
| |
Collapse
|
31
|
Clinical Application of Stem/Stromal Cells in COPD. STEM CELL-BASED THERAPY FOR LUNG DISEASE 2019. [PMCID: PMC7121219 DOI: 10.1007/978-3-030-29403-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive life-threatening disease that is significantly increasing in prevalence and is predicted to become the third leading cause of death worldwide by 2030. At present, there are no true curative treatments that can stop the progression of the disease, and new therapeutic strategies are desperately needed. Advances in cell-based therapies provide a platform for the development of new therapeutic approaches in severe lung diseases such as COPD. At present, a lot of focus is on mesenchymal stem (stromal) cell (MSC)-based therapies, mainly due to their immunomodulatory properties. Despite increasing number of preclinical studies demonstrating that systemic MSC administration can prevent or treat experimental COPD and emphysema, clinical studies have not been able to reproduce the preclinical results and to date no efficacy or significantly improved lung function or quality of life has been observed in COPD patients. Importantly, the completed appropriately conducted clinical trials uniformly demonstrate that MSC treatment in COPD patients is well tolerated and no toxicities have been observed. All clinical trials performed so far, have been phase I/II studies, underpowered for the detection of potential efficacy. There are several challenges ahead for this field such as standardized isolation and culture procedures to obtain a cell product with high quality and reproducibility, administration strategies, improvement of methods to measure outcomes, and development of potency assays. Moreover, COPD is a complex pathology with a diverse spectrum of clinical phenotypes, and therefore it is essential to develop methods to select the subpopulation of patients that is most likely to potentially respond to MSC administration. In this chapter, we will discuss the current state of the art of MSC-based cell therapy for COPD and the hurdles that need to be overcome.
Collapse
|
32
|
Zhang L, Luo Y, Lu Z, He J, Wang L, Zhang L, Zhang Y, Liu Y. Astragalus Polysaccharide Inhibits Ionizing Radiation-Induced Bystander Effects by Regulating MAPK/NF-kB Signaling Pathway in Bone Mesenchymal Stem Cells (BMSCs). Med Sci Monit 2018; 24:4649-4658. [PMID: 29976920 PMCID: PMC6069470 DOI: 10.12659/msm.909153] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background This study investigated the effect of Astragalus polysaccharides (APS) on radiation-induced bystander effects (RIBE) in human bone mesenchymal stem cells (BMSCs) induced by irradiated A549 cells. Material/Methods A549 cells were irradiated with 2 Gy X-rays to obtain conditioned medium. BMSCs were incubated with the conditioned medium or APS. The levels of reactive oxygen species (ROS) and TGF-β were detected by ELISA. Cell survival, genomic instability, and DNA damages were detected by CCK-8 assay, colony formation assay, the micronucleus test and immunofluorescence assay, respectively. The protein and phosphorylation protein expression of p38, c-Jun N-terminal kinase (JNK), extracellular regulated protein kinase (ERK1/2), P65, and cyclooxygenase-2 (COX-2) in bystander effect cells were detected by Western blot. Results The expression of COX-2 and ROS increased following stimulation with conditioned medium; this effect was inhibited by pre-exposing the cells to APS. BMSCs growth and colony formation rate decreased following stimulation with conditioned medium; this effect was suppressed by pre-exposing the cells to APS. In addition, the micronucleus rate and 53BP1 foci number increased after treatment with conditioned medium; this increase in BMSCs was inhibited by APS. The levels of phosphorylated p38, JNK, ERK1/2, NF-κB P65, and COX-2 proteins were increased by conditioned medium but were decreased by pre-treatment with APS. Conclusions RIBE in BMSCs induced by the irradiated A549 was mediated by the ROS in the conditioned medium and might be related to MAPK/NF-κB signal pathways in BMSCs. APS may block RIBE through regulating the MAPK/NF-κB pathway.
Collapse
Affiliation(s)
- Liying Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China (mainland)
| | - Yali Luo
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China (mainland)
| | - Zhiwei Lu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China (mainland)
| | - Jinpeng He
- Key Laboratory of Space Radiobiology of Gansu Province and Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China (mainland)
| | - Lei Wang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China (mainland)
| | - Lixin Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China (mainland)
| | - Yiming Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China (mainland)
| | - Yongqi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China (mainland).,Key Laboratory for Transfer of Dunhuang Medicine at the Provincial and Ministerial Level, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China (mainland)
| |
Collapse
|
33
|
Collins JJP, Lithopoulos MA, Dos Santos CC, Issa N, Möbius MA, Ito C, Zhong S, Vadivel A, Thébaud B. Impaired Angiogenic Supportive Capacity and Altered Gene Expression Profile of Resident CD146 + Mesenchymal Stromal Cells Isolated from Hyperoxia-Injured Neonatal Rat Lungs. Stem Cells Dev 2018; 27:1109-1124. [PMID: 29957134 DOI: 10.1089/scd.2017.0145] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD), the most common complication of extreme preterm birth, can be caused by oxygen-related lung injury and is characterized by impaired alveolar and vascular development. Mesenchymal stromal cells (MSCs) have lung protective effects. Conversely, BPD is associated with increased MSCs in tracheal aspirates. We hypothesized that endogenous lung (L-)MSCs are perturbed in a well-established oxygen-induced rat model mimicking BPD features. Rat pups were exposed to 21% or 95% oxygen from birth to postnatal day 10. On day 12, CD146+ L-MSCs were isolated and characterized according to the International Society for Cellular Therapy criteria. Epithelial and vascular repair potential were tested by scratch assay and endothelial network formation, respectively, immune function by mixed lymphocyte reaction assay. Microarray analysis was performed using the Affymetrix GeneChip and gene set enrichment analysis software. CD146+ L-MSCs isolated from rat pups exposed to hyperoxia had decreased CD73 expression and inhibited lung endothelial network formation. CD146+ L-MSCs indiscriminately promoted epithelial wound healing and limited T cell proliferation. Expression of potent antiangiogenic genes of the axonal guidance cue and CDC42 pathways was increased after in vivo hyperoxia, whereas genes of the anti-inflammatory Janus kinase (JAK)/signal transducer and activator of transcription (STAT) and lung/vascular growth-promoting fibroblast growth factor (FGF) pathways were decreased. In conclusion, in vivo hyperoxia exposure alters the proangiogenic effects and FGF expression of L-MSCs. In addition, decreased CD73 and JAK/STAT expression suggests decreased immune function. L-MSC function may be perturbed and contribute to BPD pathogenesis. These findings may lead to improvements in manufacturing exogenous MSCs with superior repair capabilities.
Collapse
Affiliation(s)
- Jennifer J P Collins
- 1 Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute , Ottawa, Canada .,2 Department of Cellular and Molecular Medicine, University of Ottawa , Ottawa, Canada
| | - Marissa A Lithopoulos
- 1 Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute , Ottawa, Canada .,2 Department of Cellular and Molecular Medicine, University of Ottawa , Ottawa, Canada
| | - Claudia C Dos Santos
- 3 Keenan Research Centre for Biomedical Science of St. Michael's Hospital , Toronto, Canada .,4 Interdepartmental Division of Critical Care Medicine, University of Toronto , Toronto, Canada
| | - Nahla Issa
- 1 Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute , Ottawa, Canada .,2 Department of Cellular and Molecular Medicine, University of Ottawa , Ottawa, Canada
| | - Marius A Möbius
- 1 Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute , Ottawa, Canada .,5 Department of Neonatology and Pediatric Critical Care Medicine, Medical Faculty and University Hospital Carl Gustav Carus , Technische Universität Dresden, Dresden, Germany .,6 DFG Research Center and Cluster of Excellence for Regenerative Therapies (CRTD) , Technische Universität Dresden, Dresden, Germany
| | - Caryn Ito
- 1 Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute , Ottawa, Canada .,2 Department of Cellular and Molecular Medicine, University of Ottawa , Ottawa, Canada
| | - Shumei Zhong
- 1 Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute , Ottawa, Canada
| | - Arul Vadivel
- 1 Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute , Ottawa, Canada
| | - Bernard Thébaud
- 1 Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute , Ottawa, Canada .,2 Department of Cellular and Molecular Medicine, University of Ottawa , Ottawa, Canada .,7 Children's Hospital of Eastern Ontario Research Institute , Ottawa, Canada
| |
Collapse
|
34
|
Li JH, Fan WS, Wang MM, Wang YH, Ren ZG. Effects of mesenchymal stem cells on solid tumor metastasis in experimental cancer models: a systematic review and meta-analysis. J Transl Med 2018; 16:113. [PMID: 29703232 PMCID: PMC5924448 DOI: 10.1186/s12967-018-1484-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 04/17/2018] [Indexed: 12/09/2022] Open
Abstract
Background It has been reported mesenchymal stem cells (MSCs) are recruited to and become integral parts of the tumor microenvironment. MSCs might have an active role in solid tumor progression, especially cancer metastasis. However, the contribution of MSCs in the process of cancer metastasis is still controversial. In this review, we performed a meta-analysis on the effects of MSCs administration on cancer metastasis based on published preclinical studies. Methods The PRISMA guidelines were used. A total of 42 publications met the inclusion criteria. Outcome data on the incidence and the number of cancer metastasis as well as study characteristics were extracted. Quality of the studies was assessed according to SYRCLE Risk of Bias tool. Random-effects meta-analysis was used to pool estimates. Results Of the 42 studies included, 32 reported that MSCs administration promoted outcome events (numbers or incidences of cancer metastasis), and 39 reported data suitable for meta-analysis. The median effect size (RR) was 2.04 for the incidence of cancer metastasis (95% CI 1.57–2.65, I2 = 21%), and the median effect size (SMD) was 1.23 for the number of cancer metastasis (95% CI 0.43–2.03, I2 = 89%). Heterogeneity was observed, with the greater impact based on study length and different ways of metastasis measurement and MSCs administration. Conclusion Our results suggested MSCs administration increased the number and the incidence of cancer metastasis in experimental cancer models. High heterogeneity and poor reported risk of bias limit the quality of these findings. Further preclinical studies with better design and adequate reporting are still needed. Electronic supplementary material The online version of this article (10.1186/s12967-018-1484-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jing-Huan Li
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China.,Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Wen-Shuai Fan
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Mi-Mi Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China.,Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yan-Hong Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China.,Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zheng-Gang Ren
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China. .,Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
35
|
The development of an extra-anatomic tissue-engineered artery with collateral arteries for therapeutic angiogenesis in ischemic hind limb. Sci Rep 2018; 8:4627. [PMID: 29545563 PMCID: PMC5854599 DOI: 10.1038/s41598-018-22799-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/01/2018] [Indexed: 01/12/2023] Open
Abstract
To develop tissue-engineered arteries (TEAs) with collateral arteries(CAs) in ischemic hind limb goat models(IHLMs). The IHLMs created by removing femoral arteries were divided into non-treated control group(NG); non-catheter group (NCG) in which TEA was anastomosed to external iliac artery(EIA), and surrounded with collagen sponge containing autologous MSCs and VEGF-gelatin microspheres, the distal end of TEA was ligated; catheter group(CG) which received the same procedure as NCG, also received heparin infusion through catheter in EIA. TEA patency was assessed weekly by Ultrasound. The TEA and CAs were assessed by angiography, gross examination, histology and electron microscopy. In CG, TEAs remained patent for 1 month, but became partly occluded 1 week after catheter withdrawn. In NCG, TEAs were occluded 1 week after implantation. Angiography demonstrated that communication between CAs arising from the TEAs and the native vessels was established in both groups. NCG had fewer CAs than CG (P < 0.01). At 40 days, TEAs in CG demonstrated of endothelium formation, smooth muscle cells infiltration and collagen regeneration. The CG had more capillaries and mature vessels in adventia of TEAs than NCG (P < 0.01). CG group also had more vessels around TEAs than NCG (P < 0.01) or NG (P < 0.001).
Collapse
|
36
|
Khatri M, Richardson LA. Therapeutic potential of porcine bronchoalveolar fluid-derived mesenchymal stromal cells in a pig model of LPS-induced ALI. J Cell Physiol 2018; 233:5447-5457. [PMID: 29231967 DOI: 10.1002/jcp.26397] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/04/2017] [Indexed: 12/20/2022]
Abstract
In this study, we isolated mesenchymal stromal (stem) cells (MSCs) from broncho-alveolar lavage fluid (BAL) of 2-6-week-old commercial pigs. BAL-MSCs displayed fibroblastic morphology and possessed self-renewal properties. Similar to bone-marrow MSCs, BAL-MSCs expressed mesenchymal markers and both cell types lacked the expression of hematopoetic markers. BAL-MSCs, when cultured in differentiation induction media, differentiated into adipocytes, osteocytes, and chondrocytes. Next, we examined if BAL-MSCs have the ability to treat lipopolysaccharide (LPS)-induced acute lung injury (ALI) in a pig model. Five-week-old commercial pigs were inoculated intra-tracheally with E. coli LPS (1 mg/kg body weight [b.wt.]). Twelve hours after the LPS inoculation, groups of pigs were inoculated intra-tracheally with BM-MSCs or BAL-MSCs (2 × 106 cells/kg b.wt.). Forty eight hours after the cells administration pigs were euthanized and neutrophils in BAL, lung lesions, and cytokines in lung lysates, and engraftment of MSCs in lungs were examined. Engraftment of BAL-MSCs in injured lungs was significantly higher than the BM-MSCs, however, both cell types were equally effective in attenuating LPS-induced ALI as evidenced by decreased inflammation, lung lesions, and proinflammatory cytokines in the lungs of pigs treated with BAL- or BM-MSCs. These data in a preclinical large animal model suggest that BAL-MSCs may be used in clinical settings to treat ALI in humans.
Collapse
Affiliation(s)
- Mahesh Khatri
- Department of Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, Ohio
| | - Levi A Richardson
- Department of Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, Ohio
| |
Collapse
|
37
|
Schnapper A, Christmann A, Knudsen L, Rahmanian P, Choi YH, Zeriouh M, Karavidic S, Neef K, Sterner-Kock A, Guschlbauer M, Hofmaier F, Maul AC, Wittwer T, Wahlers T, Mühlfeld C, Ochs M. Stereological assessment of the blood-air barrier and the surfactant system after mesenchymal stem cell pretreatment in a porcine non-heart-beating donor model for lung transplantation. J Anat 2017; 232:283-295. [PMID: 29193065 DOI: 10.1111/joa.12747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2017] [Indexed: 01/09/2023] Open
Abstract
More frequent utilization of non-heart-beating donor (NHBD) organs for lung transplantation has the potential to relieve the shortage of donor organs. In particular with respect to uncontrolled NHBD, concerns exist regarding the risk of ischaemia/reperfusion (IR) injury-related graft damage or dysfunction. Due to their immunomodulating and tissue-remodelling properties, bone-marrow-derived mesenchymal stem cells (MSCs) have been suspected of playing a beneficial role regarding short- and long-term survival and function of the allograft. Thus, MSC administration might represent a promising pretreatment strategy for NHBD organs. To study the initial effects of warm ischaemia and MSC application, a large animal lung transplantation model was generated, and the structural organ composition of the transplanted lungs was analysed stereologically with particular respect to the blood-gas barrier and the surfactant system. In this study, porcine lungs (n = 5/group) were analysed. Group 1 was the sham-operated control group. In pigs of groups 2-4, cardiac arrest was induced, followed by a period of 3 h of ventilated ischaemia at room temperature. In groups 3 and 4, 50 × 106 MSCs were administered intravascularly via the pulmonary artery and endobronchially, respectively, during the last 10 min of ischaemia. The left lungs were transplanted, followed by a reperfusion period of 4 h. Then, lungs were perfusion-fixed and processed for light and electron microscopy. Samples were analysed stereologically for IR injury-related structural parameters, including volume densities and absolute volumes of parenchyma components, alveolar septum components, intra-alveolar oedema, and the intracellular and intra-alveolar surfactant pool. Additionally, the volume-weighted mean volume of lamellar bodies (lbs) and their profile size distribution were determined. Three hours of ventilated warm ischaemia was tolerated without eliciting histological or ultrastructural signs of IR injury, as revealed by qualitative and quantitative assessment. However, warm ischaemia influenced the surfactant system. The volume-weighted mean volume of lbs was reduced significantly (P = 0.024) in groups subjected to ischaemia (group medians of groups 2-4: 0.180-0.373 μm³) compared with the sham control group (median 0.814 μm³). This was due to a lower number of large lb profiles (size classes 5-15). In contrast, the intra-alveolar surfactant system was not altered significantly. No significant differences were encountered comparing ischaemia alone (group 2) or ischaemia plus application of MSCs (groups 3 and 4) in this short-term model.
Collapse
Affiliation(s)
- Anke Schnapper
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,REBIRTH (From Regenerative Biology to Reconstructive Therapy), Cluster of Excellence, Hannover, Germany
| | - Astrid Christmann
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,REBIRTH (From Regenerative Biology to Reconstructive Therapy), Cluster of Excellence, Hannover, Germany
| | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,REBIRTH (From Regenerative Biology to Reconstructive Therapy), Cluster of Excellence, Hannover, Germany
| | - Parwis Rahmanian
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Yeong-Hoon Choi
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany.,Center of Molecular Medicine, University of Cologne, Cologne, Germany
| | - Mohamed Zeriouh
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Samira Karavidic
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Klaus Neef
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany.,Center of Molecular Medicine, University of Cologne, Cologne, Germany
| | - Anja Sterner-Kock
- Center for Experimental Medicine, University of Cologne, Cologne, Germany
| | - Maria Guschlbauer
- Center for Experimental Medicine, University of Cologne, Cologne, Germany.,Decentral Animal Facility, University of Cologne, Cologne, Germany
| | - Florian Hofmaier
- Center for Experimental Medicine, University of Cologne, Cologne, Germany
| | - Alexandra C Maul
- Center for Experimental Medicine, University of Cologne, Cologne, Germany
| | - Thorsten Wittwer
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany.,Center of Molecular Medicine, University of Cologne, Cologne, Germany
| | - Thorsten Wahlers
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany.,Center of Molecular Medicine, University of Cologne, Cologne, Germany
| | - Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,REBIRTH (From Regenerative Biology to Reconstructive Therapy), Cluster of Excellence, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Matthias Ochs
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,REBIRTH (From Regenerative Biology to Reconstructive Therapy), Cluster of Excellence, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| |
Collapse
|
38
|
Schubert S, Brehm W, Hillmann A, Burk J. Serum-free human MSC medium supports consistency in human but not in equine adipose-derived multipotent mesenchymal stromal cell culture. Cytometry A 2017; 93:60-72. [PMID: 28926198 DOI: 10.1002/cyto.a.23240] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
For clinical applications of multipotent mesenchymal stromal cells (MSCs), serum-free culture is preferable to standardize cell products and prevent contamination with pathogens. In contrast to human MSCs, knowledge on serum-free culture of large animal MSCs is limited, despite its relevance for preclinical studies and development of veterinary cellular therapeutics. This study aimed to evaluate the suitability of a commercially available serum-free human MSC medium for culturing equine adipose-derived MSCs in comparison with human adipose MSCs. Enzyme-free isolation by explant technique and expansion of equine and human cells in the serum-free medium were feasible. However, serum-free culture altered the morphology and complicated handling of equine MSCs, with cell aggregation and spontaneous detachment of multilayers, compared to culture in standard medium supplemented with fetal bovine serum. Furthermore, proliferation and the surface immunophenotype of equine cells were more variable compared to the controls and appeared to depend on the lot of the serum-free medium. Particularly the expression of CD90 was different between experimental groups (P < 0.05), with lower percentages of CD90+ cells found in equine MSC samples cultured in serum-free medium (5.21-83.40%) compared to standard medium (86.20-99.50%). Additionally, small subpopulations expressing MSC exclusion markers such as CD14 (0.28-11.60%), CD34 (0.00-9.87%), CD45 (0.35-10.50%), or MHCII (0.00-3.67%) were found in equine samples after serum-free culture. In contrast, human samples displayed a more consistent morphology and a consistent CD29+ (98.60-99.90%), CD73+ (94.60-98.40%), CD90+ (99.60-99.90%), and CD105+ (97.40-99.80%) immunophenotype after culture in serum-free medium. The obtained data demonstrate that the serum-free medium was suitable for human MSC culture but did not lead to entirely satisfactory results in equine MSCs. This underlines that requirements regarding serum-free culture conditions are species-specific, indicating a need for serum-free media to be optimized for MSCs from relevant animal species. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Susanna Schubert
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Philipp-Rosenthal-Straße 55, Leipzig 04103, Germany.,Faculty of Veterinary Medicine, Institute of Physiology, University of Leipzig, An den Tierkliniken 7, Leipzig 04103, Germany
| | - Walter Brehm
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Philipp-Rosenthal-Straße 55, Leipzig 04103, Germany.,Faculty of Veterinary Medicine, Large Animal Clinic for Surgery, University of Leipzig, An den Tierkliniken 21, Leipzig 04103, Germany
| | - Aline Hillmann
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Philipp-Rosenthal-Straße 55, Leipzig 04103, Germany
| | - Janina Burk
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Philipp-Rosenthal-Straße 55, Leipzig 04103, Germany.,Faculty of Veterinary Medicine, Institute of Physiology, University of Leipzig, An den Tierkliniken 7, Leipzig 04103, Germany
| |
Collapse
|
39
|
Rolandsson Enes S, Åhrman E, Palani A, Hallgren O, Bjermer L, Malmström A, Scheding S, Malmström J, Westergren-Thorsson G. Quantitative proteomic characterization of lung-MSC and bone marrow-MSC using DIA-mass spectrometry. Sci Rep 2017; 7:9316. [PMID: 28839187 PMCID: PMC5570998 DOI: 10.1038/s41598-017-09127-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/19/2017] [Indexed: 01/17/2023] Open
Abstract
Mesenchymal stromal cells (MSC) are ideal candidates for cell therapies, due to their immune-regulatory and regenerative properties. We have previously reported that lung-derived MSC are tissue-resident cells with lung-specific properties compared to bone marrow-derived MSC. Assessing relevant molecular differences between lung-MSC and bone marrow-MSC is important, given that such differences may impact their behavior and potential therapeutic use. Here, we present an in-depth mass spectrometry (MS) based strategy to investigate the proteomes of lung-MSC and bone marrow-MSC. The MS-strategy relies on label free quantitative data-independent acquisition (DIA) analysis and targeted data analysis using a MSC specific spectral library. We identified several significantly differentially expressed proteins between lung-MSC and bone marrow-MSC within the cell layer (352 proteins) and in the conditioned medium (49 proteins). Bioinformatics analysis revealed differences in regulation of cell proliferation, which was functionally confirmed by decreasing proliferation rate through Cytochrome P450 stimulation. Our study reveals important differences within proteome and matrisome profiles between lung- and bone marrow-derived MSC that may influence their behavior and affect the clinical outcome when used for cell-therapy.
Collapse
Affiliation(s)
- Sara Rolandsson Enes
- Department of Experimental Medical Science, Lung Biology Unit, Lund University, 22184, Lund, Sweden.
| | - Emma Åhrman
- Department of Experimental Medical Science, Lung Biology Unit, Lund University, 22184, Lund, Sweden.,Department of Clinical Sciences Lund, Division of Infection Medicine, Lund University, 22184, Lund, Sweden
| | - Anitha Palani
- Department of Experimental Medical Science, Matrix Biology, Lund University, 22184, Lund, Sweden
| | - Oskar Hallgren
- Department of Experimental Medical Science, Lung Biology Unit, Lund University, 22184, Lund, Sweden
| | - Leif Bjermer
- Department of Respiratory Medicine and Allergology, Lund University and Skåne University Hospital, 22184, Lund, Sweden
| | - Anders Malmström
- Department of Experimental Medical Science, Matrix Biology, Lund University, 22184, Lund, Sweden
| | - Stefan Scheding
- Lund Stem Cell Center, Lund University, 22184, Lund, Sweden.,Department of Hematology, Skåne University Hospital, 22184, Lund, Sweden
| | - Johan Malmström
- Department of Clinical Sciences Lund, Division of Infection Medicine, Lund University, 22184, Lund, Sweden
| | | |
Collapse
|