1
|
Liu K, Kang Z, Yang M, Chen F, Xia M, Dai W, Zheng S, Chen H, Lu QR, Zhou W, Lin Y. The role of oligodendrocyte progenitor cells in the spatiotemporal vascularization of the human and mouse neocortex. Glia 2025; 73:140-158. [PMID: 39392208 DOI: 10.1002/glia.24625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 08/21/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024]
Abstract
Brain vasculature formation begins with vessel invasion from the perineural vascular plexus, which expands through vessel sprouting and growth. Recent studies have indicated the existence of oligodendrocyte-vascular crosstalk during development. However, the relationship between oligodendrocyte progenitor cells (OPCs) and the ordered spatiotemporal vascularization of the neocortex has not been elucidated. Our findings suggest that OPCs play a complex role in the vessel density of the embryonic and postnatal neocortex. Analyses of normal human and mouse embryonic cerebral cortex show that vascularization and OPC distribution are tightly controlled in a spatially and temporally restricted manner, exhibiting a positive correlation. Loss of OPCs at both embryonic and postnatal stages led to a reduction in vascular density, suggesting that OPC populations play a role in vascular density. Nonetheless, dynamic observation on cultured brain slices and staining of tissue sections indicate that OPC migration is unassociated with the proximity to blood vessels, primarily occurring along radial glial cell processes. Additionally, in vitro experiments demonstrate that OPC secretions promote vascular endothelial cell (VEC) growth. Together, these observations suggest that vessel density is influenced by OPC secretions.
Collapse
Affiliation(s)
- Kaiyi Liu
- Key Laboratory of Birth Defects, Children's Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhiruo Kang
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Min Yang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Fangbing Chen
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Mingyang Xia
- Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, China
| | - Wenjuan Dai
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Shiyi Zheng
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Huiyao Chen
- Center for Molecular Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Q Richard Lu
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Wenhao Zhou
- Key Laboratory of Birth Defects, Children's Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Division of Neonatology and Center for Newborn Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yifeng Lin
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
2
|
Wang C, Liu X, Zhou J, Zhang X, Zhou Z, Zhang Q. Sensory nerves drive migration of dental pulp stem cells via the CGRP-Ramp1 axis in pulp repair. Cell Mol Life Sci 2024; 81:373. [PMID: 39196292 PMCID: PMC11358583 DOI: 10.1007/s00018-024-05400-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 07/17/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024]
Abstract
Dental pulp stem cells (DPSCs) are responsible for maintaining pulp structure and function after pulp injury. DPSCs migrate directionally to the injury site before differentiating into odontoblast-like cells, which is a prerequisite and a determinant in pulp repair. Increasing evidence suggests that sensory neuron-stem cell crosstalk is critical for maintaining normal physiological functions, and sensory nerves influence stem cells mainly by neuropeptides. However, the role of sensory nerves on DPSC behaviors after pulp injury is largely unexplored. Here, we find that sensory nerves released significant amounts of calcitonin gene-related peptide (CGRP) near the injury site, acting directly on DPSCs via receptor activity modifying protein 1 (RAMP1) to promote collective migration of DPSCs to the injury site, and ultimately promoting pulp repair. Specifically, sensory denervation leads to poor pulp repair and ectopic mineralization, in parallel with that DPSCs failed to be recruited to the injury site. Furthermore, in vitro evidence shows that sensory nerve-deficient microenvironment suppressed DPSC migration prominently among all related behaviors. Mechanistically, the CGRP-Ramp1 axis between sensory neurons and DPSCs was screened by single-cell RNA-seq analysis and immunohistochemical studies confirmed that the expression of CGRP rather than Ramp1 increases substantially near the damaged site. We further demonstrated that CGRP released by sensory nerves binds the receptor Ramp1 on DPSCs to facilitate cell collective migration by an indirect co-culture system using conditioned medium from trigeminal neurons, CGRP recombinant protein and antagonists BIBN4096. The treatment with exogenous CGRP promoted the recruitment of DPSCs, and ultimately enhanced the quality of pulp repair. Targeting the sensory nerve could therefore provide a new strategy for stem cell-based pulp repair and regeneration.
Collapse
Affiliation(s)
- Chunmeng Wang
- Department of Endodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Yanchang Middle Road, Jing'an District, Shanghai, 200072, China
| | - Xiaochen Liu
- Department of Endodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Yanchang Middle Road, Jing'an District, Shanghai, 200072, China
| | - Jiani Zhou
- Department of Endodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Yanchang Middle Road, Jing'an District, Shanghai, 200072, China
| | - Xiaoyi Zhang
- Department of Endodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Yanchang Middle Road, Jing'an District, Shanghai, 200072, China
| | - Zihao Zhou
- Department of Endodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Yanchang Middle Road, Jing'an District, Shanghai, 200072, China
| | - Qi Zhang
- Department of Endodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Yanchang Middle Road, Jing'an District, Shanghai, 200072, China.
| |
Collapse
|
3
|
Thamm JR, Jounaidi Y, Mueller ML, Rosen V, Troulis MJ, Guastaldi FPS. Temporomandibular Joint Fibrocartilage Contains CD105 Positive Mouse Mesenchymal Stem/Progenitor Cells with Increased Chondrogenic Potential. J Maxillofac Oral Surg 2023; 22:559-570. [PMID: 37534349 PMCID: PMC10390456 DOI: 10.1007/s12663-022-01721-6] [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: 06/24/2021] [Accepted: 04/08/2022] [Indexed: 10/18/2022] Open
Abstract
Objective A specific type of mesenchymal stem/progenitor cells (MSPCs), CD105+ is reported to aid in cartilage regeneration through TGF-β/Smad2-signalling. The purpose of this study was to identify and characterize CD105+ MSPCs in temporomandibular joint (TMJ) cartilage. Materials and Methods MSPCs were isolated from mouse TMJ condyle explants and evaluated for their clonogenicity and pluripotential abilities. MSPC were examined for CD105 antigen using immunohistochemistry and flow cytometry. Results Immunohistochemistry revealed presence of CD105+ MSPCs in the proliferative zone of condyle's cartilage. Only 0.2% of isolated MSPCs exhibited CD105, along with the stem cell surface markers CD44 and Sca-1. In CD105+ MSPCs, intracellular immunostaining revealed significantly higher (p < 0.05) protein levels of collagen type 1, 2, proteoglycan 4. Ability for chondrogenic differentiation was found to be significantly higher (p < 0.05) after 4 weeks compared to CD105- cells, using alcian blue staining. CD105+ cells were found to resemble an early MSPC subgroup with significantly higher gene expression of biglycan, proteoglycan 4, collagen type 2, Gli2, Sox5 (p < 0.001) and Sox9 (p < 0.05). In contrast, significantly lower levels of Runx2 (p < 0.05), Osterix, Trps1, Col10a1 (p < 0.01), Ihh (p < 0.001) related to chondrocyte senescence and commitment to osteogenic lineage, were observed compared to CD105- cells. Conclusion The study showed the existence of a CD105+ MSPC subgroup within TMJ fibrocartilage that may be activated to aid in fibrocartilage repair.
Collapse
Affiliation(s)
- Janis R. Thamm
- Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard School of Dental Medicine, Boston, MA USA
| | - Youssef Jounaidi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Max-Laurin Mueller
- Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard School of Dental Medicine, Boston, MA USA
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA USA
| | - Maria J. Troulis
- Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard School of Dental Medicine, Boston, MA USA
- Walter C. Guralnick Professor of Oral and Maxillofacial Surgery, Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard School of Dental Medicine, Boston, MA USA
| | - Fernando Pozzi Semeghini Guastaldi
- Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard School of Dental Medicine, Boston, MA USA
- Skeletal Biology Research Center, Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard School of Dental Medicine, 50 Blossom St, Thier 513A, Boston, MA 02114 USA
| |
Collapse
|
4
|
Zahmatkesh E, Khoshdel Rad N, Hossein-Khannazer N, Mohamadnejad M, Gramignoli R, Najimi M, Malekzadeh R, Hassan M, Vosough M. Cell and cell-derivative-based therapy for liver diseases: current approaches and future promises. Expert Rev Gastroenterol Hepatol 2023; 17:237-249. [PMID: 36692130 DOI: 10.1080/17474124.2023.2172398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
INTRODUCTION According to the recent updates from World Health Organization, liver diseases are the 12th most common cause of mortality. Currently, orthotopic liver transplantation (OLT) is the most effective and the only treatment for end-stage liver diseases. Owing to several shortcomings like finite numbers of healthy organ donors, lifelong immunosuppression, and complexity of the procedure, cell and cell-derivatives therapies have emerged as a potential therapeutic alternative for liver diseases. Various cell types and therapies have been proposed and their therapeutic effects evaluated in preclinical or clinical studies, including hepatocytes, hepatocyte-like cells (HLCs) derived from stem cells, human liver stem cells (HLSCs), combination therapies with various types of cells, organoids, and implantable cell-biomaterial constructs with synthetic and natural polymers or even decellularized extracellular matrix (ECM). AREAS COVERED In this review, we highlighted the current status of cell and cell-derivative-based therapies for liver diseases. Furthermore, we discussed future prospects of using HLCs, liver organoids, and their combination therapies. EXPERT OPINION Promising application of stem cell-based techniques including iPSC technology has been integrated into novel techniques such as gene editing, directed differentiation, and organoid technology. iPSCs offer promising prospects to represent novel therapeutic strategies and modeling liver diseases.
Collapse
Affiliation(s)
- Ensieh Zahmatkesh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Khoshdel Rad
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Mohamadnejad
- Cell-Based Therapies Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Roberto Gramignoli
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Reza Malekzadeh
- Digestive Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
5
|
Schmid M, Kröpfl JM, Spengler CM. Changes in Circulating Stem and Progenitor Cell Numbers Following Acute Exercise in Healthy Human Subjects: a Systematic Review and Meta-analysis. Stem Cell Rev Rep 2021; 17:1091-1120. [PMID: 33389632 PMCID: PMC8316227 DOI: 10.1007/s12015-020-10105-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 12/22/2022]
Abstract
Despite of the increasing number of investigations on the effects of acute exercise on circulating stem and progenitor cell (SC) numbers, and in particular on respective subgroups, i.e. endothelial (ESC), hematopoietic (HSC), and mesenchymal (MSC) stem and progenitor cells, a consensus regarding mechanisms and extent of these effects is still missing. The aim of this meta-analysis was to systematically evaluate the overall-effects of acute exercise on the different SC-subgroups and investigate possible subject- and intervention-dependent factors affecting the extent of SC-mobilization in healthy humans. Trials assessing SC numbers before and at least one timepoint after acute exercise, were identified in a systematic computerized search. Compared to baseline, numbers were significantly increased for early and non-specified SCs (enSCs) until up to 0.5 h after exercise (0–5 min: +0.64 [Standardized difference in means], p < 0.001; 6–20 min: +0.42, p < 0.001; 0.5 h: +0.29, p = 0.049), for ESCs until 12–48 h after exercise (0–5 min: +0.66, p < 0.001; 6–20 min: +0.43 p < 0.001; 0.5 h: +0.43, p = 0.002; 1 h: +0.58, p = 0.001; 2 h: +0.50, p = 0.002; 3–8 h: +0.70, p < 0.001; 12–48 h: +0.38, p = 0.003) and for HSCs at 0–5 min (+ 0.47, p < 0.001) and at 3 h after exercise (+ 0.68, p < 0.001). Sex, intensity and duration of the intervention had generally no influence. The extent and kinetics of the exercise-induced mobilization of SCs differ between SC-subpopulations. However, also definitions of SC-subpopulations are non-uniform. Therefore, finding a consensus with a clear definition of cell surface markers defining ESCs, HSCs and MSCs is a first prerequisite for understanding this important topic. ![]()
Collapse
Affiliation(s)
- M Schmid
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - J M Kröpfl
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - C M Spengler
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland. .,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
| |
Collapse
|
6
|
Zhang Z, Paudel S, Feltham T, Lobao MH, Schon L. Foot fat pad: Characterization by mesenchymal stromal cells in rats. Anat Rec (Hoboken) 2020; 304:1582-1591. [PMID: 33099882 DOI: 10.1002/ar.24549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/02/2020] [Accepted: 09/14/2020] [Indexed: 11/10/2022]
Abstract
Foot fat pad (FFP) is a highly functionalized fat depot of great significance for weight bearing in the foot. Mesenchymal stromal cells (MSCs) in subcutaneous adipose tissues are widely studied for regenerative potentials. MSCs in FFP, which may contribute to the physiological and pathological conditions of the foot, have not been characterized. In this study, MSCs were isolated from FFP (designated as MSCs-ffp) and subcutaneous adipose tissue (designated as MSCs-sub) from rats. The cell surface markers, proliferation, and efficiency of colony formation were compared between MSCs-ffp and MSCs-sub. In addition, MSCs-ffp were induced for osteogenic, chondrogenic, and adipogenic differentiation. The tri-lineage differentiation potentials were compared between MSCs-ffp and MSCs-sub by the expression of Runx2, Sox9, and proliferator-activated receptor gamma (PPAR-γ), respectively, using quantitative polymerized chain reaction. The expression of elastin and associated genes by MSCs-ffp were also evaluated. MSCs-ffp, like MSCs-sub, expressed CD44, CD73, and CD90. MSCs-ffp and MSCs-sub proliferated at similar rates but MSCs-ffp formed more colonies than MSCs-sub. MSCs-ffp were capable of differentiating into osteogenic, chondrogenic, and adipogenic lineages. Under the conditions of osteogenic and adipogenic differentiation, MSCs-sub expressed more Runx2 and PPAR-γ, respectively, than MSCs-ffp. The undifferentiated MSCs-ffp upregulated the expression of fibulin-5. In conclusion, MSCs-ffp shared common biology with MSCs-sub but were more efficient in colony formation, less adipogenic and osteogenic, and participated in elastogenesis. The unique features of MSCs-ffp may relate to their roles in the physiological functions of FFP.
Collapse
Affiliation(s)
- Zijun Zhang
- Center for Orthopaedic Innovation, Mercy Medical Center, Baltimore, Maryland, USA
| | - Sharada Paudel
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Tyler Feltham
- Philadelphia College of Osteopathic Medicine-GA, Suwanee, Georgia, USA
| | - Mario H Lobao
- Department of Orthopaedic Surgery, Columbia University Medical Center, New York, New York, USA
| | - Lew Schon
- Center for Orthopaedic Innovation, Mercy Medical Center, Baltimore, Maryland, USA.,Institute for Foot and Ankle Reconstruction, Mercy Medical Center, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Wu X, Wang Y, Xiao Y, Crawford R, Mao X, Prasadam I. Extracellular vesicles: Potential role in osteoarthritis regenerative medicine. J Orthop Translat 2020; 21:73-80. [PMID: 32099807 PMCID: PMC7029343 DOI: 10.1016/j.jot.2019.10.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 10/14/2019] [Accepted: 10/28/2019] [Indexed: 02/08/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent whole joint disease characterised by cartilage degradation, subchondral bone sclerosis and bone remodelling, and synovium inflammation, leading to pain, deformity, and cartilage dysfunction. Currently, there is no appropriate therapy for OA, and available treatments simply aim to reduce pain and swelling. Exosomes are membrane-bound extracellular vesicles secreted by almost all cells, receiving increasing interest because of their effect in cell-to-cell communication. Increasing evidence suggests that exosomes play an important role in cartilage physiological and pathological effects. This article reviews the potential role of exosomes in OA regenerative medicine. Special attention is given to mesenchymal stem cells-derived exosomes due to the extensive research on their cartilage repair property and their function as miRNA cargo. More investigations are needed for the effects of exosomes from synovial fluid and chondrocytes in joints. A better understanding of the mechanisms will contribute to a novel and promising therapy for OA patients. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE A better understanding of the role of extracellular vesicles in regenerative medicine will contribute to a novel and promising therapy for OA patients.
Collapse
Affiliation(s)
- Xiaoxin Wu
- Department of Orthopaedic Surgery, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011, China
- Institute of Health and Biomedical Innovation, Faculty of Science and Engineering, Queensland University of Technology, Kelvin Grove Campus, Brisbane, QLD 4059 Australia
| | - Yuewen Wang
- Xiangya School of Medicine, Central South University, 172 Tongzipo Road, Changsha, Hunan 410013, China
| | - Yin Xiao
- Institute of Health and Biomedical Innovation, Faculty of Science and Engineering, Queensland University of Technology, Kelvin Grove Campus, Brisbane, QLD 4059 Australia
- Australia–China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Ross Crawford
- Institute of Health and Biomedical Innovation, Faculty of Science and Engineering, Queensland University of Technology, Kelvin Grove Campus, Brisbane, QLD 4059 Australia
- The Prince Charles Hospital, Orthopaedic Department, Brisbane, Queensland, Australia
| | - Xinzhan Mao
- Department of Orthopaedic Surgery, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011, China
| | - Indira Prasadam
- Institute of Health and Biomedical Innovation, Faculty of Science and Engineering, Queensland University of Technology, Kelvin Grove Campus, Brisbane, QLD 4059 Australia
| |
Collapse
|
8
|
Chen F, Liu X, Chen Y, Liu JY, Lu H, Wang W, Lu X, Dean KC, Gao L, Kaplan HJ, Dean DC, Peng X, Liu Y. Sphere-induced reprogramming of RPE cells into dual-potential RPE stem-like cells. EBioMedicine 2020; 52:102618. [PMID: 31982829 PMCID: PMC6994567 DOI: 10.1016/j.ebiom.2019.102618] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 11/20/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022] Open
Abstract
Background The retinal pigment epithelium (RPE) has the potential to regenerate the entire neuroretina upon retinal injury in amphibians. In contrast, this regenerative capacity has been lost in mammals. The reprogramming of differentiated somatic cells into induced pluripotent stem cells (iPSCs) by viral transduction of exogenous stem cell factors has triggered a revolution in regenerative medicine. However, the risks of potential mutation(s) caused by random viral vector insertion in host genomes and tumor formation in recipients hamper its clinical application. One alternative is to immortalize adult stem cells with limited potential or to partially reprogram differentiated somatic cells into progenitor-like cells through non-integration protocols. Methods Sphere-induced RPE stem cells (iRPESCs) were generated from adult mouse RPE cells. Their stem cell functionality was studied in a mouse model of retinal degeneration. The molecular mechanism underlying the sphere-induced reprogramming was investigated using microarray and loss-of-function approaches. Findings We provide evidence that our sphere-induced reprogramming protocol can immortalize and transform mouse RPE cells into iRPESCs with dual potential to differentiate into cells that express either RPE or photoreceptor markers both in vitro and in vivo. When subretinally transplanted into mice with retinal degeneration, iRPESCs can integrate to the RPE and neuroretina, thereby delaying retinal degeneration in the model animals. Our molecular analyses indicate that the Hippo signaling pathway is important in iRPESC reprogramming. Interpretation The Hippo factor Yap1 is activated in the nuclei of cells at the borders of spheres. The factors Zeb1 and P300 downstream of the Hippo pathway are shown to bind to the promoters of the stemness genes Oct4, Klf4 and Sox2, thereby likely transactivate them to reprogram RPE cells into iRPESCs. Fund National Natural Science Foundation of China and the National Institute of Health USA.
Collapse
Affiliation(s)
- Fenghua Chen
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, China
| | - Xiao Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; Department of Ophthalmology, Second Affiliated Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Yao Chen
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
| | - John Y Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | - Huayi Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Wei Wang
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | - Xiaoqin Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | - Kevin C Dean
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | - Ling Gao
- Department of Ophthalmology, Second Affiliated Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Henry J Kaplan
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | - Douglas C Dean
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA; Birth Defects Center; University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.
| | - Xiaoyan Peng
- Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, China.
| | - Yongqing Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA; Birth Defects Center; University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.
| |
Collapse
|
9
|
Thong T, Forté CA, Hill EM, Colacino JA. Environmental exposures, stem cells, and cancer. Pharmacol Ther 2019; 204:107398. [PMID: 31376432 PMCID: PMC6881547 DOI: 10.1016/j.pharmthera.2019.107398] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/29/2019] [Indexed: 12/19/2022]
Abstract
An estimated 70-90% of all cancers are linked to exposure to environmental risk factors. In parallel, the number of stem cells in a tissue has been shown to be a strong predictor of risk of developing cancer in that tissue. Tumors themselves are characterized by an acquisition of "stem cell" characteristics, and a growing body of evidence points to tumors themselves being sustained and propagated by a stem cell-like population. Here, we review our understanding of the interplay between environmental exposures, stem cell biology, and cancer. We provide an overview of the role of stem cells in development, tissue homeostasis, and wound repair. We discuss the pathways and mechanisms governing stem cell plasticity and regulation of the stem cell state, and describe experimental methods for assessment of stem cells. We then review the current understanding of how environmental exposures impact stem cell function relevant to carcinogenesis and cancer prevention, with a focus on environmental and occupational exposures to chemical, physical, and biological hazards. We also highlight key areas for future research in this area, including defining whether the biological basis for cancer disparities is related to effects of complex exposure mixtures on stem cell biology.
Collapse
Affiliation(s)
- Tasha Thong
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Chanese A Forté
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Michigan Institute for Computational Discovery and Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Evan M Hill
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Justin A Colacino
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
10
|
González-Velasco Ó, De Las Rivas J, Lacal J. Proteomic and Transcriptomic Profiling Identifies Early Developmentally Regulated Proteins in Dictyostelium Discoideum. Cells 2019; 8:cells8101187. [PMID: 31581556 PMCID: PMC6830349 DOI: 10.3390/cells8101187] [Citation(s) in RCA: 5] [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: 09/17/2019] [Accepted: 09/26/2019] [Indexed: 02/06/2023] Open
Abstract
Cyclic AMP acts as a secondary messenger involving different cellular functions in eukaryotes. Here, proteomic and transcriptomic profiling has been combined to identify novel early developmentally regulated proteins in eukaryote cells. These proteomic and transcriptomic experiments were performed in Dictyostelium discoideum given the unique advantages that this organism offers as a eukaryotic model for cell motility and as a nonmammalian model of human disease. By comparing whole-cell proteome analysis of developed (cAMP-pulsed) wild-type AX2 cells and an independent transcriptomic analysis of developed wild-type AX4 cells, our results show that up to 70% of the identified proteins overlap in the two independent studies. Among them, we have found 26 proteins previously related to cAMP signaling and identified 110 novel proteins involved in calcium signaling, adhesion, actin cytoskeleton, the ubiquitin-proteasome pathway, metabolism, and proteins that previously lacked any annotation. Our study validates previous findings, mostly for the canonical cAMP-pathway, and also generates further insight into the complexity of the transcriptomic changes during early development. This article also compares proteomic data between parental and cells lacking glkA, a GSK-3 kinase implicated in substrate adhesion and chemotaxis in Dictyostelium. This analysis reveals a set of proteins that show differences in expression in the two strains as well as overlapping protein level changes independent of GlkA.
Collapse
Affiliation(s)
- Óscar González-Velasco
- Bioinformatics and Functional Genomics Research Group. Cancer Research Center (CIC-IBMCC, CSIC/USAL/IBSAL), 37007 Salamanca, Spain.
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Research Group. Cancer Research Center (CIC-IBMCC, CSIC/USAL/IBSAL), 37007 Salamanca, Spain.
| | - Jesus Lacal
- Department of Microbiology and Genetics, Faculty of Biology, University of Salamanca, 37007 Salamanca, Spain.
| |
Collapse
|
11
|
Palma A, Cerquone Perpetuini A, Ferrentino F, Fuoco C, Gargioli C, Giuliani G, Iannuccelli M, Licata L, Micarelli E, Paoluzi S, Perfetto L, Petrilli LL, Reggio A, Rosina M, Sacco F, Vumbaca S, Zuccotti A, Castagnoli L, Cesareni G. Myo-REG: A Portal for Signaling Interactions in Muscle Regeneration. Front Physiol 2019; 10:1216. [PMID: 31611808 PMCID: PMC6776608 DOI: 10.3389/fphys.2019.01216] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/06/2019] [Indexed: 12/12/2022] Open
Abstract
Muscle regeneration is a complex process governed by the interplay between several muscle-resident mononuclear cell populations. Following acute or chronic damage these cell populations are activated, communicate via cell-cell interactions and/or paracrine signals, influencing fate decisions via the activation or repression of internal signaling cascades. These are highly dynamic processes, occurring with distinct temporal and spatial kinetics. The main challenge toward a system level description of the muscle regeneration process is the integration of this plethora of inter- and intra-cellular interactions. We integrated the information on muscle regeneration in a web portal. The scientific content annotated in this portal is organized into two information layers representing relationships between different cell types and intracellular signaling-interactions, respectively. The annotation of the pathways governing the response of each cell type to a variety of stimuli/perturbations occurring during muscle regeneration takes advantage of the information stored in the SIGNOR database. Additional curation efforts have been carried out to increase the coverage of molecular interactions underlying muscle regeneration and to annotate cell-cell interactions. To facilitate the access to information on cell and molecular interactions in the context of muscle regeneration, we have developed Myo-REG, a web portal that captures and integrates published information on skeletal muscle regeneration. The muscle-centered resource we provide is one of a kind in the myology field. A friendly interface allows users to explore, approximately 100 cell interactions or to analyze intracellular pathways related to muscle regeneration. Finally, we discuss how data can be extracted from this portal to support in silico modeling experiments.
Collapse
Affiliation(s)
- Alessandro Palma
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | | | - Claudia Fuoco
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Cesare Gargioli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Giulio Giuliani
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | - Luana Licata
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Elisa Micarelli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Serena Paoluzi
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Livia Perfetto
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | - Alessio Reggio
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Marco Rosina
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Sacco
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Simone Vumbaca
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | - Luisa Castagnoli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Gianni Cesareni
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
12
|
Iansante V, Chandrashekran A, Dhawan A. Cell-based liver therapies: past, present and future. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0229. [PMID: 29786563 DOI: 10.1098/rstb.2017.0229] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2017] [Indexed: 12/16/2022] Open
Abstract
Liver transplantation represents the standard treatment for people with an end-stage liver disease and some liver-based metabolic disorders; however, shortage of liver donor tissues limits its availability. Furthermore, whole liver replacement eliminates the possibility of using native liver as a possible target for future gene therapy in case of liver-based metabolic defects. Cell therapy has emerged as a potential alternative, as cells can provide the hepatic functions and engraft in the liver parenchyma. Various options have been proposed, including human or other species hepatocytes, hepatocyte-like cells derived from stem cells or more futuristic alternatives, such as combination therapies with different cell types, organoids and cell-biomaterial combinations. In this review, we aim to give an overview of the cell therapies developed so far, highlighting preclinical and/or clinical achievements as well as the limitations that need to be overcome to make them fully effective and safe for clinical applications.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.
Collapse
Affiliation(s)
- Valeria Iansante
- Dhawan Lab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London at King's College Hospital, London SE5 9PJ, UK
| | - Anil Chandrashekran
- Dhawan Lab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London at King's College Hospital, London SE5 9PJ, UK
| | - Anil Dhawan
- Dhawan Lab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London at King's College Hospital, London SE5 9PJ, UK
| |
Collapse
|
13
|
Mendieta-Serrano MA, Mendez-Cruz FJ, Antúnez-Mojica M, Schnabel D, Alvarez L, Cárdenas L, Lomelí H, Ruiz-Santiesteban JA, Salas-Vidal E. NADPH-Oxidase-derived reactive oxygen species are required for cytoskeletal organization, proper localization of E-cadherin and cell motility during zebrafish epiboly. Free Radic Biol Med 2019; 130:82-98. [PMID: 30342187 DOI: 10.1016/j.freeradbiomed.2018.10.416] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 12/17/2022]
Abstract
Cell movements are essential for morphogenesis during animal development. Epiboly is the first morphogenetic process in zebrafish in which cells move en masse to thin and spread the deep and enveloping cell layers of the blastoderm over the yolk cell. While epiboly has been shown to be controlled by complex molecular networks, the contribution of reactive oxygen species (ROS) to this process has not previously been studied. Here, we show that ROS are required for epiboly in zebrafish. Visualization of ROS in whole embryos revealed dynamic patterns during epiboly progression. Significantly, inhibition of NADPH oxidase activity leads to a decrease in ROS formation, delays epiboly, alters E-cadherin and cytoskeleton patterns and, by 24 h post-fertilization, decreases embryo survival, effects that are rescued by hydrogen peroxide treatment. Our findings suggest that a delicate ROS balance is required during early development and that disruption of that balance interferes with cell adhesion, leading to defective cell motility and epiboly progression.
Collapse
Affiliation(s)
| | | | - Mayra Antúnez-Mojica
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Avenida Universidad #2001, Colonia Chamilpa, Cuernavaca, Morelos C.P. 62209, Mexico
| | - Denhi Schnabel
- Departamento de Genética del Desarrollo y Fisiología Molecular, Mexico
| | - Laura Alvarez
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Avenida Universidad #2001, Colonia Chamilpa, Cuernavaca, Morelos C.P. 62209, Mexico
| | - Luis Cárdenas
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad #2001, Colonia Chamilpa, Cuernavaca, Morelos C.P. 62210, Mexico
| | - Hilda Lomelí
- Departamento de Genética del Desarrollo y Fisiología Molecular, Mexico
| | | | | |
Collapse
|
14
|
Abnave P, Aboukhatwa E, Kosaka N, Thompson J, Hill MA, Aboobaker AA. Epithelial-mesenchymal transition transcription factors control pluripotent adult stem cell migration in vivo in planarians. Development 2017; 144:3440-3453. [PMID: 28893948 PMCID: PMC5665486 DOI: 10.1242/dev.154971] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/14/2017] [Indexed: 01/23/2023]
Abstract
Migration of stem cells underpins the physiology of metazoan animals. For tissues to be maintained, stem cells and their progeny must migrate and differentiate in the correct positions. This need is even more acute after tissue damage by wounding or pathogenic infection. Inappropriate migration also underpins metastasis. Despite this, few mechanistic studies address stem cell migration during repair or homeostasis in adult tissues. Here, we present a shielded X-ray irradiation assay that allows us to follow stem cell migration in planarians. We demonstrate the use of this system to study the molecular control of stem cell migration and show that snail-1, snail-2 and zeb-1 EMT transcription factor homologs are necessary for cell migration to wound sites and for the establishment of migratory cell morphology. We also observed that stem cells undergo homeostatic migration to anterior regions that lack local stem cells, in the absence of injury, maintaining tissue homeostasis. This requires the polarity determinant notum Our work establishes planarians as a suitable model for further in-depth study of the processes controlling stem cell migration in vivo.
Collapse
Affiliation(s)
- Prasad Abnave
- Department of Zoology, Tinbergen Building, South Parks Road, University of Oxford, Oxford OX1 3PS, UK
| | - Ellen Aboukhatwa
- Department of Zoology, Tinbergen Building, South Parks Road, University of Oxford, Oxford OX1 3PS, UK
| | - Nobuyoshi Kosaka
- Department of Zoology, Tinbergen Building, South Parks Road, University of Oxford, Oxford OX1 3PS, UK
| | - James Thompson
- CRUK/MRC Oxford Institute for Radiation Oncology, ORCRB Roosevelt Drive, University of Oxford, Oxford OX3 7DQ, UK
| | - Mark A Hill
- CRUK/MRC Oxford Institute for Radiation Oncology, ORCRB Roosevelt Drive, University of Oxford, Oxford OX3 7DQ, UK
| | - A Aziz Aboobaker
- Department of Zoology, Tinbergen Building, South Parks Road, University of Oxford, Oxford OX1 3PS, UK
| |
Collapse
|
15
|
A New Chapter for Mesenchymal Stem Cells: Decellularized Extracellular Matrices. Stem Cell Rev Rep 2017; 13:587-597. [DOI: 10.1007/s12015-017-9757-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
16
|
Duhart JC, Parsons TT, Raftery LA. The repertoire of epithelial morphogenesis on display: Progressive elaboration of Drosophila egg structure. Mech Dev 2017; 148:18-39. [PMID: 28433748 DOI: 10.1016/j.mod.2017.04.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 04/07/2017] [Accepted: 04/12/2017] [Indexed: 12/26/2022]
Abstract
Epithelial structures are foundational for tissue organization in all metazoans. Sheets of epithelial cells form lateral adhesive junctions and acquire apico-basal polarity perpendicular to the surface of the sheet. Genetic analyses in the insect model, Drosophila melanogaster, have greatly advanced our understanding of how epithelial organization is established, and how it is modulated during tissue morphogenesis. Major insights into collective cell migrations have come from analyses of morphogenetic movements within the adult follicular epithelium that cooperates with female germ cells to build a mature egg. Epithelial follicle cells progress through tightly choreographed phases of proliferation, patterning, reorganization and migrations, before they differentiate to form the elaborate structures of the eggshell. Distinct structural domains are organized by differential adhesion, within which lateral junctions are remodeled to further shape the organized epithelia. During collective cell migrations, adhesive interactions mediate supracellular organization of planar polarized macromolecules, and facilitate crawling over the basement membrane or traction against adjacent cell surfaces. Comparative studies with other insects are revealing the diversification of morphogenetic movements for elaboration of epithelial structures. This review surveys the repertoire of follicle cell morphogenesis, to highlight the coordination of epithelial plasticity with progressive differentiation of a secretory epithelium. Technological advances will keep this tissue at the leading edge for interrogating the precise spatiotemporal regulation of normal epithelial reorganization events, and provide a framework for understanding pathological tissue dysplasia.
Collapse
Affiliation(s)
- Juan Carlos Duhart
- School of Life Sciences, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Las Vegas, NV 89154-4004, United States
| | - Travis T Parsons
- School of Life Sciences, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Las Vegas, NV 89154-4004, United States
| | - Laurel A Raftery
- School of Life Sciences, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Las Vegas, NV 89154-4004, United States.
| |
Collapse
|
17
|
Lo Sicco C, Reverberi D, Balbi C, Ulivi V, Principi E, Pascucci L, Becherini P, Bosco MC, Varesio L, Franzin C, Pozzobon M, Cancedda R, Tasso R. Mesenchymal Stem Cell-Derived Extracellular Vesicles as Mediators of Anti-Inflammatory Effects: Endorsement of Macrophage Polarization. Stem Cells Transl Med 2017; 6:1018-1028. [PMID: 28186708 PMCID: PMC5442783 DOI: 10.1002/sctm.16-0363] [Citation(s) in RCA: 391] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/31/2016] [Accepted: 11/29/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal Stem Cells (MSCs) are effective therapeutic agents enhancing the repair of injured tissues mostly through their paracrine activity. Increasing evidences show that besides the secretion of soluble molecules, the release of extracellular vesicles (EVs) represents an alternative mechanism adopted by MSCs. Since macrophages are essential contributors toward the resolution of inflammation, which has emerged as a finely orchestrated process, the aim of the present study was to carry out a detailed characterization of EVs released by human adipose derived-MSCs to investigate their involvement as modulators of MSC anti-inflammatory effects inducing macrophage polarization. The EV-isolation method was based on repeated ultracentrifugations of the medium conditioned by MSC exposed to normoxic or hypoxic conditions (EVNormo and EVHypo ). Both types of EVs were efficiently internalized by responding bone marrow-derived macrophages, eliciting their switch from a M1 to a M2 phenotype. In vivo, following cardiotoxin-induced skeletal muscle damage, EVNormo and EVHypo interacted with macrophages recruited during the initial inflammatory response. In injured and EV-treated muscles, a downregulation of IL6 and the early marker of innate and classical activation Nos2 were concurrent to a significant upregulation of Arg1 and Ym1, late markers of alternative activation, as well as an increased percentage of infiltrating CD206pos cells. These effects, accompanied by an accelerated expression of the myogenic markers Pax7, MyoD, and eMyhc, were even greater following EVHypo administration. Collectively, these data indicate that MSC-EVs possess effective anti-inflammatory properties, making them potential therapeutic agents more handy and safe than MSCs. Stem Cells Translational Medicine 2017 Stem Cells Translational Medicine 2017;6:1018-1028.
Collapse
Affiliation(s)
- Claudia Lo Sicco
- Department of Experimental Medicine, University of Genova, Genova, Italy
- U.O. Regenerative Medicine, IRCCS AOU San Martino-IST, National Cancer Research Institute, Genova, Italy
| | - Daniele Reverberi
- U.O. Molecular Pathology, IRCCS AOU San Martino-IST, National Cancer Research Institute, Genova, Italy
| | - Carolina Balbi
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Valentina Ulivi
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Elisa Principi
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Pamela Becherini
- Molecular Biology Laboratory, Istituto Giannina Gaslini, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Maria Carla Bosco
- Molecular Biology Laboratory, Istituto Giannina Gaslini, Genova, Italy
| | - Luigi Varesio
- Molecular Biology Laboratory, Istituto Giannina Gaslini, Genova, Italy
| | - Chiara Franzin
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Michela Pozzobon
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
- Department of Women and Children Health, University of Padova, Padova, Italy
| | - Ranieri Cancedda
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Roberta Tasso
- U.O. Regenerative Medicine, IRCCS AOU San Martino-IST, National Cancer Research Institute, Genova, Italy
| |
Collapse
|
18
|
Masuzzo P, Huyck L, Simiczyjew A, Ampe C, Martens L, Van Troys M. An end-to-end software solution for the analysis of high-throughput single-cell migration data. Sci Rep 2017; 7:42383. [PMID: 28205527 PMCID: PMC5304333 DOI: 10.1038/srep42383] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 01/10/2017] [Indexed: 12/12/2022] Open
Abstract
The systematic study of single-cell migration requires the availability of software for assisting data inspection, quality control and analysis. This is especially important for high-throughput experiments, where multiple biological conditions are tested in parallel. Although the field of cell migration can count on different computational tools for cell segmentation and tracking, downstream data visualization, parameter extraction and statistical analysis are still left to the user and are currently not possible within a single tool. This article presents a completely new module for the open-source, cross-platform CellMissy software for cell migration data management. This module is the first tool to focus specifically on single-cell migration data downstream of image processing. It allows fast comparison across all tested conditions, providing automated data visualization, assisted data filtering and quality control, extraction of various commonly used cell migration parameters, and non-parametric statistical analysis. Importantly, the module enables parameters computation both at the trajectory- and at the step-level. Moreover, this single-cell analysis module is complemented by a new data import module that accommodates multiwell plate data obtained from high-throughput experiments, and is easily extensible through a plugin architecture. In conclusion, the end-to-end software solution presented here tackles a key bioinformatics challenge in the cell migration field, assisting researchers in their high-throughput data processing.
Collapse
Affiliation(s)
- Paola Masuzzo
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium.,Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Lynn Huyck
- Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Aleksandra Simiczyjew
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Christophe Ampe
- Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Lennart Martens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium.,Department of Biochemistry, Ghent University, Ghent, Belgium
| | | |
Collapse
|
19
|
Cellular Mechanisms of Liver Regeneration and Cell-Based Therapies of Liver Diseases. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8910821. [PMID: 28210629 PMCID: PMC5292184 DOI: 10.1155/2017/8910821] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 11/29/2016] [Accepted: 12/27/2016] [Indexed: 12/14/2022]
Abstract
The emerging field of regenerative medicine offers innovative methods of cell therapy and tissue/organ engineering as a novel approach to liver disease treatment. The ultimate scientific foundation of both cell therapy of liver diseases and liver tissue and organ engineering is delivered by the in-depth studies of the cellular and molecular mechanisms of liver regeneration. The cellular mechanisms of the homeostatic and injury-induced liver regeneration are unique. Restoration of the mass of liver parenchyma is achieved by compensatory hypertrophy and hyperplasia of the differentiated parenchymal cells, hepatocytes, while expansion and differentiation of the resident stem/progenitor cells play a minor or negligible role. Participation of blood-borne cells of the bone marrow origin in liver parenchyma regeneration has been proven but does not exceed 1-2% of newly formed hepatocytes. Liver regeneration is activated spontaneously after injury and can be further stimulated by cell therapy with hepatocytes, hematopoietic stem cells, or mesenchymal stem cells. Further studies aimed at improving the outcomes of cell therapy of liver diseases are underway. In case of liver failure, transplantation of engineered liver can become the best option in the foreseeable future. Engineering of a transplantable liver or its major part is an enormous challenge, but rapid progress in induced pluripotency, tissue engineering, and bioprinting research shows that it may be doable.
Collapse
|
20
|
Gamer LW, Shi RR, Gendelman A, Mathewson D, Gamer J, Rosen V. Identification and characterization of adult mouse meniscus stem/progenitor cells. Connect Tissue Res 2016; 58:238-245. [PMID: 28005443 DOI: 10.1080/03008207.2016.1271797] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Meniscal damage is a common problem that accelerates the onset of knee osteoarthritis. Stem cell-based tissue engineering treatment approaches have shown promise in preserving meniscal tissue and restoring meniscal function. The purpose of our study was to identify meniscus-derived stem/progenitor cells (MSPCs) from mouse, a model system that allows for in vivo analysis of the mechanisms underlying meniscal injury and healing. MSPCs were isolated from murine menisci grown in explant culture and characterized for stem cell properties. Flow cytometry was used to detect the presence of surface antigens related to stem cells, and qRT-PCR was used to examine the gene expression profile of MSPCs. Major proteins associated with MSPCs were localized in the adult mouse knee using immunohistochemistry. Our data show that MSPCs have universal stem cell-like properties including clonogenicity and multi-potentiality. MSPCs expressed the mesenchymal stem cell markers CD44, Sca-1, CD90, and CD73 and when cultured had elevated levels of biglycan and collagen type I, important extracellular matrix components of adult meniscus. MSPC also expressed significant levels of Lox and Igf-1, genes associated with the embryonic meniscus. Localization studies showed staining for these same proteins in the superficial and outer zones of the adult mouse meniscus, regions thought to harbor endogenous repair cells. MSPCs represent a novel resident stem cell population in the murine meniscus. Analysis of MSPCs in mice will allow for a greater understanding of the cell biology of the meniscus, essential information for enhancing therapeutic strategies for treating knee joint injury and disease.
Collapse
Affiliation(s)
- Laura W Gamer
- a Department of Developmental Biology , Harvard School of Dental Medicine , Boston , MA , USA
| | - Rui Rui Shi
- a Department of Developmental Biology , Harvard School of Dental Medicine , Boston , MA , USA
| | - Ashira Gendelman
- a Department of Developmental Biology , Harvard School of Dental Medicine , Boston , MA , USA
| | - Dylan Mathewson
- a Department of Developmental Biology , Harvard School of Dental Medicine , Boston , MA , USA
| | - Jackson Gamer
- a Department of Developmental Biology , Harvard School of Dental Medicine , Boston , MA , USA
| | - Vicki Rosen
- a Department of Developmental Biology , Harvard School of Dental Medicine , Boston , MA , USA
| |
Collapse
|