1
|
Wang Y, Yung P, Lu G, Liu Y, Ding C, Mao C, Li ZA, Tuan RS. Musculoskeletal Organs-on-Chips: An Emerging Platform for Studying the Nanotechnology-Biology Interface. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2401334. [PMID: 38491868 PMCID: PMC11733728 DOI: 10.1002/adma.202401334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Nanotechnology-based approaches are promising for the treatment of musculoskeletal (MSK) disorders, which present significant clinical burdens and challenges, but their clinical translation requires a deep understanding of the complex interplay between nanotechnology and MSK biology. Organ-on-a-chip (OoC) systems have emerged as an innovative and versatile microphysiological platform to replicate the dynamics of tissue microenvironment for studying nanotechnology-biology interactions. This review first covers recent advances and applications of MSK OoCs and their ability to mimic the biophysical and biochemical stimuli encountered by MSK tissues. Next, by integrating nanotechnology into MSK OoCs, cellular responses and tissue behaviors may be investigated by precisely controlling and manipulating the nanoscale environment. Analysis of MSK disease mechanisms, particularly bone, joint, and muscle tissue degeneration, and drug screening and development of personalized medicine may be greatly facilitated using MSK OoCs. Finally, future challenges and directions are outlined for the field, including advanced sensing technologies, integration of immune-active components, and enhancement of biomimetic functionality. By highlighting the emerging applications of MSK OoCs, this review aims to advance the understanding of the intricate nanotechnology-MSK biology interface and its significance in MSK disease management, and the development of innovative and personalized therapeutic and interventional strategies.
Collapse
Affiliation(s)
- Yuwen Wang
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Patrick Yung
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Gang Lu
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- School of Biomedical SciencesThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Yuwei Liu
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- The First Affiliated Hospital of Shenzhen UniversityShenzhen Second People's HospitalShenzhenGuangdong518037P. R. China
| | - Changhai Ding
- Clinical Research CentreZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510260China
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Chuanbin Mao
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Zhong Alan Li
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- School of Biomedical SciencesThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Key Laboratory of Regenerative MedicineMinistry of EducationSchool of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077P. R. China
- Shenzhen Research InstituteThe Chinese University of Hong KongShenzhen518172P. R. China
| | - Rocky S. Tuan
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- School of Biomedical SciencesThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| |
Collapse
|
2
|
Lv H, Xu J, Wang Y, Liu X, Chen S, Chen J, Zhai J, Zhou Y. Isolation, identification and osteogenic capability analysis of mesenchymal stem cells derived from different layers of human maxillary sinus membrane. J Clin Periodontol 2024; 51:754-765. [PMID: 38379293 DOI: 10.1111/jcpe.13956] [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: 08/30/2023] [Revised: 01/06/2024] [Accepted: 01/19/2024] [Indexed: 02/22/2024]
Abstract
AIM To discover the populations of mesenchymal stem cells (MSCs) derived from different layers of human maxillary sinus membrane (hMSM) and evaluate their osteogenic capability. MATERIALS AND METHODS hMSM was isolated into a monolayer using the combined method of physical separation and enzymatic digestion. The localization of MSCs in hMSM was performed by immunohistological staining and other techniques. Lamina propria layer-derived MSCs (LMSCs) and periosteum layer-derived MSCs (PMSCs) from hMSM were expanded using the explant cell culture method and identified by multilineage differentiation assays, colony formation assay, flow cytometry and so on. The biological characteristics of LMSCs and PMSCs were compared using RNA sequencing, reverse transcription and quantitative polymerase chain reaction, immunofluorescence staining, transwell assay, western blotting and so forth. RESULTS LMSCs and PMSCs from hMSMs were both CD73-, CD90- and CD105-positive, and CD34-, CD45- and HLA-DR-negative. LMSCs and PMSCs were identified as CD171+/CD90+ and CD171-/CD90+, respectively. LMSCs displayed stronger proliferation capability than PMSCs, and PMSCs presented stronger osteogenic differentiation capability than LMSCs. Moreover, PMSCs could recruit and promote osteogenic differentiation of LMSCs. CONCLUSIONS This study identified and isolated two different types of MSCs from hMSMs. Both MSCs served as good potential candidates for bone regeneration.
Collapse
Affiliation(s)
- Huixin Lv
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Jing Xu
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Yihan Wang
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Xiuyu Liu
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Sheng Chen
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Jingxia Chen
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Jingjie Zhai
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Yanmin Zhou
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
3
|
Chen Y, Zhang L, Shi X, Han J, Chen J, Zhang X, Xie D, Li Z, Niu X, Chen L, Yang C, Sun X, Zhou T, Su P, Li N, Greenblatt MB, Ke R, Huang J, Chen Z, Xu R. Characterization of the Nucleus Pulposus Progenitor Cells via Spatial Transcriptomics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303752. [PMID: 38311573 PMCID: PMC11095158 DOI: 10.1002/advs.202303752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 01/11/2024] [Indexed: 02/06/2024]
Abstract
Loss of refreshment in nucleus pulposus (NP) cellularity leads to intervertebral disc (IVD) degeneration. Nevertheless, the cellular sequence of NP cell differentiation remains unclear, although an increasing body of literature has identified markers of NP progenitor cells (NPPCs). Notably, due to their fragility, the physical enrichment of NP-derived cells has limited conventional transcriptomic approaches in multiple studies. To overcome this limitation, a spatially resolved transcriptional atlas of the mouse IVD is generated via the 10x Genomics Visium platform dividing NP spots into two clusters. Based on this, most reported NPPC-markers, including Cathepsin K (Ctsk), are rare and predominantly located within the NP-outer subset. Cell lineage tracing further evidence that a small number of Ctsk-expressing cells generate the entire adult NP tissue. In contrast, Tie2, which has long suggested labeling NPPCs, is actually neither expressed in NP subsets nor labels NPPCs and their descendants in mouse models; consistent with this, an in situ sequencing (ISS) analysis validated the absence of Tie2 in NP tissue. Similarly, no Tie2-cre-mediated labeling of NPPCs is observed in an IVD degenerative mouse model. Altogether, in this study, the first spatial transcriptomic map of the IVD is established, thereby providing a public resource for bone biology.
Collapse
Affiliation(s)
- Yu Chen
- The First Affiliated Hospital of Xiamen University‐ICMRS Collaborating Center for Skeletal Stem CellsState Key Laboratory of Cellular Stress BiologyFaculty of Medicine and Life SciencesSchool of MedicineXiamen UniversityXiamen361102China
- Xiamen Key Laboratory of Regeneration MedicineFujian Provincial Key Laboratory of Organ and Tissue RegenerationSchool of MedicineXiamen UniversityXiamen361102China
| | - Long Zhang
- The First Affiliated Hospital of Xiamen University‐ICMRS Collaborating Center for Skeletal Stem CellsState Key Laboratory of Cellular Stress BiologyFaculty of Medicine and Life SciencesSchool of MedicineXiamen UniversityXiamen361102China
- Xiamen Key Laboratory of Regeneration MedicineFujian Provincial Key Laboratory of Organ and Tissue RegenerationSchool of MedicineXiamen UniversityXiamen361102China
| | - Xueqing Shi
- The First Affiliated Hospital of Xiamen University‐ICMRS Collaborating Center for Skeletal Stem CellsState Key Laboratory of Cellular Stress BiologyFaculty of Medicine and Life SciencesSchool of MedicineXiamen UniversityXiamen361102China
- Xiamen Key Laboratory of Regeneration MedicineFujian Provincial Key Laboratory of Organ and Tissue RegenerationSchool of MedicineXiamen UniversityXiamen361102China
| | - Jie Han
- The First Affiliated Hospital of Xiamen University‐ICMRS Collaborating Center for Skeletal Stem CellsState Key Laboratory of Cellular Stress BiologyFaculty of Medicine and Life SciencesSchool of MedicineXiamen UniversityXiamen361102China
- Xiamen Key Laboratory of Regeneration MedicineFujian Provincial Key Laboratory of Organ and Tissue RegenerationSchool of MedicineXiamen UniversityXiamen361102China
| | - Jingyu Chen
- Gene Denovo Biotechnology CoGuangzhou510006China
| | - Xinya Zhang
- School of Medicine and School of Biomedical SciencesHuaqiao UniversityQuanzhou362000China
| | - Danlin Xie
- School of Medicine and School of Biomedical SciencesHuaqiao UniversityQuanzhou362000China
- School of Life SciencesWestlake UniversityHangzhou310030China
| | - Zan Li
- The First Affiliated Hospital of Xiamen University‐ICMRS Collaborating Center for Skeletal Stem CellsState Key Laboratory of Cellular Stress BiologyFaculty of Medicine and Life SciencesSchool of MedicineXiamen UniversityXiamen361102China
- Xiamen Key Laboratory of Regeneration MedicineFujian Provincial Key Laboratory of Organ and Tissue RegenerationSchool of MedicineXiamen UniversityXiamen361102China
| | - Xing Niu
- China Medical UniversityShenyangLiaoning110122China
| | - Lijie Chen
- China Medical UniversityShenyangLiaoning110122China
| | - Chaoyong Yang
- Department of Chemical BiologyCollege of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Xiujie Sun
- Department of Obstetrics and GynecologySchool of MedicineXiang'an Hospital of Xiamen UniversityXiamen UniversityXiamen361102China
| | - Taifeng Zhou
- Department of Spine SurgeryGuangdong Provincial Key Laboratory of Orthopedics and TraumatologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080China
| | - Peiqiang Su
- Department of Spine SurgeryGuangdong Provincial Key Laboratory of Orthopedics and TraumatologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080China
| | - Na Li
- The First Affiliated Hospital of Xiamen University‐ICMRS Collaborating Center for Skeletal Stem CellsState Key Laboratory of Cellular Stress BiologyFaculty of Medicine and Life SciencesSchool of MedicineXiamen UniversityXiamen361102China
- Xiamen Key Laboratory of Regeneration MedicineFujian Provincial Key Laboratory of Organ and Tissue RegenerationSchool of MedicineXiamen UniversityXiamen361102China
| | - Matthew B. Greenblatt
- Department of Pathology and Laboratory MedicineWeill Cornell Medical CollegeNew YorkNY10065USA
- Research DivisionHospital for Special SurgeryNew YorkNY10065USA
| | - Rongqin Ke
- School of Medicine and School of Biomedical SciencesHuaqiao UniversityQuanzhou362000China
| | - Jianming Huang
- Department of OrthopedicsChengong Hospital (the 73th Group Military Hospital of People's Liberation Army) affiliated to Xiamen UniversityXiamen361000China
| | - Zhe‐Sheng Chen
- College of Pharmacy and Health SciencesSt. John's UniversityNew YorkNY11439USA
| | - Ren Xu
- The First Affiliated Hospital of Xiamen University‐ICMRS Collaborating Center for Skeletal Stem CellsState Key Laboratory of Cellular Stress BiologyFaculty of Medicine and Life SciencesSchool of MedicineXiamen UniversityXiamen361102China
- Xiamen Key Laboratory of Regeneration MedicineFujian Provincial Key Laboratory of Organ and Tissue RegenerationSchool of MedicineXiamen UniversityXiamen361102China
| |
Collapse
|
4
|
Zineldeen DH, Mushtaq M, Haider KH. Cellular preconditioning and mesenchymal stem cell ferroptosis. World J Stem Cells 2024; 16:64-69. [PMID: 38455100 PMCID: PMC10915960 DOI: 10.4252/wjsc.v16.i2.64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 02/26/2024] Open
Abstract
In this editorial, we comment on the article published in the recent issue of the World Journal of Stem Cells. They focus on stem cell preconditioning to prevent ferroptosis by modulating the cystathionine γ-lyase/hydrogen sulfide (H2S) pathway as a novel approach to treat vascular disorders, particularly pulmonary hypertension. Preconditioned stem cells are gaining popularity in regenerative medicine due to their unique ability to survive by resisting the harsh, unfavorable microenvironment of the injured tissue. They also secrete various paracrine factors against apoptosis, necrosis, and ferroptosis to enhance cell survival. Ferroptosis, a regulated form of cell death characterized by iron accumulation and oxidative stress, has been implicated in various pathologies encompassing degenerative disorders to cancer. The lipid peroxidation cascade initiates and sustains ferroptosis, generating many reactive oxygen species that attack and damage multiple cellular structures. Understanding these intertwined mechanisms provides significant insights into developing therapeutic modalities for ferroptosis-related diseases. This editorial primarily discusses stem cell preconditioning in modulating ferroptosis, focusing on the cystathionase gamma/H2S ferroptosis pathway. Ferroptosis presents a significant challenge in mesenchymal stem cell (MSC)-based therapies; hence, the emerging role of H2S/cystathionase gamma/H2S signaling in abrogating ferroptosis provides a novel option for therapeutic intervention. Further research into understanding the precise mechanisms of H2S-mediated cytoprotection against ferroptosis is warranted to enhance the therapeutic potential of MSCs in clinical settings, particularly vascular disorders.
Collapse
Affiliation(s)
- Doaa Hussein Zineldeen
- Basic Sciences, Sulaiman AlRajhi University, Albukairiyah 52736, AlQaseem, Saudi Arabia
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Tanta University, Tanta 6632110, Egypt
| | - Mazhar Mushtaq
- Basic Sciences, Sulaiman AlRajhi University, Albukairiyah 52736, AlQaseem, Saudi Arabia
| | - Khawaja Husnain Haider
- Basic Sciences, Sulaiman AlRajhi University, Albukairiyah 52736, AlQaseem, Saudi Arabia.
| |
Collapse
|
5
|
Zaib S, Areeba, Khan I. Purinergic Signaling and its Role in the Stem Cell Differentiation. Mini Rev Med Chem 2024; 24:863-883. [PMID: 37828668 DOI: 10.2174/0113895575261206231003151416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 07/30/2023] [Accepted: 08/30/2023] [Indexed: 10/14/2023]
Abstract
Purinergic signaling is a mechanism in which extracellular purines and pyrimidines interact with specialized cell surface receptors known as purinergic receptors. These receptors are divided into two families of P1 and P2 receptors, each responding to different nucleosides and nucleotides. P1 receptors are activated by adenosine, while P2 receptors are activated by pyrimidine and purines. P2X receptors are ligand-gated ion channels, including seven subunits (P2X1-7). However, P2Y receptors are the G-protein coupled receptors comprising eight subtypes (P2Y1/2/4/6/11/12/13/14). The disorder in purinergic signaling leads to various health-related issues and diseases. In various aspects, it influences the activity of non-neuronal cells and neurons. The molecular mechanism of purinergic signaling provides insight into treating various human diseases. On the contrary, stem cells have been investigated for therapeutic applications. Purinergic signaling has shown promising effect in stem cell engraftment. The immune system promotes the autocrine and paracrine mechanisms and releases the significant factors essential for successful stem cell therapy. Each subtype of purinergic receptor exerts a beneficial effect on the damaged tissue. The most common effect caused by purinergic signaling is the proliferation and differentiation that treat different health-related conditions.
Collapse
Affiliation(s)
- Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Areeba
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Imtiaz Khan
- Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, United Kingdom
| |
Collapse
|
6
|
Chepeleva EV. Cell Therapy in the Treatment of Coronary Heart Disease. Int J Mol Sci 2023; 24:16844. [PMID: 38069167 PMCID: PMC10706847 DOI: 10.3390/ijms242316844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Heart failure is a leading cause of death in patients who have suffered a myocardial infarction. Despite the timely use of modern reperfusion therapies such as thrombolysis, surgical revascularization and balloon angioplasty, they are sometimes unable to prevent the development of significant areas of myocardial damage and subsequent heart failure. Research efforts have focused on developing strategies to improve the functional status of myocardial injury areas. Consequently, the restoration of cardiac function using cell therapy is an exciting prospect. This review describes the characteristics of various cell types relevant to cellular cardiomyoplasty and presents findings from experimental and clinical studies investigating cell therapy for coronary heart disease. Cell delivery methods, optimal dosage and potential treatment mechanisms are discussed.
Collapse
Affiliation(s)
- Elena V. Chepeleva
- Federal State Budgetary Institution National Medical Research Center Named after Academician E.N. Meshalkin of the Ministry of Health of the Russian Federation, 15, Rechkunovskaya Str., 630055 Novosibirsk, Russia;
- Research Institute of Clinical and Experimental Lymphology—Branch of the Institute of Cytology and Genetics Siberian Branch of Russian Academy of Sciences, 2, Timakova Str., 630060 Novosibirsk, Russia
| |
Collapse
|
7
|
Abubakr S, Hazem NM, Sherif RN, Elhawary AA, Botros KG. Correlation between SDF-1α, CD34 positive hematopoietic stem cells and CXCR4 expression with liver fibrosis in CCl4 rat model. BMC Gastroenterol 2023; 23:323. [PMID: 37730560 PMCID: PMC10512633 DOI: 10.1186/s12876-023-02932-y] [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: 04/22/2023] [Accepted: 08/25/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND One of the most frequent disorders is liver fibrosis. An improved understanding of the different events during the process of liver fibrosis & its reversibility could be helpful in its staging and in finding potential therapeutic agents. AIM The goal of this research was to evaluate the relationship among CD34 + HPSCs, SDF-1α, and CXCR4 receptor expression with the percentage of the area of hepatic fibrosis. MATERIALS AND METHODS Thirty-six male Sprague-Dawley rats were separated into the control group, liver injury group & spontaneous reversion group. The liver injury was induced by using 2 ml/kg CCl4 twice a week. Flow cytometric examination of CD34 + cells in the blood & liver was performed. Bone marrow & liver samples were taken for evaluation of the SDF-1α mRNA by PCR. Liver specimens were stained for histopathological and CXCR4 immuno-expression evaluation. RESULTS In the liver injury group, the hepatic enzymes, fibrosis area percentage, CXCR4 receptor expression in the liver, CD34 + cells in the blood and bone marrow & the level SDF-1α in the liver and its concentration gradient were statistically significantly elevated with the progression of the liver fibrosis. On the contrary, SDF-1α in the bone marrow was statistically significantly reduced with the development of liver fibrosis. During the spontaneous reversion group, all the studied parameters apart from SDF-1α in the bone marrow were statistically substantially decreased compared with the liver injury group. We found a statistically substantial positive correlation between fibrosis area and all of the following: liver enzymes, CXCR4 receptor expression in the liver, CD34 + cells in the blood and liver, and SDF- 1α in the liver and its concentration gradient. In conclusion, in CCl4 rat model, the fibrosis area is significantly correlated with many parameters in the blood, bone marrow, and liver, which can be used during the process of follow-up during the therapeutic interventions.
Collapse
Affiliation(s)
- Sara Abubakr
- Human Anatomy & Embryology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Noha M Hazem
- Medical Biochemistry and Molecular Biology Department, Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Algomhoria Street, Mansoura, 35516, Egypt.
- Pathological Sciences Department, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia.
| | - R N Sherif
- Human Anatomy & Embryology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Adel Abdelmohdy Elhawary
- Human Anatomy & Embryology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Kamal G Botros
- Human Anatomy & Embryology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
8
|
Bujko K, Ciechanowicz AK, Kucia M, Ratajczak MZ. Molecular analysis and comparison of CD34 + and CD133 + very small embryonic-like stem cells purified from umbilical cord blood. Cytometry A 2023; 103:703-711. [PMID: 37246957 DOI: 10.1002/cyto.a.24767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 05/30/2023]
Abstract
Very small embryonic like stem cells (VSELs) are a dormant population of stem cells that, as proposed, are deposited during embryogenesis in various tissues, including bone marrow (BM). These cells are released under steady state conditions from their tissue locations and circulate at a low level in peripheral blood (PB). Their number increases in response to stressors as well as tissue/organ damage. This increase is evident during neonatal delivery, as delivery stress prompts enrichment of umbilical cord blood (UCB) with VSELs. These cells could be purified from BM, PB, and UCB by multiparameter sorting as a population of very small CXCR4+ Lin- CD45- cells that express the CD34 or CD133 antigen. In this report, we evaluated a number of CD34+ Lin- CD45- and CD133+ Lin- CD45- UCB-derived VSELs. We also performed initial molecular characterization of both cell populations for expression of selected pluripotency markers and compared these cells at the proteomic level. We noticed that CD133+ Lin- CD45- population is more rare and express, at a higher level, mRNA for pluripotency markers Oct-4 and Nanog as well as the stromal-derived factor-1 (SDF-1) CXCR4 receptor that regulates trafficking of these cells, however both cells population did not significantly differ in the expression of proteins assigned to main biological processes.
Collapse
Affiliation(s)
- Kamila Bujko
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | | | - Magdalena Kucia
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Mariusz Z Ratajczak
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
9
|
Rashid K, Ahmad A, Meerasa SS, Khan AQ, Wu X, Liang L, Cui Y, Liu T. Cancer stem cell-derived exosome-induced metastatic cancer: An orchestra within the tumor microenvironment. Biochimie 2023; 212:1-11. [PMID: 37011805 DOI: 10.1016/j.biochi.2023.03.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 02/20/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Although the mechanisms as well as pathways associated with cancer stem cell (CSC) maintenance, expansion, and tumorigenicity have been extensively studied and the role of tumor cell (TC)-derived exosomes in this process is well understood, there is a paucity of research focusing specifically on the functional mechanisms of CSC-derived exosomes (CSC-Exo)/-exosomal-ncRNAs and their impact on malignancy. This shortcoming needs to be addressed, given that these vesicular and molecular components of CSCs could have a great impact on the cancer initiation, progression, and recurrence through their interaction with other key tumor microenvironment (TME) components, such as MSCs/MSC-Exo and CAFs/CAF-Exo. In particular, understanding CSCs/CSC-Exo and its crosstalk with MSCs/MSC-Exo or CAFs/CAF-Exo that are associated with the proliferation, migration, differentiation, angiogenesis, and metastasis through an enhanced process of self-renewal, chemotherapy as well as radiotherapy resistance may aid cancer treatment. This review contributes to this endeavor by summarizing the characteristic features and functional mechanisms of CSC-Exo/MSC-Exo/CAF-Exo and their mutual impact on cancer progression and therapy resistance.
Collapse
Affiliation(s)
- Khalid Rashid
- Department of Cancer Biology, Faculty of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| | - Aqeel Ahmad
- Department of Medical Biochemistry, College of Medicine, Shaqra University, Shaqra, Saudi Arabia.
| | - Semmal Syed Meerasa
- Department of Physiology, College of Medicine, Shaqra University, Shaqra, Saudi Arabia
| | - Abdul Q Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Xiaobo Wu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Li Liang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuehong Cui
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Expression of CD117 (c-Kit) on Circulating B Cells in Pediatric Schistosomiasis. Infect Immun 2022; 90:e0016022. [PMID: 35862720 PMCID: PMC9387214 DOI: 10.1128/iai.00160-22] [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: 01/09/2023] Open
Abstract
Few B cells express CD27, the primary marker for memory B cells, in pediatric schistosomiasis, suggesting B cell malfunction. This study further demonstrates unexpected high expression of CD117 on circulating B cells in children highly exposed to Schistosoma mansoni infectious larvae. CD117 is expressed by immature or lymphoma B cells, but not by mature, circulating cells. We therefore sought to define the significance of CD117 on blood B cells. We found that CD117-positive (CD117+) B cells increased with the intensity of schistosome infection. In addition, CD117 expression was reduced on CD23+ B cells previously shown to correlate with resistance to infection. Stimulation with a panel of cytokines demonstrated that CD117 levels were upregulated in response to a combination of interleukin 4 (IL-4) and stem cell factor (SCF), the ligand for CD117, whereas IL-2 led to a reduction. In addition, stimulation with SCF generally reduced B cell activation levels. Upon further investigation, it was established that multiple circulating cells expressed increased levels of CD117, including monocytes, neutrophils, and eosinophils, and expression levels correlated with that of B cells. Finally, we identified a population of large circulating cells with features of reticulocytes. Overall, our results suggest that hyperexposure to intravascular parasitic worms elicits immature cells from the bone marrow. Levels of SCF were shown to reduce as children began to transition through puberty. The study results pose an explanation for the inability of children to develop significant immunity to infection until after puberty.
Collapse
|
11
|
Ischemic Brain Stroke and Mesenchymal Stem Cells: An Overview of Molecular Mechanisms and Therapeutic Potential. Stem Cells Int 2022; 2022:5930244. [PMID: 35663353 PMCID: PMC9159823 DOI: 10.1155/2022/5930244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 10/12/2021] [Accepted: 05/04/2022] [Indexed: 12/15/2022] Open
Abstract
Ischemic brain injury is associated with a high rate of mortality and disability with no effective therapeutic strategy. Recently, a growing number of studies are focusing on mesenchymal stem cell-based therapies for neurodegenerative disorders. However, despite having the promising outcome of preclinical studies, the clinical application of stem cell therapy remained elusive due to little or no progress in clinical trials. The objective of this study was to provide a generalized critique for the role of mesenchymal stem cell therapy in ischemic stroke injury, its underlying mechanisms, and constraints on its preclinical and clinical applications. Thus, we attempted to present an overview of previously published reports to evaluate the progress and provide molecular basis of mesenchymal stem cells (MSCs) therapy and its application in preclinical and clinical settings, which could aid in designing an effective regenerative therapeutic strategy in the future.
Collapse
|
12
|
Enrichment of SOX2-Positive Cells in BRAF V600E Mutated and Recurrent Ameloblastoma. J Pers Med 2022; 12:jpm12010077. [PMID: 35055392 PMCID: PMC8780877 DOI: 10.3390/jpm12010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/01/2022] [Accepted: 01/05/2022] [Indexed: 11/17/2022] Open
Abstract
Ameloblastoma is the most common benign odontogenic neoplasm, but with an aggressive behavior and a high recurrence rate. Nowadays wide surgical resection is the current recommended treatment, which can cause further loss of function and esthetics. Recent studies point to the stem/progenitor cells as both initiators and propagators of the tumors. Elucidation of the cellular and molecular mechanisms underlying the tumor stem cells is of broad interest for understanding tumorigenesis and for developing effective targeted therapies. SRY related HMG box gene 2 (SOX2) is a transcription factor that plays important roles in development, stem cell renewal, and cancer formation. Few studies have revealed increased SOX2 expression in atypical ameloblastoma and ameloblastic carcinoma. For the development of personalized medicine for ameloblastoma, biomarkers that provide prognostic or predictive information regarding a tumor’s nature or its response to treatment are essential. Thus, in this study, we aimed to study if SOX2-positive cells exist in ameloblastomas and their correlation with the clinicopathologic parameters. Our data suggested BRAF(V600E) mutation might contribute to the expansion of SOX2-positive cells. The identification of BRAF(V600E) mutation and the amplification of SOX2-positive cells in ameloblastomas imply the possible benefit of applying BRAF and SOX2 inhibitors in recurrent and un-resectable ameloblastomas.
Collapse
|
13
|
Ali S, Haque N, Azhar Z, Saeinasab M, Sefat F. Regenerative Medicine of Liver: Promises, Advances and Challenges. Biomimetics (Basel) 2021; 6:biomimetics6040062. [PMID: 34698078 PMCID: PMC8544204 DOI: 10.3390/biomimetics6040062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/06/2021] [Accepted: 10/14/2021] [Indexed: 12/16/2022] Open
Abstract
Liver tissue engineering is a rapidly developing field which combines the novel use of liver cells, appropriate biochemical factors, and engineering principles, in order to replace or regenerate damaged liver tissue or the organ. The aim of this review paper is to critically investigate different possible methods to tackle issues related with liver diseases/disorders mainly using regenerative medicine. In this work the various regenerative treatment options are discussed, for improving the prognosis of chronic liver disorders. By reviewing existing literature, it is apparent that the current popular treatment option is liver transplantation, although the breakthroughs of stem cell-based therapy and bioartificial liver technology make them a promising alternative.
Collapse
Affiliation(s)
- Saiful Ali
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD7 1DP, UK; (S.A.); (N.H.); (Z.A.)
| | - Nasira Haque
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD7 1DP, UK; (S.A.); (N.H.); (Z.A.)
| | - Zohya Azhar
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD7 1DP, UK; (S.A.); (N.H.); (Z.A.)
| | - Morvarid Saeinasab
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran;
| | - Farshid Sefat
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD7 1DP, UK; (S.A.); (N.H.); (Z.A.)
- Interdisciplinary Research Centre in Polymer Science & Technology (Polymer IRC), University of Bradford, Bradford BD7 1DP, UK
- Correspondence: ; Tel.: +44-(0)-1274-233679 or +44-(0)-781-381-7460
| |
Collapse
|
14
|
Rijsbergen LC, van Dijk LLA, Engel MFM, de Vries RD, de Swart RL. In Vitro Modelling of Respiratory Virus Infections in Human Airway Epithelial Cells - A Systematic Review. Front Immunol 2021; 12:683002. [PMID: 34489934 PMCID: PMC8418200 DOI: 10.3389/fimmu.2021.683002] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
Respiratory tract infections (RTI) are a major cause of morbidity and mortality in humans. A large number of RTIs is caused by viruses, often resulting in more severe disease in infants, elderly and the immunocompromised. Upon viral infection, most individuals experience common cold-like symptoms associated with an upper RTI. However, in some cases a severe and sometimes life-threatening lower RTI may develop. Reproducible and scalable in vitro culture models that accurately reflect the human respiratory tract are needed to study interactions between respiratory viruses and the host, and to test novel therapeutic interventions. Multiple in vitro respiratory cell culture systems have been described, but the majority of these are based on immortalized cell lines. Although useful for studying certain aspects of viral infections, such monomorphic, unicellular systems fall short in creating an understanding of the processes that occur at an integrated tissue level. Novel in vitro models involving primary human airway epithelial cells and, more recently, human airway organoids, are now in use. In this review, we describe the evolution of in vitro cell culture systems and their characteristics in the context of viral RTIs, starting from advances after immortalized cell cultures to more recently developed organoid systems. Furthermore, we describe how these models are used in studying virus-host interactions, e.g. tropism and receptor studies as well as interactions with the innate immune system. Finally, we provide an outlook for future developments in this field, including co-factors that mimic the microenvironment in the respiratory tract.
Collapse
Affiliation(s)
- Laurine C. Rijsbergen
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Laura L. A. van Dijk
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Maarten F. M. Engel
- Medical Library, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Rory D. de Vries
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Rik L. de Swart
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
15
|
Kim IG, Cho H, Shin J, Cho JH, Cho SW, Chung EJ. Regeneration of irradiation-damaged esophagus by local delivery of mesenchymal stem-cell spheroids encapsulated in a hyaluronic-acid-based hydrogel. Biomater Sci 2021; 9:2197-2208. [PMID: 33506817 DOI: 10.1039/d0bm01655a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Radiation therapy (RT) is a typical treatment for head and neck cancers. Generally, prolonged irradiation of the esophagus causes esophageal fibrosis due to increased reactive oxygen species and proinflammatory cytokines. This study was designed to determine whether catechol-functionalized hyaluronic acid (HA-CA) hydrogel-encapsulated human mesenchymal stem-cell spheroids (MSC-SPs) could ameliorate damage to the esophagus in a mouse model of radiation-induced esophageal fibrosis. MSC-SPs were cultured in concave microwells 600 μm in diameter at a cell density of 1 × 106 cells per mL. Most cells formed spheroids with a 100-300 μm size distribution in concave microwells. MSC-SPs were well maintained in the HA gel, and live-dead staining confirmed that most cells survived. The HA gel containing the MSC-SPs was then injected into the damaged esophageal layer. Inflammatory signs or adverse tissue reactions were not observed after esophageal injection of HA-gel-encapsulated MSC-SPs. Based on Masson's trichrome staining at 4 and 12 weeks postinjection, the inner esophageal layer (IEL) was significantly thinner in the MSC-SP + HA gel group compared to those in the other experimental groups. While the saline and HA gel treatments made the esophageal muscles loose and thick, the MSC-SP + HA gel group showed bundles of tightly packed esophageal muscles, as assayed by desmin immunostaining. qPCR analysis showed that epithelial genes tended to increase over time in the MSC-SP + HA gel group, and the expression of most fibrosis-related genes decreased. This study proposes the potential of using HA-CA-hydrogel-encapsulated MSC-SPs as a promising therapy against radiation-induced esophageal fibrosis.
Collapse
Affiliation(s)
- In Gul Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
16
|
Avian Satellite Cell Plasticity. Animals (Basel) 2020; 10:ani10081322. [PMID: 32751789 PMCID: PMC7459542 DOI: 10.3390/ani10081322] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary Adult muscle regeneration and reconstruction is dependent on a population of adult stem cells, known as satellite cells. These cells were suggested to exhibit a certain degree of plasticity, being able to differentiate into lineages unassociated with muscle cells. In this study, we have used a range of visualization methods, as well as PCR, to identify a population of satellite cells obtained from samples of chicken muscles. Then, the cells, expressing a previously introduced detectable transgene, were introduced into chicken embryos and detected after three and eighteen days of their development. The traces of cell populations derived from the introduced satellite cells were detected in a range of embryonic tissues in both of the studied timeframes. The results of this study give further proof of the plasticity of muscle satellite cells, showing the potential locations of their migration during embryonic development. Abstract Adult myogenesis is dependent on a population of precursor cells, located between the sarcolemma and the basal lamina of the muscle fiber. These satellite cells, usually present in a quiescent state, become activated in response to mechanical muscle strain, differentiating and fusing to add new nuclei to enlarging muscles. As their myogenic lineage commitment is induced on demand, muscle satellite cells exhibit a certain amount of plasticity, possibly being able to be directed to differentiate into non-myogenic fates. In this study, myosatellite cells were isolated from chicken muscle samples, characterized in vitro and introduced into developing blastoderms. They were further investigated using fluorescence microscopy, immunohistochemistry and PCR, to determine their location in embryos after three and eighteen days. The results of the in vitro analysis confirmed that the cells obtained from the Pectoralis thoracicus are highly myogenic, based on the expression of Pax7, Myogenin, MyoD, Desmin and the myotube assay. Furthermore, the investigation of satellite cells within the embryo showed their migration to the regions of Pectoralis thoracicus, heart, liver, gizzard, proventriculus, intestine and brain. Overall, the results of the study proved the high myogenicity of chicken Pectoralis thoracicus cell isolates, as well as provided new information about their migration pathways following introduction into the blastocyst. The presence of the introduced LacZ or eGFP transgenes across the embryo, even 20 days after myosatellite cell injection, further supports the notion that satellite cells exhibit significant plasticity, potentially transdifferentiating into non-muscle lineages.
Collapse
|
17
|
Shahid A, Bharadwaj M. The connection between the Th17 cell related cytokines and cancer stem cells in cancer: Novel therapeutic targets. Immunol Lett 2019; 213:9-20. [PMID: 31278971 DOI: 10.1016/j.imlet.2019.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/25/2019] [Accepted: 07/02/2019] [Indexed: 02/08/2023]
Abstract
Cancer Stem Cells (CSCs) are the subpopulation of cells present in the different types of cancers with capabilities of self-renewal, differentiation, and tumorigenicity when transplanted into an animal host. The research work on the CSC has been providing a promising approach for the improvement of cancer therapies in the future. The CSCs have a close connection with the cytokines related with the T helper 17 (Th17) cell and other factors present in the tumor microenvironment, and these play a pivotal role in tumor progression and metastasis. The properties of CSCs are well defined in various type of tumor which is mainly developed by chemically and spontaneously in murine cancer model but in human defined primarily on acute myeloid leukemia, glioma, and breast cancer. The role of Th1, Th2, Natural Killer cells are well described in the cancer biology, but the Th17 cells are the subset which is recently exploited, and lots of research are going on. In this Review, we summarize current findings of the characteristics and functions of the Th17 cell and its signature cytokines in different cancers and their interconnections with cancer stem cells and with their markers. We have also discussed the functional properties of CSCs and how the CSCs markers can be distinguished from normal stem cells markers. We have also talked about the strategies that are efficiently targeting of CSCs and Th17 cells in different cancers.
Collapse
Affiliation(s)
- Ayaz Shahid
- Molecular Biology Group, National Institute of Cancer Prevention and Research, Indian Council of Medical Research (ICMR), Department of Health Research, Noida, 201301, India
| | - Mausumi Bharadwaj
- Molecular Biology Group, National Institute of Cancer Prevention and Research, Indian Council of Medical Research (ICMR), Department of Health Research, Noida, 201301, India.
| |
Collapse
|
18
|
Park J, Lee H, Lee HJ, Kim GC, Kim SS, Han S, Song K. Non-thermal atmospheric pressure plasma is an excellent tool to activate proliferation in various mesoderm-derived human adult stem cells. Free Radic Biol Med 2019; 134:374-384. [PMID: 30685405 DOI: 10.1016/j.freeradbiomed.2019.01.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 02/08/2023]
Abstract
Adult stem cells are capable of self-renewal and differentiation into specific cell types in tissues and have high potential for stem cell therapy. Mesenchymal and hematopoietic stem cells are easily attainable from the human body and have become applicable tools for adult stem cell therapy. However, there are still technical barriers for the application of mesenchymal and hematopoietic stem cells for therapy, such as the small number of cell populations, high risk of contamination, and loss of their stemness properties in vitro. In our previous study, we showed that non-thermal atmospheric pressure plasma (NTAPP) promoted the proliferation of adipose tissue-derived stem cells (ASCs) by 1.6-fold on average, while maintaining their stemness. Here, we examined the feasibility of NTAPP as a tool to activate the proliferation of mesenchymal and hematopoietic stem cells in vitro without affecting their stem cell characteristics. NTAPP increased the proliferation of bone marrow-derived stem cells (BM-MSCs) and hematopoietic stem cells (HSCs) by 1.8- and 2-fold, respectively, when compared to that of untreated cells. As observed in ASCs, NTAPP exposure also activated the expression of stem cell-specific surface markers, CD44 and CD105, by 5-fold in BM-MSCs, when compared to that in unexposed control cells in a low glucose medium with a low concentration of basic fibroblast growth factor (b-FGF). In addition, NTAPP exposure highly augmented the mRNA expression of well-known pluripotent genes for stemness, such as Oct4, Sox2, and Nanog in ASCs and BM-MSCs when compared to that in unexposed control cells. When cell cycle progression was examined, the G1-S shift was accelerated, and expression of PCNA was increased in NTAPP-exposed ASCs when compared to that in untreated control cells, suggesting that NTAPP activated G1-S transition. Taken together, these results demonstrated that NTAPP activated the proliferation of various mesodermal-derived human adult stem cells by accelerating the G1-S transition while maintaining their pluripotency and stemness, strongly suggesting that NTAPP can be an efficient tool for expanding the population of various adult stem cells in vitro for medical applications.
Collapse
Affiliation(s)
- Jeongyeon Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyunyoung Lee
- Department of Electrical Engineering, Pusan National University, Pusan, 46241, Republic of Korea
| | - Hae June Lee
- Department of Electrical Engineering, Pusan National University, Pusan, 46241, Republic of Korea
| | - Gyoo Cheon Kim
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Sung-Soo Kim
- Department of Anatomy, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Sungbum Han
- Batang Plastic Surgery Center, Seoul, 06120, Republic of Korea
| | - Kiwon Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
19
|
Harrington L, Pucci F. In medio stat virtus: unanticipated consequences of telomere dysequilibrium. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2016.0444. [PMID: 29335368 PMCID: PMC5784064 DOI: 10.1098/rstb.2016.0444] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2017] [Indexed: 12/13/2022] Open
Abstract
The integrity of chromosome ends, or telomeres, depends on myriad processes that must balance the need to compact and protect the telomeric, G-rich DNA from detection as a double-stranded DNA break, and yet still permit access to enzymes that process, replicate and maintain a sufficient reserve of telomeric DNA. When unable to maintain this equilibrium, erosion of telomeres leads to perturbations at or near the telomeres themselves, including loss of binding by the telomere protective complex, shelterin, and alterations in transcription and post-translational modifications of histones. Although the catastrophic consequences of full telomere de-protection are well described, recent evidence points to other, less obvious perturbations that arise when telomere length equilibrium is altered. For example, critically short telomeres also perturb DNA methylation and histone post-translational modifications at distal sites throughout the genome. In murine stem cells for example, this dysregulated chromatin leads to inappropriate suppression of pluripotency regulator factors such as Nanog. This review summarizes these recent findings, with an emphasis on how these genome-wide, telomere-induced perturbations can have profound consequences on cell function and fate. This article is part of the theme issue ‘Understanding diversity in telomere dynamics’.
Collapse
Affiliation(s)
- Lea Harrington
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, College of Science and Engineering, University of Edinburgh, Mayfield Road, Edinburgh EH9 3JR, UK
| | - Fabio Pucci
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, College of Science and Engineering, University of Edinburgh, Mayfield Road, Edinburgh EH9 3JR, UK
| |
Collapse
|
20
|
Takeda H, Dondzillo A, Randall JA, Gubbels SP. Challenges in Cell-Based Therapies for the Treatment of Hearing Loss. Trends Neurosci 2018; 41:823-837. [PMID: 30033182 DOI: 10.1016/j.tins.2018.06.008] [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] [Received: 01/08/2018] [Revised: 06/20/2018] [Accepted: 06/21/2018] [Indexed: 12/17/2022]
Abstract
Hearing loss in mammals is an irreversible process caused by degeneration of the hair cells of the inner ear. Current therapies for hearing loss include hearing aids and cochlear implants that provide substantial benefits to most patients, but also have several shortcomings. There is great interest in the development of regenerative therapies to treat deafness in the future. Cell-based therapies, based either on adult, multipotent stem, or other types of pluripotent cells, offer promise for generating differentiated cell types to replace lost or damaged hair cells of the inner ear. In this review, we focus on the methods proposed and avenues for research that seem the most promising for stem cell-based auditory sensory cell regeneration, from work collected over the past 15 years.
Collapse
Affiliation(s)
- Hiroki Takeda
- Department of Otolaryngology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Department of Otolaryngology-Head and Neck Surgery, Kumamoto University Graduate School of Medicine, Kumamoto City, Japan; These authors contributed equally to this work
| | - Anna Dondzillo
- Department of Otolaryngology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; These authors contributed equally to this work
| | - Jessica A Randall
- Department of Otolaryngology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Samuel P Gubbels
- Department of Otolaryngology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
21
|
Liu Y, Yang F, Liang S, Liu Q, Fu S, Wang Z, Yang C, Lin J. N-Cadherin Upregulation Promotes the Neurogenic Differentiation of Menstrual Blood-Derived Endometrial Stem Cells. Stem Cells Int 2018; 2018:3250379. [PMID: 29692815 PMCID: PMC5859830 DOI: 10.1155/2018/3250379] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/19/2017] [Accepted: 12/04/2017] [Indexed: 02/07/2023] Open
Abstract
Peripheral nerve injuries are typically caused by either trauma or medical disorders, and recently, stem cell-based therapies have provided a promising treatment approach. Menstrual blood-derived endometrial stem cells (MenSCs) are considered an ideal therapeutic option for peripheral nerve repair due to a noninvasive collection procedure and their high proliferation rate and immunological tolerance. Here, we successfully isolated MenSCs and examined their biological characteristics including their morphology, multipotency, and immunophenotype. Subsequent in vitro studies demonstrated that MenSCs express high levels of neurotrophic factors, such as NT3, NT4, BDNF, and NGF, and are capable of transdifferentiating into glial-like cells under conventional induction conditions. Moreover, upregulation of N-cadherin (N-cad) mRNA and protein expression was observed after neurogenic differentiation. In vivo studies clearly showed that N-cad knockdown via in utero electroporation perturbed the migration and maturation of mouse neural precursor cells (NPCs). Finally, a further transfection assay also confirmed that N-cad upregulation in MenSCs results in the expression of S100. Collectively, our results confirmed the paracrine effect of MenSCs on neuroprotection as well as their potential for transdifferentiation into glial-like cells and demonstrated that N-cad upregulation promotes the neurogenic differentiation of MenSCs, thereby providing support for transgenic MenSC-based therapy for peripheral nerve injury.
Collapse
Affiliation(s)
- Yanli Liu
- Stem Cell Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang 453003, China
| | - Fen Yang
- Stem Cell Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Shengying Liang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang 453003, China
| | - Qing Liu
- Stem Cell Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Sulei Fu
- College of Biomedical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Zhenyu Wang
- School of Biological and Chemical Engineering, Liaoning Institute of Science and Technology, Benxi 117004, China
| | - Ciqing Yang
- Stem Cell Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang 453003, China
| | - Juntang Lin
- Stem Cell Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang 453003, China
- College of Biomedical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| |
Collapse
|
22
|
Current Perspectives Regarding Stem Cell-Based Therapy for Liver Cirrhosis. Can J Gastroenterol Hepatol 2018; 2018:4197857. [PMID: 29670867 PMCID: PMC5833156 DOI: 10.1155/2018/4197857] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/16/2018] [Indexed: 12/12/2022] Open
Abstract
Liver cirrhosis is a major cause of mortality and a common end of various progressive liver diseases. Since the effective treatment is currently limited to liver transplantation, stem cell-based therapy as an alternative has attracted interest due to promising results from preclinical and clinical studies. However, there is still much to be understood regarding the precise mechanisms of action. A number of stem cells from different origins have been employed for hepatic regeneration with different degrees of success. The present review presents a synopsis of stem cell research for the treatment of patients with liver cirrhosis according to the stem cell type. Clinical trials to date are summarized briefly. Finally, issues to be resolved and future perspectives are discussed with regard to clinical applications.
Collapse
|
23
|
Impact of Antibiotics on the Proliferation and Differentiation of Human Adipose-Derived Mesenchymal Stem Cells. Int J Mol Sci 2017; 18:ijms18122522. [PMID: 29186789 PMCID: PMC5751125 DOI: 10.3390/ijms18122522] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 12/17/2022] Open
Abstract
Adipose tissue is a promising source of mesenchymal stem cells. Their potential to differentiate and regenerate other types of tissues may be affected by several factors. This may be due to in vitro cell-culture conditions, especially the supplementation with antibiotics. The aim of our study was to evaluate the effects of a penicillin-streptomycin mixture (PS), amphotericin B (AmB), a complex of AmB with copper (II) ions (AmB-Cu2+) and various combinations of these antibiotics on the proliferation and differentiation of adipose-derived stem cells in vitro. Normal human adipose-derived stem cells (ADSC, Lonza) were routinely maintained in a Dulbecco’s Modified Eagle Medium (DMEM) that was either supplemented with selected antibiotics or without antibiotics. The ADSC that were used for the experiment were at the second passage. The effect of antibiotics on proliferation was analyzed using the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) and sulforhodamine-B (SRB) tests. Differentiation was evaluated based on Alizarin Red staining, Oil Red O staining and determination of the expression of ADSC, osteoblast and adipocyte markers by real-time RT-qPCR. The obtained results indicate that the influence of antibiotics on adipose-derived stem cells depends on the duration of exposure and on the combination of applied compounds. We show that antibiotics alter the proliferation of cells and also promote natural osteogenesis, and adipogenesis, and that this effect is also noticeable in stimulated osteogenesis.
Collapse
|
24
|
Tang BL. The use of mesenchymal stem cells (MSCs) for amyotrophic lateral sclerosis (ALS) therapy – a perspective on cell biological mechanisms. Rev Neurosci 2017; 28:725-738. [DOI: 10.1515/revneuro-2017-0018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/04/2017] [Indexed: 12/12/2022]
Abstract
AbstractRecent clinical trials of mesenchymal stem cells (MSCs) transplantation have demonstrated procedural safety and clinical proof of principle with a modest indication of benefit in patients with amyotrophic lateral sclerosis (ALS). While replacement therapy remained unrealistic, the clinical efficacy of this therapeutic option could be potentially enhanced if we could better decipher the mechanisms underlying some of the beneficial effects of transplanted cells, and work toward augmenting or combining these in a strategic manner. Novel ways whereby MSCs could act in modifying disease progression should also be explored. In this review, I discuss the known, emerging and postulated mechanisms of action underlying effects that transplanted MSCs may exert to promote motor neuron survival and/or to encourage regeneration in ALS. I shall also speculate on how transplanted cells may alter the diseased environment so as to minimize non-neuron cell autonomous damages by immune cells and astrocytes.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Medical Drive, Singapore 117597, Singapore
| |
Collapse
|
25
|
Ishii T, Sakai D, Schol J, Nakai T, Suyama K, Watanabe M. Sciatic nerve regeneration by transplantation of in vitro differentiated nucleus pulposus progenitor cells. Regen Med 2017. [DOI: 10.2217/rme-2016-0168] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Aim: To assess the applicability of mouse intervertebral disc-derived nucleus pulposus (NP) progenitor cells as a cell source for sciatic nerve regeneration. Materials & methods: P0-Cre/Floxed-EGFP-transgenic mouse-derived NP progenitor cells were differentiated to Schwann-like cells in conventional induction medium. Schwann-like cells were subsequently transplanted into a mouse model of sciatic nerve transection, and nerve regeneration assessed by immunohistochemistry, electron microscopy and functional walking track analysis and heat stimulus reflex. Results & conclusion: NP progenitor cells differentiated into Schwann-like cells. Transplantation of these cells promoted myelinated axon formation, morphology restoration and nerve function improvement. NP progenitor cells have the capacity to differentiate into neuronal cells and are candidates for peripheral nerve regeneration therapy.
Collapse
Affiliation(s)
- Takayuki Ishii
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine; 143 Shimokasuya, Isehara, Kanagawa, 259–1143, Japan
| | - Daisuke Sakai
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine; 143 Shimokasuya, Isehara, Kanagawa, 259–1143, Japan
| | - Jordy Schol
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine; 143 Shimokasuya, Isehara, Kanagawa, 259–1143, Japan
| | - Tomoko Nakai
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine; 143 Shimokasuya, Isehara, Kanagawa, 259–1143, Japan
| | - Kaori Suyama
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine; 143 Shimokasuya, Isehara, Kanagawa, 259–1143, Japan
| | - Masahiko Watanabe
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine; 143 Shimokasuya, Isehara, Kanagawa, 259–1143, Japan
| |
Collapse
|
26
|
Kang KJ, Ko SY, Ryu CJ, Jang YJ. A monoclonal antibody recognizes undifferentiation-specific carbohydrate moieties expressed on cell surface of the human dental pulp cells. Stem Cell Res 2017; 21:85-93. [DOI: 10.1016/j.scr.2017.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/30/2017] [Accepted: 04/02/2017] [Indexed: 12/29/2022] Open
|
27
|
|
28
|
Wei Y, Hu Z, Gu W, Liu G, Shi B, Liu E, Liu T. CD117 +CD44 + Stem T Cells Develop in the Thymus and Potently Suppress T-cell Proliferation by Modulating the CTLA-4 Pathway. Stem Cell Res Ther 2017; 8:56. [PMID: 28279199 PMCID: PMC5345162 DOI: 10.1186/s13287-017-0495-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 12/10/2016] [Accepted: 02/09/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND CD117 is expressed on double-negative (DN; CD4-CD8-) cells (Nat Rev Immunol 14:529-545; 2014), but whether it is expressed in other stages and its subsequent functions are unclear. We used an improved method of flow cytometry to analyze different populations of thymocytes (Sci Rep 4:5781; 2014). The expression of CD117 and CTLA-4 were directly assayed in the early stage of thymocytes. METHODS Flow cytometry was used to analyze different populations of thymocytes, and T-cell proliferation assays, RT-PCR, and real-time RT-PCR were used to characterize the stem cells and examine the function of CD44+CD117+ cells. RESULTS In DN cells, CD117 expression was greatest on CD44+CD25+ cells (DN2), followed by CD44+CD25- (DN1), CD44-CD25+ (DN3), and CD44-CD25- (DN4) cells. In thymocytes, CD117 expression was highest in DN cells, followed by single-positive (SP; CD4 or CD8) and double-positive (DP; CD4+CD8+) cells. Especially, CD117 expression was positively associated with CD44 and CTLA-4 expression. CTLA-4 expression was highest in DN cells, followed by SP and DP cells. CTLA-4 expression was positively associated with CD25, CD44, and Foxp3 expression. CD44+CD117+ T cells expressed more CTLA-4, which suppressed T-cell proliferation and blocked CTLA-4 to cause antibody-induced T-cell proliferation. CONCLUSION These results suggest that CD44+CD117+ T cells are stem cells and a specific T-cell phenotype that initially develops in the thymus, but they do not progress through DN3 and DN4 stages, lack a DP stage, and potently suppress T-cell proliferation and modulate the CTLA-4 pathway.
Collapse
Affiliation(s)
- Yang Wei
- Immunology and Tumor Research Instituted, the First Affiliate Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China.,Core Research Laboratory, the Second Affiliated Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710049, China
| | - Zhansheng Hu
- Immunology and Tumor Research Instituted, the First Affiliate Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China.,The First Affiliated Hospital, Jinzhou Medical University, Liaoning, 121004, People's Republic of China
| | - Wen Gu
- Immunology and Tumor Research Instituted, the First Affiliate Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Gang Liu
- Clinical Research Center, Guangdong Medical Collage, Zhanjiang, Guangdong, 524001, China
| | - Bingyin Shi
- Immunology and Tumor Research Instituted, the First Affiliate Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Enqi Liu
- The School of Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Tie Liu
- Immunology and Tumor Research Instituted, the First Affiliate Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China.
| |
Collapse
|
29
|
Soundara Rajan T, Giacoppo S, Scionti D, Diomede F, Grassi G, Pollastro F, Piattelli A, Bramanti P, Mazzon E, Trubiani O. Cannabidiol Activates Neuronal Precursor Genes in Human Gingival Mesenchymal Stromal Cells. J Cell Biochem 2016; 118:1531-1546. [PMID: 27918106 DOI: 10.1002/jcb.25815] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 11/28/2016] [Indexed: 12/18/2022]
Abstract
In the last years, mesenchymal stromal cells (MSCs) from oral tissues have received considerable interest in regenerative medicine since they can be obtained with minimal invasive procedure and exhibit immunomodulatory properties. This study was aimed to investigate whether in vitro pre-treatment of MSCs obtained from human gingiva (hGMSCs) with Cannabidiol (CBD), a cannabinoid component produced by the plant Cannabis sativa, may promote human gingiva derived MSCs to differentiate toward neuronal precursor cells. Specifically, we have treated the hGMSCs with CBD (5 µM) for 24 h in order to evaluate the expression of genes involved in cannabidiol signaling, cell proliferation, self-renewal and multipotency, and neural progenitor cells differentiation. Next generation sequencing (NGS) demonstrated that CBD activates genes associated with G protein coupled receptor signaling in hGMSCs. Genes involved in DNA replication, cell cycle, proliferation, and apoptosis were regulated. Moreover, genes associated with the biological process of neuronal progenitor cells (NCPs) proliferation, neuron differentiation, neurogenesis, and nervous system development were significantly modulated. From our results, we hypothesize that human gingiva-derived MSCs conditioned with CBD could represent a valid method for improving the hGMSCs phenotype and thus might be a potential therapeutic tool in the treatment of neurodegenerative diseases. J. Cell. Biochem. 118: 1531-1546, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | | | | | - Francesca Diomede
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti 66100, Italy
| | - Gianpaolo Grassi
- Council for Research and Experimentation in Agriculture-Research Centre for Industrial Crops (CRA-CIN), Rovigo, Italy
| | - Federica Pollastro
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale, Novara 28100, Italy
| | - Adriano Piattelli
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti 66100, Italy
| | | | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Messina 98124, Italy
| | - Oriana Trubiani
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti 66100, Italy
| |
Collapse
|
30
|
Skog MS, Nystedt J, Korhonen M, Anderson H, Lehti TA, Pajunen MI, Finne J. Expression of neural cell adhesion molecule and polysialic acid in human bone marrow-derived mesenchymal stromal cells. Stem Cell Res Ther 2016; 7:113. [PMID: 27528376 PMCID: PMC4986182 DOI: 10.1186/s13287-016-0373-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 06/28/2016] [Accepted: 07/21/2016] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND In order to develop novel clinical applications and to gain insights into possible therapeutic mechanisms, detailed molecular characterization of human bone marrow-derived mesenchymal stromal cells (hBM-MSCs) is needed. Neural cell adhesion molecule (NCAM, CD56) is a transmembrane glycoprotein modulating cell-cell and cell-matrix interactions. An additional post-translational modification of NCAM is the α2,8-linked polysialic acid (polySia). Because of its background, NCAM is often considered a marker of neural lineage commitment. Generally, hBM-MSCs are considered to be devoid of NCAM expression, but more rigorous characterization is needed. METHODS We have studied NCAM and polySia expression in five hBM-MSC lines at mRNA and protein levels. Cell surface localization was confirmed by immunofluorescence staining and expression frequency in the donor-specific lines by flow cytometry. For the detection of poorly immunogenic polySia, a fluorochrome-tagged catalytically defective enzyme was employed. RESULTS All five known NCAM isoforms are expressed in these cells at mRNA level and the three main isoforms are present at protein level. Both polysialyltransferases, generally responsible for NCAM polysialylation, are expressed at mRNA level, but only very few cells express polySia at the cell surface. CONCLUSIONS Our results underline the need for a careful control of methods and conditions in the characterization of MSCs. This study shows that, against the generally held view, clinical-grade hBM-MSCs do express NCAM. In contrast, although both polysialyltransferase genes are transcribed in these cells, very few express polySia at the cell surface. NCAM and polySia represent new candidate molecules for influencing MSC interactions.
Collapse
Affiliation(s)
- Maria S Skog
- Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland.
| | - Johanna Nystedt
- Cell Therapy Services, Finnish Red Cross Blood Service, Kivihaantie 7, FI-00310, Helsinki, Finland
| | - Matti Korhonen
- Cell Therapy Services, Finnish Red Cross Blood Service, Kivihaantie 7, FI-00310, Helsinki, Finland
| | - Heidi Anderson
- Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland.,Present Address: Genoscoper Laboratories Oy, P.O. Box 1040, FI-00251, Helsinki, Finland
| | - Timo A Lehti
- Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Maria I Pajunen
- Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland.,Present Address: Department of Bacteriology and Immunology, Medicum, Research Programs Unit, Immunobiology, University of Helsinki, P.O. Box 21, FI-00014, Helsinki, Finland
| | - Jukka Finne
- Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| |
Collapse
|
31
|
In Vitro Validation of a Closed Device Enabling the Purification of the Fluid Portion of Liposuction Aspirates. Plast Reconstr Surg 2016; 137:1157-1167. [DOI: 10.1097/prs.0000000000002014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
32
|
Liu Y, Kumar VS, Zhang W, Rehman J, Malik AB. Activation of type II cells into regenerative stem cell antigen-1(+) cells during alveolar repair. Am J Respir Cell Mol Biol 2015; 53:113-24. [PMID: 25474582 DOI: 10.1165/rcmb.2013-0497oc] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The alveolar epithelium is composed of two cell types: type I cells comprise 95% of the gas exchange surface area, whereas type II cells secrete surfactant, while retaining the ability to convert into type I cells to induce alveolar repair. Using lineage-tracing analyses in the mouse model of Pseudomonas aeruginosa-induced lung injury, we identified a population of stem cell antigen (Sca)-1-expressing type II cells with progenitor cell properties that mediate alveolar repair. These cells were shown to be distinct from previously reported Sca-1-expressing bronchioalveolar stem cells. Microarray and Wnt reporter studies showed that surfactant protein (Sp)-C(+)Sca-1(+) cells expressed Wnt signaling pathway genes, and inhibiting Wnt/β-catenin signaling prevented the regenerative function of Sp-C(+)Sca-1(+) cells in vitro. Thus, P. aeruginosa-mediated lung injury induces the generation of a Sca-1(+) subset of type II cells. The progenitor phenotype of the Sp-C(+)Sca-1(+) cells that mediates alveolar epithelial repair might involve Wnt signaling.
Collapse
Affiliation(s)
| | | | | | - Jalees Rehman
- Departments of 1 Pharmacology.,3 Medicine, University of Illinois College of Medicine, Chicago, Illinois
| | | |
Collapse
|
33
|
Adipose-derived Mesenchymal Stem Cells and Their Reparative Potential in Ischemic Heart Disease. ACTA ACUST UNITED AC 2015; 68:599-611. [DOI: 10.1016/j.rec.2015.02.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/23/2015] [Indexed: 12/21/2022]
|
34
|
Badimon L, Oñate B, Vilahur G. Células madre mesenquimales derivadas de tejido adiposo y su potencial reparador en la enfermedad isquémica coronaria. Rev Esp Cardiol 2015. [DOI: 10.1016/j.recesp.2015.02.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
35
|
Rusu MC, Vrapciu AD, Hostiuc S, Hariga CS. Brown adipocytes, cardiac protection and a common adipo- and myogenic stem precursor in aged human hearts. Med Hypotheses 2015; 85:212-4. [PMID: 25956736 DOI: 10.1016/j.mehy.2015.04.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 04/18/2015] [Accepted: 04/25/2015] [Indexed: 12/25/2022]
Abstract
New data on adult stem cells (ASCs) are continuously added by research for use in regenerative medicine. However organ-specific ASC markers are incompletely explored. It was demonstrated that in non-cardiac brown adipose tissue (BAT) CD133+ cells differentiate in cardiomyocytes, and such BAT-derived cells induce bone marrow-derived cells into cardiomyocytes, thus being a promising source for cardiac stem cell therapy. During embryogenesis the subepicardial fat derives from BAT. Although it was not specifically investigated in human adult or aged hearts, it is actually known that metabolically active BAT can be found in many adult humans, is related to antiobesity effects, and it may derive from stem/progenitor cells. Stro-1 can safely identify in situ cardiac stem cells (CSCs) with myogenic and adipogenic potential. It was therefore raised the hypothesis of subepicardial differentiation of CSCs in BAT in adult/aged hearts, which could be viewed, such as in infants, as a mechanism of protection. This could be determined by the reactivation of an embryologic differentiation pattern in which brown adipocytes and muscle cells derive from a common stem ancestor. Such quiescent common stem ancestors could be suggested in adult, or aged, human hearts, when subepicardial BAT is found, and if a Stro-1+/CD133+/Isl-1+ phenotype of CSCs is determined.
Collapse
Affiliation(s)
- M C Rusu
- Division of Anatomy, Faculty of Dental Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania; MEDCENTER, Center of Excellence in Laboratory Medicine and Pathology, Bucharest, Romania; International Society of Regenerative Medicine and Surgery (ISRMS), Romania.
| | - A D Vrapciu
- Division of Anatomy, Faculty of Dental Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - S Hostiuc
- Division of Legal Medicine and Bioethics, Department 2 Morphological Sciences, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania; National Institute of Legal Medicine, Bucharest, Romania
| | - C S Hariga
- Department 11 Surgery, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
36
|
Alexanian AR. Epigenetic modulators promote mesenchymal stem cell phenotype switches. Int J Biochem Cell Biol 2015; 64:190-4. [PMID: 25936755 DOI: 10.1016/j.biocel.2015.04.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 04/14/2015] [Accepted: 04/21/2015] [Indexed: 01/18/2023]
Abstract
Discoveries in recent years have suggested that some tissue specific adult stem cells in mammals might have the ability to differentiate into cell types from different germ layers. This phenomenon has been referred to as stem cell transdifferentiation or plasticity. Despite controversy, the current consensus holds that transdifferentiation does occur in mammals, but only within a limited range. Understanding the mechanisms that underlie the switches in phenotype and development of the methods that will promote such type of conversions can open up endless possibilities for regenerative medicine. Epigenetic control contributes to various processes that lead to cellular plasticity and DNA and histone covalent modifications play a key role in these processes. Recently, we have been able to convert human mesenchymal stem cells (hMSCs) into neural-like cells by exposing cells to epigenetic modifiers and neural inducing factors. The goal of this study was to investigate the stability and plasticity of these transdifferentiated cells. To this end, neurally induced MSCs (NI-hMSCs) were exposed to adipocyte inducing factors. Grown for 24-48 h in fat induction media NI-hMSCs reversed their morphology into fibroblast-like cells and regained their proliferative properties. After 3 weeks approximately 6% of hMSCs differentiated into multilocular or plurivacuolar adipocyte cells that demonstrated by Oil Red O staining. Re-exposure of these cultures or the purified adipocytes to neural induction medium induced the cells to re-differentiate into neuronal-like cells. These data suggest that cell plasticity can be manipulated by the combination of small molecule modulators of chromatin modifying enzymes and specific cell signaling pathways.
Collapse
Affiliation(s)
- Arshak R Alexanian
- Cell Reprogramming & Therapeutics LLC, W229 N1870 Westwood Drive, Waukesha, WI 53186 United States.
| |
Collapse
|
37
|
Wu R, Duan L, Cui F, Cao J, Xiang Y, Tang Y, Zhou L. S100A9 promotes human hepatocellular carcinoma cell growth and invasion through RAGE-mediated ERK1/2 and p38 MAPK pathways. Exp Cell Res 2015; 334:228-38. [PMID: 25907296 DOI: 10.1016/j.yexcr.2015.04.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 04/06/2015] [Accepted: 04/12/2015] [Indexed: 01/27/2023]
Abstract
S100A9 belongs to the S100 family of calcium-binding proteins and is over-expressed in many human tumors including hepatocellular carcinoma (HCC). Recent study demonstrated that S100A9 is significantly elevated and is associated with tumor differentiation and vascular invasion in HCC. The functional role of S100A9 is, however, poorly understood. Here, we demonstrated that S100A9 treatment increased viability, invasiveness and clone formation in three HCC cell lines (HepG2, SMMC-7721 and Huh7). S100A9 also promoted tumor growth in vivo by a xenograft mouse model. In addition, we observed a co-localization of S100A9 with receptor for advanced glycation end products (RAGE) in human HCC intratumoral tissues and an interaction of S100A9 with RAGE in vitro. Treatment with RAGE blocking antibody blocked the enhanced viability, invasion, clone formation and tumor growth in vivo resulted by S100A9, suggesting that these effects were mediated via RAGE ligation. In order to investigate the signaling pathways, mitogen-activated protein kinase (MAPK) phosphorylation was characterized. S100A9 caused a significant increase in p-p38 and p-ERK1/2 levels, and inhibition of which blocked enhanced invasion and viability resulted by S100A9, respectively. Furthermore, treatment with RAGE blocking antibodies also abrogated the S100A9-induced p38 and ERK1/2 activation, suggesting that S100A9-induced MAPK activation is mediated via RAGE ligation. Our data demonstrate that S100A9 binds to RAGE and stimulates RAGE-dependent MAPK signaling cascades, promoting cell growth and invasion in HCC.
Collapse
Affiliation(s)
- Rui Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Liang Duan
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Fang Cui
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ju Cao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Xiang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yishu Tang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lan Zhou
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
38
|
Stecklum M, Wulf-Goldenberg A, Purfürst B, Siegert A, Keil M, Eckert K, Fichtner I. Cell differentiation mediated by co-culture of human umbilical cord blood stem cells with murine hepatic cells. In Vitro Cell Dev Biol Anim 2015; 51:183-91. [PMID: 25270685 DOI: 10.1007/s11626-014-9817-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 08/28/2014] [Indexed: 12/27/2022]
Abstract
In the present study, purified human cord blood stem cells were co-cultivated with murine hepatic alpha mouse liver 12 (AML12) cells to compare the effect on endodermal stem cell differentiation by either direct cell-cell interaction or by soluble factors in conditioned hepatic cell medium. With that approach, we want to mimic in vitro the situation of preclinical transplantation experiments using human cells in mice. Cord blood stem cells, cultivated with hepatic conditioned medium, showed a low endodermal differentiation but an increased connexin 32 (Cx32) and Cx43, and cytokeratin 8 (CK8) and CK19 expression was monitored by reverse transcription polymerase chain reaction (RT-PCR). Microarray profiling indicated that in cultivated cord blood cells, 604 genes were upregulated 2-fold, with the highest expression for epithelial CK19 and epithelial cadherin (E-cadherin). On ultrastructural level, there were no major changes in the cellular morphology, except a higher presence of phago(ly)some-like structures observed. Direct co-culture of AML12 cells with cord blood cells led to less incisive differentiation with increased sex-determining region Y-box 17 (SOX17), Cx32 and Cx43, as well as epithelial CK8 and CK19 expressions. On ultrastructural level, tight cell contacts along the plasma membranes were revealed. FACS analysis in co-cultivated cells quantified dye exchange on low level, as also proved by time relapse video-imaging of labelled cells. Modulators of gap junction formation influenced dye transfer between the co-cultured cells, whereby retinoic acid increased and 3-heptanol reduced the dye transfer. The study indicated that the cell-co-cultured model of human umbilical cord blood cells and murine AML12 cells may be a suitable approach to study some aspects of endodermal/hepatic cell differentiation induction.
Collapse
Affiliation(s)
- Maria Stecklum
- Max Delbrück Center for Molecular Medicine, Berlin-Buch, Robert-Rössle-Str. 10, 13125, Berlin, Germany,
| | | | | | | | | | | | | |
Collapse
|
39
|
Li Y, Müller AL, Ngo MA, Sran K, Bellan D, Arora RC, Kirshenbaum LA, Freed DH. Statins impair survival of primary human mesenchymal progenitor cells via mevalonate depletion, NF-κB signaling, and Bnip3. J Cardiovasc Transl Res 2014; 8:96-105. [PMID: 25547946 DOI: 10.1007/s12265-014-9603-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/12/2014] [Indexed: 12/20/2022]
Abstract
Circulating progenitor cells of bone marrow origin have been implicated in transplant cardiac allograft vasculopathy (CAV) and cardiac fibrosis. HMG-CoA reductase inhibitors, called "statins," have been shown to impair the progression of CAV and improve patient survival. We examined the in vitro effects of three HMG-CoA reductase inhibitors atorvastatin, simvastatin, and pravastatin on the viability of MSCs and expression of nuclear factor kappa B (NF-κB). Mesenchymal stem cells (MSCs) isolated from human patients were treated with atorvastatin, simvastatin, and pravastatin at 0.1, 1.0, or 10 μM ± mevalonate. Human MSC treatment with 1 and 10 μM simvastatin or atorvastatin resulted in progressively reduced cell viability, which was associated with a decline in NF-κB p65. Viability was rescued by co-incubation with mevalonate or by pretreatment with Inhibitor of nuclear factor kappa-B kinase subunit beta (Iκκ-β). Pravastatin did not affect MSC viability or NF-κB expression. Mevalonate depletion through HMG-CoA reductase inhibition impairs the viability of primary human MSC through down-regulating NF-κB.
Collapse
Affiliation(s)
- Yun Li
- Institute of Cardiovascular Sciences, St. Boniface Research Centre, Department of Physiology, University of Manitoba, Winnipeg, MB, Canada
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Kaebisch C, Schipper D, Babczyk P, Tobiasch E. The role of purinergic receptors in stem cell differentiation. Comput Struct Biotechnol J 2014; 13:75-84. [PMID: 26900431 PMCID: PMC4720018 DOI: 10.1016/j.csbj.2014.11.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 10/31/2014] [Accepted: 11/03/2014] [Indexed: 12/20/2022] Open
Abstract
A major challenge modern society has to face is the increasing need for tissue regeneration due to degenerative diseases or tumors, but also accidents or warlike conflicts. There is great hope that stem cell-based therapies might improve current treatments of cardiovascular diseases, osteochondral defects or nerve injury due to the unique properties of stem cells such as their self-renewal and differentiation potential. Since embryonic stem cells raise severe ethical concerns and are prone to teratoma formation, adult stem cells are still in the focus of research. Emphasis is placed on cellular signaling within these cells and in between them for a better understanding of the complex processes regulating stem cell fate. One of the oldest signaling systems is based on nucleotides as ligands for purinergic receptors playing an important role in a huge variety of cellular processes such as proliferation, migration and differentiation. Besides their natural ligands, several artificial agonists and antagonists have been identified for P1 and P2 receptors and are already used as drugs. This review outlines purinergic receptor expression and signaling in stem cells metabolism. We will briefly describe current findings in embryonic and induced pluripotent stem cells as well as in cancer-, hematopoietic-, and neural crest-derived stem cells. The major focus will be placed on recent findings of purinergic signaling in mesenchymal stem cells addressed in in vitro and in vivo studies, since stem cell fate might be manipulated by this system guiding differentiation towards the desired lineage in the future.
Collapse
Affiliation(s)
| | | | | | - Edda Tobiasch
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, Von-Liebig-Str. 20, 53359 Rheinbach, Germany
| |
Collapse
|
41
|
Mosaad YM. Hematopoietic stem cells: an overview. Transfus Apher Sci 2014; 51:68-82. [PMID: 25457002 DOI: 10.1016/j.transci.2014.10.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 10/06/2014] [Accepted: 10/08/2014] [Indexed: 12/15/2022]
Abstract
Considerable efforts have been made in recent years in understanding the mechanisms that govern hematopoietic stem cell (HSC) origin, development, differentiation, self-renewal, aging, trafficking, plasticity and transdifferentiation. Hematopoiesis occurs in sequential waves in distinct anatomical locations during development and these shifts in location are accompanied by changes in the functional status of the stem cells and reflect the changing needs of the developing organism. HSCs make a choice of either self-renewal or committing to differentiation. The balance between self-renewal and differentiation is considered to be critical to the maintenance of stem cell numbers. It is still under debate if HSC can rejuvenate infinitely or if they do not possess ''true" self-renewal and undergo replicative senescence such as any other somatic cell. Gene therapy applications that target HSCs offer a great potential for the treatment of hematologic and immunologic diseases. However, the clinical success has been limited by many factors. This review is intended to summarize the recent advances made in the human HSC field, and will review the hematopoietic stem cell from definition through development to clinical applications.
Collapse
Affiliation(s)
- Youssef Mohamed Mosaad
- Clinical Immunology Unit, Clinical Pathology Department & Mansoura Research Center for Cord Stem Cell (MARC_CSC), Mansoura Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
42
|
Cicione C, Muiños-López E, Hermida-Gómez T, Fuentes-Boquete I, Díaz-Prado S, Blanco FJ. Alternative protocols to induce chondrogenic differentiation: transforming growth factor-β superfamily. Cell Tissue Bank 2014; 16:195-207. [DOI: 10.1007/s10561-014-9472-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 08/26/2014] [Indexed: 12/22/2022]
|
43
|
Vainshtein JM, Kabarriti R, Mehta KJ, Roy-Chowdhury J, Guha C. Bone marrow-derived stromal cell therapy in cirrhosis: clinical evidence, cellular mechanisms, and implications for the treatment of hepatocellular carcinoma. Int J Radiat Oncol Biol Phys 2014; 89:786-803. [PMID: 24969793 DOI: 10.1016/j.ijrobp.2014.02.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 02/09/2014] [Accepted: 02/12/2014] [Indexed: 01/18/2023]
Abstract
Current treatment options for hepatocellular carcinoma (HCC) are often limited by the presence of underlying liver disease. In patients with liver cirrhosis, surgery, chemotherapy, and radiation therapy all carry a high risk of hepatic complications, ranging from ascites to fulminant liver failure. For patients receiving radiation therapy, cirrhosis dramatically reduces the already limited radiation tolerance of the liver and represents the most important clinical risk factor for the development of radiation-induced liver disease. Although improvements in conformal radiation delivery techniques have improved our ability to safely irradiate confined areas of the liver to increasingly higher doses with excellent local disease control, patients with moderate-to-severe liver cirrhosis continue to face a shortage of treatment options for HCC. In recent years, evidence has emerged supporting the use of bone marrow-derived stromal cells (BMSCs) as a promising treatment for liver cirrhosis, with several clinical studies demonstrating sustained improvement in clinical parameters of liver function after autologous BMSC infusion. Three predominant populations of BMSCs, namely hematopoietic stem cells, mesenchymal stem cells, and endothelial progenitor cells, seem to have therapeutic potential in liver injury and cirrhosis. Preclinical studies of BMSC transplantation have identified a range of mechanisms through which these cells mediate their therapeutic effects, including hepatocyte transdifferentiation and fusion, paracrine stimulation of hepatocyte proliferation, inhibition of activated hepatic stellate cells, enhancement of fibrolytic matrix metalloproteinase activity, and neovascularization of regenerating liver. By bolstering liver function in patients with underlying Child's B or C cirrhosis, autologous BMSC infusion holds great promise as a therapy to improve the safety, efficacy, and utility of surgery, chemotherapy, and hepatic radiation therapy in the treatment of HCC.
Collapse
Affiliation(s)
| | - Rafi Kabarriti
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Keyur J Mehta
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Jayanta Roy-Chowdhury
- Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York; Department of Genetics, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Chandan Guha
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York; Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
44
|
Chun SY, Kwon JB, Chae SY, Lee JK, Bae JS, Kim BS, Kim HT, Yoo ES, Lim JO, Yoo JJ, Kim WJ, Kim BW, Kwon TG. Combined injection of three different lineages of early-differentiating human amniotic fluid-derived cells restores urethral sphincter function in urinary incontinence. BJU Int 2014; 114:770-83. [PMID: 24841807 DOI: 10.1111/bju.12815] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To investigate whether a triple combination of early-differentiated cells derived from human amniotic fluid stem cells (hAFSCs) would show synergistic effects in urethral sphincter regeneration. MATERIALS AND METHODS We early-differentiated hAFSCs into muscle, neuron and endothelial progenitor cells and then injected them into the urethral sphincter region of pudendal neurectomized ICR mice, as single-cell, double-cell or triple-cell combinations. Urodynamic studies and histological, immunohistochemical and molecular analyses were performed. RESULTS Urodynamic study showed significantly improved leak point pressure in the triple-cell-combination group compared with the single-cell- or double-cell-combination groups. These functional results were confirmed by histological and immunohistochemical analyses, as evidenced by the formation of new striated muscle fibres and neuromuscular junctions at the cell injection site. Molecular analysis showed higher target marker expression in the retrieved urethral tissue of the triple-cell-combination group. The injection of early-differentiated hAFSCs suppressed in vivo host CD8 lymphocyte aggregations and did not form teratoma. The nanoparticle-labelled early-differentiated hAFSCs could be tracked in vivo with optical imaging for up to 14 days after injection. CONCLUSION Our novel concept of triple-combined early-differentiated cell therapy for the damaged sphincter may provide a viable option for incontinence treatment.
Collapse
Affiliation(s)
- So Young Chun
- Joint Institute for Regenerative Medicine, Kyungpook National University Hospital, Daegu, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Higashi K, Asano K, Yagi M, Yamada K, Arakawa T, Ehashi T, Mori T, Sumida K, Kushida M, Ando S, Kinoshita M, Kakehi K, Tachibana T, Saito K. Expression of the clustered NeuAcα2-3Galβ O-glycan determines the cell differentiation state of the cells. J Biol Chem 2014; 289:25833-43. [PMID: 25074924 DOI: 10.1074/jbc.m114.550848] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Human embryonic stem cells (hESCs) are pluripotent stem cells from early embryos, and their self-renewal capacity depends on the sustained expression of hESC-specific molecules and the suppressed expression of differentiation-associated genes. To discover novel molecules expressed on hESCs, we generated a panel of monoclonal antibodies against undifferentiated hESCs and evaluated their ability to mark cancer cells, as well as hESCs. MAb7 recognized undifferentiated hESCs and showed a diffuse band with molecular mass of >239 kDa in the lysates of hESCs. Although some amniotic epithelial cells expressed MAb7 antigen, its expression was barely detected in normal human keratinocytes, fibroblasts, or endothelial cells. The expression of MAb7 antigen was observed only in pancreatic and gastric cancer cells, and its levels were elevated in metastatic and poorly differentiated cancer cell lines. Analyses of MAb7 antigen suggested that the clustered NeuAcα2-3Galβ O-linked oligosaccharides on DMBT1 (deleted in malignant brain tumors 1) were critical for MAb7 binding in cancer cells. Although features of MAb7 epitope were similar with those of TRA-1-60, distribution of MAb7 antigen in cancer cells was different from that of TRA-1-60 antigen. Exposure of a histone deacetylase inhibitor to differentiated gastric cancer MKN74 cells evoked the expression of MAb7 antigen, whereas DMBT1 expression remained unchanged. Cell sorting followed by DNA microarray analyses identified the down-regulated genes responsible for the biosynthesis of MAb7 antigen in MKN74 cells. In addition, treatment of metastatic pancreatic cancer cells with MAb7 significantly abrogated the adhesion to endothelial cells. These results raised the possibility that MAb7 epitope is a novel marker for undifferentiated cells such as hESCs and cancer stem-like cells and plays a possible role in the undifferentiated cells.
Collapse
Affiliation(s)
- Kiyoshi Higashi
- From the Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Osaka 554-8558, Japan,
| | - Kouji Asano
- From the Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Osaka 554-8558, Japan
| | - Masaki Yagi
- the Department of Bioengineering, Graduate School of Engineering, Osaka City University, Osaka 558-8585, Japan
| | - Keita Yamada
- the Laboratory of Toxicology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka 584-8540, Japan, and
| | - Tatsuhiko Arakawa
- the Department of Bioengineering, Graduate School of Engineering, Osaka City University, Osaka 558-8585, Japan
| | - Tomo Ehashi
- From the Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Osaka 554-8558, Japan
| | - Takashi Mori
- From the Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Osaka 554-8558, Japan
| | - Kayo Sumida
- From the Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Osaka 554-8558, Japan
| | - Masahiko Kushida
- From the Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Osaka 554-8558, Japan
| | - Satoshi Ando
- From the Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Osaka 554-8558, Japan
| | | | - Kazuaki Kakehi
- the School of Pharmacy, Kinki University, Higashi-Osaka 577-8502, Japan
| | - Taro Tachibana
- the Department of Bioengineering, Graduate School of Engineering, Osaka City University, Osaka 558-8585, Japan
| | - Koichi Saito
- From the Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Osaka 554-8558, Japan
| |
Collapse
|
46
|
Ismail F, Winkler DA. Getting to the Source: Selective Drug Targeting of Cancer Stem Cells. ChemMedChem 2014; 9:885-98. [DOI: 10.1002/cmdc.201400068] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 04/02/2014] [Indexed: 12/22/2022]
|
47
|
Cieślar-Pobuda A, Bäck M, Magnusson K, Jain MV, Rafat M, Ghavami S, Nilsson KPR, Łos MJ. Cell type related differences in staining with pentameric thiophene derivatives. Cytometry A 2014; 85:628-35. [DOI: 10.1002/cyto.a.22437] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 12/04/2013] [Accepted: 12/24/2013] [Indexed: 11/09/2022]
Affiliation(s)
- Artur Cieślar-Pobuda
- Department of Clinical and Experimental Medicine (IKE); Division of Cell Biology, and Integrative Regenerative Medicine Center (IGEN), Linköping University; Linköping Sweden
- Biosystems Group; Institute of Automatic Control; Silesian University of Technology; Gliwice Poland
| | - Marcus Bäck
- Department of Chemistry; IFM, Linköping University; Linköping Sweden
| | - Karin Magnusson
- Department of Chemistry; IFM, Linköping University; Linköping Sweden
| | - Mayur V. Jain
- Department of Clinical and Experimental Medicine (IKE); Division of Cell Biology, and Integrative Regenerative Medicine Center (IGEN), Linköping University; Linköping Sweden
| | - Mehrdad Rafat
- Department of Clinical and Experimental Medicine (IKE); Division of Cell Biology, and Integrative Regenerative Medicine Center (IGEN), Linköping University; Linköping Sweden
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science; Manitoba Institute of Child Health; Manitoba Canada
- Department of Physiology; St. Boniface Research Centre; University of Manitoba; Manitoba Canada
| | | | - Marek J. Łos
- Department of Clinical and Experimental Medicine (IKE); Division of Cell Biology, and Integrative Regenerative Medicine Center (IGEN), Linköping University; Linköping Sweden
- Department of Pathology; Pomeranian Medical University; Szczecin Poland
| |
Collapse
|
48
|
Ghavami S, Shojaei S, Yeganeh B, Ande SR, Jangamreddy JR, Mehrpour M, Christoffersson J, Chaabane W, Moghadam AR, Kashani HH, Hashemi M, Owji AA, Łos MJ. Autophagy and apoptosis dysfunction in neurodegenerative disorders. Prog Neurobiol 2013; 112:24-49. [PMID: 24211851 DOI: 10.1016/j.pneurobio.2013.10.004] [Citation(s) in RCA: 758] [Impact Index Per Article: 63.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 10/08/2013] [Accepted: 10/15/2013] [Indexed: 12/12/2022]
Abstract
Autophagy and apoptosis are basic physiologic processes contributing to the maintenance of cellular homeostasis. Autophagy encompasses pathways that target long-lived cytosolic proteins and damaged organelles. It involves a sequential set of events including double membrane formation, elongation, vesicle maturation and finally delivery of the targeted materials to the lysosome. Apoptotic cell death is best described through its morphology. It is characterized by cell rounding, membrane blebbing, cytoskeletal collapse, cytoplasmic condensation, and fragmentation, nuclear pyknosis, chromatin condensation/fragmentation, and formation of membrane-enveloped apoptotic bodies, that are rapidly phagocytosed by macrophages or neighboring cells. Neurodegenerative disorders are becoming increasingly prevalent, especially in the Western societies, with larger percentage of members living to an older age. They have to be seen not only as a health problem, but since they are care-intensive, they also carry a significant economic burden. Deregulation of autophagy plays a pivotal role in the etiology and/or progress of many of these diseases. Herein, we briefly review the latest findings that indicate the involvement of autophagy in neurodegenerative diseases. We provide a brief introduction to autophagy and apoptosis pathways focusing on the role of mitochondria and lysosomes. We then briefly highlight pathophysiology of common neurodegenerative disorders like Alzheimer's diseases, Parkinson's disease, Huntington's disease and Amyotrophic lateral sclerosis. Then, we describe functions of autophagy and apoptosis in brain homeostasis, especially in the context of the aforementioned disorders. Finally, we discuss different ways that autophagy and apoptosis modulation may be employed for therapeutic intervention during the maintenance of neurodegenerative disorders.
Collapse
Affiliation(s)
- Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Canada; Manitoba Institute of Child Health, Department of Physiology, University of Manitoba, Winnipeg, Canada; St. Boniface Research Centre, University of Manitoba, Winnipeg, Canada
| | - Shahla Shojaei
- Department of Biochemistry, Recombinant Protein Laboratory, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behzad Yeganeh
- Manitoba Institute of Child Health, Department of Physiology, University of Manitoba, Winnipeg, Canada; Hospital for Sick Children Research Institute, Department of Physiology and Experimental Medicine, University of Toronto, Canada
| | - Sudharsana R Ande
- Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - Jaganmohan R Jangamreddy
- Department of Clinical and Experimental Medicine (IKE), Integrative Regenerative Medicine Center (IGEN), Division of Cell Biology, Linkoping University, Linkoping, Sweden
| | - Maryam Mehrpour
- INSERM U845, Research Center "Growth & Signaling" Paris Descartes University Medical School, France
| | - Jonas Christoffersson
- Department of Clinical and Experimental Medicine (IKE), Integrative Regenerative Medicine Center (IGEN), Division of Cell Biology, Linkoping University, Linkoping, Sweden
| | - Wiem Chaabane
- Department of Clinical and Experimental Medicine (IKE), Integrative Regenerative Medicine Center (IGEN), Division of Cell Biology, Linkoping University, Linkoping, Sweden; Department of Biology, Faculty of Sciences, Tunis University, Tunis, Tunisia
| | | | - Hessam H Kashani
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Canada; Manitoba Institute of Child Health, Department of Physiology, University of Manitoba, Winnipeg, Canada
| | - Mohammad Hashemi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran; Cellular and Molecular Biology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ali A Owji
- Department of Biochemistry, Recombinant Protein Laboratory, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Marek J Łos
- Department of Clinical and Experimental Medicine (IKE), Integrative Regenerative Medicine Center (IGEN), Division of Cell Biology, Linkoping University, Linkoping, Sweden.
| |
Collapse
|
49
|
Oñate B, Vilahur G, Camino-López S, Díez-Caballero A, Ballesta-López C, Ybarra J, Moscatiello F, Herrero J, Badimon L. Stem cells isolated from adipose tissue of obese patients show changes in their transcriptomic profile that indicate loss in stemcellness and increased commitment to an adipocyte-like phenotype. BMC Genomics 2013; 14:625. [PMID: 24040759 PMCID: PMC3848661 DOI: 10.1186/1471-2164-14-625] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 09/11/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The adipose tissue is an endocrine regulator and a risk factor for atherosclerosis and cardiovascular disease when by excessive accumulation induces obesity. Although the adipose tissue is also a reservoir for stem cells (ASC) their function and "stemcellness" has been questioned. Our aim was to investigate the mechanisms by which obesity affects subcutaneous white adipose tissue (WAT) stem cells. RESULTS Transcriptomics, in silico analysis, real-time polymerase chain reaction (PCR) and western blots were performed on isolated stem cells from subcutaneous abdominal WAT of morbidly obese patients (ASCmo) and of non-obese individuals (ASCn). ASCmo and ASCn gene expression clustered separately from each other. ASCmo showed downregulation of "stemness" genes and upregulation of adipogenic and inflammatory genes with respect to ASCn. Moreover, the application of bioinformatics and Ingenuity Pathway Analysis (IPA) showed that the transcription factor Smad3 was tentatively affected in obese ASCmo. Validation of this target confirmed a significantly reduced Smad3 nuclear translocation in the isolated ASCmo. CONCLUSIONS The transcriptomic profile of the stem cells reservoir in obese subcutaneous WAT is highly modified with significant changes in genes regulating stemcellness, lineage commitment and inflammation. In addition to body mass index, cardiovascular risk factor clustering further affect the ASC transcriptomic profile inducing loss of multipotency and, hence, capacity for tissue repair. In summary, the stem cells in the subcutaneous WAT niche of obese patients are already committed to adipocyte differentiation and show an upregulated inflammatory gene expression associated to their loss of stemcellness.
Collapse
Affiliation(s)
- Blanca Oñate
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Effects of severe hypoxia on bone marrow mesenchymal stem cells differentiation potential. Stem Cells Int 2013; 2013:232896. [PMID: 24082888 PMCID: PMC3777136 DOI: 10.1155/2013/232896] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Revised: 06/27/2013] [Accepted: 06/30/2013] [Indexed: 02/06/2023] Open
Abstract
Background. The interests in mesenchymal stem cells (MSCs) and their application in cell therapy have resulted in a better understanding of the basic biology of these cells. Recently hypoxia has been indicated as crucial for complete chondrogenesis. We aimed at analyzing bone marrow MSCs (BM-MSCs) differentiation capacity under normoxic and severe hypoxic culture conditions. Methods. MSCs were characterized by flow cytometry and differentiated towards adipocytes, osteoblasts, and chondrocytes under normoxic or severe hypoxic conditions. The differentiations were confirmed comparing each treated point with a control point made of cells grown in DMEM and fetal bovine serum (FBS). Results. BM-MSCs from the donors displayed only few phenotypical differences in surface antigens expressions. Analyzing marker genes expression levels of the treated cells compared to their control point for each lineage showed a good differentiation in normoxic conditions and the absence of this differentiation capacity in severe hypoxic cultures. Conclusions. In our experimental conditions, severe hypoxia affects the in vitro differentiation potential of BM-MSCs. Adipogenic, osteogenic, and chondrogenic differentiations are absent in severe hypoxic conditions. Our work underlines that severe hypoxia slows cell differentiation by means of molecular mechanisms since a decrease in the expression of adipocyte-, osteoblast-, and chondrocyte-specific genes was observed.
Collapse
|