1
|
Guo H, Sheng A, Qi X, Zhu L, Wang G, Zou Y, Guan Q, Lu Y, Tang H, Hou X. Depot-specific differences and heterogeneity of adipose-derived stem cells in diet-induced obesity. Obesity (Silver Spring) 2024. [PMID: 39496515 DOI: 10.1002/oby.24149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 11/06/2024]
Abstract
OBJECTIVE Obesity is a global health concern. Studying the heterogeneity of adipose-derived stem cells (ADSCs) plays a pivotal role in understanding metabolic disorders, such as obesity. METHODS Mass cytometry was used to determine the depot-specific differences and heterogeneity of ADSCs and their alterations at the single-cell level in a diet-induced-obesity (DIO) model in which mice were treated with liraglutide. RESULTS We characterized the relationship among ADSC markers and found that CD26 and CD142 could identify the most representative heterogeneous ADSCs in subcutaneous adipose tissue and visceral adipose tissue. Specifically, CD26+CD142- and CD26+CD142+ ADSCs were exclusive to subcutaneous adipose tissue and visceral adipose tissue, respectively, whereas CD26-CD142+ ADSCs were present in both. RNA analysis explored the potential functions of these three subgroups. In the visceral adipose tissue of DIO mice, we observed a substantial downregulation of CD26+CD142+ ADSCs and upregulation of CD26-CD142+ ADSCs, both of which were mitigated by liraglutide treatment. CONCLUSIONS Our study highlights the depot-specific differences and heterogeneity of ADSCs and their alterations under DIO conditions, which can potentially be reversed by liraglutide treatment. This study provides new insights into the identification of more specific ADSC subgroups to explore the etiology of metabolism-related diseases.
Collapse
Affiliation(s)
- Honglin Guo
- Department of Pathology, Shandong Provincial Hospital, Shandong University, Jinan, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, China
- Department of Center Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Ailing Sheng
- Department of Endocrinology, Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Xiangyu Qi
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lin Zhu
- Department of Endocrinology, University Town hospital, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guanyu Wang
- Department of Internal Medicine, No. 2 People's Hospital of Lixia District, Jinan, China
| | - Yizhou Zou
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qingbo Guan
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuntao Lu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Stem Cell Clinical Institute, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Hui Tang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Stem Cell Clinical Institute, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xu Hou
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Stem Cell Clinical Institute, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
2
|
Zhang J, Zhang N, Mai Q, Zhou C. The frontier of precision medicine: application of single-cell multi-omics in preimplantation genetic diagnosis. Brief Funct Genomics 2024:elae041. [PMID: 39486398 DOI: 10.1093/bfgp/elae041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/03/2024] [Indexed: 11/04/2024] Open
Abstract
The advent of single-cell multi-omics technologies has revolutionized the landscape of preimplantation genetic diagnosis (PGD), offering unprecedented insights into the genetic, transcriptomic, and proteomic profiles of individual cells in early-stage embryos. This breakthrough holds the promise of enhancing the accuracy, efficiency, and scope of PGD, thereby significantly improving outcomes in assisted reproductive technologies (ARTs) and genetic disease prevention. This review provides a comprehensive overview of the importance of PGD in the context of precision medicine and elucidates how single-cell multi-omics technologies have transformed this field. We begin with a brief history of PGD, highlighting its evolution and application in detecting genetic disorders and facilitating ART. Subsequently, we delve into the principles, methodologies, and applications of single-cell genomics, transcriptomics, and proteomics in PGD, emphasizing their role in improving diagnostic precision and efficiency. Furthermore, we review significant recent advances within this domain, including key experimental designs, findings, and their implications for PGD practices. The advantages and limitations of these studies are analyzed to assess their potential impact on the future development of PGD technologies. Looking forward, we discuss the emerging research directions and challenges, focusing on technological advancements, new application areas, and strategies to overcome existing limitations. In conclusion, this review underscores the pivotal role of single-cell multi-omics in PGD, highlighting its potential to drive the progress of precision medicine and personalized treatment strategies, thereby marking a new era in reproductive genetics and healthcare.
Collapse
Affiliation(s)
- Jinglei Zhang
- Reproductive Medical Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Nan Zhang
- General Surgery, The First Affiliated Hospital of Henan University of CM, Zhengzhou 450052, China
| | - Qingyun Mai
- Reproductive Medical Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Canquan Zhou
- Reproductive Medical Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| |
Collapse
|
3
|
Chen S, Liang B, Xu J. Unveiling heterogeneity in MSCs: exploring marker-based strategies for defining MSC subpopulations. J Transl Med 2024; 22:459. [PMID: 38750573 PMCID: PMC11094970 DOI: 10.1186/s12967-024-05294-5] [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: 11/28/2023] [Accepted: 05/11/2024] [Indexed: 05/19/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) represent a heterogeneous cell population distributed throughout various tissues, demonstrating remarkable adaptability to microenvironmental cues and holding immense promise for disease treatment. However, the inherent diversity within MSCs often leads to variability in therapeutic outcomes, posing challenges for clinical applications. To address this heterogeneity, purification of MSC subpopulations through marker-based isolation has emerged as a promising approach to ensure consistent therapeutic efficacy. In this review, we discussed the reported markers of MSCs, encompassing those developed through candidate marker strategies and high-throughput approaches, with the aim of explore viable strategies for addressing the heterogeneity of MSCs and illuminate prospective research directions in this field.
Collapse
Affiliation(s)
- Si Chen
- Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, People's Republic of China
| | - Bowei Liang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, People's Republic of China
| | - Jianyong Xu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Fuqiang Avenue 1001, Shenzhen, 518060, Guangdong, People's Republic of China.
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China.
| |
Collapse
|
4
|
Zhu X, Zeng C, Yu B. White adipose tissue in metabolic associated fatty liver disease. Clin Res Hepatol Gastroenterol 2024; 48:102336. [PMID: 38604293 DOI: 10.1016/j.clinre.2024.102336] [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: 12/05/2023] [Revised: 03/28/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND Metabolic associated fatty liver disease (MAFLD) is a prevalent chronic liver condition globally, currently lacking universally recognized therapeutic drugs, thereby increasing the risk of cirrhosis and hepatocellular carcinoma. Research has reported an association between white adipose tissue and MAFLD. SCOPE OF REVIEW White adipose tissue (WAT) is involved in lipid metabolism and can contribute to the progression of MAFLD by mediating insulin resistance, inflammation, exosomes, autophagy, and other processes. This review aims to elucidate the mechanisms through which WAT plays a role in the development of MAFLD. MAJOR CONCLUSIONS WAT participates in the occurrence and progression of MAFLD by mediating insulin resistance, inflammation, autophagy, and exosome secretion. Fibrosis and restricted expansion of adipose tissue can lead to the release of more free fatty acids (FFA), exacerbating the progression of MAFLD. WAT-secreted TNF-α and IL-1β, through the promotion of JNK/JKK/p38MAPK expression, interfere with insulin receptor serine and tyrosine phosphorylation, worsening insulin resistance. Adiponectin, by inhibiting the TLR-4-NF-κB pathway and suppressing M2 to M1 transformation, further inhibits the secretion of IL-6, IL-1β, and TNF-α, improving insulin resistance in MAFLD patients. Various gene expressions within WAT, such as MBPAT7, Nrf2, and Ube4A, can ameliorate insulin resistance in MAFLD patients. Autophagy-related gene Atg7 promotes the expression of fibrosis-related genes, worsening MAFLD. Non-pharmacological treatments, including diabetes-related medications and exercise, can improve MAFLD.
Collapse
Affiliation(s)
- Xiaoqin Zhu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, Hubei, 430000, PR China
| | - Chuanfei Zeng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, Hubei, 430000, PR China
| | - Baoping Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, Hubei, 430000, PR China.
| |
Collapse
|
5
|
Zhang Q, Lu C, Lu F, Liao Y, Cai J, Gao J. Challenges and opportunities in obesity: the role of adipocytes during tissue fibrosis. Front Endocrinol (Lausanne) 2024; 15:1365156. [PMID: 38686209 PMCID: PMC11056552 DOI: 10.3389/fendo.2024.1365156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Obesity is a chronic disease that affects the energy balance of the whole body. In addition to increasing fat mass, tissue fibrosis occurred in white adipose tissue in obese condition. Fibrosis is the over-activation of fibroblasts leading to excessive accumulation of extracellular matrix, which could be caused by various factors, including the status of adipocytes. The morphology of adipocytes responds rapidly and dynamically to nutrient fluctuations. Adaptive hypertrophy of normal adipocytes protects peripheral organs from damage from lipotoxicity. However, the biological behavior of hypertrophic adipocytes in chronic obesity is abnormally altered. Adipocytes lead to fibrotic remodeling of the extracellular matrix by inducing unresolved chronic inflammation, persistent hypoxia, and increasing myofibroblast numbers. Moreover, adipocyte-induced fibrosis not only restricts the flexible expansion and contraction of adipose tissue but also initiates the development of various diseases through cellular autonomic and paracrine effects. Regarding anti-fibrotic therapy, dysregulated intracellular signaling and epigenetic changes represent potential candidate targets. Thus, modulation of adipocytes may provide potential therapeutic avenues for reversing pathological fibrosis in adipose tissue and achieving the anti-obesity purpose.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chongxuan Lu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunjun Liao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junrong Cai
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Shuster-Hyman H, Siddiqui F, Gallagher D, Gauthier-Fisher A, Librach CL. Time course and mechanistic analysis of human umbilical cord perivascular cell mitigation of lipopolysaccharide-induced systemic and neurological inflammation. Cytotherapy 2023; 25:125-137. [PMID: 36473795 DOI: 10.1016/j.jcyt.2022.10.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 10/11/2022] [Accepted: 10/20/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND AIMS Because of their potent immunomodulatory and anti-inflammatory properties, mesenchymal stromal cells are a major focus in the field of stem cell therapy. However, the precise mechanisms underlying this are not entirely understood. Human umbilical cord perivascular cells (HUCPVCs) are a promising cell therapy candidate. This study was designed to evaluate the time course and mechanisms by which HUCPVCs mitigate lipopolysaccharide (LPS)-induced systemic and neurological inflammation in immunocompetent mice. To explore the underlying mechanisms, the authors investigated the biodistribution and fate of HUCPVCs. METHODS Male C57BL/6 mice were randomly allocated to four groups: control, LPS, HUCPVCs or LPS + HUCPVCs. Quantitative polymerase chain reaction, enzyme-linked immunosorbent assay and cytokine arrays were used to assess changes in pro-inflammatory mediators systemically and in the brain. Depressive-like behavioral changes were evaluated via a forced swim test. Quantum dot (qDot) labeling and immunohistochemistry were used to assess the biodistribution and fate of HUCPVCs and interactions with recipient innate immune cells. RESULTS A single intravenously delivered dose of HUCPVCs significantly reduced the systemic inflammation induced by LPS within the first 24 h after administration. HUCPVC treatment abrogated the upregulated expression of pro-inflammatory genes in the hippocampus and cortex and attenuated depressive-like behavior induced by LPS treatment. Biodistribution analysis revealed that HUCPVC-derived qDots rapidly accumulated in the lungs and demonstrated limited in vivo persistence. Furthermore, qDot signals were associated with major recipient innate immune cells and promoted a shift in macrophages toward a regulatory phenotype in the lungs. CONCLUSIONS Overall, this study demonstrates that HUCPVCs can successfully reduce systemic and neurological inflammation induced by LPS within the first 24 h after administration. Biodistribution and fate analyses suggest a critical role for the innate immune system in the HUCPVC-based immunomodulation mechanism.
Collapse
Affiliation(s)
- Hannah Shuster-Hyman
- CReATe Fertility Center, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | - Clifford L Librach
- CReATe Fertility Center, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
7
|
Qu Q, Fu B, Long Y, Liu ZY, Tian XH. Current Strategies for Promoting the Large-scale Production of Exosomes. Curr Neuropharmacol 2023; 21:1964-1979. [PMID: 36797614 PMCID: PMC10514529 DOI: 10.2174/1570159x21666230216095938] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/21/2022] [Accepted: 11/05/2022] [Indexed: 02/18/2023] Open
Abstract
Exosomes, as nanoscale biological vesicles, have been shown to have great potential for biomedical applications. However, the low yield of exosomes limits their application. In this review, we focus on methods to increase exosome yield. Two main strategies are used to increase exosome production, one is based on genetic manipulation of the exosome biogenesis and release pathway, and the other is by pretreating parent cells, changing the culture method or adding different components to the medium. By applying these strategies, exosomes can be produced on a large scale to facilitate their practical application in the clinic.
Collapse
Affiliation(s)
- Qing Qu
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, 77 Puhe Avenue, Shenbei New District, Shenyang, 110122, China
| | - Bin Fu
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, 77 Puhe Avenue, Shenbei New District, Shenyang, 110122, China
| | - Yong Long
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, 77 Puhe Avenue, Shenbei New District, Shenyang, 110122, China
| | - Zi-Yu Liu
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, 77 Puhe Avenue, Shenbei New District, Shenyang, 110122, China
| | - Xiao-Hong Tian
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, 77 Puhe Avenue, Shenbei New District, Shenyang, 110122, China
| |
Collapse
|
8
|
Cryobanking European Mink (Mustela lutreola) Mesenchymal Stem Cells and Oocytes. Int J Mol Sci 2022; 23:ijms23169319. [PMID: 36012583 PMCID: PMC9408899 DOI: 10.3390/ijms23169319] [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: 07/07/2022] [Revised: 08/01/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022] Open
Abstract
The European mink (Mustela lutreola) is one of Europe’s most endangered species, and it is on the brink of extinction in the Iberian Peninsula. The species’ precarious situation requires the application of new ex situ conservation methodologies that complement the existing ex situ and in situ conservation measures. Here, we report for the first time the establishment of a biobank for European mink mesenchymal stem cells (emMSC) and oocytes from specimens found dead in the Iberian Peninsula, either free or in captivity. New emMSC lines were isolated from different tissues: bone marrow (emBM-MSC), oral mucosa (emOM-MSc), dermal skin (emDS-MSC), oviduct (emO-MSc), endometrium (emE-MSC), testicular (emT-MSC), and adipose tissue from two different adipose depots: subcutaneous (emSCA-MSC) and ovarian (emOA-MSC). All eight emMSC lines showed plastic adhesion, a detectable expression of characteristic markers of MSCs, and, when cultured under osteogenic and adipogenic conditions, differentiation capacity to these lineages. Additionally, we were able to keep 227 Cumulus-oocyte complexes (COCs) in the biobank, 97 of which are grade I or II. The European mink MSC and oocyte biobank will allow for the conservation of the species’ genetic variability, the application of assisted reproduction techniques, and the development of in vitro models for studying the molecular mechanisms of infectious diseases that threaten the species’ precarious situation.
Collapse
|
9
|
Kay AG, Fox JM, Hewitson JP, Stone AP, Robertson S, James S, Wang XN, Kapasa E, Yang XB, Genever PG. CD317-Positive Immune Stromal Cells in Human "Mesenchymal Stem Cell" Populations. Front Immunol 2022; 13:903796. [PMID: 35734183 PMCID: PMC9207511 DOI: 10.3389/fimmu.2022.903796] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/04/2022] [Indexed: 12/31/2022] Open
Abstract
Heterogeneity of bone marrow mesenchymal stromal cells (MSCs, frequently referred to as "mesenchymal stem cells") clouds biological understanding and hampers their clinical development. In MSC cultures most commonly used in research and therapy, we have identified an MSC subtype characterized by CD317 expression (CD317pos (29.77 ± 3.00% of the total MSC population), comprising CD317dim (28.10 ± 4.60%) and CD317bright (1.67 ± 0.58%) MSCs) and a constitutive interferon signature linked to human disease. We demonstrate that CD317pos MSCs induced cutaneous tissue damage when applied a skin explant model of inflammation, whereas CD317neg MSCs had no effect. Only CD317neg MSCs were able to suppress proliferative cycles of activated human T cells in vitro, whilst CD317pos MSCs increased polarization towards pro-inflammatory Th1 cells and CD317neg cell lines did not. Using an in vivo peritonitis model, we found that CD317neg and CD317pos MSCs suppressed leukocyte recruitment but only CD317neg MSCs suppressed macrophage numbers. Using MSC-loaded scaffolds implanted subcutaneously in immunocompromised mice we were able to observe tissue generation and blood vessel formation with CD317neg MSC lines, but not CD317pos MSC lines. Our evidence is consistent with the identification of an immune stromal cell, which is likely to contribute to specific physiological and pathological functions and influence clinical outcome of therapeutic MSCs.
Collapse
Affiliation(s)
- Alasdair G. Kay
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom,*Correspondence: Paul G. Genever, ; Alasdair G. Kay,
| | - James M. Fox
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - James P. Hewitson
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - Andrew P. Stone
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - Sophie Robertson
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - Sally James
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - Xiao-nong Wang
- Translational and Clinical Research Institute, Newcastle University, Newcastle, United Kingdom
| | - Elizabeth Kapasa
- Department of Oral Biology, School of Dentistry, University of Leeds, St James’s University Hospital, Leeds, United Kingdom
| | - Xuebin B. Yang
- Department of Oral Biology, School of Dentistry, University of Leeds, St James’s University Hospital, Leeds, United Kingdom
| | - Paul G. Genever
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom,*Correspondence: Paul G. Genever, ; Alasdair G. Kay,
| |
Collapse
|
10
|
Rubina KA, Semina EV, Kalinina NI, Sysoeva VY, Balatskiy AV, Tkachuk VA. Revisiting the multiple roles of T-cadherin in health and disease. Eur J Cell Biol 2021; 100:151183. [PMID: 34798557 DOI: 10.1016/j.ejcb.2021.151183] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/08/2021] [Accepted: 11/08/2021] [Indexed: 01/02/2023] Open
Abstract
As a non-canonical member of cadherin superfamily, T-cadherin was initially described as a molecule involved in homophilic recognition in the nervous and vascular systems. The ensuing decades clearly demonstrated that T-cadherin is a remarkably multifunctional molecule. It was validated as a bona fide receptor for both: LDL exerting adverse atherogenic action and adiponectin mediating many protective metabolic and cardiovascular effects. Motivated by the latest progress and accumulated data unmasking important roles of T-cadherin in blood vessel function and tissue regeneration, here we revisit the original function of T-cadherin as a guidance receptor for the growing axons and blood vessels, consider the recent data on T-cadherin-induced exosomes' biogenesis and their role in myocardial regeneration and revascularization. The review expands upon T-cadherin contribution to mesenchymal stem/stromal cell compartment in adipose tissue. We also dwell upon T-cadherin polymorphisms (SNP) and their possible therapeutic applications. Furthermore, we scrutinize the molecular hub of insulin and adiponectin receptors (AdipoR1 and AdipoR2) conveying signals to their downstream targets in quest for defining a putative place of T-cadherin in this molecular circuitry.
Collapse
Affiliation(s)
- K A Rubina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia.
| | - E V Semina
- Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - N I Kalinina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - V Yu Sysoeva
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - A V Balatskiy
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - V A Tkachuk
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| |
Collapse
|
11
|
Shklyaev SS, Melnichenko GA, Volevodz NN, Falaleeva NA, Ivanov SA, Kaprin AD, Mokrysheva NG. Adiponectin: a pleiotropic hormone with multifaceted roles. PROBLEMY ENDOKRINOLOGII 2021; 67:98-112. [PMID: 35018766 PMCID: PMC9753852 DOI: 10.14341/probl12827] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 10/22/2021] [Indexed: 05/28/2023]
Abstract
Adipose tissue mostly composed of different types of fat is one of the largest endocrine organs in the body playing multiple intricate roles including but not limited to energy storage, metabolic homeostasis, generation of heat, participation in immune functions and secretion of a number of biologically active factors known as adipokines. The most abundant of them is adiponectin. This adipocite-derived hormone exerts pleiotropic actions and exhibits insulin-sensitizing, antidiabetic, anti-obesogenic, anti-inflammatory, antiatherogenic, cardio- and neuroprotective properties. Contrariwise to its protective effects against various pathological events in different cell types, adiponectin may have links to several systemic diseases and malignances. Reduction in adiponectin levels has an implication in COVID-19-associated respiratory failure, which is attributed mainly to a phenomenon called 'adiponectin paradox'. Ample evidence about multiple functions of adiponectin in the body was obtained from animal, mostly rodent studies. Our succinct review is entirely about multifaceted roles of adiponectin and mechanisms of its action in different physiological and pathological states.
Collapse
Affiliation(s)
- S. S. Shklyaev
- National Research Center for Endocrinology of the Ministry of Health of the Russian Federation;
A. Tsyb Medical Radiological Research Center — Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation
| | - G. A. Melnichenko
- National Research Center for Endocrinology of the Ministry of Health of the Russian Federatio
| | - N. N. Volevodz
- National Research Center for Endocrinology of the Ministry of Health of the Russian Federatio
| | - N. A. Falaleeva
- A. Tsyb Medical Radiological Research Center — Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation
| | - S. A. Ivanov
- A. Tsyb Medical Radiological Research Center — Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation
| | - A. D. Kaprin
- A. Tsyb Medical Radiological Research Center — Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation
| | - N. G. Mokrysheva
- National Research Center for Endocrinology of the Ministry of Health of the Russian Federation
| |
Collapse
|
12
|
Calcat-i-Cervera S, Sanz-Nogués C, O'Brien T. When Origin Matters: Properties of Mesenchymal Stromal Cells From Different Sources for Clinical Translation in Kidney Disease. Front Med (Lausanne) 2021; 8:728496. [PMID: 34616756 PMCID: PMC8488400 DOI: 10.3389/fmed.2021.728496] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022] Open
Abstract
Advanced therapy medicinal products (ATMPs) offer new prospects to improve the treatment of conditions with unmet medical needs. Kidney diseases are a current major health concern with an increasing global prevalence. Chronic renal failure appears after many years of impairment, which opens a temporary window to apply novel therapeutic approaches to delay or halt disease progression. The immunomodulatory, anti-inflammatory, and pro-regenerative properties of mesenchymal stromal cells (MSCs) have sparked interest for their use in cell-based regenerative therapies. Currently, several early-phase clinical trials have been completed and many are ongoing to explore MSC safety and efficacy in a wide range of nephropathies. However, one of the current roadblocks to the clinical translation of MSC therapies relates to the lack of standardization and harmonization of MSC manufacturing protocols, which currently hinders inter-study comparability. Studies have shown that cell culture processing variables can have significant effects on MSC phenotype and functionality, and these are highly variable across laboratories. In addition, heterogeneity within MSC populations is another obstacle. Furthermore, MSCs may be isolated from several sources which adds another variable to the comparative assessment of outcomes. There is now a growing body of literature highlighting unique and distinctive properties of MSCs according to the tissue origin, and that characteristics such as donor, age, sex and underlying medical conditions may alter the therapeutic effect of MSCs. These variables must be taken into consideration when developing a cell therapy product. Having an optimal scale-up strategy for MSC manufacturing is critical for ensuring product quality while minimizing costs and time of production, as well as avoiding potential risks. Ideally, optimal scale-up strategies must be carefully considered and identified during the early stages of development, as making changes later in the bioprocess workflow will require re-optimization and validation, which may have a significant long-term impact on the cost of the therapy. This article provides a summary of important cell culture processing variables to consider in the scale-up of MSC manufacturing as well as giving a comprehensive review of tissue of origin-specific biological characteristics of MSCs and their use in current clinical trials in a range of renal pathologies.
Collapse
Affiliation(s)
| | | | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), CÚRAM, Biomedical Science Building, National University of Ireland, Galway, Ireland
| |
Collapse
|
13
|
Lim KM, Dayem AA, Choi Y, Lee Y, An J, Gil M, Lee S, Kwak HJ, Vellingirl B, Shin HJ, Cho SG. High Therapeutic and Esthetic Properties of Extracellular Vesicles Produced from the Stem Cells and Their Spheroids Cultured from Ocular Surgery-Derived Waste Orbicularis Oculi Muscle Tissues. Antioxidants (Basel) 2021; 10:antiox10081292. [PMID: 34439540 PMCID: PMC8389225 DOI: 10.3390/antiox10081292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are paracrine factors that mediate stem cell therapeutics. We aimed at evaluating the possible therapeutic and esthetic applications of EVs prepared from the waste human facial tissue-derived orbicularis oculi muscle stem cells (OOM-SCs). OOM-SCs were isolated from the ocular tissues (from elders and youngsters) after upper eyelid blepharoplasty or epiblepharon surgeries. EVs were prepared from the OOM-SCs (OOM-SC-EVs) and their three-dimensional spheroids. OOM-SCs showed a spindle-like morphology with trilineage differentiation capacity, positive expression of CD105, CD 90, and CD73, and negative expression of CD45 and CD34, and their stem cell properties were compared with other adult mesenchymal stem cells. OOM-SC-EVs showed a high inhibitory effect on melanin synthesis in B16F10 cells by blocking tyrosinase activity. OOM-SC-EVs treatment led to a significant attenuation of senescence-associated changes, a decrease in reactive oxygen species generation, and an upregulation of antioxidant genes. We demonstrated the regeneration activity of OOM-SC-EVs in in vitro wound healing of normal human dermal fibroblasts and upregulation of anti-wrinkle-related genes and confirmed the therapeutic potential of OOM-SC-EVs in the healing of the in vivo wound model. Our study provides promising therapeutic and esthetic applications of OOM-SC-EVs, which can be obtained from the ocular surgery-derived waste human facial tissues.
Collapse
Affiliation(s)
- Kyung Min Lim
- Molecular & Cellular Reprogramming Center (MCRC), Department of Stem Cell & Regenerative Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.M.L.); (A.A.D.); (Y.C.); (Y.L.); (J.A.); (M.G.); (S.L.); (H.J.K.)
| | - Ahmed Abdal Dayem
- Molecular & Cellular Reprogramming Center (MCRC), Department of Stem Cell & Regenerative Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.M.L.); (A.A.D.); (Y.C.); (Y.L.); (J.A.); (M.G.); (S.L.); (H.J.K.)
| | - Yujin Choi
- Molecular & Cellular Reprogramming Center (MCRC), Department of Stem Cell & Regenerative Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.M.L.); (A.A.D.); (Y.C.); (Y.L.); (J.A.); (M.G.); (S.L.); (H.J.K.)
| | - Yoonjoo Lee
- Molecular & Cellular Reprogramming Center (MCRC), Department of Stem Cell & Regenerative Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.M.L.); (A.A.D.); (Y.C.); (Y.L.); (J.A.); (M.G.); (S.L.); (H.J.K.)
| | - Jongyub An
- Molecular & Cellular Reprogramming Center (MCRC), Department of Stem Cell & Regenerative Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.M.L.); (A.A.D.); (Y.C.); (Y.L.); (J.A.); (M.G.); (S.L.); (H.J.K.)
| | - Minchan Gil
- Molecular & Cellular Reprogramming Center (MCRC), Department of Stem Cell & Regenerative Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.M.L.); (A.A.D.); (Y.C.); (Y.L.); (J.A.); (M.G.); (S.L.); (H.J.K.)
| | - Soobin Lee
- Molecular & Cellular Reprogramming Center (MCRC), Department of Stem Cell & Regenerative Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.M.L.); (A.A.D.); (Y.C.); (Y.L.); (J.A.); (M.G.); (S.L.); (H.J.K.)
| | - Hee Jeong Kwak
- Molecular & Cellular Reprogramming Center (MCRC), Department of Stem Cell & Regenerative Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.M.L.); (A.A.D.); (Y.C.); (Y.L.); (J.A.); (M.G.); (S.L.); (H.J.K.)
| | - Balachandar Vellingirl
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641-046, India;
| | - Hyun Jin Shin
- Department of Ophthalmology, Research Institute of Medical Science, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Korea
- Correspondence: (H.J.S.); (S.-G.C.)
| | - Ssang-Goo Cho
- Molecular & Cellular Reprogramming Center (MCRC), Department of Stem Cell & Regenerative Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.M.L.); (A.A.D.); (Y.C.); (Y.L.); (J.A.); (M.G.); (S.L.); (H.J.K.)
- Correspondence: (H.J.S.); (S.-G.C.)
| |
Collapse
|
14
|
Kita S, Shimomura I. Stimulation of exosome biogenesis by adiponectin, a circulating factor secreted from adipocytes. J Biochem 2021; 169:173-179. [PMID: 32979268 DOI: 10.1093/jb/mvaa105] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/25/2020] [Indexed: 01/08/2023] Open
Abstract
Adiponectin is an adipocyte-derived circulating factor that protects various organs and tissues. Such a pleiotropic action mechanism has not yet been fully explained. Clinically important multimer adiponectin existing in serum bound to cells expressing T-cadherin, a glycosylphosphatidylinositol-anchored cadherin, but not to the cells expressing other known receptors, AdipoRs or calreticulin. Adiponectin bound to the cell-surface, accumulated inside of multivesicular bodies through T-cadherin, and increased exosome biogenesis and secretion from the cells. Such increased exosome production accompanied the reduction of cellular ceramides in endothelial cells and mouse aorta, and enhanced skeletal muscle regeneration. Significantly lower plasma exosome levels were found in mice genetically deficient in either adiponectin or T-cadherin. Therapeutic effects of mesenchymal stem cells (MSCs) for a pressure overload-induced heart failure in mice required the presence of adiponectin in plasma, T-cadherin expression and exosome biogenesis in MSCs themselves, accompanying an increase of plasma exosomes. Essentially all organs seem to have MSCs and/or their related somatic stem cells expressing T-cadherin. Our recent studies suggested the importance of exosome-stimulation by multimer adiponectin in its well-known pleiotropic organ protections.
Collapse
Affiliation(s)
- Shunbun Kita
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, 2-2 Suita, Osaka 565-0871, Japan.,Department of Adipose Management, Graduate School of Medicine, Osaka University, 2-2 Suita, Osaka 565-0871, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, 2-2 Suita, Osaka 565-0871, Japan
| |
Collapse
|
15
|
Okuchi Y, Reeves J, Ng SS, Doro DH, Junyent S, Liu KJ, El Haj AJ, Habib SJ. Wnt-modified materials mediate asymmetric stem cell division to direct human osteogenic tissue formation for bone repair. NATURE MATERIALS 2021; 20:108-118. [PMID: 32958876 DOI: 10.1038/s41563-020-0786-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 07/27/2020] [Indexed: 06/11/2023]
Abstract
The maintenance of human skeletal stem cells (hSSCs) and their progeny in bone defects is a major challenge. Here, we report on a transplantable bandage containing a three-dimensional Wnt-induced osteogenic tissue model (WIOTM). This bandage facilitates the long-term viability of hSSCs (8 weeks) and their progeny, and enables bone repair in an in vivo mouse model of critical-sized calvarial defects. The newly forming bone is structurally comparable to mature cortical bone and consists of human and murine cells. Furthermore, we show that the mechanism of WIOTM formation is governed by Wnt-mediated asymmetric cell division of hSSCs. Covalently immobilizing Wnts onto synthetic materials can polarize single dividing hSSCs, orient the spindle and simultaneously generate a Wnt-proximal hSSC and a differentiation-prone Wnt-distal cell. Our results provide insight into the regulation of human osteogenesis and represent a promising approach to deliver human osteogenic constructs that can survive in vivo and contribute to bone repair.
Collapse
Affiliation(s)
- Yoshihisa Okuchi
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - Joshua Reeves
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - Soon Seng Ng
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - Daniel H Doro
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Sergi Junyent
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - Karen J Liu
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Alicia J El Haj
- Healthcare Technology Institute, Institute of Translational Medicine, University of Birmingham, Birmingham, UK
| | - Shukry J Habib
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK.
| |
Collapse
|
16
|
Anastasio A, Gergues M, Lebhar MS, Rameshwar P, Fernandez-Moure J. Isolation and characterization of mesenchymal stem cells in orthopaedics and the emergence of compact bone mesenchymal stem cells as a promising surgical adjunct. World J Stem Cells 2020; 12:1341-1353. [PMID: 33312402 PMCID: PMC7705465 DOI: 10.4252/wjsc.v12.i11.1341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/26/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
The potential clinical and economic impact of mesenchymal stem cell (MSC) therapy is immense. MSCs act through multiple pathways: (1) as “trophic” cells, secreting various factors that are immunomodulatory, anti-inflammatory, anti-apoptotic, proangiogenic, proliferative, and chemoattractive; (2) in conjunction with cells native to the tissue they reside in to enhance differentiation of surrounding cells to facilitate tissue regrowth. Researchers have developed methods for the extraction and expansion of MSCs from animal and human tissues. While many sources of MSCs exist, including adipose tissue and iliac crest bone graft, compact bone (CB) MSCs have shown great potential for use in orthopaedic surgery. CB MSCs exert powerful immunomodulatory effects in addition to demonstrating excellent regenerative capacity for use in filling boney defects. CB MSCs have been shown to have enhanced response to hypoxic conditions when compared with other forms of MSCs. More work is needed to continue to characterize the potential applications for CB MSCs in orthopaedic trauma.
Collapse
Affiliation(s)
- Albert Anastasio
- Department of Orthopedic Surgery, Duke University Health System, Durham, NC 27710, United States
| | - Marina Gergues
- Department of Medicine, Hematology/Oncology, Rutgers University, New Jersey Medical School, Newark, NJ 07103, United States
| | - Michael S Lebhar
- School of Medicine, Duke University School of Medicine, Durham, NC 27710, United States
| | - Pranela Rameshwar
- Department of Medicine-Hematology/Oncology, Rutgers School of Biomedical Health Science, Newark, NJ 07103, United States
| | - Joseph Fernandez-Moure
- Department of Surgery, Division of Trauma, Acute, and Critical Care Surgery, Duke University School of Medicine, Durham, NC 27710, United States
| |
Collapse
|
17
|
Ravikumar M, Smith RAA, Nurcombe V, Cool SM. Heparan Sulfate Proteoglycans: Key Mediators of Stem Cell Function. Front Cell Dev Biol 2020; 8:581213. [PMID: 33330458 PMCID: PMC7710810 DOI: 10.3389/fcell.2020.581213] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are an evolutionarily ancient subclass of glycoproteins with exquisite structural complexity. They are ubiquitously expressed across tissues and have been found to exert a multitude of effects on cell behavior and the surrounding microenvironment. Evidence has shown that heterogeneity in HSPG composition is crucial to its functions as an essential scaffolding component in the extracellular matrix as well as a vital cell surface signaling co-receptor. Here, we provide an overview of the significance of HSPGs as essential regulators of stem cell function. We discuss the various roles of HSPGs in distinct stem cell types during key physiological events, from development through to tissue homeostasis and regeneration. The contribution of aberrant HSPG production to altered stem cell properties and dysregulated cellular homeostasis characteristic of cancer is also reviewed. Finally, we consider approaches to better understand and exploit the multifaceted functions of HSPGs in influencing stem cell characteristics for cell therapy and associated culture expansion strategies.
Collapse
Affiliation(s)
- Maanasa Ravikumar
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Raymond Alexander Alfred Smith
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Victor Nurcombe
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, Singapore, Singapore
| | - Simon M Cool
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
18
|
Chen W, Zhang Z, Chang C, Yang Z, Wang P, Fu H, Wei X, Chen E, Tan S, Huang W, Sun L, Ni T, Yang Y, Wang Y. A bioenergetic shift is required for spermatogonial differentiation. Cell Discov 2020; 6:56. [PMID: 32864161 PMCID: PMC7431567 DOI: 10.1038/s41421-020-0183-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/22/2020] [Indexed: 12/22/2022] Open
Abstract
A bioenergetic balance between glycolysis and mitochondrial respiration is particularly important for stem cell fate specification. It however remains to be determined whether undifferentiated spermatogonia switch their preference for bioenergy production during differentiation. In this study, we found that ATP generation in spermatogonia was gradually increased upon retinoic acid (RA)-induced differentiation. To accommodate this elevated energy demand, RA signaling concomitantly switched ATP production in spermatogonia from glycolysis to mitochondrial respiration, accompanied by increased levels of reactive oxygen species. Disrupting mitochondrial respiration significantly blocked spermatogonial differentiation. Inhibition of glucose conversion to glucose-6-phosphate or pentose phosphate pathway also repressed the formation of c-Kit+ differentiating germ cells, suggesting that metabolites produced from glycolysis are required for spermatogonial differentiation. We further demonstrated that the expression levels of several metabolic regulators and enzymes were significantly altered upon RA-induced differentiation, with both RNA-seq and quantitative proteomic analyses. Taken together, our data unveil a critically regulated bioenergetic balance between glycolysis and mitochondrial respiration that is required for spermatogonial proliferation and differentiation.
Collapse
Affiliation(s)
- Wei Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241 China
| | - Zhaoran Zhang
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824 USA
| | - Chingwen Chang
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824 USA
| | - Zhichang Yang
- Department of Chemistry, College of Natural Science, Michigan State University, East Lansing, MI 48824 USA
| | - Pengxiang Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241 China
| | - Haihui Fu
- State Key Laboratory of Genetic Engineering & MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Xiao Wei
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241 China
| | - Eric Chen
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824 USA
| | - Suxu Tan
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824 USA
| | - Wen Huang
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824 USA
| | - Liangliang Sun
- Department of Chemistry, College of Natural Science, Michigan State University, East Lansing, MI 48824 USA
| | - Ting Ni
- State Key Laboratory of Genetic Engineering & MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Yi Yang
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237 China
| | - Yuan Wang
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824 USA
| |
Collapse
|
19
|
Adiponectin Stimulates Exosome Release to Enhance Mesenchymal Stem-Cell-Driven Therapy of Heart Failure in Mice. Mol Ther 2020; 28:2203-2219. [PMID: 32652045 PMCID: PMC7351027 DOI: 10.1016/j.ymthe.2020.06.026] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/18/2020] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are cultured adult stem cells that originally reside in virtually all tissues, and the gain of MSCs by transplantation has become the leading form of cell therapy in various diseases. However, there is limited knowledge on the alteration of its efficacy by factors in recipients. Here, we report that the cardioprotective properties of intravenously injected MSCs in a mouse model of pressure-overload heart failure largely depend on circulating adiponectin, an adipocyte-secreted factor. The injected MSCs exert their function through exosomes, extracellular vesicles of endosome origin. Adiponectin stimulated exosome biogenesis and secretion through binding to T-cadherin, a unique glycosylphosphatidylinositol-anchored cadherin, on MSCs. A pharmacological or adenovirus-mediated genetic increase in plasma adiponectin enhanced the therapeutic efficacy of MSCs. Our findings provide novel insights into the importance of adiponectin in mesenchymal-progenitor-mediated organ protections.
Collapse
|
20
|
Gomzikova MO, Aimaletdinov AM, Bondar OV, Starostina IG, Gorshkova NV, Neustroeva OA, Kletukhina SK, Kurbangaleeva SV, Vorobev VV, Garanina EE, Persson JL, Jeyapalan J, Mongan NP, Khaiboullina SF, Rizvanov AA. Immunosuppressive properties of cytochalasin B-induced membrane vesicles of mesenchymal stem cells: comparing with extracellular vesicles derived from mesenchymal stem cells. Sci Rep 2020; 10:10740. [PMID: 32612100 PMCID: PMC7330035 DOI: 10.1038/s41598-020-67563-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/05/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles derived from mesenchymal stem cells (MSCs) represent a novel approach for regenerative and immunosuppressive therapy. Recently, cytochalasin B-induced microvesicles (CIMVs) were shown to be effective drug delivery mediators. However, little is known about their immunological properties. We propose that the immunophenotype and molecular composition of these vesicles could contribute to the therapeutic efficacy of CIMVs. To address this issue, CIMVs were generated from murine MSC (CIMVs-MSCs) and their cytokine content and surface marker expression determined. For the first time, we show that CIMVs-MSCs retain parental MSCs phenotype (Sca-1+, CD49e+, CD44+, CD45−). Also, CIMVs-MSCs contained a cytokine repertoire reflective of the parental MSCs, including IL-1β, IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, IL-12(p40), IL-13, IL-17, CCL2, CCL3, CCL4, CCL5, CCL11, G-CSF, GM-CSF and TNF-α. Next, we evaluated the immune-modulating properties of CIMVs-MSCs in vivo using standard preclinical tests. MSCs and CIMVs-MSCs reduced serum levels of anti-sheep red blood cell antibody and have limited effects on neutrophil and peritoneal macrophage activity. We compared the immunomodulatory effect of MSCs, CIMVs and EVs. We observed no immunosuppression in mice pretreated with natural EVs, whereas MSCs and CIMVs-MSCs suppressed antibody production in vivo. Additionally, we have investigated the biodistribution of CIMVs-MSCs in vivo and demonstrated that CIMVs-MSCs localized in liver, lung, brain, heart, spleen and kidneys 48 h after intravenous injection and can be detected 14 days after subcutaneous and intramuscular injection. Collectively our data demonstrates immunomodulatory efficacy of CIMVs and supports their further preclinical testing as an effective therapeutic delivery modality.
Collapse
Affiliation(s)
- M O Gomzikova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia, 420008. .,M.M. Shemyakin-Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia, 117997.
| | - A M Aimaletdinov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia, 420008
| | - O V Bondar
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia, 420008
| | - I G Starostina
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia, 420008
| | - N V Gorshkova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia, 420008
| | - O A Neustroeva
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia, 420008
| | - S K Kletukhina
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia, 420008
| | - S V Kurbangaleeva
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia, 420008
| | - V V Vorobev
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia, 420008
| | - E E Garanina
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia, 420008
| | - J L Persson
- Department of Translational Medicine, Lund University, 205 02, Malmö, Sweden.,Department of Molecular Biology, Umeå University, Umeå, 901 87, USA
| | - J Jeyapalan
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, LE12 5RD, UK
| | - N P Mongan
- Department of Translational Medicine, Lund University, 205 02, Malmö, Sweden.,Department of Pharmacology, Weill Cornell Medicine, 1300 York Ave., New York, NY, 10065, USA
| | - S F Khaiboullina
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia, 420008.,Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, NV, USA
| | - A A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia, 420008. .,M.M. Shemyakin-Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia, 117997.
| |
Collapse
|
21
|
Primary cilia-dependent signaling is involved in regulating mesenchymal stem cell proliferation and pluripotency maintenance. J Mol Histol 2020; 51:241-250. [PMID: 32399704 PMCID: PMC7253378 DOI: 10.1007/s10735-020-09876-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022]
Abstract
Using a large-scale quantitative mesenchymal stem cells (MSCs) membrane proteomics analysis, we identified a large group of ciliary proteins in the MSCs membrane fraction, which prompted us to examine the cilia, intricate organelles that were originally discovered approximately 100 years ago. Here we characterize their primary structure and function in MSCs. We first characterized the primary cilia on undifferentiated human MSCs by immunostaining and verified these observation with scanning and 3D electronic microscopy. To investigate the function of the primary cilia of the MSCs we induced loss of function by means of siRNA knockdown (targeted to two known ciliary proteins: IFT172 and KIF3A). After either of these two proteins was knocked down by the respective siRNA, the MSCs showed fewer and shortened primary cilia. The MSCs proliferation assays showed increased cell proliferative activity under confluent conditions after the siRNA knockdown of IFT172 or KIF3A; among these MSCs, the proportion in S phase was increased in the IFT172 siRNA knockdown group. The expression of stem cell markers on the MSCs, namely, Oct4, Nanog and Sox2, were downregulated after the siRNA-induced knockdown of IFT172 or KIF3A, and the gene expression upregulation of ectoderm lineage markers was notable. Furthermore, manipulation of the primary cilia-dependent Shh pathway, using the Shh activator SAG (smoothened agonist), upregulated the gene expression of pluripotency markers, including Nanog and Oct4, and transcriptional target genes in the Shh pathway. This study confirms that MSCs have primary cilia and provides evidence that primary cilia-dependent signaling pathways play functional roles in MSCs proliferation and stemness maintenance.
Collapse
|
22
|
Brinkhof B, Zhang B, Cui Z, Ye H, Wang H. ALCAM (CD166) as a gene expression marker for human mesenchymal stromal cell characterisation. Gene X 2020; 763S:100031. [PMID: 32550557 PMCID: PMC7285916 DOI: 10.1016/j.gene.2020.100031] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/08/2020] [Indexed: 02/06/2023] Open
Abstract
Background Human mesenchymal stromal cells (MSCs) phenotypically share their positive expression of the International Society for Cell and Gene Therapy (ISCT) markers CD73, CD90 and CD105 with fibroblasts. Fibroblasts are often co-isolated as an unwanted by-product from biopsy and they can rapidly overgrow the MSCs in culture. Indeed, many other surface markers have been proposed, though no unique MSC specific marker has been identified yet. Quantitative PCR (qPCR) is a precise, efficient and rapid method for gene expression analysis. To identify a marker suitable for accurate MSC characterisation, qPCR was exploited. Methods and results Two commercially obtained bone marrow (BM) derived MSCs and an hTERT immortalised BM-MSC line (MSC-TERT) have been cultured for different days and at different oxygen levels before RNA extraction. Together with RNA samples previous extracted from umbilical cord derived MSCs and MSC-TERT cells cultured in 2D or 3D, this heterogeneous sample set was quantitatively analysed for the expression levels of 18 candidate MSC marker genes. The expression levels in MSCs were compared with the expression levels in fibroblasts to verify the differentiation capability of these genes between MSCs and fibroblasts. None of the ISCT markers could differentiate between fibroblasts and MSCs. A total of six other genes (ALCAM, CLIC1, EDIL3, EPHA2, NECTIN2, and TMEM47) were identified as possible biomarkers for accurate identification of MSCs. Conclusion Justified by considerations on expression level, reliability and specificity, Activated-Leukocyte Cell Adhesion Molecule (ALCAM) was the best candidate for improving the biomarker set of MSC identification.
Collapse
Key Words
- (q)PCR, (quantitative) polymerase chain reaction
- AD, adipose
- AF, Amniotic Fluid
- ALCAM, Activated-Leukocyte Cell Adhesion Molecule
- Activated-leukocyte cell adhesion molecule
- BM, bone marrow
- BSG, Basigin
- Biomarker
- CD, cluster of differentiation
- CLIC1, chloride intracellular channel 1
- CLIC4, chloride intracellular channel 4
- Cq, Quantification cycle
- DF, Dermal Fibroblasts
- DP, Dental Pulp
- EDIL3, EGF like repeats and discoidin domains 3
- ENG, Endoglin
- EPHA2, EPH receptor A2
- ER, Endoplasmatic Reticulum
- FACS, Fluorescence Assisted Cell Sorting
- FN1, Fibronectin 1
- IGFBP7, insulin like growth factor binding protein 7
- ISCT, International Society for Cell and Gene Therapy
- ITGA1, integrin subunit alpha 1
- LAMP1, lysosomal associated membrane protein 1
- LRRC59, leucine rich repeat containing 59
- MCAM, melanoma cell adhesion molecule
- MM, Multiple Myeloma
- MPC, Mesenchymal Progenitor Cell
- MSC
- MSC, Mesenchymal Stromal Cells
- NECTIN2, nectin cell adhesion molecule 2
- NK, Natural Killer
- NT5E, 5′-nucleotidase ecto
- OS, Osteosarcoma
- PL, Placenta
- PPIA, peptidylprolyl isomerase A
- PUM1, pumilio RNA binding family member 1
- RM, Regenerative Medicine
- RNA
- RNA-seq, RNA sequencing
- RT, Reverse Transcriptase
- Regenerative medicine
- SEM, Standard Error of the Mean
- TBP, TATA-box binding protein
- TCF, Tissue Culture Plate
- TE, Tissue Engineering
- TFRC, transferrin receptor
- THY1, Thy-1 cell surface antigen
- TLN1, Talin 1
- TMEM47, transmembrane protein 47
- UC, umbilical cord
- YWHAZ, tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta
- cDNA, DNA complementary to RNA
- qPCR
Collapse
Affiliation(s)
- Bas Brinkhof
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Bo Zhang
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Hui Wang
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom.,Oxford Suzhou Centre for Advanced Research, Suzhou Industrial Park, Jiangsu 215123, China
| |
Collapse
|
23
|
Cooper TT, Sherman SE, Bell GI, Ma J, Kuljanin M, Jose SE, Lajoie GA, Hess DA. Characterization of a Vimentin high /Nestin high proteome and tissue regenerative secretome generated by human pancreas-derived mesenchymal stromal cells. Stem Cells 2020; 38:666-682. [PMID: 31904137 DOI: 10.1002/stem.3143] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/14/2019] [Indexed: 12/11/2022]
Abstract
Multipotent/mesenchymal stromal cells (MSCs) exist within a variety of postnatal tissues; however, global proteomic analyses comparing tissue-specific MSC are limited. Using human bone marrow (BM)-derived MSCs as a gold standard, we used label-free mass spectrometry and functional assays to characterize the proteome, secretome, and corresponding function of human pancreas-derived MSCs (Panc-MSCs) with a classical phenotype (CD90+/CD73+/CD105+/CD45-/CD31-). Both MSC subtypes expressed mesenchymal markers vimentin, α-SMA, and STRO-1; however, expression of nestin was increased in Panc-MSCs. Accordingly, these Vimentinhigh /Nestinhigh cells were isolated from fresh human pancreatic islet and non-islet tissues. Next, we identified expression of >60 CD markers shared between Panc-MSCs and BM-MSCs, including validated expression of CD14. An additional 19 CD markers were differentially expressed, including reduced pericyte-marker CD146 expression on Panc-MSCs. Panc-MSCs also showed reduced expression of proteins involved in lipid and retinoid metabolism. Accordingly, Panc-MSCs showed restricted responses to adipogenic stimuli in vitro, although both MSC types demonstrated trilineage differentiation. In contrast, Panc-MSCs demonstrated accelerated growth kinetics and competency to pro-neurogenic stimuli in vitro. The secretome of Panc-MSCs was highly enriched for proteins associated with vascular development, wound healing and chemotaxis. Similar to BM-MSCs, Panc-MSCs conditioned media augmented endothelial cell survival, proliferation, and tubule formation in vitro. Importantly, the secretome of both MSC types was capable of stimulating chemotactic infiltration of murine endothelial cells in vivo and reduced hyperglycemia in STZ-treated mice following intrapancreatic injection. Overall, this study provides foundational knowledge to develop Panc-MSCs as a unique MSC subtype with functional properties beneficial in regenerative medicine for diabetes and vascular disease.
Collapse
Affiliation(s)
- Tyler T Cooper
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada.,Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada.,Department of Biochemistry, Don Rix Protein Identification Facility, Western University, London, Ontario, Canada
| | - Stephen E Sherman
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada.,Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada
| | - Gillian I Bell
- Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada
| | - Jun Ma
- Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada.,Department of Biochemistry, Don Rix Protein Identification Facility, Western University, London, Ontario, Canada
| | - Miljan Kuljanin
- Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada.,Department of Biochemistry, Don Rix Protein Identification Facility, Western University, London, Ontario, Canada
| | - Shauna E Jose
- Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada
| | - Gilles A Lajoie
- Department of Biochemistry, Don Rix Protein Identification Facility, Western University, London, Ontario, Canada
| | - David A Hess
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada.,Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada
| |
Collapse
|
24
|
Pearson S, Guo B, Pierce A, Azadbakht N, Brazzatti JA, Patassini S, Mulero-Navarro S, Meyer S, Flotho C, Gelb BD, Whetton AD. Proteomic Analysis of an Induced Pluripotent Stem Cell Model Reveals Strategies to Treat Juvenile Myelomonocytic Leukemia. J Proteome Res 2020; 19:194-203. [PMID: 31657576 PMCID: PMC6942217 DOI: 10.1021/acs.jproteome.9b00495] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
![]()
Juvenile
myelomonocytic leukemia (JMML) is an aggressive myeloproliferative
neoplasm of early childhood with a poor survival rate, thus there
is a requirement for improved treatment strategies. Induced pluripotent
stem cells offer the ability to model disease and develop new treatment
strategies. JMML is frequently associated with mutations in PTPN11. Children with Noonan syndrome, a development disorder,
have an increased incidence of JMML associated with specific germline
mutations in PTPN11. We undertook a proteomic assessment
of myeloid cells derived from induced pluripotent stem cells obtained
from Noonan syndrome patients with PTPN11 mutations,
either associated or not associated with an increased incidence of
JMML. We report that the proteomic perturbations induced by the leukemia-associated PTPN11 mutations are associated with TP53 and NF-Kκb
signaling. We have previously shown that MYC is involved in the differential
gene expression observed in Noonan syndrome patients associated with
an increased incidence of JMML. Thus, we employed drugs to target
these pathways and demonstrate differential effects on clonogenic
hematopoietic cells derived from Noonan syndrome patients, who develop
JMML and those who do not. Further, we demonstrated these small molecular
inhibitors, JQ1 and CBL0137, preferentially extinguish primitive hematopoietic
cells from sporadic JMML patients as opposed to cells from healthy
individuals.
Collapse
Affiliation(s)
- Stella Pearson
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre , The University of Manchester, Wolfson Molecular Imaging Centre , 27 Palatine Road , Withington, Manchester M20 3LJ , U.K
| | - Baoqiang Guo
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre , The University of Manchester, Wolfson Molecular Imaging Centre , 27 Palatine Road , Withington, Manchester M20 3LJ , U.K
| | - Andrew Pierce
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre , The University of Manchester, Wolfson Molecular Imaging Centre , 27 Palatine Road , Withington, Manchester M20 3LJ , U.K
| | - Narges Azadbakht
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre , The University of Manchester, Wolfson Molecular Imaging Centre , 27 Palatine Road , Withington, Manchester M20 3LJ , U.K
| | - Julie A Brazzatti
- Stoller Biomarker Discovery Centre, Manchester Academic Health Science Centre , University of Manchester , Manchester M13 9NQ , U.K
| | - Stefano Patassini
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre , The University of Manchester, Wolfson Molecular Imaging Centre , 27 Palatine Road , Withington, Manchester M20 3LJ , U.K
| | | | - Stefan Meyer
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre , The University of Manchester, Wolfson Molecular Imaging Centre , 27 Palatine Road , Withington, Manchester M20 3LJ , U.K
| | - Christian Flotho
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine , University of Freiburg , 79106 Freiburg , Germany
| | - Bruce D Gelb
- The Mindich Child Health and Development Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Anthony D Whetton
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre , The University of Manchester, Wolfson Molecular Imaging Centre , 27 Palatine Road , Withington, Manchester M20 3LJ , U.K.,Stoller Biomarker Discovery Centre, Manchester Academic Health Science Centre , University of Manchester , Manchester M13 9NQ , U.K
| |
Collapse
|
25
|
Molecular phenotyping of the surfaceome of migratory chondroprogenitors and mesenchymal stem cells using biotinylation, glycocapture and quantitative LC-MS/MS proteomic analysis. Sci Rep 2019; 9:9018. [PMID: 31227739 PMCID: PMC6588563 DOI: 10.1038/s41598-019-44957-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 04/24/2019] [Indexed: 12/16/2022] Open
Abstract
The complement of cell surface proteins, collectively referred to as the surfaceome, is a useful indicator of normal differentiation processes, and the development of pathologies such as osteoarthritis (OA). We employed biochemical and proteomic tools to explore the surfaceome and to define biomarkers in chondrogenic progenitor cells (CPC) derived from human OA knee articular cartilage. These cells have great therapeutic potential, but their unexplored biology limits their clinical application. We performed biotinylation combined with glycocapture and high throughput shotgun proteomics to define the surface proteome of human bone marrow mesenchymal stem cells (MSCs) and human CPCs. We prepared cell surface protein-enriched fractions from MSCs and CPCs, and then a proteomic approach was used to compare and evaluate protein changes between undifferentiated MSCs and CPCs. 1256 proteins were identified in the study, of which 791 (63%) were plasma membrane, cell surface or extracellular matrix proteins. Proteins constituting the surfaceome were annotated and categorized. Our results provide, for the first time, a repository of quantitative proteomic data on the surfaceome of two closely related cell types relevant to cartilage biology and OA. These results may provide novel insights into the transformation of the surfaceome during chondrogenic differentiation and phenotypic changes during OA development.
Collapse
|
26
|
Wiese DM, Ruttan CC, Wood CA, Ford BN, Braid LR. Accumulating Transcriptome Drift Precedes Cell Aging in Human Umbilical Cord-Derived Mesenchymal Stromal Cells Serially Cultured to Replicative Senescence. Stem Cells Transl Med 2019; 8:945-958. [PMID: 30924318 DOI: 10.1002/sctm.18-0246] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 01/22/2019] [Indexed: 12/13/2022] Open
Abstract
In preclinical studies, mesenchymal stromal cells (MSCs) exhibit robust potential for numerous applications. To capitalize on these benefits, cell manufacturing and delivery protocols have been scaled up to facilitate clinical trials without adequately addressing the impact of these processes on cell utility nor inevitable regulatory requirements for consistency. Growing evidence indicates that culture-aged MSCs, expanded to the limits of replicative exhaustion to generate human doses, are not equivalent to early passage cells, and their use may underpin reportedly underwhelming or inconsistent clinical outcomes. Here, we sought to define the maximum expansion boundaries for human umbilical cord-derived MSCs, cultured in chemically defined xeno- and serum-free media, that yield consistent cell batches comparable to early passage cells. Two male and two female donor populations, recovered from cryostorage at mean population doubling level (mPDL) 10, were serially cultivated until replicative exhaustion (senescence). At each passage, growth kinetics, cell morphology, and transcriptome profiles were analyzed. All MSC populations displayed comparable growth trajectories through passage 9 (P9; mPDL 45) and variably approached senescence after P10 (mPDL 49). Transcription profiles of 14,500 human genes, generated by microarray, revealed a nonlinear evolution of culture-adapted MSCs. Significant expression changes occurred only after P5 (mPDL 27) and accumulated rapidly after P9 (mPDL 45), preceding other cell aging metrics. We report that cryobanked umbilical cord-derived MSCs can be reliably expanded to clinical human doses by P4 (mPDL 23), before significant transcriptome drift, and thus represent a mesenchymal cell source suited for clinical translation of cellular therapies. Stem Cells Translational Medicine 2019;8:945&958.
Collapse
Affiliation(s)
| | | | | | - Barry N Ford
- Casualty Management Section, DRDC Suffield Research Centre, Medicine Hat, Alberta, Canada
| | | |
Collapse
|
27
|
Trivanović D, Drvenica I, Kukolj T, Obradović H, Okić Djordjević I, Mojsilović S, Krstić J, Bugarski B, Jauković A, Bugarski D. Adipoinductive effect of extracellular matrix involves cytoskeleton changes and SIRT1 activity in adipose tissue stem/stromal cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S370-S382. [PMID: 30198336 DOI: 10.1080/21691401.2018.1494183] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Adipose tissue (AT) homeostasis and expansion are dependent on complex crosstalk between resident adipose stromal/stem cells (ASCs) and AT extracellular matrix (ECM). Although adipose tissue ECM (atECM) is one of the key players in the stem cell niche, data on bidirectional interaction of ASCs and atECM are still scarce. Here, we investigated how atECM guides ASCs' differentiation. atECM altered shape and cytoskeleton organization of ASCs without changing their proliferation, β-galactosidase activity and adhesion. Cytoskeleton modifications occurred due to fostered parallel organization of F-actin and elevated expression of Vimentin in ASCs. After seven-day cultivation, atECM impaired osteogenesis of ASCs, simultaneously decreasing expression of Runx2. In addition, atECM accelerated early adipogenesis concomitantly with altered Vimentin organization in ASCs, slightly increasing PPARγ, while elevated Adiponectin and Vimentin mRNA expression. Early adipogenesis triggered by atECM was followed by upregulated mitochondrial activity and Sirtuin 1 (SIRT1) expression in ASCs. Proadipogenic events induced by atECM were mediated by SIRT1, indicating the supportive role of atECM in adipogenesis-related metabolic state of ASCs. These results provide a closer look at the effects of atECM on ASC physiology and may support the advancement of engineering design in soft tissue reconstruction and fundamental research of AT.
Collapse
Affiliation(s)
- Drenka Trivanović
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Ivana Drvenica
- b Laboratory for Immunology , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Tamara Kukolj
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Hristina Obradović
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Ivana Okić Djordjević
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Slavko Mojsilović
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Jelena Krstić
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Branko Bugarski
- c Department of Chemical Engineering, Faculty of Technology and Metallurgy , University of Belgrade , Belgrade , Serbia
| | - Aleksandra Jauković
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| | - Diana Bugarski
- a Laboratory for Experimental Hematology and Stem Cells , Institute for Medical Research, University of Belgrade , Belgrade , Serbia
| |
Collapse
|
28
|
Hayes AJ, Smith SM, Caterson B, Melrose J. Concise Review: Stem/Progenitor Cell Proteoglycans Decorated with 7-D-4, 4-C-3, and 3-B-3(-) Chondroitin Sulfate Motifs Are Morphogenetic Markers of Tissue Development. Stem Cells 2018; 36:1475-1486. [PMID: 29893019 PMCID: PMC6381390 DOI: 10.1002/stem.2860] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/02/2018] [Accepted: 05/11/2018] [Indexed: 01/02/2023]
Abstract
This study reviewed the occurrence of chondroitin sulfate (CS) motifs 4-C-3, 7-D-4, and 3-B-3(-), which are expressed by progenitor cells in tissues undergoing morphogenesis. These motifs have a transient early expression pattern during tissue development and also appear in mature tissues during pathological remodeling and attempted repair processes by activated adult stem cells. The CS motifs are information and recognition modules, which may regulate cellular behavior and delineate stem cell niches in developmental tissues. One of the difficulties in determining the precise role of stem cells in tissue development and repair processes is their short engraftment period and the lack of specific markers, which differentiate the activated stem cell lineages from the resident cells. The CS sulfation motifs 7-D-4, 4-C-3, and 3-B-3 (-) decorate cell surface proteoglycans on activated stem/progenitor cells and appear to identify these cells in transitional areas of tissue development and in tissue repair and may be applicable to determining a more precise role for stem cells in tissue morphogenesis. Stem Cells 2018;36:1475-1486.
Collapse
Affiliation(s)
- Anthony J Hayes
- Bioimaging Research Hub, Cardiff School of Biosciences, Cardiff University, Cardiff, Wales, United Kingdom
| | - Susan M Smith
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Royal North Shore Hospital and University of Sydney, St. Leonards, New South Wales, Australia
| | - Bruce Caterson
- School of Biosciences, Cardiff University, Cardiff, Wales, United Kingdom
| | - James Melrose
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Royal North Shore Hospital and University of Sydney, St. Leonards, New South Wales, Australia.,Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
29
|
Colunga T, Dalton S. Building Blood Vessels with Vascular Progenitor Cells. Trends Mol Med 2018; 24:630-641. [PMID: 29802036 PMCID: PMC6050017 DOI: 10.1016/j.molmed.2018.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022]
Abstract
Vascular progenitor cells have been identified from perivascular cell fractions and peripheral blood and bone marrow mononuclear fractions. These vascular progenitors share the ability to generate some of the vascular lineages, including endothelial cells, smooth muscle cells, and pericytes. The potential therapeutic uses for vascular progenitor cells are broad and relate to stroke, ischemic disease, and to the engineering of whole organs and tissues that require a vascular component. This review summarizes the best-characterized sources of vascular progenitor cells and discusses advances in 3D printing and electrospinning using blended polymers for the creation of biomimetic vascular grafts. These advances are pushing the field of regenerative medicine closer to the creation of small-diameter vascular grafts with long-term clinical utility.
Collapse
Affiliation(s)
- Thomas Colunga
- Center for Molecular Medicine, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA; Department of Biochemistry and Molecular Biology, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA
| | - Stephen Dalton
- Center for Molecular Medicine, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA; Department of Biochemistry and Molecular Biology, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA.
| |
Collapse
|
30
|
Baker LA, Rosa GJM, Hao Z, Piazza A, Hoffman C, Binversie EE, Sample SJ, Muir P. Multivariate genome-wide association analysis identifies novel and relevant variants associated with anterior cruciate ligament rupture risk in the dog model. BMC Genet 2018; 19:39. [PMID: 29940858 PMCID: PMC6019516 DOI: 10.1186/s12863-018-0626-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023] Open
Abstract
Background Anterior cruciate ligament rupture (ACLR) is a debilitating and potentially life-changing condition in humans, as there is a high prevalence of early-onset osteoarthritis after injury. Identification of high-risk individuals before they become patients is important, as post-treatment lifetime burden of ACLR in the USA ranges from $7.6 to $17.7 billion annually. ACLR is a complex disease with multiple risk factors including genetic predisposition. Naturally occurring ACLR in the dog is an excellent model for human ACLR, as risk factors and disease characteristics in humans and dogs are similar. In a univariate genome-wide association study (GWAS) of 237 Labrador Retrievers, we identified 99 ACLR candidate loci. It is likely that additional variants remain to be identified. Joint analysis of multiple correlated phenotypes is an underutilized technique that increases statistical power, even when only one phenotype is associated with the trait. Proximal tibial morphology has been shown to affect ACLR risk in both humans and dogs. In the present study, tibial plateau angle (TPA) and relative tibial tuberosity width (rTTW) were measured on bilateral radiographs from purebred Labrador Retrievers that were recruited to our initial GWAS. We performed a multivariate genome wide association analysis of ACLR status, TPA, and rTTW. Results Our analysis identified 3 loci with moderate evidence of association that were not previously associated with ACLR. A locus on Chr1 associated with both ACLR and rTTW is located within ROR2, a gene important for cartilage and bone development. A locus on Chr4 associated with both ACLR and TPA resides within DOCK2, a gene that has been shown to promote immune cell migration and invasion in synovitis, an important predictor of ACLR. A third locus on Chr23 associated with only ACLR is located near a long non-coding RNA (lncRNA). LncRNA’s are important for regulation of gene transcription and translation. Conclusions These results did not overlap with our previous GWAS, which is reflective of the different methods used, and supports the need for further work. The results of the present study are highly relevant to ACLR pathogenesis, and identify potential drug targets for medical treatment. Electronic supplementary material The online version of this article (10.1186/s12863-018-0626-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lauren A Baker
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, Wisconsin, USA
| | - Guilherme J M Rosa
- Department of Animal Sciences, College of Agricultural and Life Sciences, University of Wisconsin-Madison, 1675 Observatory Drive, Madison, Wisconsin, USA
| | - Zhengling Hao
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, Wisconsin, USA
| | - Alexander Piazza
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, Wisconsin, USA
| | - Christopher Hoffman
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, Wisconsin, USA
| | - Emily E Binversie
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, Wisconsin, USA
| | - Susannah J Sample
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, Wisconsin, USA
| | - Peter Muir
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, Wisconsin, USA.
| |
Collapse
|
31
|
Hamidian Jahromi S, Li Y, Davies JE. Effect of Tumor Necrosis Factor Alpha Dose and Exposure Time on Tumor Necrosis Factor-Induced Gene-6 Activation by Neonatal and Adult Mesenchymal Stromal Cells. Stem Cells Dev 2017; 27:44-54. [PMID: 29121823 DOI: 10.1089/scd.2017.0179] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tumor necrosis factor alpha (TNF-α) induced protein 6 is a major anti-inflammatory mediator released by activated mesenchymal stromal cells (MSCs). Neonatal MSCs are considered more metabolically active than cells derived from adult tissues, and potentially less heterogeneous. We hypothesized that a TNF-α-activated neonatal MSC population [human umbilical cord perivascular cells (HUCPVCs)] would show an enhanced level of TSG-6 activation compared with adult bone marrow MSCs (BMMSCs). Thus, we stimulated HUCPVCs, and both human BMMSCs (hBMMSCs) and mouse BMMSCs (mBMMSCs) with 1, 10, 50, and 100 ng/mL of recombinant TNF-α over various exposure times. Supernatant, and total RNA, of the cells were collected for measurement of both TSG-6 RNA expression, and secreted TSG-6 protein. To compare gene levels, quantification was done by normalizing the expression levels of TSG-6 to the geometric mean of the three most stable reference genes, out of a cohort of 30 tested genes, using the Pfaffl method. We found that HUCPVCs exhibited both an enhanced and more rapid response to low dose (1 ng/mL) TNF-α exposure resulting in ∼11.5-fold increase in TSG-6 expression within the first 30 min. In contrast, hBMMSCs showed 2-fold increase by 1 h that increased to 9.5-fold with a higher (50 ng/mL) TNF-α exposure for the same time. mBMMSCs showed a two-fold increase after 24 h that was independent of TNF-α concentration. Thus, although TSG-6 expression level varied among donors, both hMSC populations exhibited enhanced TSG-6 upregulation, upon TNF-α stimulation, compared with mBMMSCs. In conclusion, HUCPVCs showed higher sensitivity, and a prompter response to TNF-α stimulation compared with hBMMSCs. Thus, neonatal MSCs may be a stronger candidate population than those derived from adult bone marrow to treat inflammatory diseases.
Collapse
Affiliation(s)
- Shiva Hamidian Jahromi
- 1 Institute of Biomaterials and Biomedical Engineering, University of Toronto , Toronto, Ontario, Canada .,2 Faculty of Dentistry, University of Toronto , Toronto, Ontario, Canada
| | - Yunqing Li
- 1 Institute of Biomaterials and Biomedical Engineering, University of Toronto , Toronto, Ontario, Canada
| | - John E Davies
- 1 Institute of Biomaterials and Biomedical Engineering, University of Toronto , Toronto, Ontario, Canada .,2 Faculty of Dentistry, University of Toronto , Toronto, Ontario, Canada
| |
Collapse
|
32
|
Vertès AA. Methods and practices to diversify cell-based products. Regen Med 2017; 12:997-1013. [PMID: 29243940 DOI: 10.2217/rme-2017-0093] [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: 11/21/2022] Open
Abstract
Medicinal signaling cell (MSC)-based products represent emerging treatments in various therapeutic areas including cardiometabolic, inflammation, autoimmunity, orthopedics, wound healing and oncology. Exploring innovation beyond minimally manipulated plastic-adherent ex vivo expanded allogeneic MSCs enables product delineation. Product delineation is on the critical path to maximize clinical benefits and market access. An innovation framework is presented here along various innovation dimensions comprising composition-of-matter by means of positive cell surface markers, formulation varying for example the cell dose or the preservation mode and medium, manufacturing to adapt the secretome of MSCs to the condition of interest, the mode of delivery and corresponding delivery devices, as well as molecular engineering and biomarkers. The rationale of the innovation space thus described applies generally to all cell-based therapies.
Collapse
Affiliation(s)
- Alain A Vertès
- London Business School, UK & NxR Biotechnologies GmbH, Basel, Switzerland
| |
Collapse
|
33
|
Fernandez-Rebollo E, Mentrup B, Ebert R, Franzen J, Abagnale G, Sieben T, Ostrowska A, Hoffmann P, Roux PF, Rath B, Goodhardt M, Lemaitre JM, Bischof O, Jakob F, Wagner W. Human Platelet Lysate versus Fetal Calf Serum: These Supplements Do Not Select for Different Mesenchymal Stromal Cells. Sci Rep 2017; 7:5132. [PMID: 28698620 PMCID: PMC5506010 DOI: 10.1038/s41598-017-05207-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 05/25/2017] [Indexed: 01/23/2023] Open
Abstract
Culture medium of mesenchymal stromal cells (MSCs) is usually supplemented with either human platelet lysate (HPL) or fetal calf serum (FCS). Many studies have demonstrated that proliferation and cellular morphology are affected by these supplements – it is therefore important to determine if they favor outgrowth of different subpopulations and thereby impact on the heterogeneous composition of MSCs. We have isolated and expanded human bone marrow-derived MSCs in parallel with HPL or FCS and demonstrated that HPL significantly increases proliferation and leads to dramatic differences in cellular morphology. Remarkably, global DNA-methylation profiles did not reveal any significant differences. Even at the transcriptomic level, there were only moderate changes in pairwise comparison. Furthermore, the effects on proliferation, cytoskeletal organization, and focal adhesions were reversible by interchanging to opposite culture conditions. These results indicate that cultivation of MSCs with HPL or FCS has no systematic bias for specific cell types.
Collapse
Affiliation(s)
- Eduardo Fernandez-Rebollo
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, 52074, Germany. .,Institute for Biomedical Technology - Cell Biology, RWTH Aachen University Medical School, Aachen, 52074, Germany.
| | - Birgit Mentrup
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, 97074, Germany
| | - Regina Ebert
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, 97074, Germany
| | - Julia Franzen
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, 52074, Germany.,Institute for Biomedical Technology - Cell Biology, RWTH Aachen University Medical School, Aachen, 52074, Germany
| | - Giulio Abagnale
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, 52074, Germany.,Institute for Biomedical Technology - Cell Biology, RWTH Aachen University Medical School, Aachen, 52074, Germany
| | - Torsten Sieben
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, 52074, Germany.,Institute for Biomedical Technology - Cell Biology, RWTH Aachen University Medical School, Aachen, 52074, Germany
| | - Alina Ostrowska
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, 52074, Germany.,Institute for Biomedical Technology - Cell Biology, RWTH Aachen University Medical School, Aachen, 52074, Germany
| | - Per Hoffmann
- Department of Genomics, Institute of Human Genetics, University of Bonn, Bonn, 53127, Germany.,Human Genomics Research Group, Department of Biomedicine, University of Basel, Basel, 4031, Switzerland
| | - Pierre-François Roux
- Laboratory of Nuclear Organization and Oncogenesis, Department of Cell Biology and Infection, INSERM U.993, Institute Pasteur, 75015, Paris, France
| | - Björn Rath
- Department for Orthopedics, RWTH Aachen University Medical School, Aachen, 52074, Germany
| | - Michele Goodhardt
- Institut Universitaire d'Hématologie, INSERM UMRS-1126, University Paris Diderot, 75010, Paris, France
| | - Jean-Marc Lemaitre
- Institute of Regenerative Medicine and Biotherapies (IRMB), INSERM U1183, University of Montpellier, Montpellier, Cedex 05, 34295, France
| | - Oliver Bischof
- Laboratory of Nuclear Organization and Oncogenesis, Department of Cell Biology and Infection, INSERM U.993, Institute Pasteur, 75015, Paris, France
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, 97074, Germany
| | - Wolfgang Wagner
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, 52074, Germany. .,Institute for Biomedical Technology - Cell Biology, RWTH Aachen University Medical School, Aachen, 52074, Germany.
| |
Collapse
|
34
|
Marcelin G, Ferreira A, Liu Y, Atlan M, Aron-Wisnewsky J, Pelloux V, Botbol Y, Ambrosini M, Fradet M, Rouault C, Hénégar C, Hulot JS, Poitou C, Torcivia A, Nail-Barthelemy R, Bichet JC, Gautier EL, Clément K. A PDGFRα-Mediated Switch toward CD9 high Adipocyte Progenitors Controls Obesity-Induced Adipose Tissue Fibrosis. Cell Metab 2017; 25:673-685. [PMID: 28215843 DOI: 10.1016/j.cmet.2017.01.010] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 11/24/2016] [Accepted: 01/19/2017] [Indexed: 02/06/2023]
Abstract
Obesity-induced white adipose tissue (WAT) fibrosis is believed to accelerate WAT dysfunction. However, the cellular origin of WAT fibrosis remains unclear. Here, we show that adipocyte platelet-derived growth factor receptor-α-positive (PDGFRα+) progenitors adopt a fibrogenic phenotype in obese mice prone to visceral WAT fibrosis. More specifically, a subset of PDGFRα+ cells with high CD9 expression (CD9high) originates pro-fibrotic cells whereas their CD9low counterparts, committed to adipogenesis, are almost completely lost in the fibrotic WAT. PDGFRα pathway activation promotes a phenotypic shift toward PDGFRα+CD9high fibrogenic cells, driving pathological remodeling and altering WAT function in obesity. These findings translated to human obesity as the frequency of CD9high progenitors in omental WAT (oWAT) correlates with oWAT fibrosis level, insulin-resistance severity, and type 2 diabetes. Collectively, our data demonstrate that in addition to representing a WAT adipogenic niche, different PDGFRα+ cell subsets modulate obesity-induced WAT fibrogenesis and are associated with loss of metabolic fitness.
Collapse
Affiliation(s)
- Geneviève Marcelin
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital, F-75013 Paris, France; INSERM, UMRS 1166 (teams 2, 4, and 6 NutriOmics), F-75013 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMRS 1166, F-75013 Paris, France.
| | - Adaliene Ferreira
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital, F-75013 Paris, France; INSERM, UMRS 1166 (teams 2, 4, and 6 NutriOmics), F-75013 Paris, France; Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Yuejun Liu
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital, F-75013 Paris, France; INSERM, UMRS 1166 (teams 2, 4, and 6 NutriOmics), F-75013 Paris, France; Assistance Publique Hopitaux de Paris, AP-HP, Pitié-Salpêtrière Hospital, Nutrition and Endocrinology Department and Hepato-biliary and Digestive Surgery Department, F-75013 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMRS 1166, F-75013 Paris, France
| | - Michael Atlan
- Assistance Publique Hôpitaux de Paris, Aesthetic Plastic Reconstructive Unit, Tenon Hospital, Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1166, 75020 Paris, France
| | - Judith Aron-Wisnewsky
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital, F-75013 Paris, France; INSERM, UMRS 1166 (teams 2, 4, and 6 NutriOmics), F-75013 Paris, France; Assistance Publique Hopitaux de Paris, AP-HP, Pitié-Salpêtrière Hospital, Nutrition and Endocrinology Department and Hepato-biliary and Digestive Surgery Department, F-75013 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMRS 1166, F-75013 Paris, France
| | - Véronique Pelloux
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital, F-75013 Paris, France; INSERM, UMRS 1166 (teams 2, 4, and 6 NutriOmics), F-75013 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMRS 1166, F-75013 Paris, France
| | - Yair Botbol
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Marc Ambrosini
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital, F-75013 Paris, France; INSERM, UMRS 1166 (teams 2, 4, and 6 NutriOmics), F-75013 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMRS 1166, F-75013 Paris, France
| | - Magali Fradet
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| | - Christine Rouault
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital, F-75013 Paris, France; INSERM, UMRS 1166 (teams 2, 4, and 6 NutriOmics), F-75013 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMRS 1166, F-75013 Paris, France
| | - Corneliu Hénégar
- HudsonAlpha Institute for Biotechnology, 601 Genome Way, Huntsville, AL 35806, USA
| | - Jean-Sébastien Hulot
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital, F-75013 Paris, France; INSERM, UMRS 1166 (teams 2, 4, and 6 NutriOmics), F-75013 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMRS 1166, F-75013 Paris, France
| | - Christine Poitou
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital, F-75013 Paris, France; INSERM, UMRS 1166 (teams 2, 4, and 6 NutriOmics), F-75013 Paris, France; Assistance Publique Hopitaux de Paris, AP-HP, Pitié-Salpêtrière Hospital, Nutrition and Endocrinology Department and Hepato-biliary and Digestive Surgery Department, F-75013 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMRS 1166, F-75013 Paris, France
| | - Adriana Torcivia
- Assistance Publique Hopitaux de Paris, AP-HP, Pitié-Salpêtrière Hospital, Nutrition and Endocrinology Department and Hepato-biliary and Digestive Surgery Department, F-75013 Paris, France
| | - Raphael Nail-Barthelemy
- Assistance Publique Hôpitaux de Paris, Aesthetic Plastic Reconstructive Unit, Tenon Hospital, Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1166, 75020 Paris, France
| | - Jean-Christophe Bichet
- Assistance Publique Hôpitaux de Paris, Plastic Surgery and Mammary Cancer Department, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| | - Emmanuel L Gautier
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital, F-75013 Paris, France; INSERM, UMRS 1166 (teams 2, 4, and 6 NutriOmics), F-75013 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMRS 1166, F-75013 Paris, France
| | - Karine Clément
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital, F-75013 Paris, France; INSERM, UMRS 1166 (teams 2, 4, and 6 NutriOmics), F-75013 Paris, France; Assistance Publique Hopitaux de Paris, AP-HP, Pitié-Salpêtrière Hospital, Nutrition and Endocrinology Department and Hepato-biliary and Digestive Surgery Department, F-75013 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMRS 1166, F-75013 Paris, France.
| |
Collapse
|
35
|
de Almeida DC, Ferreira MRP, Franzen J, Weidner CI, Frobel J, Zenke M, Costa IG, Wagner W. Epigenetic Classification of Human Mesenchymal Stromal Cells. Stem Cell Reports 2016; 6:168-75. [PMID: 26862701 PMCID: PMC4750140 DOI: 10.1016/j.stemcr.2016.01.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/04/2016] [Accepted: 01/07/2016] [Indexed: 01/05/2023] Open
Abstract
Standardization of mesenchymal stromal cells (MSCs) is hampered by the lack of a precise definition for these cell preparations; for example, there are no molecular markers to discern MSCs and fibroblasts. In this study, we followed the hypothesis that specific DNA methylation (DNAm) patterns can assist classification of MSCs. We utilized 190 DNAm profiles to address the impact of tissue of origin, donor age, replicative senescence, and serum supplements on the epigenetic makeup. Based on this, we elaborated a simple epigenetic signature based on two CpG sites to classify MSCs and fibroblasts, referred to as the Epi-MSC-Score. Another two-CpG signature can distinguish between MSCs from bone marrow and adipose tissue, referred to as the Epi-Tissue-Score. These assays were validated by site-specific pyrosequencing analysis in 34 primary cell preparations. Furthermore, even individual subclones of MSCs were correctly classified by our epigenetic signatures. In summary, we propose an alternative concept to use DNAm patterns for molecular definition of cell preparations, and our epigenetic scores facilitate robust and cost-effective quality control of MSC cultures.
Collapse
Affiliation(s)
- Danilo Candido de Almeida
- Division of Stem Cell Biology and Cellular Engineering, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University Medical School, Pauwelsstraße 20, 52074 Aachen, Germany; Department of Cell Biology, Institute for Biomedical Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany; Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Marcelo R P Ferreira
- Department of Cell Biology, IZKF Research Group Bioinformatics, Institute for Biomedical Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany; Department of Statistics, Centre for Natural and Exact Sciences, Federal University of Paraiba, João Pessoa 58051-900, Brazil
| | - Julia Franzen
- Division of Stem Cell Biology and Cellular Engineering, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University Medical School, Pauwelsstraße 20, 52074 Aachen, Germany; Department of Cell Biology, Institute for Biomedical Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany
| | - Carola I Weidner
- Division of Stem Cell Biology and Cellular Engineering, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University Medical School, Pauwelsstraße 20, 52074 Aachen, Germany; Department of Cell Biology, Institute for Biomedical Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany
| | - Joana Frobel
- Division of Stem Cell Biology and Cellular Engineering, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University Medical School, Pauwelsstraße 20, 52074 Aachen, Germany; Department of Cell Biology, Institute for Biomedical Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany
| | - Martin Zenke
- Department of Cell Biology, Institute for Biomedical Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany; Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany
| | - Ivan G Costa
- Department of Cell Biology, IZKF Research Group Bioinformatics, Institute for Biomedical Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany
| | - Wolfgang Wagner
- Division of Stem Cell Biology and Cellular Engineering, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University Medical School, Pauwelsstraße 20, 52074 Aachen, Germany; Department of Cell Biology, Institute for Biomedical Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany.
| |
Collapse
|
36
|
EphA2 is a biomarker of hMSCs derived from human placenta and umbilical cord. Taiwan J Obstet Gynecol 2016; 54:749-56. [PMID: 26700997 DOI: 10.1016/j.tjog.2015.10.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2015] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE The heterogeneous nature of mesenchymal stem cells (MSCs) and the absence of known MSC-specific biomarkers make it challenging to define MSC phenotypes and characteristics. In this study, we compared the phenotypic and functional features of human placenta-derived MSCs with those of human dermal fibroblasts in vitro in order to identify a biomarker that can be used to increase the purity of MSCs in a primary culture of placenta-derived cells. MATERIALS AND METHODS Liquid chromatography-tandem mass spectrometry analysis was used to analyze and compare the proteome of human placenta-derived MSCs with that of fibroblasts. Quantitative real-time polymerase chain reaction, immunofluorescence, and flow cytometry were used to determine expression levels of EphA2 in placenta-derived MSCs. EphA2-positive cells were enriched by magnetic-activated cell sorting or with a cell sorter. An shRNA-mediated EphA2 knockdown was used to assess the role of EphA2 in MSC response to Tumor necrosis factor (TNF)-α stimulation. RESULTS Analysis of proteomics data from MSCs and fibroblasts resulted in the identification of the EphA2 surface protein biomarker, which could reliably distinguish MSCs from fibroblasts. EphA2 was significantly upregulated in placenta-derived MSCs when compared to fibroblasts. EphA2 played an important role in MSC migration in response to inflammatory stimuli, such as TNF-α. EphA2-enriched MSCs were also more responsive to inflammatory stimuli in vitro when compared to unsorted MSCs, indicating a role for EphA2 in the immunomodulatory functionality of MSCs. CONCLUSION EphA2 can be used to distinguish and isolate MSCs from a primary culture of placenta-derived cells. EphA2-sorted MSCs exhibited superior responsiveness to TNF-α signaling in an inflammatory environment compared with unsorted MSCs or MSC-like cells.
Collapse
|
37
|
Billing AM, Ben Hamidane H, Bhagwat AM, Cotton RJ, Dib SS, Kumar P, Hayat S, Goswami N, Suhre K, Rafii A, Graumann J. Complementarity of SOMAscan to LC-MS/MS and RNA-seq for quantitative profiling of human embryonic and mesenchymal stem cells. J Proteomics 2016; 150:86-97. [PMID: 27613379 DOI: 10.1016/j.jprot.2016.08.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/01/2016] [Accepted: 08/29/2016] [Indexed: 11/16/2022]
Abstract
Dynamic range limitations are challenging to proteomics, particularly in clinical samples. Affinity proteomics partially overcomes this, yet suffers from dependence on reagent quality. SOMAscan, an aptamer-based platform for over 1000 proteins, avoids that issue using nucleic acid binders. Targets include low expressed proteins not easily accessible by other approaches. Here we report on the potential of SOMAscan for the study of differently sourced mesenchymal stem cells (MSC) in comparison to LC-MS/MS and RNA sequencing. While targeting fewer analytes, SOMAscan displays high precision and dynamic range coverage, allowing quantification of proteins not measured by the other platforms. Expression between cell types (ESC and MSC) was compared across techniques and uncovered the expected large differences. Sourcing was investigated by comparing subtypes: bone marrow-derived, standard in clinical studies, and ESC-derived MSC, thought to hold similar potential but devoid of inter-donor variability and proliferating faster in vitro. We confirmed subtype-equivalency, as well as vesicle and extracellular matrix related processes in MSC. In contrast, the proliferative nature of ESC was captured less by SOMAscan, where nuclear proteins are underrepresented. The complementary of SOMAscan allowed the comprehensive exploration of CD markers and signaling molecules, not readily accessible otherwise and offering unprecedented potential in subtype characterization. SIGNIFICANCE Mesenchymal stem cells (MSC) represent promising stem cell-derived therapeutics as indicated by their application in >500 clinical trials currently registered with the NIH. Tissue-derived MSC require invasive harvesting and imply donor-to-donor differences, to which embryonic stem cell (ESC)-derived MSC may provide an alternative and thus warrant thorough characterization. In continuation of our previous study where we compared in depth embryonic stem cells (ESC) and MSC from two sources (bone marrow and ESC-derived), we included the aptamer-based SOMAscan assay, complementing LC-MS/MS and RNA-seq data. Furthermore, SOMAscan, a targeted proteomics platform developed for analyzing clinical samples, has been benchmarked against established analytical platforms (LC-MS/MS and RNA-seq) using stem cell comparisons as a model.
Collapse
Affiliation(s)
- Anja M Billing
- Research Division, Weill Cornell Medical College in Qatar, Doha, Qatar.
| | | | - Aditya M Bhagwat
- Research Division, Weill Cornell Medical College in Qatar, Doha, Qatar.
| | - Richard J Cotton
- Research Division, Weill Cornell Medical College in Qatar, Doha, Qatar.
| | - Shaima S Dib
- Research Division, Weill Cornell Medical College in Qatar, Doha, Qatar.
| | - Pankaj Kumar
- Research Division, Weill Cornell Medical College in Qatar, Doha, Qatar.
| | - Shahina Hayat
- Research Division, Weill Cornell Medical College in Qatar, Doha, Qatar.
| | - Neha Goswami
- Research Division, Weill Cornell Medical College in Qatar, Doha, Qatar.
| | - Karsten Suhre
- Research Division, Weill Cornell Medical College in Qatar, Doha, Qatar.
| | - Arash Rafii
- Research Division, Weill Cornell Medical College in Qatar, Doha, Qatar.
| | - Johannes Graumann
- Research Division, Weill Cornell Medical College in Qatar, Doha, Qatar.
| |
Collapse
|
38
|
Rohart F, Mason EA, Matigian N, Mosbergen R, Korn O, Chen T, Butcher S, Patel J, Atkinson K, Khosrotehrani K, Fisk NM, Lê Cao KA, Wells CA. A molecular classification of human mesenchymal stromal cells. PeerJ 2016; 4:e1845. [PMID: 27042394 PMCID: PMC4811172 DOI: 10.7717/peerj.1845] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/03/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cells (MSC) are widely used for the study of mesenchymal tissue repair, and increasingly adopted for cell therapy, despite the lack of consensus on the identity of these cells. In part this is due to the lack of specificity of MSC markers. Distinguishing MSC from other stromal cells such as fibroblasts is particularly difficult using standard analysis of surface proteins, and there is an urgent need for improved classification approaches. Transcriptome profiling is commonly used to describe and compare different cell types; however, efforts to identify specific markers of rare cellular subsets may be confounded by the small sample sizes of most studies. Consequently, it is difficult to derive reproducible, and therefore useful markers. We addressed the question of MSC classification with a large integrative analysis of many public MSC datasets. We derived a sparse classifier (The Rohart MSC test) that accurately distinguished MSC from non-MSC samples with >97% accuracy on an internal training set of 635 samples from 41 studies derived on 10 different microarray platforms. The classifier was validated on an external test set of 1,291 samples from 65 studies derived on 15 different platforms, with >95% accuracy. The genes that contribute to the MSC classifier formed a protein-interaction network that included known MSC markers. Further evidence of the relevance of this new MSC panel came from the high number of Mendelian disorders associated with mutations in more than 65% of the network. These result in mesenchymal defects, particularly impacting on skeletal growth and function. The Rohart MSC test is a simple in silico test that accurately discriminates MSC from fibroblasts, other adult stem/progenitor cell types or differentiated stromal cells. It has been implemented in the www.stemformatics.org resource, to assist researchers wishing to benchmark their own MSC datasets or data from the public domain. The code is available from the CRAN repository and all data used to generate the MSC test is available to download via the Gene Expression Omnibus or the Stemformatics resource.
Collapse
Affiliation(s)
- Florian Rohart
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Elizabeth A. Mason
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | - Nicholas Matigian
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Rowland Mosbergen
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Othmar Korn
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | - Tyrone Chen
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Suzanne Butcher
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Jatin Patel
- The University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
| | - Kerry Atkinson
- The University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
| | - Kiarash Khosrotehrani
- The University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
- Centre for Advanced Prenatal Care, Royal Brisbane & Women’s Hospital, Brisbane, Queensland, Australia
| | - Nicholas M. Fisk
- The University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
- Centre for Advanced Prenatal Care, Royal Brisbane & Women’s Hospital, Brisbane, Queensland, Australia
| | - Kim-Anh Lê Cao
- The University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Christine A. Wells
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
39
|
Comprehensive transcriptomic and proteomic characterization of human mesenchymal stem cells reveals source specific cellular markers. Sci Rep 2016; 6:21507. [PMID: 26857143 PMCID: PMC4746666 DOI: 10.1038/srep21507] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 01/26/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSC) are multipotent cells with great potential in therapy, reflected by more than 500 MSC-based clinical trials registered with the NIH. MSC are derived from multiple tissues but require invasive harvesting and imply donor-to-donor variability. Embryonic stem cell-derived MSC (ESC-MSC) may provide an alternative, but how similar they are to ex vivo MSC is unknown. Here we performed an in depth characterization of human ESC-MSC, comparing them to human bone marrow-derived MSC (BM-MSC) as well as human embryonic stem cells (hESC) by transcriptomics (RNA-seq) and quantitative proteomics (nanoLC-MS/MS using SILAC). Data integration highlighted and validated a central role of vesicle-mediated transport and exosomes in MSC biology and also demonstrated, through enrichment analysis, their versatility and broad application potential. Particular emphasis was placed on comparing profiles between ESC-MSC and BM-MSC and assessing their equivalency. Data presented here shows that differences between ESC-MSC and BM-MSC are similar in magnitude to those reported for MSC of different origin and the former may thus represent an alternative source for therapeutic applications. Finally, we report an unprecedented coverage of MSC CD markers, as well as membrane associated proteins which may benefit immunofluorescence-based applications and contribute to a refined molecular description of MSC.
Collapse
|
40
|
Prospective isolation of resident adult human mesenchymal stem cell population from multiple organs. Int J Hematol 2015; 103:138-44. [PMID: 26676805 DOI: 10.1007/s12185-015-1921-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 12/01/2015] [Accepted: 12/02/2015] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem/stromal cells (MSCs) have the potential to form colonies in culture and reside in adult tissues. Because MSCs have been defined using cells cultured in vitro, discrepancies have arisen between studies concerning their properties. There are also differences between populations obtained using different isolation methods. This review article focuses on recent developments in the identification of novel MSC markers for the in vivo localization and prospective isolation of human MSCs. The prospective isolation method described in this study represents an important strategy for the isolation of MSCs in a short period of time, and may find applications for regenerative medicine. Purified MSCs can be tailored according to their intended clinical therapeutic applications. Lineage tracing methods define the MSC phenotype and can be used to investigate the physiological roles of MSCs in vivo. These findings may facilitate the development of effective stem cell treatments.
Collapse
|
41
|
Myocardial regeneration strategy using Wharton's jelly mesenchymal stem cells as an off-the-shelf 'unlimited' therapeutic agent: results from the Acute Myocardial Infarction First-in-Man Study. ADVANCES IN INTERVENTIONAL CARDIOLOGY 2015; 11:100-7. [PMID: 26161101 PMCID: PMC4495125 DOI: 10.5114/pwki.2015.52282] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 04/18/2015] [Accepted: 06/08/2015] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION In large-animal acute myocardial infarction (AMI) models, Wharton's jelly (umbilical cord matrix) mesenchymal stem cells (WJMSCs) effectively promote angiogenesis and drive functional myocardial regeneration. Human data are lacking. AIM To evaluate the feasibility and safety of a novel myocardial regeneration strategy using human WJMSCs as a unique, allogenic but immuno-privileged, off-the-shelf cellular therapeutic agent. MATERIAL AND METHODS The inclusion criterion was first, large (LVEF ≤ 45%, CK-MB > 100 U/l) AMI with successful infarct-related artery primary percutaneous coronary intervention reperfusion (TIMI ≥ 2). Ten consecutive patients (age 32-65 years, peak hs-troponin T 17.3 ±9.1 ng/ml and peak CK-MB 533 ±89 U/l, sustained echo LVEF reduction to 37.6 ±2.6%, cMRI LVEF 40.3 ±2.7% and infarct size 20.1 ±2.8%) were enrolled. RESULTS 30 × 10(6) WJMSCs were administered (LAD/Cx/RCA in 6/3/1) per protocol at ≈ 5-7 days using a cell delivery-dedicated, coronary-non-occlusive method. No clinical symptoms or ECG signs of myocardial ischemia occurred. There was no epicardial flow or myocardial perfusion impairment (TIMI-3 in all; cTFC 45 ±8 vs. 44 ±9, p = 0.51), and no patient showed hs-troponin T elevation (0.92 ±0.29 ≤ 24 h before vs. 0.89 ±0.28 ≤ 24 h after; decrease, p = 0.04). One subject experienced, 2 days after cell transfer, a transient temperature rise (38.9°C); this was reactive to paracetamol with no sequel. No other adverse events and no significant arrhythmias (ECG Holter) occurred. Up to 12 months there was one new, non-index territory lethal AMI but no adverse events that might be attributable to WJMSC treatment. CONCLUSIONS This study demonstrated the feasibility and procedural safety of WJMSC use as off-the-shelf cellular therapy in human AMI and suggested further clinical safety of WJMSC cardiac transfer, providing a basis for randomized placebo-controlled endpoint-powered evaluation.
Collapse
|