1
|
Das S, Thompson W, Papoutsakis ET. Engineered and hybrid human megakaryocytic extracellular vesicles for targeted non-viral cargo delivery to hematopoietic (blood) stem and progenitor cells. Front Bioeng Biotechnol 2024; 12:1435228. [PMID: 39386042 PMCID: PMC11461334 DOI: 10.3389/fbioe.2024.1435228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Native and engineered extracellular vesicles generated from human megakaryocytes (huMkEVs) or from the human megakaryocytic cell line CHRF (CHEVs) interact with tropism delivering their cargo to both human and murine hematopoietic stem and progenitor cells (HSPCs). To develop non-viral delivery vectors to HSPCs based on MkEVs, we first confirmed, using NOD-scid IL2Rγnull (NSG™) mice, the targeting potential of the large EVs, enriched in microparticles (huMkMPs), chosen for their large cargo capacity. 24 h post intravenous infusion into NSG mice, huMkEVs induced a nearly 50% increase in murine platelet counts. PKH26-labeled huMkEVs or CHEVs localized to the HSPC-rich bone marrow preferentially interacting with murine HSPCs, thus confirming their receptor-mediated tropism for NSG HSPCs, and their potential to treat thromobocytopenias. We explored this tropism to functionally deliver synthetic cargo, notably plasmid DNA coding for a fluorescent reporter, to NSG HSPCs both in vitro and in vivo. We loaded huMkEVs with plasmid DNA either through electroporation or by generating hybrid particles with preloaded liposomes. Both methods facilitated successful functional targeted delivery of pDNA, as tissue weight-normalized fluorescence intensity of the expressed fluorescent reporter was significantly higher in bone marrow than other tissues. Furthermore, the fraction of fluorescent CD117+ HSPCs was nearly 19-fold higher than other cell types within the bone marrow 72-h following administration of the hybrid particles, further supporting that HSPC tropism is retained when using hybrid particles. These data demonstrate the potential of these EVs as a non-viral, HSPC-specific cargo vehicle for gene therapy applications to treat hematological diseases.
Collapse
Affiliation(s)
- Samik Das
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, United States
| | - Will Thompson
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, United States
| | - Eleftherios Terry Papoutsakis
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, United States
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| |
Collapse
|
2
|
Ying Z, Lyu L, Xu X, Wen Z, Xue J, Chen M, Li Z, Jiang L, Chen T. Resident vascular Sca1 + progenitors differentiate into endothelial cells in vascular remodeling via miR-145-5p/ERG signaling pathway. iScience 2024; 27:110080. [PMID: 38883819 PMCID: PMC11176791 DOI: 10.1016/j.isci.2024.110080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/17/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
Endothelial cell (EC) damage or dysfunction serves as the initial event in the pathogenesis of various cardiovascular diseases. Progenitor cells have been postulated to be able to differentiate into ECs, facilitate endothelial regeneration, and alleviate vascular pathological remodeling. However, the precise cellular origins and underlying mechanisms remain elusive. Through single-cell RNA sequencing (scRNA-seq), we identified an increasing population of progenitors expressing stem cell antigen 1 (Sca1) during vascular remodeling in mice. Using both mouse femoral artery injury and vein graft models, we determined that Sca1+ cells differentiate into ECs, restored endothelium in arterial and venous remodeling processes. Notably, we have observed that the differentiation of Sca1+ cells into ECs is negatively regulated by the microRNA-145-5p (miR-145-5p)-Erythroblast transformation-specific-related gene (ERG) pathway. Inhibiting miR-145-5p promoted Sca1+ cell differentiation and reduced neointimal formation after vascular injury. Finally, a similar downregulation of miR-145-5p in human arteriovenous fistula was observed comparing to healthy veins.
Collapse
Affiliation(s)
- Zhangquan Ying
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lingxia Lyu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiaodong Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zuoshi Wen
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jianing Xue
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Mengjia Chen
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhoubin Li
- Department of Lung Transplantation and General Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Liujun Jiang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ting Chen
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Affiliated First Hospital of Ningbo University, Ningbo 315010, China
| |
Collapse
|
3
|
Nagel M, Niestroj M, Bansal R, Fleck D, Lampert A, Stopkova R, Stopka P, Ben-Shaul Y, Spehr M. Deciphering the chemical language of inbred and wild mouse conspecific scents. eLife 2024; 12:RP90529. [PMID: 38747258 PMCID: PMC11095937 DOI: 10.7554/elife.90529] [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] [Indexed: 05/18/2024] Open
Abstract
In most mammals, conspecific chemosensory communication relies on semiochemical release within complex bodily secretions and subsequent stimulus detection by the vomeronasal organ (VNO). Urine, a rich source of ethologically relevant chemosignals, conveys detailed information about sex, social hierarchy, health, and reproductive state, which becomes accessible to a conspecific via vomeronasal sampling. So far, however, numerous aspects of social chemosignaling along the vomeronasal pathway remain unclear. Moreover, since virtually all research on vomeronasal physiology is based on secretions derived from inbred laboratory mice, it remains uncertain whether such stimuli provide a true representation of potentially more relevant cues found in the wild. Here, we combine a robust low-noise VNO activity assay with comparative molecular profiling of sex- and strain-specific mouse urine samples from two inbred laboratory strains as well as from wild mice. With comprehensive molecular portraits of these secretions, VNO activity analysis now enables us to (i) assess whether and, if so, how much sex/strain-selective 'raw' chemical information in urine is accessible via vomeronasal sampling; (ii) identify which chemicals exhibit sufficient discriminatory power to signal an animal's sex, strain, or both; (iii) determine the extent to which wild mouse secretions are unique; and (iv) analyze whether vomeronasal response profiles differ between strains. We report both sex- and, in particular, strain-selective VNO representations of chemical information. Within the urinary 'secretome', both volatile compounds and proteins exhibit sufficient discriminative power to provide sex- and strain-specific molecular fingerprints. While total protein amount is substantially enriched in male urine, females secrete a larger variety at overall comparatively low concentrations. Surprisingly, the molecular spectrum of wild mouse urine does not dramatically exceed that of inbred strains. Finally, vomeronasal response profiles differ between C57BL/6 and BALB/c animals, with particularly disparate representations of female semiochemicals.
Collapse
Affiliation(s)
- Maximilian Nagel
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
| | - Marco Niestroj
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
| | - Rohini Bansal
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University of JerusalemJerusalemIsrael
| | - David Fleck
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
| | - Angelika Lampert
- Institute of Neurophysiology, Uniklinik RWTH Aachen UniversityAachenGermany
- Research Training Group 2416 MultiSenses – MultiScales, RWTH Aachen UniversityAachenGermany
| | - Romana Stopkova
- BIOCEV group, Department of Zoology, Faculty of Science, Charles UniversityPragueCzech Republic
| | - Pavel Stopka
- BIOCEV group, Department of Zoology, Faculty of Science, Charles UniversityPragueCzech Republic
| | - Yoram Ben-Shaul
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University of JerusalemJerusalemIsrael
| | - Marc Spehr
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
- Research Training Group 2416 MultiSenses – MultiScales, RWTH Aachen UniversityAachenGermany
| |
Collapse
|
4
|
Xue G, Zhang X, Li W, Zhang L, Zhang Z, Zhou X, Zhang D, Zhang L, Li Z. A logic-incorporated gene regulatory network deciphers principles in cell fate decisions. eLife 2024; 12:RP88742. [PMID: 38652107 PMCID: PMC11037919 DOI: 10.7554/elife.88742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
Organisms utilize gene regulatory networks (GRN) to make fate decisions, but the regulatory mechanisms of transcription factors (TF) in GRNs are exceedingly intricate. A longstanding question in this field is how these tangled interactions synergistically contribute to decision-making procedures. To comprehensively understand the role of regulatory logic in cell fate decisions, we constructed a logic-incorporated GRN model and examined its behavior under two distinct driving forces (noise-driven and signal-driven). Under the noise-driven mode, we distilled the relationship among fate bias, regulatory logic, and noise profile. Under the signal-driven mode, we bridged regulatory logic and progression-accuracy trade-off, and uncovered distinctive trajectories of reprogramming influenced by logic motifs. In differentiation, we characterized a special logic-dependent priming stage by the solution landscape. Finally, we applied our findings to decipher three biological instances: hematopoiesis, embryogenesis, and trans-differentiation. Orthogonal to the classical analysis of expression profile, we harnessed noise patterns to construct the GRN corresponding to fate transition. Our work presents a generalizable framework for top-down fate-decision studies and a practical approach to the taxonomy of cell fate decisions.
Collapse
Affiliation(s)
- Gang Xue
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
| | - Xiaoyi Zhang
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
| | - Wanqi Li
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
| | - Lu Zhang
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
| | - Zongxu Zhang
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
| | - Xiaolin Zhou
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
| | - Di Zhang
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
| | - Lei Zhang
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
- Beijing International Center for Mathematical Research, Center for Machine Learning Research, Peking UniversityBeijingChina
| | - Zhiyuan Li
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
| |
Collapse
|
5
|
Tao T, Du L, Teng P, Guo Y, Wang X, Hu Y, Zhao H, Xu Q, Ma L. Stem cell antigen-1 +cell-derived fibroblasts are crucial for cardiac fibrosis during heart failure. Cell Mol Life Sci 2023; 80:300. [PMID: 37740736 PMCID: PMC11073062 DOI: 10.1007/s00018-023-04957-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/21/2023] [Accepted: 09/07/2023] [Indexed: 09/25/2023]
Abstract
AIMS Mesenchymal stem cells (MSCs) present in the heart cannot differentiate into cardiomyocytes, but may play a role in pathological conditions. Therefore, the aim of this study was to scrutinise the role and mechanism of MSC differentiation in vivo during heart failure. METHODS AND RESULTS We performed single-cell RNA sequencing of total non-cardiomyocytes from murine and adult human hearts. By analysing the transcriptomes of single cells, we illustrated the dynamics of the cell landscape during the progression of heart hypertrophy, including those of stem cell antigen-1 (Sca1)+ stem/progenitor cells and fibroblasts. By combining genetic lineage tracing and bone marrow transplantation models, we demonstrated that non-bone marrow-derived Sca1+ cells give rise to fibroblasts. Interestingly, partial depletion of Sca1+ cells alleviated the severity of myocardial fibrosis and led to a significant improvement in cardiac function in Sca1-CreERT2;Rosa26-eGFP-DTA mice. Similar non-cardiomyocyte cell composition and heterogeneity were observed in human patients with heart failure. Mechanistically, our study revealed that Sca1+ cells can transform into fibroblasts and affect the severity of fibrosis through the Wnt4-Pdgfra pathway. CONCLUSIONS Our study describes the cellular landscape of hypertrophic hearts and reveals that fibroblasts derived from Sca1+ cells with a non-bone marrow source largely account for cardiac fibrosis. These findings provide novel insights into the pathogenesis of cardiac fibrosis and have potential therapeutic implications for heart failure. Non-bone marrow-derived Sca1+ cells differentiate into fibroblasts involved in cardiac fibrosis via Wnt4-PDGFRα pathway.
Collapse
Affiliation(s)
- Tingting Tao
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Luping Du
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Peng Teng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Yan Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Xuyang Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Yanhua Hu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Haige Zhao
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China.
| | - Liang Ma
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China.
| |
Collapse
|
6
|
Yang F, Nourse C, Helgason GV, Kirschner K. Unraveling Heterogeneity in the Aging Hematopoietic Stem Cell Compartment: An Insight From Single-cell Approaches. Hemasphere 2023; 7:e895. [PMID: 37304939 PMCID: PMC10256339 DOI: 10.1097/hs9.0000000000000895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/18/2023] [Indexed: 06/13/2023] Open
Abstract
Specific cell types and, therefore, organs respond differently during aging. This is also true for the hematopoietic system, where it has been demonstrated that hematopoietic stem cells alter a variety of features, such as their metabolism, and accumulate DNA damage, which can lead to clonal outgrowth over time. In addition, profound changes in the bone marrow microenvironment upon aging lead to senescence in certain cell types such as mesenchymal stem cells and result in increased inflammation. This heterogeneity makes it difficult to pinpoint the molecular drivers of organismal aging gained from bulk approaches, such as RNA sequencing. A better understanding of the heterogeneity underlying the aging process in the hematopoietic compartment is, therefore, needed. With the advances of single-cell technologies in recent years, it is now possible to address fundamental questions of aging. In this review, we discuss how single-cell approaches can and indeed are already being used to understand changes observed during aging in the hematopoietic compartment. We will touch on established and novel methods for flow cytometric detection, single-cell culture approaches, and single-cell omics.
Collapse
Affiliation(s)
- Fei Yang
- School of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Craig Nourse
- School of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - G. Vignir Helgason
- School of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Kristina Kirschner
- School of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| |
Collapse
|
7
|
Shay TF, Sullivan EE, Ding X, Chen X, Ravindra Kumar S, Goertsen D, Brown D, Crosby A, Vielmetter J, Borsos M, Wolfe DA, Lam AW, Gradinaru V. Primate-conserved carbonic anhydrase IV and murine-restricted LY6C1 enable blood-brain barrier crossing by engineered viral vectors. SCIENCE ADVANCES 2023; 9:eadg6618. [PMID: 37075114 PMCID: PMC10115422 DOI: 10.1126/sciadv.adg6618] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The blood-brain barrier (BBB) presents a major challenge for delivering large molecules to study and treat the central nervous system. This is due in part to the scarcity of targets known to mediate BBB crossing. To identify novel targets, we leverage a panel of adeno-associated viruses (AAVs) previously identified through mechanism-agnostic directed evolution for improved BBB transcytosis. Screening potential cognate receptors for enhanced BBB crossing, we identify two targets: murine-restricted LY6C1 and widely conserved carbonic anhydrase IV (CA-IV). We apply AlphaFold-based in silico methods to generate capsid-receptor binding models to predict the affinity of AAVs for these identified receptors. Demonstrating how these tools can unlock target-focused engineering strategies, we create an enhanced LY6C1-binding vector, AAV-PHP.eC, that, unlike our prior PHP.eB, also works in Ly6a-deficient mouse strains such as BALB/cJ. Combined with structural insights from computational modeling, the identification of primate-conserved CA-IV enables the design of more specific and potent human brain-penetrant chemicals and biologicals, including gene delivery vectors.
Collapse
Affiliation(s)
- Timothy F. Shay
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Corresponding author. (T.F.S.); (V.G.)
| | - Erin E. Sullivan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Xiaozhe Ding
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Xinhong Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Sripriya Ravindra Kumar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - David Goertsen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - David Brown
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Anaya Crosby
- California State Polytechnic University, Pomona, Pomona, CA, USA
| | - Jost Vielmetter
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Máté Borsos
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Damien A. Wolfe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Annie W. Lam
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Corresponding author. (T.F.S.); (V.G.)
| |
Collapse
|
8
|
Redman RS, Alvarez-Martinez JC. Identifying stem cells in the main excretory ducts of rat major salivary glands: adventures with commercial antibodies. Biotech Histochem 2023; 98:280-290. [PMID: 36779267 DOI: 10.1080/10520295.2023.2177348] [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: 02/14/2023] Open
Abstract
We investigated the entire length of the main excretory ducts (MED) of the major sublingual, parotid and submandibular salivary glands of mature laboratory rats for mucous (goblet) and luminal ciliated cells, biomarkers of cell proliferation, apoptosis, and five biomarkers of stem cells. Spleen and testis were used as positive controls. We used formalin fixed, paraffin embedded tissues. No mucous cells or cells with luminal cilia were observed in hematoxylin and eosin, alcian blue or periodic acid-Schiff stained sections. Immunohistochemistry using rabbit anti-rat antibodies produced anomalous reactions with cleaved caspase-3 for apoptosis, Ki-67 for proliferative activity and Sox 2. Following antigen retrieval, no primary antibody and all three negative controls, labeled macrophages appeared in the spleen. TUNEL staining revealed a few cells per section undergoing apoptosis. Reactions deemed valid occurred in MED with cytokeratin-5 and c-Kit and stem cell antigen 1 (Sca-1) mostly in the gland and middle segments. Other ducts, but not acini or myoepithelial cells, also were variably stained with c-Kit and Sca-1.
Collapse
Affiliation(s)
- Robert S Redman
- Oral Pathology Research Laboratory, Department of Veterans Affairs Medical Center, Washington, DC, USA.,Dental Service, Department of Veterans Affairs Medical Center, Washington, DC, USA
| | | |
Collapse
|
9
|
Ni Z, Lyu L, Gong H, Du L, Wen Z, Jiang H, Yang H, Hu Y, Zhang B, Xu Q, Guo X, Chen T. Multilineage commitment of Sca-1 + cells in reshaping vein grafts. Theranostics 2023; 13:2154-2175. [PMID: 37153747 PMCID: PMC10157743 DOI: 10.7150/thno.77735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 03/23/2023] [Indexed: 05/10/2023] Open
Abstract
Vein graft failure remains a significant clinical problem. Similar to other vascular diseases, stenosis of vein grafts is caused by several cell lines; however, the sources of these cells remain unclear. The objective of this study was to investigate the cellular sources that reshape vein grafts. By analyzing transcriptomics data and constructing inducible lineage-tracing mouse models, we investigated the cellular components of vein grafts and their fates. The sc-RNAseq data suggested that Sca-1+ cells were vital players in vein grafts and might serve as progenitors for multilineage commitment. By generating a vein graft model in which the venae cavae from C57BL/6J wild-type mice were transplanted adjacent to the carotid arteries of Sca-1(Ly6a)-CreERT2; Rosa26-tdTomato mice, we demonstrated that the recipient Sca-1+ cells dominated reendothelialization and the formation of adventitial microvessels, especially at the perianastomotic regions. In turn, using chimeric mouse models, we confirmed that the Sca-1+ cells that participated in reendothelialization and the formation of adventitial microvessels all had a non-bone-marrow origin, whereas bone-marrow-derived Sca-1+ cells differentiated into inflammatory cells in vein grafts. Furthermore, using a parabiosis mouse model, we confirmed that non-bone-marrow-derived circulatory Sca-1+ cells were vital for the formation of adventitial microvessels, whereas Sca-1+ cells derived from local carotid arteries were the source of endothelium restoration. Using another mouse model in which venae cavae from Sca-1 (Ly6a)-CreERT2; Rosa26-tdTomato mice were transplanted adjacent to the carotid arteries of C57BL/6J wild-type mice, we confirmed that the donor Sca-1+ cells were mainly responsible for smooth muscle cells commitment in the neointima, particularly at the middle bodies of vein grafts. In addition, we provided evidence that knockdown/knockout of Pdgfrα in Sca-1+ cells decreased the cell potential to generate SMCs in vitro and decreased number of intimal SMCs in vein grafts. Our findings provided cell atlases of vein grafts, which demonstrated that recipient carotid arteries, donor veins, non-bone-marrow circulation, and the bone marrow provided diverse Sca-1+ cells/progenitors that participated in the reshaping of vein grafts.
Collapse
Affiliation(s)
- Zhichao Ni
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lingxia Lyu
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hui Gong
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Luping Du
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zuoshi Wen
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hua Jiang
- Department of kidney disease center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, PR China
| | - Hao Yang
- Department of kidney disease center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, PR China
| | - Yanhua Hu
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Bohuan Zhang
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- ✉ Corresponding authors: Qingbo Xu, MD. PhD. , Tel: +86 571-87236500, Fax: +86 571 4008306430 Department of Cardiology, the First Affiliated Hospital, Zhejiang University Medical School, 79 Qingchun Road, Hangzhou 310003, Hangzhou, China. Or Xiaogang Guo, MD. PhD. , Tel: +86 571-87236500 Department of Cardiology, the First Affiliated Hospital, Zhejiang University Medical School, 79 Qingchun Road, Hangzhou 310003, Hangzhou, China. Or Ting Chen, MD. PhD. , Tel: +86 15067127900 Mailing Address: Department of Cardiology, the First Affiliated Hospital, Zhejiang University Medical School, 79 Qingchun Road, Hangzhou 310003, Hangzhou, China
| | - Xiaogang Guo
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- ✉ Corresponding authors: Qingbo Xu, MD. PhD. , Tel: +86 571-87236500, Fax: +86 571 4008306430 Department of Cardiology, the First Affiliated Hospital, Zhejiang University Medical School, 79 Qingchun Road, Hangzhou 310003, Hangzhou, China. Or Xiaogang Guo, MD. PhD. , Tel: +86 571-87236500 Department of Cardiology, the First Affiliated Hospital, Zhejiang University Medical School, 79 Qingchun Road, Hangzhou 310003, Hangzhou, China. Or Ting Chen, MD. PhD. , Tel: +86 15067127900 Mailing Address: Department of Cardiology, the First Affiliated Hospital, Zhejiang University Medical School, 79 Qingchun Road, Hangzhou 310003, Hangzhou, China
| | - Ting Chen
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
- ✉ Corresponding authors: Qingbo Xu, MD. PhD. , Tel: +86 571-87236500, Fax: +86 571 4008306430 Department of Cardiology, the First Affiliated Hospital, Zhejiang University Medical School, 79 Qingchun Road, Hangzhou 310003, Hangzhou, China. Or Xiaogang Guo, MD. PhD. , Tel: +86 571-87236500 Department of Cardiology, the First Affiliated Hospital, Zhejiang University Medical School, 79 Qingchun Road, Hangzhou 310003, Hangzhou, China. Or Ting Chen, MD. PhD. , Tel: +86 15067127900 Mailing Address: Department of Cardiology, the First Affiliated Hospital, Zhejiang University Medical School, 79 Qingchun Road, Hangzhou 310003, Hangzhou, China
| |
Collapse
|
10
|
Weng X, Li J, Guan Q, Zhao H, Wang Z, Gleave ME, Nguan CY, Du C. The functions of clusterin in renal mesenchymal stromal cells: Promotion of cell growth and regulation of macrophage activation. Exp Cell Res 2022; 413:113081. [PMID: 35218723 DOI: 10.1016/j.yexcr.2022.113081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/31/2022] [Accepted: 02/15/2022] [Indexed: 12/24/2022]
Abstract
Clusterin (CLU) increases resistance to renal ischemia-reperfusion injury and promotes renal tissue repair. However, the mechanisms underlying of the renal protection of CLU remain unknown. Mesenchymal stromal cells (MSCs) may contribute to kidney cell turnover and injury repair. This study investigated the in vitro functions of CLU in kidney mesenchymal stromal cells (KMSCs). KMSCs were grown in plastic culture plates. Cell surface markers, apoptosis and phagocytosis were determined by flow cytometry, and CLU protein by Western blot. There were no differences in the expression of MSC markers (positive: CD133, Sca-1, CD44, CD117 and NG2, and negative: CD34, CD45, CD163, CD41, CD276, CD138, CD79a, CD146 and CD140b) and in the trilineage differentiation to chondrocytes, adipocytes and osteocytes between wild type (WT) and CLU knockout (KO) KMSCs. CLU was expressed intracellularly and secreted by WT KMSCs, and it was up-regulated by hypoxia. CLU did not prevent hypoxia-induced cell apoptosis but promoted cell growth in KMSC cultures. Furthermore, incubation with CLU-containing culture medium from WT KMSCs increased CD206 expression and phagocytic capacity of macrophages. In conclusion, our data for the first time demonstrate the function of CLU in the promotion of KMSCs proliferation, and it may be required for KMSCs-regulated macrophage M2 polarization and phagocytic activity.
Collapse
Affiliation(s)
- Xiaodong Weng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China; Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jing Li
- Department of Ophthamology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430060, China
| | - Qiunong Guan
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Haimei Zhao
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada; College of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Zihuan Wang
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada; First Clinical Medical School, Southern Medical University, Guangzhou, 510000, China
| | - Martin E Gleave
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Christopher Yc Nguan
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Caigan Du
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
11
|
Tang J, Zhu H, Liu S, Wang H, Huang X, Yan Y, Wang L, Zhou B. Sca1 marks a reserve endothelial progenitor population that preferentially expand after injury. Cell Discov 2021; 7:88. [PMID: 34580277 PMCID: PMC8476504 DOI: 10.1038/s41421-021-00303-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 06/30/2021] [Indexed: 11/10/2022] Open
Affiliation(s)
- Juan Tang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Huan Zhu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Shaoyan Liu
- Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haixiao Wang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xiuzhen Huang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yan Yan
- Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lixin Wang
- Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, China. .,School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
12
|
Du X, Gu H, Sun Y, Hu Y. Ly-6D of Japanese flounder (Paralichthys olivaceus) functions as a complement regulator and promotes host clearance of pathogen. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 122:104104. [PMID: 33891970 DOI: 10.1016/j.dci.2021.104104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 06/12/2023]
Abstract
The Lymphocyte antigen-6 (Ly-6) superfamily has been considered to play an important role in the innate immunity of mammals. The functions of Ly-6 proteins are diverse since their low sequence homology. Currently, the function of Ly-6D, a member of Ly-6 family proteins, is completely unknown in teleost. In the present study, we identified and characterized a Ly-6D homologue (named PoLy-6D) from the teleost fish Paralichthys olivaceus and examined its immune function. PoLy-6D possesses a hydrophobic signal peptide, a LU domain including a conserved "LXCXXC" motif in N-terminus and a "CCXXXXCN" motif in C-terminus. Under normal physiological condition, PoLy-6D expression distributes in all the examined tissues, the highest three tissues are successively spleen, head kidney, and blood. When infected by extracellular and intracellular bacterial pathogens and viral pathogen, PoLy-6D expression was induced and the patterns vary with different types of microbial pathogens infection and different immune tissues. In vitro experiment showed recombinant PoLy-6D (rPoLy-6D) inhibited the lysis of rabbit red blood cells by serum and selectively improved bacterial survival in serum. After serum were treated by antibody of rPoLy-6D, bacteriostatic effect of serum was obviously enhanced. These results indicate the importance of PoLy-6D as a complement regulator. rPoLy-6D possessed the binding activity to multiple bacteria but did not exhibit antimicrobial activities. The interaction between rPoLy-6D and bacteria suggests that PoLy-6D is involved in host clearance of pathogens probably by serving as a receptor for pathogens. Overexpression of PoLy-6D in vivo promoted the host defense against invading E. piscicida. These findings add new insights into the regulation mechanism of the complement system in teleost and emphasize the importance of Ly-6D products for the control of pathogen infection.
Collapse
Affiliation(s)
- Xiangyu Du
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, Hainan, 570228, PR China; Institute of Tropical Bioscience and Biotechnology, Hainan Academy of Tropical Agricultural Resource, CATAS, Haikou, 571101, China
| | - Hanjie Gu
- Institute of Tropical Bioscience and Biotechnology, Hainan Academy of Tropical Agricultural Resource, CATAS, Haikou, 571101, China; Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Haikou, 571101, China
| | - Yun Sun
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, Hainan, 570228, PR China.
| | - Yonghua Hu
- Institute of Tropical Bioscience and Biotechnology, Hainan Academy of Tropical Agricultural Resource, CATAS, Haikou, 571101, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China; Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Haikou, 571101, China.
| |
Collapse
|
13
|
So HS, Kim MG, Lee JC, Kook SH. Glucose oxidase induces mobilization of long-term repopulating hematopoietic cells in mice. Stem Cells Transl Med 2021; 10:1446-1453. [PMID: 34160898 PMCID: PMC8459634 DOI: 10.1002/sctm.20-0514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 05/16/2021] [Accepted: 06/06/2021] [Indexed: 12/17/2022] Open
Abstract
Hematopoietic stem progenitor cells (HSPCs) mobilized to peripheral blood, rather than those remaining in the bone marrow (BM), are commonly used as stem cell source in the clinic. As reactive oxygen species (ROS) are suggested as mediator of HSPC mobilization, we examined the impacts of glucose oxidase (GO) on peripheral mobilization of BM HSPCs and the associated mechanisms. Intravenous injection of GO induced HSPC mobilization even by single treatment, and the GO‐mobilized cells maintained their long‐term reconstituting and differentiating potentials in conditioned recipients. GO‐injected mice lived a normal life without adverse effects such as stem cell senescence, hematopoietic disorders, and blood parameter alteration. The mobilization effect of GO was even evident in animal models showing poor mobilization, such as old, 5‐fluorouracil‐treated, or alendronate‐treated mice. Importantly, combined injection of GO with granulocyte colony‐stimulating factor (G‐CSF) and/or AMD3100 enhanced more greatly HSPC mobilization than did G‐CSF, AMD3100, or both. The GO‐stimulated HSPC mobilization was almost completely attenuated by n‐acetyl‐L‐cysteine treatment. Collectively, our results not only highlight the potential role of GO in HSPC mobilization via ROS signaling, but also provide a GO‐based new strategy to improve HSPC mobilization in poorly mobilizing allogeneic or autologous donors via combination with G‐CSF and/or AMD3100.
Collapse
Affiliation(s)
- Han-Sol So
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| | - Min-Guk Kim
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea.,Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
| | - Jeong-Chae Lee
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea.,Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
| | - Sung-Ho Kook
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| |
Collapse
|
14
|
Tan YY, Zhang Y, Li B, Ou YW, Xie SJ, Chen PP, Mei SQ, Huang QJ, Zheng LL, Qu LH. PERK Signaling Controls Myoblast Differentiation by Regulating MicroRNA Networks. Front Cell Dev Biol 2021; 9:670435. [PMID: 34124052 PMCID: PMC8193987 DOI: 10.3389/fcell.2021.670435] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 03/31/2021] [Indexed: 01/29/2023] Open
Abstract
The unfolded protein response (UPR) plays important roles in various cells that have a high demand for protein folding, which are involved in the process of cell differentiation and development. Here, we separately knocked down the three sensors of the UPR in myoblasts and found that PERK knockdown led to a marked transformation in myoblasts from a fusiform to a rounded morphology, which suggests that PERK is required for early myoblast differentiation. Interestingly, knocking down PERK induced reprogramming of C2C12 myoblasts into stem-like cells by altering the miRNA networks associated with differentiation and stemness maintenance, and the PERK-ATF4 signaling pathway transactivated muscle differentiation-associated miRNAs in the early stage of myoblast differentiation. Furthermore, we identified Ppp1cc as a direct target gene of miR-128 regulated by the PERK signaling pathway and showed that its repression is critical for a feedback loop that regulates the activity of UPR-associated signaling pathways, leading to cell migration, cell fusion, endoplasmic reticulum expansion, and myotube formation during myoblast differentiation. Subsequently, we found that the RNA-binding protein ARPP21, encoded by the host gene of miR-128-2, antagonized miR-128 activity by competing with it to bind to the 3' untranslated region (UTR) of Ppp1cc to maintain the balance of the differentiation state. Together, these results reveal the crucial role of PERK signaling in myoblast maintenance and differentiation and identify the mechanism underlying the role of UPR signaling as a major regulator of miRNA networks during early differentiation of myoblasts.
Collapse
Affiliation(s)
- Ye-Ya Tan
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yin Zhang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Research Center of Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bin Li
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yang-Wen Ou
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Guangzhou, University of Chinese Medicine, Guangzhou, China
| | - Shu-Juan Xie
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Pei-Pei Chen
- AMI Key Laboratory of Chinese Medicine in Guangzhou, Guangdong Province Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Science, Guangzhou, China
| | - Shi-Qiang Mei
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Qiao-Juan Huang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ling-Ling Zheng
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Liang-Hu Qu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
15
|
Doshida Y, Sano H, Iwabuchi S, Aigaki T, Yoshida M, Hashimoto S, Ishigami A. Age-associated changes in the transcriptomes of non-cultured adipose-derived stem cells from young and old mice assessed via single-cell transcriptome analysis. PLoS One 2020; 15:e0242171. [PMID: 33237970 PMCID: PMC7688117 DOI: 10.1371/journal.pone.0242171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Adipose-derived stem cells (ASCs) exhibit self-renewal and pluripotency. The differentiation potency of ASCs has been reported to deteriorate with aging; however, relevant studies used ASCs that were isolated and subcultured several times. It is still unclear whether subcultured ASCs accurately reflect the in vivo state. To address this question, we used freshly isolated stromal vascular fractions (SVFs) and performed comprehensive single-cell transcriptome analysis. In this study, we identified three cell populations as putative ASC candidates in SVFs and three novel ASC-related genes: Adamts7, Snai2, and Tgfbr1, that are reported to be negative regulators of cell differentiation. Moreover, we identified age-associated high gene expression levels of Adamts7, Egfr, and Igfbp4 in the earliest differentiation stage of ASCs. These results suggest that aging may make it impossible to maintain the stringency of the regulation of the expression of some genes related to ASC differentiation.
Collapse
Affiliation(s)
- Yuta Doshida
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
- Department of Biological Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Haruka Sano
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
- Department of Life Science and Bioethics, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sadahiro Iwabuchi
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Toshiro Aigaki
- Department of Biological Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Masayuki Yoshida
- Department of Life Science and Bioethics, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinichi Hashimoto
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
- Department of Biological Sciences, Tokyo Metropolitan University, Tokyo, Japan
| |
Collapse
|
16
|
Zhang L, Mack R, Breslin P, Zhang J. Molecular and cellular mechanisms of aging in hematopoietic stem cells and their niches. J Hematol Oncol 2020; 13:157. [PMID: 33228751 PMCID: PMC7686726 DOI: 10.1186/s13045-020-00994-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 02/08/2023] Open
Abstract
Aging drives the genetic and epigenetic changes that result in a decline in hematopoietic stem cell (HSC) functioning. Such changes lead to aging-related hematopoietic/immune impairments and hematopoietic disorders. Understanding how such changes are initiated and how they progress will help in the development of medications that could improve the quality life for the elderly and to treat and possibly prevent aging-related hematopoietic diseases. Here, we review the most recent advances in research into HSC aging and discuss the role of HSC-intrinsic events, as well as those that relate to the aging bone marrow niche microenvironment in the overall processes of HSC aging. In addition, we discuss the potential mechanisms by which HSC aging is regulated.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Ryan Mack
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Peter Breslin
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA.,Departments of Molecular/Cellular Physiology and Department of Biology, Loyola University Medical Center and Loyola University Chicago, Chicago, IL, 60660, USA
| | - Jiwang Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA. .,Department of Pathology, Loyola University Medical Center, Maywood, IL, 60153, USA.
| |
Collapse
|
17
|
Christensen R, Gunnarsson AP, Jensen UB. The role of stem cell antigen-1/Lymphocyte antigen 6A-2/6E-1 knock out in murine epidermis. Stem Cell Res 2020; 49:102047. [PMID: 33157392 DOI: 10.1016/j.scr.2020.102047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/30/2020] [Accepted: 10/09/2020] [Indexed: 11/27/2022] Open
Abstract
Stem Cell Antigen-1 (SCA-1) is a central positive marker for isolating stem cells in several tissues in the mouse. However, for the epidermis, this appears to be the opposite since lack of SCA-1 has been shown to identify keratinocyte populations with progenitor characteristics. This study investigates the effect of SCA-1 knockout in murine keratinocytes. We compared Sca-1EGFP/EGFP knockout and wildtype mice with respect to the three-dimensional morphology of the epidermis, performed functional assays, and generated gene expression profiles on FACS sorted cells. There were no morphological abnormalities on skin, fur, or hair follicles in transgenic knockout mice compared to wild type mice. SCA-1 knockout keratinocytes showed significantly reduced colony-forming efficiency, colony size and proliferation rate in vitro, however, SCA-1 knockout did not alter wound healing efficiency or keratinocyte proliferation rate in vivo. Moreover, gene expression profiling shows that the effect from knockout of SCA-1 in keratinocytes is dissimilar from what has been observed in other tissues. Additionally, tumor assay indicated that SCA-1 knockout decreases the number of formed papillomas. The results indicate a more complex role for SCA-1, which might differ between epidermal keratinocytes during homeostasis and activated conditions.
Collapse
Affiliation(s)
- Rikke Christensen
- Department of Clinical Genetics, Aarhus University Hospital, Brendstrupgaardsvej 21C, 8200 Aarhus N, Denmark; Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark.
| | - Anders Patrik Gunnarsson
- Department of Clinical Genetics, Aarhus University Hospital, Brendstrupgaardsvej 21C, 8200 Aarhus N, Denmark; Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark.
| | - Uffe Birk Jensen
- Department of Clinical Genetics, Aarhus University Hospital, Brendstrupgaardsvej 21C, 8200 Aarhus N, Denmark.
| |
Collapse
|
18
|
Cho J, Park E. Ferulic acid maintains the self-renewal capacity of embryo stem cells and adipose-derived mesenchymal stem cells in high fat diet-induced obese mice. J Nutr Biochem 2019; 77:108327. [PMID: 31926451 DOI: 10.1016/j.jnutbio.2019.108327] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/18/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022]
Abstract
Self-renewal is required for embryo stem cells (ESCs) and adipose-derived mesenchymal stem cells (ADMSCs). This study examined the ability of ferulic acid in mouse ESCs and ADMSCs, in a high fat diet-induced mouse model. Initially, five natural compounds of ferulic acid, xanthohumol, curcumin, ascorbic acid, and quercetin were screened in ESCs using an alkaline phosphate +(AP+) assay, as a self-renewal biomarker. A ferulic acid treatment was the highest AP+ staining in hop-hit screening compounds. Also a ferulic acid increased Nanog mRNA levels in ESCs. The in vivo effects of ferulic acid were next examined in an obese mouse model. C57BL/6 J male mice were fed either a high fat diet (HFD) or control diet with ferulic acid (5 g/kg diet) for 8 weeks. Ferulic acid exhibited weight loss and improved glucose homeostasis, lipid profiling, and hepatic steatosis in a HFD-induced mouse model. Next, ADMSCs (Sca-1+CD45-), a hallmark of fat stem cells, were then isolated and quantified from mouse abdominal adipose tissue. A HFD decreased the Sca-1+CD45- cell population of ADMSCs, but HFD-induced obese mice given ferulic acid showed an increased the Sca-1+CD45- cell population of ADMSCs. Moreover, ferulic acid enhanced NANOG mRNA levels in human ADMSCs and its related gene mRNA expression. Overall, this study suggests that ferulic acid preserves self-renewal in ESCs, and contributes to ADMSCs self-renewal and effective weight control in obesity.
Collapse
Affiliation(s)
- Jinkyung Cho
- Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, NY., USA
| | - Eunmi Park
- Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, NY., USA; Department of Food and Nutrition, Hannam University, Daejeon 306-791, Republic of Korea.
| |
Collapse
|
19
|
Yu J, Murthy V, Liu SL. Relating GPI-Anchored Ly6 Proteins uPAR and CD59 to Viral Infection. Viruses 2019; 11:E1060. [PMID: 31739586 PMCID: PMC6893729 DOI: 10.3390/v11111060] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/10/2019] [Accepted: 11/12/2019] [Indexed: 12/30/2022] Open
Abstract
The Ly6 (lymphocyte antigen-6)/uPAR (urokinase-type plasminogen activator receptor) superfamily protein is a group of molecules that share limited sequence homology but conserved three-fingered structures. Despite diverse cellular functions, such as in regulating host immunity, cell adhesion, and migration, the physiological roles of these factors in vivo remain poorly characterized. Notably, increasing research has focused on the interplays between Ly6/uPAR proteins and viral pathogens, the results of which have provided new insight into viral entry and virus-host interactions. While LY6E (lymphocyte antigen 6 family member E), one key member of the Ly6E/uPAR-family proteins, has been extensively studied, other members have not been well characterized. Here, we summarize current knowledge of Ly6/uPAR proteins related to viral infection, with a focus on uPAR and CD59. Our goal is to provide an up-to-date view of the Ly6/uPAR-family proteins and associated virus-host interaction and viral pathogenesis.
Collapse
Affiliation(s)
- Jingyou Yu
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA; (J.Y.); (V.M.)
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA
| | - Vaibhav Murthy
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA; (J.Y.); (V.M.)
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA
| | - Shan-Lu Liu
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA; (J.Y.); (V.M.)
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
- Viruses and Emerging Pathogens Program, Infectious Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
20
|
Zhang L, Sultana N, Yan J, Yang F, Chen F, Chepurko E, Yang FC, Du Q, Zangi L, Xu M, Bu L, Cai CL. Cardiac Sca-1 + Cells Are Not Intrinsic Stem Cells for Myocardial Development, Renewal, and Repair. Circulation 2019; 138:2919-2930. [PMID: 30566018 DOI: 10.1161/circulationaha.118.035200] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND For more than a decade, Sca-1+ cells within the mouse heart have been widely recognized as a stem cell population with multipotency that can give rise to cardiomyocytes, endothelial cells, and smooth muscle cells in vitro and after cardiac grafting. However, the developmental origin and authentic nature of these cells remain elusive. METHODS Here, we used a series of high-fidelity genetic mouse models to characterize the identity and regenerative potential of cardiac resident Sca-1+ cells. RESULTS With these novel genetic tools, we found that Sca-1 does not label cardiac precursor cells during early embryonic heart formation. Postnatal cardiac resident Sca-1+ cells are in fact a pure endothelial cell population. They retain endothelial properties and exhibit minimal cardiomyogenic potential during development, normal aging and upon ischemic injury. CONCLUSIONS Our study provides definitive insights into the nature of cardiac resident Sca-1+ cells. The observations challenge the current dogma that cardiac resident Sca-1+ cells are intrinsic stem cells for myocardial development, renewal, and repair, and suggest that the mechanisms of transplanted Sca-1+ cells in heart repair need to be reassessed.
Collapse
Affiliation(s)
- Lu Zhang
- Riley Heart Research Center and Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L. Zhang, F.Y., C.-L.C.).,Department of Developmental and Regenerative Biology and The Black Family Stem Cell Institute (L. Zhang, N.S., F.Y., C.-L.C.), Icahn School of Medicine at Mount Sinai, New York
| | - Nishat Sultana
- Department of Developmental and Regenerative Biology and The Black Family Stem Cell Institute (L. Zhang, N.S., F.Y., C.-L.C.), Icahn School of Medicine at Mount Sinai, New York.,Department of Medicine and Cardiovascular Research Center (N.S., E.C., L. Zangi), Icahn School of Medicine at Mount Sinai, New York
| | - Jianyun Yan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, and Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, China (J.Y.)
| | - Fan Yang
- Riley Heart Research Center and Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L. Zhang, F.Y., C.-L.C.).,Department of Developmental and Regenerative Biology and The Black Family Stem Cell Institute (L. Zhang, N.S., F.Y., C.-L.C.), Icahn School of Medicine at Mount Sinai, New York
| | - Fuxue Chen
- College of Life Sciences, Shanghai University, China (F.C.)
| | - Elena Chepurko
- Department of Medicine and Cardiovascular Research Center (N.S., E.C., L. Zangi), Icahn School of Medicine at Mount Sinai, New York
| | - Feng-Chun Yang
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, FL (F.-C.Y., Q.D., M.X.)
| | - Qinghua Du
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, FL (F.-C.Y., Q.D., M.X.)
| | - Lior Zangi
- Department of Medicine and Cardiovascular Research Center (N.S., E.C., L. Zangi), Icahn School of Medicine at Mount Sinai, New York
| | - Mingjiang Xu
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, FL (F.-C.Y., Q.D., M.X.)
| | - Lei Bu
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY (L.B.)
| | - Chen-Leng Cai
- Riley Heart Research Center and Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L. Zhang, F.Y., C.-L.C.).,Department of Developmental and Regenerative Biology and The Black Family Stem Cell Institute (L. Zhang, N.S., F.Y., C.-L.C.), Icahn School of Medicine at Mount Sinai, New York
| |
Collapse
|
21
|
Vagnozzi RJ, Sargent MA, Lin SCJ, Palpant NJ, Murry CE, Molkentin JD. Genetic Lineage Tracing of Sca-1 + Cells Reveals Endothelial but Not Myogenic Contribution to the Murine Heart. Circulation 2019; 138:2931-2939. [PMID: 29991486 DOI: 10.1161/circulationaha.118.035210] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND The adult mammalian heart displays a cardiomyocyte turnover rate of ≈1%/y throughout postnatal life and after injuries such as myocardial infarction (MI), but the question of which cell types drive this low level of new cardiomyocyte formation remains contentious. Cardiac-resident stem cells marked by stem cell antigen-1 (Sca-1, gene name Ly6a) have been proposed as an important source of cardiomyocyte renewal. However, the in vivo contribution of endogenous Sca-1+ cells to the heart at baseline or after MI has not been investigated. METHODS Here we generated Ly6a gene-targeted mice containing either a constitutive or an inducible Cre recombinase to perform genetic lineage tracing of Sca-1+ cells in vivo. RESULTS We observed that the contribution of endogenous Sca-1+ cells to the cardiomyocyte population in the heart was <0.005% throughout all of cardiac development, with aging, or after MI. In contrast, Sca-1+ cells abundantly contributed to the cardiac vasculature in mice during physiological growth and in the post-MI heart during cardiac remodeling. Specifically, Sca-1 lineage-traced endothelial cells expanded postnatally in the mouse heart after birth and into adulthood. Moreover, pulse labeling of Sca-1+ cells with an inducible Ly6a-MerCreMer allele also revealed a preferential expansion of Sca-1 lineage-traced endothelial cells after MI injury in the mouse. CONCLUSIONS Cardiac-resident Sca-1+ cells are not significant contributors to cardiomyocyte renewal in vivo. However, cardiac Sca-1+ cells represent a subset of vascular endothelial cells that expand postnatally with enhanced responsiveness to pathological stress in vivo.
Collapse
Affiliation(s)
- Ronald J Vagnozzi
- Department of Pediatrics (R.J.V., M.A.S., S.-C.J.L., J.D.M.), Cincinnati Children's Hospital Medical Center, OH
| | - Michelle A Sargent
- Department of Pediatrics (R.J.V., M.A.S., S.-C.J.L., J.D.M.), Cincinnati Children's Hospital Medical Center, OH
| | - Suh-Chin J Lin
- Department of Pediatrics (R.J.V., M.A.S., S.-C.J.L., J.D.M.), Cincinnati Children's Hospital Medical Center, OH
| | - Nathan J Palpant
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia (N.J.P.)
| | - Charles E Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle (C.E.M.)
| | - Jeffery D Molkentin
- Department of Pediatrics (R.J.V., M.A.S., S.-C.J.L., J.D.M.), Cincinnati Children's Hospital Medical Center, OH.,Howard Hughes Medical Institute (J.D.M.), Cincinnati Children's Hospital Medical Center, OH
| |
Collapse
|
22
|
Langrzyk A, Nowak WN, Stępniewski J, Jaźwa A, Florczyk-Soluch U, Józkowicz A, Dulak J. Critical View on Mesenchymal Stromal Cells in Regenerative Medicine. Antioxid Redox Signal 2018; 29:169-190. [PMID: 28874054 DOI: 10.1089/ars.2017.7159] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE The belief in the potency of stem cells has resulted in the medical applications of numerous cell types for organ repair, often with the low adherence to methodological stringency. Such uncritical enthusiasm is mainly presented in the approaches employing so-called mesenchymal stem cells (MSC), for the treatment of numerous, unrelated conditions. However, it should be stressed that such broad clinical applications of MSC are mostly based on the belief that MSC can efficiently differentiate into multiple cell types, not only osteoblasts, chondrocytes and adipose cells. Recent Advances: Studies employing lineage tracing established more promising markers to characterize MSC identity and localization in vivo and confirmed the differences between MSC isolated from various organs. Furthermore, preclinical and clinical experiments proved that transdifferentiation of MSC is unlikely to contribute to repair of numerous tissues, including the heart. Therefore, the salvage hypotheses, like MSC fusion with cells in target organs or the paracrine mechanisms, were proposed to justify the widespread application of MSC and to explain transient, if any, effects. CRITICAL ISSUES The lack of standardization concerning the cells markers, their origin and particularly the absence of stringent functional characterization of MSC, leads to propagation of the worrying hype despite the lack of convincing therapeutic efficiency of MSC. FUTURE DIRECTIONS The adherence to rigorous methodological rules is necessary to prevent the application of procedures which can be dangerous for patients and scientific research on the medical application of stem cells. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
| | - Witold N Nowak
- 2 Cardiovascular Division, King's College London , London, United Kingdom .,3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Jacek Stępniewski
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Agnieszka Jaźwa
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Urszula Florczyk-Soluch
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Alicja Józkowicz
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Józef Dulak
- 1 Kardio-Med Silesia , Zabrze, Poland .,3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| |
Collapse
|
23
|
Tokoro Y, Yamada Y, Takayanagi SI, Hagiwara T. 57R2A, a newly established monoclonal antibody against mouse GPR56, marks long-term repopulating hematopoietic stem cells. Exp Hematol 2017; 59:51-59.e1. [PMID: 29225194 DOI: 10.1016/j.exphem.2017.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 11/29/2017] [Accepted: 12/01/2017] [Indexed: 11/25/2022]
Abstract
GPR56 molecule, a G-protein-coupled receptor, was suggested to be expressed in mouse hematopoietic stem cells (HSCs) by gene expression analyses. However, little is known about the cell surface expression of GPR56 protein in mouse HSCs due to the absence of an appropriate monoclonal antibody against GPR56 for flow cytometry analyses. In the present study, we established a novel monoclonal antibody against mouse GPR56 (57R2A) to examine the expression and distribution of GPR56 protein in HSCs. A flow cytometry analysis using 57R2A showed that GPR56 was highly expressed in the CD34-, c-Kit+, Sca-1+, lineage-negative (Lin-) fraction, which are highly enriched with HSCs. The competitive long-term repopulation (LTR) assay showed that LTR cells were included only within the GPR56+ fraction (≤15%) of bone marrow mononuclear cells (BMMNCs), but not within the remaining GPR56- fraction (85%), suggesting that all HSCs express GPR56 protein on their surface. Furthermore, we showed that double staining of BMMNCs with only 57R2A and AMM2 (monoclonal antibody against the HSC marker MPL) enabled enrichment of all LTR cells in the double-positive fraction (0.8% of BMMNCs), within which the LTR potency was consistent with the expression of both GPR56 and MPL. In conclusion, these findings for 57R2A suggest that all HSCs in mouse BMMNCs express GPR56 protein on their surface and that GPR56 is a positive marker for HSCs.
Collapse
Affiliation(s)
- Yusuke Tokoro
- Kyowa Hakko Kirin Co., Ltd., Tokyo, Japan; Graduate School of Agriculture, Kyoto University, Kyoto, Japan.
| | | | | | | |
Collapse
|
24
|
Constitutive transgene expression of Stem Cell Antigen-1 in the hair follicle alters the sensitivity to tumor formation and progression. Stem Cell Res 2017; 23:109-118. [DOI: 10.1016/j.scr.2017.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/06/2017] [Indexed: 02/05/2023] Open
|
25
|
James AW, Shen J, Tsuei R, Nguyen A, Khadarian K, Meyers CA, Pan HC, Li W, Kwak JH, Asatrian G, Culiat CT, Lee M, Ting K, Zhang X, Soo C. NELL-1 induces Sca-1+ mesenchymal progenitor cell expansion in models of bone maintenance and repair. JCI Insight 2017; 2:92573. [PMID: 28614787 PMCID: PMC5470886 DOI: 10.1172/jci.insight.92573] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 05/05/2017] [Indexed: 12/25/2022] Open
Abstract
NELL-1 is a secreted, osteogenic protein first discovered to control ossification of the cranial skeleton. Recently, NELL-1 has been implicated in bone maintenance. However, the cellular determinants of NELL-1's bone-forming effects are still unknown. Here, recombinant human NELL-1 (rhNELL-1) implantation was examined in a clinically relevant nonhuman primate lumbar spinal fusion model. Prolonged rhNELL-1 protein release was achieved using an apatite-coated β-tricalcium phosphate carrier, resulting in a local influx of stem cell antigen-1-positive (Sca-1+) mesenchymal progenitor cells (MPCs), and complete osseous fusion across all samples (100% spinal fusion rate). Murine studies revealed that Nell-1 haploinsufficiency results in marked reductions in the numbers of Sca-1+CD45-CD31- bone marrow MPCs associated with low bone mass. Conversely, rhNELL-1 systemic administration in mice showed a marked anabolic effect accompanied by increased numbers of Sca-1+CD45-CD31- bone marrow MPCs. Mechanistically, rhNELL-1 induces Sca-1 transcription among MPCs, in a process requiring intact Wnt/β-catenin signaling. In summary, NELL-1 effectively induces bone formation across small and large animal models either via local implantation or intravenous delivery. NELL-1 induces an expansion of a bone marrow subset of MPCs with Sca-1 expression. These findings provide compelling justification for the clinical translation of a NELL-1-based therapy for local or systemic bone formation.
Collapse
Affiliation(s)
- Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA.,UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California, USA
| | - Jia Shen
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Rebecca Tsuei
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Alan Nguyen
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Kevork Khadarian
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Carolyn A Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hsin Chuan Pan
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Weiming Li
- Department of Orthopedics, The First Clinical Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jin H Kwak
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Greg Asatrian
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | | | - Min Lee
- Section of Biomaterials, School of Dentistry, UCLA, Los Angeles, California, USA
| | - Kang Ting
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Xinli Zhang
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Chia Soo
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California, USA.,Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| |
Collapse
|
26
|
Diet-induced obesity regulates adipose-resident stromal cell quantity and extracellular matrix gene expression. Stem Cell Res 2016; 17:181-90. [DOI: 10.1016/j.scr.2016.07.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 06/30/2016] [Accepted: 07/03/2016] [Indexed: 01/10/2023] Open
|
27
|
Organization, evolution and functions of the human and mouse Ly6/uPAR family genes. Hum Genomics 2016; 10:10. [PMID: 27098205 PMCID: PMC4839075 DOI: 10.1186/s40246-016-0074-2] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/14/2016] [Indexed: 01/08/2023] Open
Abstract
Members of the lymphocyte antigen-6 (Ly6)/urokinase-type plasminogen activator receptor (uPAR) superfamily of proteins are cysteine-rich proteins characterized by a distinct disulfide bridge pattern that creates the three-finger Ly6/uPAR (LU) domain. Although the Ly6/uPAR family proteins share a common structure, their expression patterns and functions vary. To date, 35 human and 61 mouse Ly6/uPAR family members have been identified. Based on their subcellular localization, these proteins are further classified as GPI-anchored on the cell membrane, or secreted. The genes encoding Ly6/uPAR family proteins are conserved across different species and are clustered in syntenic regions on human chromosomes 8, 19, 6 and 11, and mouse Chromosomes 15, 7, 17, and 9, respectively. Here, we review the human and mouse Ly6/uPAR family gene and protein structure and genomic organization, expression, functions, and evolution, and introduce new names for novel family members.
Collapse
|
28
|
Baustian C, Hanley S, Ceredig R. Isolation, selection and culture methods to enhance clonogenicity of mouse bone marrow derived mesenchymal stromal cell precursors. Stem Cell Res Ther 2015; 6:151. [PMID: 26303631 PMCID: PMC4549076 DOI: 10.1186/s13287-015-0139-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 07/24/2015] [Accepted: 07/24/2015] [Indexed: 11/10/2022] Open
Abstract
Introduction Conventionally cultured mouse bone marrow mesenchymal stromal cells (mBM-MSC) are a heterogeneous population that often initially contain contaminating haematopoietic cells. Variability in isolation methods, culture protocols and the lack of specific mBM MSC markers might explain this heterogeneity. The aim of this study is to optimise the isolation, culture conditions and selection of mBM-MSC. Methods Mouse BM-MSCs were isolated from crushed long bones (cBM) or flushed bone marrow (fBM) from 6–8 week old C57Bl/6 mice. These subpopulations were analysed by flow cytometry using commonly used mBM-MSC cell surface marker, e.g. Sca-1, CD29 and CD44. Cells were cultured and expanded in vitro in hypoxic conditions of either 2 % or 5 % oxygen. Cell sorting and qRT-PCR was used to determine transcript levels of stem cell and lineage related genes in individual subpopulations. Results During early passaging not only do contaminating haematopoietic cells disappear, but there is a change in the phenotype of mBM-MSC affecting particularly CD44 and Sca-1 expression. By fluorescence activated cell sorting of CD45−/Ter119− mBM stroma based on Sca-1 expression and expansion in hypoxic conditions, we show that Sca-1+ cells had higher CFU-F frequencies and showed enhanced proliferation compared with Sca-1− cells. As evaluated by in vitro assays and qRT-PCR, these cells presented in vitro tri-lineage differentiation along osteocyte, chondrocyte, and adipocyte lineages. Finally, by prospective isolation of Sca-1+PDGFRα+CD90+ cells we have isolated mBM-MSC on a single cell level, achieving a CFU-F frequency of 1/4. Functional investigations demonstrated that these MSC clones inhibited T-lymphocyte proliferation. Conclusion By positive selection using a combination of antibodies to Sca-1, CD90 and PDGFRα and culturing in hypoxia, we have found a subpopulation of BM cells from C57Bl/6 mice with a CFU-F cloning efficiency of 1/4. To our knowledge these results represent the highest frequencies of mouse MSC cloning from C57Bl/6 mice yet reported. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0139-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Claas Baustian
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and School of Medicine, National University of Ireland, Galway, Ireland.
| | - Shirley Hanley
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and School of Medicine, National University of Ireland, Galway, Ireland.
| | - Rhodri Ceredig
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and School of Medicine, National University of Ireland, Galway, Ireland. .,Biosciences, National University of Ireland Galway, Newcastle Road, Dangan, Galway, Ireland.
| |
Collapse
|
29
|
Camarata TD, Weaver GC, Vasilyev A, Arnaout MA. Negative Regulation of TGFβ Signaling by Stem Cell Antigen-1 Protects against Ischemic Acute Kidney Injury. PLoS One 2015; 10:e0129561. [PMID: 26053644 PMCID: PMC4460127 DOI: 10.1371/journal.pone.0129561] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/10/2015] [Indexed: 12/22/2022] Open
Abstract
Acute kidney injury, often caused by an ischemic insult, is associated with significant short-term morbidity and mortality, and increased risk of chronic kidney disease. The factors affecting the renal response to injury following ischemia and reperfusion remain to be clarified. We found that the Stem cell antigen-1 (Sca-1), commonly used as a stem cell marker, is heavily expressed in renal tubules of the adult mouse kidney. We evaluated its potential role in the kidney using Sca-1 knockout mice submitted to acute ischemia reperfusion injury (IRI), as well as cultured renal proximal tubular cells in which Sca-1 was stably silenced with shRNA. IRI induced more severe injury in Sca-1 null kidneys, as assessed by increased expression of Kim-1 and Ngal, rise in serum creatinine, abnormal pathology, and increased apoptosis of tubular epithelium, and persistent significant renal injury at day 7 post IRI, when recovery of renal function in control animals was nearly complete. Serum creatinine, Kim-1 and Ngal were slightly but significantly elevated even in uninjured Sca-1-/- kidneys. Sca-1 constitutively bound both TGFβ receptors I and II in cultured normal proximal tubular epithelial cells. Its genetic loss or silencing lead to constitutive TGFβ receptor—mediated activation of canonical Smad signaling even in the absence of ligand and to KIM-1 expression in the silenced cells. These studies demonstrate that by normally repressing TGFβ-mediated canonical Smad signaling, Sca-1 plays an important in renal epithelial cell homeostasis and in recovery of renal function following ischemic acute kidney injury.
Collapse
Affiliation(s)
- Troy D. Camarata
- Leukocyte Biology & Inflammation Program, Renal Division and Department of Medicine Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Grant C. Weaver
- Leukocyte Biology & Inflammation Program, Renal Division and Department of Medicine Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Alexandr Vasilyev
- Leukocyte Biology & Inflammation Program, Renal Division and Department of Medicine Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - M. Amin Arnaout
- Leukocyte Biology & Inflammation Program, Renal Division and Department of Medicine Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- Center For Regenerative Medicine, Department of Medicine, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
30
|
Harada K, Harada T, Ferdous T, Takenawa T, Ueyama Y. Osteogenic cell fractions isolated from mouse tongue muscle. Mol Med Rep 2015; 12:31-6. [PMID: 25684092 PMCID: PMC4438915 DOI: 10.3892/mmr.2015.3350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 11/07/2014] [Indexed: 01/06/2023] Open
Abstract
The use of stem cells represents a promising approach for the treatment of bone defects. However, successful treatments rely upon the availability of cells that are easily obtained and that appropriately differentiate into osteoblasts. The tongue potentially represents a source of autologous cells for such purposes. In the present study, the ability of stem cell antigen-1 (Sca-1) positive cells derived from tongue muscle to differentiate into osteoblasts was investigated. The tongue muscles were excised from Jcl-ICR mice and tongue muscle-derived Sca-1-positive cells (TDSCs) were isolated from the tongue muscle using a magnetic cell separation system with microbeads. TDSCs were cultured in plastic dishes or gelatin sponges of β-tricalcium phosphate (β-TCP) with bone differentiation-inducing medium. The expression of osteogenic markers (Runx2, osterix, alkaline phosphatase, fibronectin, osteocalcin, osteonectin and osteopontin) was investigated in cultured TDSCs by western blot analysis. The formation of mineralized matrices was examined using alizarin red S and Von Kossa staining. Bone formation was investigated in cultured TDSCs by hematoxylin-eosin staining and immunohistochemstry. In the present study, the expression of Sca-1 in mouse tongue muscle was demonstrated and TDSCs were isolated at high purity. TDSCs differentiated into cells of osteoblast lineage, as demonstrated by the upregulation of osteoblastic marker expression. The formation of mineralized matrices was confirmed by alizarin red S or Von Kossa staining in vitro. Bone formation was observed in the gelatin sponges of β-TCP, which were subsequently implanted under the skin of the backs of nude mice. These results suggested that TDSCs retain their osteogenic differentiation potential and therefore the tongue muscle may be used as a source of stem cells for bone regeneration.
Collapse
Affiliation(s)
- Koji Harada
- Department of Oral and Maxillofacial Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755‑8505, Japan
| | - Toyoko Harada
- Department of Oral and Maxillofacial Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755‑8505, Japan
| | - Tarannum Ferdous
- Department of Oral and Maxillofacial Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755‑8505, Japan
| | - Takanori Takenawa
- Department of Oral and Maxillofacial Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755‑8505, Japan
| | - Yoshiya Ueyama
- Department of Oral and Maxillofacial Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755‑8505, Japan
| |
Collapse
|
31
|
Crisostomo V, Casado JG, Baez-Diaz C, Blazquez R, Sanchez-Margallo FM. Allogeneic cardiac stem cell administration for acute myocardial infarction. Expert Rev Cardiovasc Ther 2015; 13:285-99. [DOI: 10.1586/14779072.2015.1011621] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
32
|
Ge Z, Lal S, Le TYL, Dos Remedios C, Chong JJH. Cardiac stem cells: translation to human studies. Biophys Rev 2014; 7:127-139. [PMID: 28509972 DOI: 10.1007/s12551-014-0148-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/13/2014] [Indexed: 02/08/2023] Open
Abstract
The discovery of multiple classes of cardiac progenitor cells in the adult mammalian heart has generated hope for their use as a therapeutic in heart failure. However, successful results from animal models have not always yielded similar findings in human studies. Recent Phase I/II trials of c-Kit (SCIPIO) and cardiosphere-based (CADUCEUS) cardiac progenitor cells have demonstrated safety and some therapeutic efficacy. Gaps remain in our understanding of the origins, function and relationships between the different progenitor cell families, many of which are heterogeneous populations with overlapping definitions. Another challenge lies in the limitations of small animal models in replicating the human heart. Cryopreserved human cardiac tissue provides a readily available source of cardiac progenitor cells and may help address these questions. We review important findings and relative unknowns of the main classes of cardiac progenitor cells, highlighting differences between animal and human studies.
Collapse
Affiliation(s)
- Zijun Ge
- Bosch Institute, The University of Sydney, Sydney, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Sean Lal
- Bosch Institute, The University of Sydney, Sydney, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Thi Y L Le
- Department of Cardiology Westmead Hospital, Sydney, NSW, Australia.,Centre for Heart Research, Westmead Millennium Institute for Medical Research, 176 Hawkesbury Road, Westmead, Sydney, NSW, Australia, 2145
| | | | - James J H Chong
- Department of Cardiology Westmead Hospital, Sydney, NSW, Australia. .,Sydney Medical School, University of Sydney, Sydney, NSW, Australia. .,Centre for Heart Research, Westmead Millennium Institute for Medical Research, 176 Hawkesbury Road, Westmead, Sydney, NSW, Australia, 2145.
| |
Collapse
|
33
|
Abstract
Various stem cell types have been tested for regenerating damaged myocardium after myocardial infarction. However, the results of clinical trials have not been consistent, with only some of the trials reporting small improvements in cardiac function. It seems that engraftment and survival of injected cells is limited and transplanted stem cells either do not differentiate into cardiac cells or differentiate into only limited number of cardiac cells. The exact mechanism(s) of cardiac functional improvement by cell therapy are unclear, but paracrine effect may play a central role. The resident cardiac progenitor cells identified within the adult myocardium have distinct advantages over other stem cell types for cardiac cell therapy, as they are likely precommitted to the cardiovascular fate. However, isolating and expanding these cells from cardiac biopsies is a challenge. More recently, direct reprogramming of fibroblasts into cardiomyocytes has given new hope for myocardial regeneration. Here we will review different stem cells used in cardiac cell therapy with a focus on the native cardiac progenitor cells and briefly outline future directions of cardiac cell therapy.
Collapse
|
34
|
Valente M, Nascimento DS, Cumano A, Pinto-do-Ó P. Sca-1+ cardiac progenitor cells and heart-making: a critical synopsis. Stem Cells Dev 2014; 23:2263-73. [PMID: 24926741 DOI: 10.1089/scd.2014.0197] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The identification, in the adult, of cardiomyocyte turnover events and of cardiac progenitor cells (CPCs) has revolutionized the field of cardiovascular medicine. However, the low rate of CPCs differentiation events reported both in vitro and in vivo, even after injury, raised concerns on the biological significance of these subsets. In this Comprehensive Review, we discuss the current understanding of cardiac Lin(-)Sca-1(+) cells in light of what is also known for cellular compartments with similar phenotypes in other organs. The Lin(-)Sca-1(+) heart subset is heterogeneous and displays a mesenchymal profile, characterized by a limited ability to generate cardiomyocytes in vitro and in vivo, even after injury. There is no evidence for Sca-1 expression in embryonic cardiovascular progenitors. In other organs, Sca-1 expression is mainly observed on mesoderm-derived cells, although it is not restricted to stem/progenitor cell populations. It is urgent to determine, at a single cell level, to which extent cardiac Lin(-)Sca-1(+) cells overlap with the fibroblast compartment.
Collapse
Affiliation(s)
- Mariana Valente
- 1 Stem-Cell Microenvironments in Repair/Regeneration Team, Microenvironments for NewTherapies Group, INEB-Instituto Nacional de Engenharia Biomédica, Universidade do Porto , Porto, Portugal
| | | | | | | |
Collapse
|
35
|
Park KS, Shin SW, Choi JW, Um SH. Specific protein markers for stem cell cross-talk with neighboring cells in the environment. Int J Stem Cells 2014; 6:75-86. [PMID: 24386551 DOI: 10.15283/ijsc.2013.6.2.75] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2013] [Indexed: 01/04/2023] Open
Abstract
A stem cell interacts with the neighboring cells in its environment. To maintain a living organism's metabolism, either cell-cell or cell-environment interactions may be significant. Usually, these cells communicate with each other through biological signaling by interactive behaviors of primary proteins or complementary chemicals. The signaling intermediates offer the stem cell's functionality on its metabolism. With the rapid advent of omics technologies, various specific markers by which stem cells cooperate with their surroundings have been discovered and established. In this article, we review several stem cell markers used to communicate with either cancer or immune cells in the human body.
Collapse
Affiliation(s)
- Kyung Soo Park
- Department of Chemical and Biomolecular Engineering and Sogang University, Seoul, Korea
| | - Seung Won Shin
- School of Chemical Engineering and Sungkyunkwan University, Suwon
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering and Sogang University, Seoul, Korea ; Graduate School of Management of Technology, Sogang University, Seoul, Korea
| | - Soong Ho Um
- School of Chemical Engineering and Sungkyunkwan University, Suwon ; SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon
| |
Collapse
|
36
|
Malecki M, Putzer E, Sabo C, Foorohar A, Quach C, Stampe C, Beauchaine M, Tombokan X, Malecki R, Anderson M. Directed cardiomyogenesis of autologous human induced pluripotent stem cells recruited to infarcted myocardium with bioengineered antibodies. MOLECULAR AND CELLULAR THERAPIES 2014; 2:13. [PMID: 25132967 PMCID: PMC4131312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 04/01/2014] [Indexed: 11/21/2023]
Abstract
OBJECTIVE Myocardial infarctions constitute a major factor contributing to non-natural mortality world-wide. Clinical trials of myocardial regenerative therapy, currently pursued by cardiac surgeons, involve administration of stem cells into the hearts of patients suffering from myocardial infarctions. Unfortunately, surgical acquisition of these cells from bone marrow or heart is traumatic, retention of these cells to sites of therapeutic interventions is low, and directed differentiation of these cells in situ into cardiomyocytes is difficult. The specific aims of this work were: (1) to generate autologous, human, pluripotent, induced stem cells (ahiPSCs) from the peripheral blood of the patients suffering myocardial infarctions; (2) to bioengineer heterospecific antibodies (htAbs) and use them for recruitment of the ahiPSCs to infarcted myocardium; (3) to initiate in situ directed cardiomyogenesis of the ahiPSCs retained to infarcted myocardium. METHODS Peripheral blood was drawn from six patients scheduled for heart transplants. Mononuclear cells were isolated and reprogrammed, with plasmids carrying six genes (NANOG, POU5F1, SOX2, KLF4, LIN28A, MYC), to yield the ahiPSCs. Cardiac tissues were excised from the injured hearts of the patients, who received transplants during orthotopic surgery. These tissues were used to prepare in vitro models of stem cell therapy of infarcted myocardium. The htAbs were bioengineered, which simultaneously targeted receptors displayed on pluripotent stem cells (SSEA-4, SSEA-3, TRA-1-60, TRA-1-81) and proteins of myocardial sarcomeres (myosin, α-actinin, actin, titin). They were used to bridge the ahiPSCs to the infarcted myocardium. The retained ahiPSCs were directed with bone morphogenetic proteins and nicotinamides to differentiate towards myocardial lineage. RESULTS The patients' mononuclear cells were efficiently reprogrammed into the ahiPSCs. These ahiPSCs were administered to infarcted myocardium in in vitro models. They were recruited to and retained at the treated myocardium with higher efficacy and specificity, if were preceded with the htAbs, than with isotype antibodies or plain buffers. The retained cells differentiated into cardiomyocytes. CONCLUSIONS The proof of concept has been attained, for reprogramming the patients' blood mononuclear cells (PBMCs) into the ahiPSCs, recruiting these cells to infarcted myocardium, and initiating their cardiomyogenesis. This novel strategy is ready to support the ongoing clinical trials aimed at regeneration of infarcted myocardium.
Collapse
Affiliation(s)
- Marek Malecki
- />Phoenix Biomolecular Engineering Foundation, San Francisco, CA USA
- />National Magnetic Resonance Facility, National Institutes of Health, Madison, WI USA
- />University of Wisconsin, Madison, Madison, WI USA
| | - Emily Putzer
- />University of Wisconsin, Madison, Madison, WI USA
- />Latin American Youth Center, Washington, DC USA
| | - Chelsea Sabo
- />University of Wisconsin, Madison, Madison, WI USA
- />University of Sheffield, Sheffield, EU UK
| | - Afsoon Foorohar
- />Phoenix Biomolecular Engineering Foundation, San Francisco, CA USA
- />Western University, Lebanon, OR USA
| | - Carol Quach
- />Phoenix Biomolecular Engineering Foundation, San Francisco, CA USA
- />Western University, Pomona, CA USA
| | | | | | | | - Raf Malecki
- />San Francisco State University, San Francisco, CA USA
| | - Mark Anderson
- />National Magnetic Resonance Facility, National Institutes of Health, Madison, WI USA
- />University of Wisconsin, Madison, Madison, WI USA
| |
Collapse
|
37
|
Tokunaga M, Inoue M, Jiang Y, Barnes RH, Buchner DA, Chun TH. Fat depot-specific gene signature and ECM remodeling of Sca1(high) adipose-derived stem cells. Matrix Biol 2014; 36:28-38. [PMID: 24726953 DOI: 10.1016/j.matbio.2014.03.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 03/25/2014] [Accepted: 03/26/2014] [Indexed: 12/19/2022]
Abstract
Stem cell antigen-1 (Sca1 or Ly6A/E) is a cell surface marker that is widely expressed in mesenchymal stem cells, including adipose-derived stem cells (ASCs). We hypothesized that the fat depot-specific gene signature of Sca1(high) ASCs may play the major role in defining adipose tissue function and extracellular matrix (ECM) remodeling in a depot-specific manner. Herein we aimed to characterize the unique gene signature and ECM remodeling of Sca1(high) ASCs isolated from subcutaneous (inguinal) and visceral (epididymal) adipose tissues. Sca1(high) ASCs are found in the adventitia and perivascular areas of adipose tissues. Sca1(high) ASCs purified with magnetic-activated cell sorting (MACS) demonstrate dendrite or round shape with the higher expression of cytokines and chemokines (e.g., Il6, Cxcl1) and the lower expression of a glucose transporter (Glut1). Subcutaneous and visceral fat-derived Sca1(high) ASCs particularly differ in the gene expressions of adhesion and ECM molecules. While the expression of the major membrane-type collagenase (MMP14) is comparable between the groups, the expressions of secreted collagenases (MMP8 and MMP13) are higher in visceral Sca1(high) ASCs than in subcutaneous ASCs. Consistently, slow but focal MMP-dependent collagenolysis was observed with subcutaneous adipose tissue-derived vascular stromal cells, whereas rapid and bulk collagenolysis was observed with visceral adipose tissue-derived cells in MMP-dependent and -independent manners. These results suggest that the fat depot-specific gene signatures of ASCs may contribute to the distinct patterns of ECM remodeling and adipose function in different fat depots.
Collapse
Affiliation(s)
- Masakuni Tokunaga
- Department of Internal Medicine, Division of MEND, University of Michigan Medical School, United States
| | - Mayumi Inoue
- Department of Internal Medicine, Division of MEND, University of Michigan Medical School, United States
| | - Yibin Jiang
- Department of Internal Medicine, Division of MEND, University of Michigan Medical School, United States; Biointerfaces Institute, University of Michigan, United States
| | - Richard H Barnes
- Department of Internal Medicine, Division of MEND, University of Michigan Medical School, United States; Biointerfaces Institute, University of Michigan, United States
| | - David A Buchner
- Department of Genetics and Genome Sciences, Case Western Reserve University, United States
| | - Tae-Hwa Chun
- Department of Internal Medicine, Division of MEND, University of Michigan Medical School, United States; Biointerfaces Institute, University of Michigan, United States.
| |
Collapse
|
38
|
Donangelo I, Ren SG, Eigler T, Svendsen C, Melmed S. Sca1⁺ murine pituitary adenoma cells show tumor-growth advantage. Endocr Relat Cancer 2014; 21:203-16. [PMID: 24481638 PMCID: PMC3978815 DOI: 10.1530/erc-13-0229] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The role of tumor stem cells in benign tumors such as pituitary adenomas remains unclear. In this study, we investigated whether the cells within pituitary adenomas that spontaneously develop in Rb+/- mice are hierarchically distributed with a subset being responsible for tumor growth. Cells derived directly from such tumors grew as spheres in serum-free culture medium supplemented with epidermal growth factor and basic fibroblast growth factor. Some cells within growing pituitary tumor spheres (PTS) expressed common stem cell markers (Sca1, Sox2, Nestin, and CD133), but were devoid of hormone-positive differentiated cells. Under subsequent differentiating conditions (matrigel-coated growth surface), PTS expressed all six pituitary hormones. We next searched for specific markers of the stem cell population and isolated a Sca1(+) cell population that showed increased sphere formation potential, lower mRNA hormone expression, higher expression of stem cell markers (Notch1, Sox2, and Nestin), and increased proliferation rates. When transplanted into non-obese diabetic-severe combined immunodeficiency gamma mice brains, Sca1(+) pituitary tumor cells exhibited higher rates of tumor formation (brain tumors observed in 11/11 (100%) vs 7/12 (54%) of mice transplanted with Sca1(+) and Sca1(-) cells respectively). Magnetic resonance imaging and histological analysis of brain tumors showed that tumors derived from Sca1(+) pituitary tumor cells were also larger and plurihormonal. Our findings show that Sca1(+) cells derived from benign pituitary tumors exhibit an undifferentiated expression profile and tumor-proliferative advantages, and we propose that they could represent putative pituitary tumor stem/progenitor cells.
Collapse
Affiliation(s)
| | | | | | - Clive Svendsen
- Regenerative Medicine Institute, Cedars-Sinai Medical Center
| | | |
Collapse
|
39
|
Furtado MB, Costa MW, Pranoto EA, Salimova E, Pinto AR, Lam NT, Park A, Snider P, Chandran A, Harvey RP, Boyd R, Conway SJ, Pearson J, Kaye DM, Rosenthal NA. Cardiogenic genes expressed in cardiac fibroblasts contribute to heart development and repair. Circ Res 2014; 114:1422-34. [PMID: 24650916 DOI: 10.1161/circresaha.114.302530] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
RATIONALE Cardiac fibroblasts are critical to proper heart function through multiple interactions with the myocardial compartment, but appreciation of their contribution has suffered from incomplete characterization and lack of cell-specific markers. OBJECTIVE To generate an unbiased comparative gene expression profile of the cardiac fibroblast pool, identify and characterize the role of key genes in cardiac fibroblast function, and determine their contribution to myocardial development and regeneration. METHODS AND RESULTS High-throughput cell surface and intracellular profiling of cardiac and tail fibroblasts identified canonical mesenchymal stem cell and a surprising number of cardiogenic genes, some expressed at higher levels than in whole heart. While genetically marked fibroblasts contributed heterogeneously to interstitial but not cardiomyocyte compartments in infarcted hearts, fibroblast-restricted depletion of one highly expressed cardiogenic marker, T-box 20, caused marked myocardial dysmorphology and perturbations in scar formation on myocardial infarction. CONCLUSIONS The surprising transcriptional identity of cardiac fibroblasts, the adoption of cardiogenic gene programs, and direct contribution to cardiac development and repair provoke alternative interpretations for studies on more specialized cardiac progenitors, offering a novel perspective for reinterpreting cardiac regenerative therapies.
Collapse
Affiliation(s)
- Milena B Furtado
- From the Australian Regenerative Medicine Institute (M.B.F., M.W.C., E.A.P., E.S., A.R.P., A.C., N.A.R.), Department of Anatomy and Developmental Biology (A.R.P., R.B.), and Monash Biomedical Imaging (J.P.), Monash University, Melbourne, Victoria, Australia; Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.T.L., D.M.K.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (P.S., S.J.C.); and Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia (R.P.H.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hsu YC, Mildenstein K, Hunter K, Tkachenko O, Mullen CA. Acute lymphoid leukemia cells with greater stem cell antigen-1 (Ly6a/Sca-1) expression exhibit higher levels of metalloproteinase activity and are more aggressive in vivo. PLoS One 2014; 9:e88966. [PMID: 24586463 PMCID: PMC3930640 DOI: 10.1371/journal.pone.0088966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 01/16/2014] [Indexed: 12/13/2022] Open
Abstract
Stem cell antigen-1 (Ly6a/Sca-1) is a gene that is expressed in activated lymphocytes, hematopoietic stem cells and stem cells of a variety of tissues in mice. Despite decades of study its functions remain poorly defined. These studies explored the impact of expression of this stem cell associated gene in acute lymphoid leukemia. Higher levels of Ly6a/Sca-1 expression led to more aggressive leukemia growth in vivo and earlier death of hosts. Leukemias expressing higher levels of Ly6a/Sca-1 exhibited higher levels of matrix metalloproteinases. The results suggest the hypothesis that the more aggressive behavior of Ly6a/Sca-1 expressing leukemias is due at least in part to greater capacity to degrade microenvironmental stroma and invade tissues.
Collapse
Affiliation(s)
- Yu-Chiao Hsu
- Department of Pediatrics, University of Rochester, Rochester, New York, United States of America
| | - Kurt Mildenstein
- Department of Pediatrics, University of Rochester, Rochester, New York, United States of America
| | - Kordell Hunter
- Department of Pediatrics, University of Rochester, Rochester, New York, United States of America
| | - Olena Tkachenko
- Department of Pediatrics, University of Rochester, Rochester, New York, United States of America
| | - Craig A. Mullen
- Department of Pediatrics, University of Rochester, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
41
|
van Vliet P, Sluijter JPG, Doevendans PA, Goumans MJ. Isolation and expansion of resident cardiac progenitor cells. Expert Rev Cardiovasc Ther 2014; 5:33-43. [PMID: 17187455 DOI: 10.1586/14779072.5.1.33] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
After myocardial infarction, loss of viable cardiomyocytes severely impairs cardiac function. Recently, stem cell transplantation has been put forward as a promising approach to repair the damaged heart. Although several clinical trials have already been performed, the dominant beneficial effects are probably due to neoangiogenesis and arteriogenesis. However, replacement of cardiomyocytes is vital to improve cardiac function in the long term. Stem cells and progenitor cells, with the capacity to differentiate into cardiomyocytes, have been described in both embryonic and adult tissues. Upon stimulation, cardiac progenitor cells proliferate and differentiate into cardiomyocytes, vascular smooth muscle cells, and endothelial cells. Currently however, high proliferation rates and differentiation of cardiac progenitor cells beyond the fetal stage have not yet been achieved. Full differentiation into adult cardiomyocytes in vitro and in vivo might be important for efficient integration with the host environment and therefore more research is needed to study factors that influence proliferation and differentiation. Here we will discuss the isolation of cardiac progenitor cells, their potential to differentiate into various cell types needed for cardiac repair, the possible mechanisms behind these events, and how these cells may be implemented in future clinical settings.
Collapse
Affiliation(s)
- Patrick van Vliet
- The Interuniversity Cardiology Institute of The Netherlands, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
42
|
Malecki M, Putzer E, Sabo C, Foorohar A, Quach C, Stampe C, Beauchaine M, Malecki R, Tombokan X, Anderson M. Directed cardiomyogenesis of autologous human induced pluripotent stem cells recruited to infarcted myocardium with bioengineered antibodies. MOLECULAR AND CELLULAR THERAPIES 2014; 2. [PMID: 25132967 PMCID: PMC4131312 DOI: 10.1186/2052-8426-2-13] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objective Myocardial infarctions constitute a major factor contributing to non-natural mortality world-wide. Clinical trials ofmyocardial regenerative therapy, currently pursued by cardiac surgeons, involve administration of stem cells into the hearts of patients suffering from myocardial infarctions. Unfortunately, surgical acquisition of these cells from bone marrow or heart is traumatic, retention of these cells to sites of therapeutic interventions is low, and directed differentiation of these cells in situ into cardiomyocytes is difficult. The specific aims of this work were: (1) to generate autologous, human, pluripotent, induced stem cells (ahiPSCs) from the peripheral blood of the patients suffering myocardial infarctions; (2) to bioengineer heterospecific tetravalent antibodies (htAbs) and use them for recruitment of the ahiPSCs to infarcted myocardium; (3) to initiate in situ directed cardiomyogenesis of the ahiPSCs retained to infarcted myocardium. Methods Peripheral blood was drawn from six patients scheduled for heart transplants. Mononuclear cells were isolated and reprogrammed, with plasmids carrying six genes (NANOG, POU5F1, SOX2, KLF4, LIN28A, MYC), to yield the ahiPSCs. Cardiac tissues were excised from the injured hearts of the patients, who received transplants during orthotopic surgery. These tissues were used to prepare in vitro model of stem cell therapy of infarcted myocardium. The htAbs were bioengineered, which simultaneously targeted receptors displayed on pluripotent stem cells (SSEA-4, SSEA-3, TRA-1-60, TRA-1-81) and proteins of myocardial sarcomeres (myosin, α-actinin, actin, titin). They were used to bridge the ahiPSCs to the infarcted myocardium. The retained ahiPSCs were directed with bone morphogenetic proteins and nicotinamides to differentiate towards myocardial lineage. Results The patients’ mononuclear cells were efficiently reprogrammed into the ahiPSCs. These ahiPSCs were administered to infarcted myocardium in in vitro models. They were recruited to and retained at the treated myocardium with higher efficacy and specificity, if were preceded the htAbs, than with isotype antibodies or plain buffers. The retained cells differentiated into cardiomyocytes. Conclusions The proof of concept has been attained, for reprogramming the patients’ blood mononuclear cells (PBMCs) into the ahiPSCs, recruiting these cells to infarcted myocardium, and initiating their cardiomyogenesis. This novel strategy is ready to support the ongoing clinical trials aimed at regeneration of infarcted myocardium.
Collapse
Affiliation(s)
- Marek Malecki
- Phoenix Biomolecular Engineering Foundation, San Francisco, CA, USA ; National Magnetic Resonance Facility, National Institutes of Health, Madison, WI, USA ; University of Wisconsin, Madison, WI, USA
| | - Emily Putzer
- University of Wisconsin, Madison, WI, USA ; American Youth Center, Washington, DC, USA
| | - Chelsea Sabo
- University of Wisconsin, Madison, WI, USA ; University of Sheffield, Sheffield, UK, EU
| | - Afsoon Foorohar
- Phoenix Biomolecular Engineering Foundation, San Francisco, CA, USA ; Western University, Lebanon, OR, USA
| | - Carol Quach
- Phoenix Biomolecular Engineering Foundation, San Francisco, CA, USA ; Western University, Pomona, CA, USA
| | | | | | - Raf Malecki
- San Francisco State University, San Francisco, CA, USA
| | | | - Mark Anderson
- National Magnetic Resonance Facility, National Institutes of Health, Madison, WI, USA ; University of Wisconsin, Madison, WI, USA
| |
Collapse
|
43
|
Osada M, Singh VJ, Wu K, Sant’Angelo DB, Pezzano M. Label retention identifies a multipotent mesenchymal stem cell-like population in the postnatal thymus. PLoS One 2013; 8:e83024. [PMID: 24340075 PMCID: PMC3858364 DOI: 10.1371/journal.pone.0083024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/29/2013] [Indexed: 02/06/2023] Open
Abstract
Thymic microenvironments are essential for the proper development and selection of T cells critical for a functional and self-tolerant adaptive immune response. While significant turnover occurs, it is unclear whether populations of adult stem cells contribute to the maintenance of postnatal thymic epithelial microenvironments. Here, the slow cycling characteristic of stem cells and their property of label-retention were used to identify a K5-expressing thymic stromal cell population capable of generating clonal cell lines that retain the capacity to differentiate into a number of mesenchymal lineages including adipocytes, chondrocytes and osteoblasts suggesting a mesenchymal stem cell-like phenotype. Using cell surface analysis both culture expanded LRCs and clonal thymic mesenchymal cell lines were found to express Sca1, PDGFRα, PDGFRβ,CD29, CD44, CD49F, and CD90 similar to MSCs. Sorted GFP-expressing stroma, that give rise to TMSC lines, contribute to thymic architecture when reaggregated with fetal stroma and transplanted under the kidney capsule of nude mice. Together these results show that the postnatal thymus contains a population of mesenchymal stem cells that can be maintained in culture and suggests they may contribute to the maintenance of functional thymic microenvironments.
Collapse
Affiliation(s)
- Masako Osada
- Department of Biology, The City College of New York, CUNY, New York, New York, United States of America
| | - Varan J. Singh
- Department of Biology, The City College of New York, CUNY, New York, New York, United States of America
| | - Kenmin Wu
- Department of Biology, The City College of New York, CUNY, New York, New York, United States of America
| | - Derek B. Sant’Angelo
- Child Health Institute of New Jersey, Department of Pediatrics, Rutgers, Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America
| | - Mark Pezzano
- Department of Biology, The City College of New York, CUNY, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
44
|
Kulkarni-Datar K, Orsulic S, Foster R, Rueda BR. Ovarian tumor initiating cell populations persist following paclitaxel and carboplatin chemotherapy treatment in vivo. Cancer Lett 2013; 339:237-46. [PMID: 23791886 DOI: 10.1016/j.canlet.2013.06.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 06/06/2013] [Accepted: 06/10/2013] [Indexed: 01/06/2023]
Abstract
Development of recurrent platinum resistant disease following chemotherapy presents a challenge in managing ovarian cancer. Using tumors derived from genetically defined mouse ovarian cancer cells, we investigated the stem cell properties of residual cells post-chemotherapy. Utilizing CD133 and Sca-1 as markers of candidate tumor initiating cells (TIC), we determined that the relative levels of CD133+ and Sca-1+ cells were unaltered following chemotherapy. CD133+ and Sca-1+ cells exhibited increased stem cell-related gene expression, were enriched in G0/G1-early S phase and exhibited increased tumor initiating capacity, giving rise to heterogeneous tumors. Our findings suggest that residual TICs may contribute to recurrent disease.
Collapse
Affiliation(s)
- Kashmira Kulkarni-Datar
- Vincent Center for Reproductive Biology, Department of Vincent Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
45
|
Lee PY, Wang JX, Parisini E, Dascher CC, Nigrovic PA. Ly6 family proteins in neutrophil biology. J Leukoc Biol 2013; 94:585-94. [PMID: 23543767 DOI: 10.1189/jlb.0113014] [Citation(s) in RCA: 229] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The murine Ly6 complex was identified 35 years ago using antisera to lymphocytes. With advances in mAb development, molecular cloning, and genome sequencing, >20 structurally related genes have been identified within this complex on chromosome 15. All members of the Ly6 family and their human homologues share the highly conserved LU domain and most also possess a GPI anchor. Interestingly, many Ly6 proteins are expressed in a lineage-specific fashion, and their expression often correlates with stages of differentiation. As a result, Ly6 proteins are frequently used as surface markers for leukocyte subset identification and targets for antibody-mediated depletion. Murine neutrophils display prominent surface expression of several Ly6 proteins, including Ly6B, Ly6C, and Ly6G. Although the physiology of most Ly6 proteins is not well understood, a role in neutrophil functions, such as migration, is recognized increasingly. In this review, we will provide an overview of the Ly6 complex and discuss, in detail, the specific Ly6 proteins implicated in neutrophil biology.
Collapse
Affiliation(s)
- Pui Y Lee
- 1.Immunology, and Allergy, Brigham and Women's Hospital, One Jimmy Fund Way, Smith 516c, Boston, MA 02115, USA.
| | | | | | | | | |
Collapse
|
46
|
Abstract
Since the resident cardiac stem/progenitor cells were discovered, their ability to maintain the architecture and functional integrity of adult heart has been broadly explored. The methods for isolation and purification of the cardiac stem cells are crucial for the precise analysis of their developmental origin and intrinsic potential as tissue stem cells. Stem cell antigen-1 (Sca-1) is one of the useful cell surface markers to purify the cardiac progenitor cells. Another purification strategy is based on the high efflux ability of the dye, which is a common feature of tissue stem cells. These dye-extruding cells have been called side population cells because they locate in the side of dye-retaining cells after fluorescent cell sorting. In this chapter, we describe the methodology for the isolation of cardiac SP cells and Sca-1 positive cells.
Collapse
Affiliation(s)
- Toshio Nagai
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan
| | | | | |
Collapse
|
47
|
Zhao X, Huang L. Cardiac stem cells: A promising treatment option for heart failure. Exp Ther Med 2012; 5:379-383. [PMID: 23407679 PMCID: PMC3570189 DOI: 10.3892/etm.2012.854] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 10/18/2012] [Indexed: 12/25/2022] Open
Abstract
Cardiovascular diseases are the most common cause of death in the world. The development of heart failure is mainly due to the loss of cardiomyocytes following myocardial infarction and the absence of endogenous myocardial repair. Numerous studies have focused on cardiac stem cells (CSCs) due to their therapeutic benefit, particularly in the treatment of heart failure. It has previously been demonstrated that CSCs are able to promote the regeneration of cardiomyocytes in animals following myocardial infarction. However, the underlying mechanism(s) remain unclear. This review mainly discusses the cardioprotective effect of CSCs and the effect of CSCs on the function of cardiomyocytes, and compares the efficacies of CSCs from rats, mice and humans, thereby contributing to an improved understanding of CSCs as a promising treatment option for heart failure.
Collapse
Affiliation(s)
- Xiaohui Zhao
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | | |
Collapse
|
48
|
Abstract
Mammary gland stem cells (MaSC) have not been identified in spite of extensive research spanning over several decades. This has been primarily due to the complexity of mammary gland structure and its development, cell heterogeneity in the mammary gland and the insufficient knowledge about MaSC markers. At present, Lin (-) CD29 (i) CD49f (i) CD24 (+/mod) Sca- 1 (-) cells of the mammary gland have been reported to be enriched with MaSCs. We suggest that the inclusion of stem cell markers like Oct4, Sox2, Nanog and the mammary gland differentiation marker BRCA-1 may further narrow down the search for MaSCs. In addition, we have discussed some of the other unresolved puzzles on the mammary gland stem cells, such as their similarities and/or differences with mammary cancer stem cells, use of milk as source of mammary stem cells and the possibility of in vitro differentiation of embryonic stem (ES) cells into functional mammary gland structures in this review. Nevertheless, it is the lack of identity for a MaSC that is curtailing the advances in some of the above and other related areas.
Collapse
Affiliation(s)
- Suneesh Kaimala
- CSIR - Centre for Cellular and Molecular Biology, Uppal Road, Habsiguda, Hyderabad 500 007, India
| | | | | |
Collapse
|
49
|
Azalea-Romero M, González-Mendoza M, Cáceres-Pérez AA, Lara-Padilla E, Cáceres-Cortés JR. Low expression of stem cell antigen-1 on mouse haematopoietic precursors is associated with erythroid differentiation. Cell Immunol 2012; 279:187-95. [PMID: 23246681 DOI: 10.1016/j.cellimm.2012.10.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 09/18/2012] [Accepted: 10/04/2012] [Indexed: 11/28/2022]
Abstract
Sca1 is a surface marker of haematopoietic stem cell but its role in erythropoiesis is still largely unknown. In this work we evaluated the ability of Sca1⁺ cells to differentiate into cells of the erythrocytic lineage. We performed FACS analysis of complete and purified Sca1⁺ bone marrow cells from C3H/HeNHsd mice and measured the expression of CD71 and Terr119 to evaluate the stages in erythroid development. Definitive erythropoiesis was evident within the complete bone marrow, while only proerythroblasts were found in Sca1⁺ cells, suggesting that Sca1 is a negative regulator of erythropoiesis. We also used FDCP-mix cells and their PU.1 and SCL transfectants. The PU.1 transfectant showed significantly increased expression of Sca1 and was not induced to differentiate into red blood cells, while the SCL transfectant showed significantly lower expression of Sca1 and produced red blood cells. The results of this study suggest that increased Sca1 expression on erythropoietic precursors inhibits erythroid differentiation.
Collapse
Affiliation(s)
- Mirna Azalea-Romero
- Laboratory of Cancer and Hematopoiesis, Superior School of Medicine, National Polytechnic Institute, C.P. 11340 México, Mexico
| | | | | | | | | |
Collapse
|
50
|
Generation of functional blood vessels from a single c-kit+ adult vascular endothelial stem cell. PLoS Biol 2012; 10:e1001407. [PMID: 23091420 PMCID: PMC3473016 DOI: 10.1371/journal.pbio.1001407] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 09/05/2012] [Indexed: 12/22/2022] Open
Abstract
Adult vascular endothelial stem cells are shown to reside in the blood vessel wall endothelium. When isolated, these cells are capable of clonal expansion and generate functional blood vessels in vivo. In adults, the growth of blood vessels, a process known as angiogenesis, is essential for organ growth and repair. In many disorders including cancer, angiogenesis becomes excessive. The cellular origin of new vascular endothelial cells (ECs) during blood vessel growth in angiogenic situations has remained unknown. Here, we provide evidence for adult vascular endothelial stem cells (VESCs) that reside in the blood vessel wall endothelium. VESCs constitute a small subpopulation within CD117+ (c-kit+) ECs capable of undergoing clonal expansion while other ECs have a very limited proliferative capacity. Isolated VESCs can produce tens of millions of endothelial daughter cells in vitro. A single transplanted c-kit-expressing VESC by the phenotype lin−CD31+CD105+Sca1+CD117+ can generate in vivo functional blood vessels that connect to host circulation. VESCs also have long-term self-renewal capacity, a defining functional property of adult stem cells. To provide functional verification on the role of c-kit in VESCs, we show that a genetic deficit in endothelial c-kit expression markedly decreases total colony-forming VESCs. In vivo, c-kit expression deficit resulted in impaired EC proliferation and angiogenesis and retardation of tumor growth. Isolated VESCs could be used in cell-based therapies for cardiovascular repair to restore tissue vascularization after ischemic events. VESCs also provide a novel cellular target to block pathological angiogenesis and cancer growth. Angiogenesis—the growth of blood vessels—is essential for organ growth and repair, but also occurs during tumorigenesis and in certain inflammatory disorders. All blood vessels are lined by endothelial cells (ECs)—thin, flattened cells that form a continuous monolayer throughout the entire circulatory system. The cellular origin of new vascular ECs during blood vessel growth in angiogenic situations in adults is a matter of debate. New ECs could develop, in principle, from as yet undiscovered stem cells, as is well documented for the differentiated cells of skin or epithelia, or by the duplication of existing differentiated ECs. Here, we provide evidence for the existence of vascular endothelial stem cells (VESCs) that reside in the adult blood vessel wall endothelium. VESCs constitute a small subpopulation of ECs capable of clonal expansion, while other ECs have a very limited proliferative capacity. When isolated, these VESCs can produce tens of millions of endothelial daughter cells, and a single transplanted VESC can generate in vivo functional blood vessels that connect to host blood circulation. Isolated VESCs could be used in cell-based therapies for cardiovascular repair to restore tissue vascularization following ischemia and could also be pursued as a novel cellular target of inhibition to block pathological angiogenesis, for example during tumor growth.
Collapse
|