1
|
Duparc H, Muller D, Gilles L, Chédeville AL, El Khoury M, Guignard R, Debili N, Wittner M, Kauskot A, Pasquier F, Antony-Debré I, Marty C, Vainchenker W, Plo I, Raslova H. Deregulation of the p19/CDK4/CDK6 axis in Jak2 V617F megakaryocytes accelerates the development of myelofibrosis. Leukemia 2024; 38:898-902. [PMID: 38378843 DOI: 10.1038/s41375-024-02170-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/22/2024]
Affiliation(s)
- Hélène Duparc
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris Cité, Paris, France
| | - Delphine Muller
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Laure Gilles
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Agathe L Chédeville
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris Cité, Paris, France
| | - Mira El Khoury
- INSERM UMRS 938, PARIS, Sorbonne Université, Centre de Recherche Saint- Antoine, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Rose Guignard
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris Cité, Paris, France
| | - Najet Debili
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Monika Wittner
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Alexandre Kauskot
- INSERM U1176, Hemostasis, Inflammation & Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Florence Pasquier
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Département d'Hématologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Iléana Antony-Debré
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Caroline Marty
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - William Vainchenker
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Hana Raslova
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France.
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France.
- Gustave Roussy, Villejuif, France.
| |
Collapse
|
2
|
Abdallah MG, Teoh VSI, Dutta B, Yokomizo T, Osato M. Childhood hematopoietic stem cells constitute the permissive window for RUNX1-ETO leukemogenesis. Int J Hematol 2023; 117:830-838. [PMID: 37129801 DOI: 10.1007/s12185-023-03605-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
Cancer is a very rare event at the cellular level, although it is a common disease at the body level as one third of humans die of cancer. A small subset of cells in the body harbor the cellular features that constitute a permissive window for a particular genetic change to induce cancer. The significance of a permissive window is ironically best shown by a large number of failures in generating the animal model for acute myeloid leukemia (AML) with t(8;21). Over the decades, the RUNX1-ETO fusion gene created by t(8;21) has been introduced into various types of hematopoietic cells, largely at adult stage, in mice; however, all the previous attempts failed to generate tractable AML models. In stark contrast, we recently succeeded in inducing AML with the clinical features seen in human patients by specifically introducing RUNX1-ETO in childhood hematopoietic stem cells (HSCs). This result in mice is consistent with adolescent and young adult (AYA) onset in human t(8;21) patients, and suggests that childhood HSCs constitute the permissive window for RUNX1-ETO leukemogenesis. If loss of a permissive window is induced pharmacologically, cancer cells might be selectively targeted. Such a permissive window modifier may serve as a novel therapeutic drug.
Collapse
Affiliation(s)
- Mohamed Gaber Abdallah
- Department of Medical Biochemistry, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Vania Swee Imm Teoh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Bibek Dutta
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Tomomasa Yokomizo
- Department of Microscopic and Developmental Anatomy, Tokyo Women's Medical University, Tokyo, Japan
| | - Motomi Osato
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-Ku, Kumamoto, 860-0811, Japan.
- Department of General Internal Medicine, Kumamoto Kenhoku Hospital, Tamana, Japan.
| |
Collapse
|
3
|
rhTPO Ameliorates Radiation-Induced Long-Term Hematopoietic Stem Cell Injury in Mice. Molecules 2023; 28:molecules28041953. [PMID: 36838940 PMCID: PMC9961369 DOI: 10.3390/molecules28041953] [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: 01/04/2023] [Revised: 02/06/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Exposure to medium and high doses of ionizing radiation (IR) can induce long-term bone marrow (BM) suppression. We previously showed that recombinant human thrombopoietin (rhTPO) significantly promotes recovery from hematopoietic-acute radiation syndrome, but its effect on long-term BM suppression remains unknown. C57BL/6 mice were exposed to 6.5 Gy γ-rays of total body irradiation (TBI) at a dose-rate of 63.01 cGy per minute, and the mice were treated with rhTPO (100 μg; intramuscular injection) or vehicle at 2 h after TBI. All mice were killed one or two months after TBI for analysis of peripheral blood cell counts, long-term hematopoietic stem cell (HSC) frequency, and BM-derived clonogenic activity. The HSC self-renewal capacity was analyzed by BM transplantation. The levels of reactive oxygen species (ROS) production and ratios of γH2AX+ and p16, p53, and p21 mRNA in HSCs were measured by flow cytometry and real-time polymerase chain reaction, respectively. Treatment with rhTPO reduced long-term myelosuppression by improving long-term hematopoietic reconstitution (p < 0.05) after transplantation and resting state maintenance of HSCs (p < 0.05). Moreover, rhTPO treatment was associated with a sustained reduction in long-term ROS production, reduction of long-term DNA damage, diminished p53/p21 mRNA expression, and prevention of senescence after TBI. This study suggests rhTPO is an effective agent for treating IR-induced long-term BM injury because it regulates hematopoietic remodeling and HSC cycle disorder through the ROS/p53/p21/p16 pathway long term after IR.
Collapse
|
4
|
D'costa M, Bothe A, Das S, Udhaya Kumar S, Gnanasambandan R, George Priya Doss C. CDK regulators—Cell cycle progression or apoptosis—Scenarios in normal cells and cancerous cells. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 135:125-177. [PMID: 37061330 DOI: 10.1016/bs.apcsb.2022.11.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Serine/threonine kinases called cyclin-dependent kinases (CDKs) interact with cyclins and CDK inhibitors (CKIs) to control the catalytic activity. CDKs are essential controllers of RNA transcription and cell cycle advancement. The ubiquitous overactivity of the cell cycle CDKs is caused by a number of genetic and epigenetic processes in human cancer, and their suppression can result in both cell cycle arrest and apoptosis. This review focused on CDKs, describing their kinase activity, their role in phosphorylation inhibition, and CDK inhibitory proteins (CIP/KIP, INK 4, RPIC). We next compared the role of different CDKs, mainly p21, p27, p57, p16, p15, p18, and p19, in the cell cycle and apoptosis in cancer cells with respect to normal cells. The current work also draws attention to the use of CDKIs as therapeutics, overcoming the pharmacokinetic barriers of pan-CDK inhibitors, analyze new chemical classes that are effective at attacking the CDKs that control the cell cycle (cdk4/6 or cdk2). It also discusses CDKI's drawbacks and its combination therapy against cancer patients. These findings collectively demonstrate the complexity of cancer cell cycles and the need for targeted therapeutic intervention. In order to slow the progression of the disease or enhance clinical outcomes, new medicines may be discovered by researching the relationship between cell death and cell proliferation.
Collapse
Affiliation(s)
- Maria D'costa
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Anusha Bothe
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Soumik Das
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - S Udhaya Kumar
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - R Gnanasambandan
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India.
| | - C George Priya Doss
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India.
| |
Collapse
|
5
|
Cell-intrinsic factors governing quiescence vis-à-vis activation of adult hematopoietic stem cells. Mol Cell Biochem 2022; 478:1361-1382. [PMID: 36309884 DOI: 10.1007/s11010-022-04594-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/13/2022] [Indexed: 10/31/2022]
Abstract
Hematopoiesis is a highly complex process, regulated by both intrinsic and extrinsic factors. Often, these two regulatory arms work in tandem to maintain the steady-state condition of hematopoiesis. However, at times, certain intrinsic attributes of hematopoietic stem cells (HSCs) override the external stimuli and dominate the outcome. These could be genetic events like mutations or environmentally induced epigenetic or transcriptomic changes. Since leukemic stem cells (LSCs) share molecular pathways that also regulate normal HSCs, identifying specific, dominantly acting intrinsic factors could help in the development of novel therapeutic approaches. Here we have reviewed such dominantly acting intrinsic factors governing quiescence vis-à-vis activation of the HSCs in the face of external forces acting on them. For brevity, we have restricted our review to the articles dealing with adult HSCs of human and mouse origin that have been published in the last 10 years. Hematopoietic stem cells (HSCs) are closely associated with various stromal cells in their microenvironment and, thus, constantly receive signaling cues from them. The illustration depicts some dominantly acting intrinsic or cell-autonomous factors operative in the HSCs. These fall into various categories, such as epigenetic regulators, transcription factors, cell cycle regulators, tumor suppressor genes, signaling pathways, and metabolic regulators, which counteract the outcome of extrinsic signaling exerted by the HSC niche.
Collapse
|
6
|
Schirripa A, Sexl V, Kollmann K. Cyclin-dependent kinase inhibitors in malignant hematopoiesis. Front Oncol 2022; 12:916682. [PMID: 36033505 PMCID: PMC9403899 DOI: 10.3389/fonc.2022.916682] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
The cell-cycle is a tightly orchestrated process where sequential steps guarantee cellular growth linked to a correct DNA replication. The entire cell division is controlled by cyclin-dependent kinases (CDKs). CDK activation is balanced by the activating cyclins and CDK inhibitors whose correct expression, accumulation and degradation schedule the time-flow through the cell cycle phases. Dysregulation of the cell cycle regulatory proteins causes the loss of a controlled cell division and is inevitably linked to neoplastic transformation. Due to their function as cell-cycle brakes, CDK inhibitors are considered as tumor suppressors. The CDK inhibitors p16INK4a and p15INK4b are among the most frequently altered genes in cancer, including hematopoietic malignancies. Aberrant cell cycle regulation in hematopoietic stem cells (HSCs) bears severe consequences on hematopoiesis and provokes hematological disorders with a broad array of symptoms. In this review, we focus on the importance and prevalence of deregulated CDK inhibitors in hematological malignancies.
Collapse
|
7
|
Mayer IM, Hoelbl-Kovacic A, Sexl V, Doma E. Isolation, Maintenance and Expansion of Adult Hematopoietic Stem/Progenitor Cells and Leukemic Stem Cells. Cancers (Basel) 2022; 14:1723. [PMID: 35406494 PMCID: PMC8996967 DOI: 10.3390/cancers14071723] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 12/12/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are rare, self-renewing cells that perch on top of the hematopoietic tree. The HSCs ensure the constant supply of mature blood cells in a tightly regulated process producing peripheral blood cells. Intense efforts are ongoing to optimize HSC engraftment as therapeutic strategy to treat patients suffering from hematopoietic diseases. Preclinical research paves the way by developing methods to maintain, manipulate and expand HSCs ex vivo to understand their regulation and molecular make-up. The generation of a sufficient number of transplantable HSCs is the Holy Grail for clinical therapy. Leukemia stem cells (LSCs) are characterized by their acquired stem cell characteristics and are responsible for disease initiation, progression, and relapse. We summarize efforts, that have been undertaken to increase the number of long-term (LT)-HSCs and to prevent differentiation towards committed progenitors in ex vivo culture. We provide an overview and compare methods currently available to isolate, maintain and enrich HSC subsets, progenitors and LSCs and discuss their individual advantages and drawbacks.
Collapse
Affiliation(s)
| | | | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (I.M.M.); (A.H.-K.); (E.D.)
| | | |
Collapse
|
8
|
Li YR, Fu M, Song YQ, Li SL, Ge XY. Long noncoding RNA MRPL23-AS1 suppresses anoikis in salivary adenoid cystic carcinoma in vitro. Oral Dis 2022; 29:1588-1601. [PMID: 35175670 DOI: 10.1111/odi.14156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/18/2021] [Accepted: 10/19/2021] [Indexed: 11/27/2022]
Abstract
Distant lung metastasis is the main factor that affects the survival rate of patients with salivary adenoid cystic carcinoma (SACC). Anoikis resistance is a feature of tumor cells that easily metastasize. The long non-coding RNA (lncRNA) MRPL23 antisense RNA 1 (MPRL23-AS1) is related to lung metastasis in SACC, but its role in anoikis resistance is unknown.After altering MPRL23-AS1 expression in SACC cells, anoikis resistance was detected by calcein AM/PI staining and annexin V/PI flow cytometry. The apoptosis marker activated caspase-3 and the bcl-2/bax ratio were detected by Western blotting. The relationship between MPRL23-AS1 and the promoter of the potential downstream target gene p19INK4D was identified by chromatin immunoprecipitation (ChIP)-PCR assay. p19INK4D expression in patient tissues was determined using qRT-PCR and immunohistochemistry.The functional experiments showed that MPRL23-AS1 could promote anoikis resistance in vitro. MRPL23-AS1 recruited the EZH2 to the promoter region of p19INK4D, inhibited p19INK4D expression, and promoted tumor cell anoikis resistance. p19INK4D overexpression did not affect anoikis in attached cells; however, it attenuated the anoikis resistance effect of MPRL23-AS1 in suspension cells. p19INK4D expression was significantly lower in SACC tissues than in normal tissues.The novel MRPL23-AS1/p19INK4D axis may be a potential SACC biomarker or therapeutic target.
Collapse
Affiliation(s)
- Yin-Ran Li
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry of Health & NMPA Key Laboratory for Dental Material.,Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P.R. China
| | - Min Fu
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry of Health & NMPA Key Laboratory for Dental Material.,Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P.R. China
| | - Ye-Qing Song
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry of Health & NMPA Key Laboratory for Dental Material
| | - Sheng-Lin Li
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry of Health & NMPA Key Laboratory for Dental Material.,Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P.R. China
| | - Xi-Yuan Ge
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry of Health & NMPA Key Laboratory for Dental Material.,Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P.R. China
| |
Collapse
|
9
|
Opportunities and Challenges in Stem Cell Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1341:143-175. [PMID: 33748933 DOI: 10.1007/5584_2021_624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Studying aging, as a physiological process that can cause various pathological phenotypes, has attracted lots of attention due to its increasing burden and prevalence. Therefore, understanding its mechanism to find novel therapeutic alternatives for age-related disorders such as neurodegenerative and cardiovascular diseases is essential. Stem cell senescence plays an important role in aging. In the context of the underlying pathways, mitochondrial dysfunction, epigenetic and genetic alterations, and other mechanisms have been studied and as a consequence, several rejuvenation strategies targeting these mechanisms like pharmaceutical interventions, genetic modification, and cellular reprogramming have been proposed. On the other hand, since stem cells have great potential for disease modeling, they have been useful for representing aging and its associated disorders. Accordingly, the main mechanisms of senescence in stem cells and promising ways of rejuvenation, along with some examples of stem cell models for aging are introduced and discussed. This review aims to prepare a comprehensive summary of the findings by focusing on the most recent ones to shine a light on this area of research.
Collapse
|
10
|
Chen H, Liu O, Chen S, Zhou Y. Aging and Mesenchymal Stem Cells: Therapeutic Opportunities and Challenges in the Older Group. Gerontology 2021; 68:339-352. [PMID: 34161948 DOI: 10.1159/000516668] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/07/2021] [Indexed: 11/19/2022] Open
Abstract
With aging, a portion of cells, including mesenchymal stem cells (MSCs), become senescent, and these senescent cells accumulate and promote various age-related diseases. Therefore, the older age group has become a major population for MSC therapy, which is aimed at improving tissue regeneration and function of the aged body. However, the application of MSC therapy is often unsatisfying in the aged group. One reasonable conjecture for this correlation is that aging microenvironment reduces the number and function of MSCs. Cellular senescence also plays an important role in MSC function impairment. Thus, it is necessary to explore the relationship between senescence and MSCs for improving the application of MSCs in the elderly. Here, we present the influence of aging on MSCs and the characteristics and functional changes of senescent MSCs. Furthermore, current therapeutic strategies for improving MSC therapy in the elderly group are also discussed.
Collapse
Affiliation(s)
- Huan Chen
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, and Xiangya School of Stomatology, Central South University, Changsha, China
| | - Ousheng Liu
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, and Xiangya School of Stomatology, Central South University, Changsha, China
| | - Sijia Chen
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, and Xiangya School of Stomatology, Central South University, Changsha, China
| | - Yueying Zhou
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, and Xiangya School of Stomatology, Central South University, Changsha, China
| |
Collapse
|
11
|
Han X, Kuang Y, Chen H, Liu T, Zhang J, Liu J. p19INK4d: More than Just a Cyclin-Dependent Kinase Inhibitor. Curr Drug Targets 2021; 21:96-102. [PMID: 31400265 DOI: 10.2174/1389450120666190809161901] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/11/2019] [Accepted: 07/17/2019] [Indexed: 12/28/2022]
Abstract
Cyclin-dependent kinase inhibitors (CDKIs) are important cell cycle regulators. The CDKI family is composed of the INK4 family and the CIP/KIP family. p19INK4d belongs to the INK4 gene family and is involved in a series of normal physiological activities and the pathogenesis of diseases. Many factors play regulatory roles in the p19INK4d gene expression at the transcriptional and posttranscriptional levels. p19INK4d not only regulates the cell cycle but also plays regulatory roles in apoptosis, DNA damage repair, cell differentiation of hematopoietic cells, and cellular senescence. In this review, the regulatory network of the p19INK4d gene expression and its biological functions are summarized, which provides a basis for further study of p19INK4d as a drug target for disease treatment.
Collapse
Affiliation(s)
- Xu Han
- Molecular Biology Research Center and the Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Yijin Kuang
- Molecular Biology Research Center and the Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Huiyong Chen
- Molecular Biology Research Center and the Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Ting Liu
- Department of Rheumatology, the First Affiliated Hospital of South China University, Hengyang, Hunan, China
| | - Ji Zhang
- Department of Rheumatology, the First Affiliated Hospital of South China University, Hengyang, Hunan, China
| | - Jing Liu
- Molecular Biology Research Center and the Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| |
Collapse
|
12
|
Li Y, He M, Zhang W, Yang M, Ding Y, Xu S, Gu J, Li Y, Yin J, Gao Y. Antioxidant Small Molecule Compound Chrysin Promotes the Self-Renewal of Hematopoietic Stem Cells. Front Pharmacol 2020; 11:399. [PMID: 32300303 PMCID: PMC7142222 DOI: 10.3389/fphar.2020.00399] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 03/16/2020] [Indexed: 12/11/2022] Open
Abstract
There is an increasing demand for the expansion of functional human hematopoietic stem cells (hHSCs) for various clinical applications. Based on our primary screening of antioxidant small molecule compounds library, a small molecule compound C2968 (chrysin) was identificated to expand cord blood CD34+ cells in vitro. Then we further verified the optimum concentration and explored its effect on hHSCs phenotype and biological function. C2968 could significantly increase the proportion and absolute number of CD34+CD38−CD49f+ and CD34+CD38−CD45RA−CD90+ cells under 2.5 μM. Furthermore, the total number of colony-forming units and the frequency of LT-HSCs in C2968-treated group were significantly higher than control, indicating the multipotency and long-term activity of hematopoietic stem and progenitor cells were sustained. Additionally, C2968 treatment could maintain transplantable HSCs that preserve balanced multilineage potential and promote rapid engraftment after transplantation in immunodeficient (NOG) mice. Mechanistically, the activity of chrysin might be mediated through multiple mechanisms namely delaying HSC differentiation, inhibiting ROS-activated apoptosis, and modulating of cyclin-dependent kinase inhibitors. Overall, chrysin showed good ex vivo expansion effect on hHSCs, which could maintain the self-renewal and multilineage differentiation potential of hHSCs. Through further research on its antioxidant mechanism, it may become a promising tool for further fundamental research and clinical umbilical cord blood transplantation of hHSCs.
Collapse
Affiliation(s)
- Yinghui Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Mei He
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Wenshan Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Ming Yang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yahui Ding
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China
| | - Shiqi Xu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jiali Gu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yafang Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jingjing Yin
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yingdai Gao
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
13
|
Maupin KA, Himes ER, Plett AP, Chua HL, Singh P, Ghosh J, Mohamad SF, Abeysekera I, Fisher A, Sampson C, Hong JM, Childress P, Alvarez M, Srour EF, Bruzzaniti A, Pelus LM, Orschell CM, Kacena MA. Aging negatively impacts the ability of megakaryocytes to stimulate osteoblast proliferation and bone mass. Bone 2019; 127:452-459. [PMID: 31299382 PMCID: PMC6708771 DOI: 10.1016/j.bone.2019.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/15/2019] [Accepted: 07/08/2019] [Indexed: 12/11/2022]
Abstract
Osteoblast number and activity decreases with aging, contributing to the age-associated decline of bone mass, but the mechanisms underlying changes in osteoblast activity are not well understood. Here, we show that the age-associated bone loss critically depends on impairment of the ability of megakaryocytes (MKs) to support osteoblast proliferation. Co-culture of osteoblast precursors with young MKs is known to increase osteoblast proliferation and bone formation. However, co-culture of osteoblast precursors with aged MKs resulted in significantly fewer osteoblasts compared to co-culture with young MKs, and this was associated with the downregulation of transforming growth factor beta. In addition, the ability of MKs to increase bone mass was attenuated during aging as transplantation of GATA1low/low hematopoietic donor cells (which have elevated MKs/MK precursors) from young mice resulted in an increase in bone mass of recipient mice compared to transplantation of young wild-type donor cells, whereas transplantation of GATA1low/low donor cells from old mice failed to enhance bone mass in recipient mice compared to transplantation of old wild-type donor cells. These findings suggest that the preservation or restoration of the MK-mediated induction of osteoblast proliferation during aging may hold the potential to prevent age-associated bone loss and resulting fractures.
Collapse
Affiliation(s)
| | - Evan R Himes
- Indiana University School of Medicine, Indiana, USA
| | | | - Hui Lin Chua
- Indiana University School of Medicine, Indiana, USA
| | | | | | | | | | - Alexa Fisher
- Indiana University School of Medicine, Indiana, USA
| | | | - Jung-Min Hong
- Indiana University School of Dentistry, Indiana, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
Mammal megakaryocytes (MK) undergo polyploidization during their differentiation. This process leads to a marked increase in the MK size and of their cytoplasm. Contrary to division by classical mitosis, ploidization allows an economical manner to produce platelets as they arise from the fragmentation of the MK cytoplasm. The platelet production in vivo correlates to the entire MK cytoplasm mass that depends both upon the number of MKs and their size. Polyploidization occurs by several rounds of DNA replication with at the end of each round an aborted mitosis at late phase of cytokinesis. As there is also a defect in karyokinesis, MKs are giant cells with a single polylobulated nucleus with a 2xN ploidy. However, polyploidization per se does not increase platelet production because it requires a parallel development of MK organelles such as mitochondria, granules and the demarcation membrane system. MK polyploidization is regulated by extrinsic factors, more particularly by thrombopoietin (TPO), which during a platelet stress increases first polyploidization before enhancing the MK number and by transcription factors such as RUNX1, GATA1, and FLI1 that regulate MK differentiation explaining why polyploidization and cytoplasmic maturation are intermingled. MK polyploidization is ontogenically regulated and is markedly altered in malignant myeloid disorders such as acute megakaryoblastic leukemia and myeloproliferative disorders as well as in hereditary thrombocytopenia, more particularly those involving transcription factors or signaling pathways. In addition, MKs arising from progenitors in vitro have a much lower ploidy in vitro than in vivo leading to a low yield of platelet production in vitro. Thus, it is tempting to find approaches to increase MK polyploidization in vitro. However, these approaches require molecules that are able to simultaneously increase MK polyploidization and to induce terminal differentiation. Here, we will focus on the regulation by extrinsic and intrinsic factors of MK polyploidization during development and pathological conditions.
Collapse
Affiliation(s)
- William Vainchenker
- UMR 1170, Institut National de la Santé et de la Recherche Médicale, Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy Cancer Campus, Equipe Labellisée Ligue Nationale Contre le Cancer , Villejuif, France
| | - Hana Raslova
- UMR 1170, Institut National de la Santé et de la Recherche Médicale, Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy Cancer Campus, Equipe Labellisée Ligue Nationale Contre le Cancer , Villejuif, France
| |
Collapse
|
15
|
Paczulla AM, Rothfelder K, Raffel S, Konantz M, Steinbacher J, Wang H, Tandler C, Mbarga M, Schaefer T, Falcone M, Nievergall E, Dörfel D, Hanns P, Passweg JR, Lutz C, Schwaller J, Zeiser R, Blazar BR, Caligiuri MA, Dirnhofer S, Lundberg P, Kanz L, Quintanilla-Martinez L, Steinle A, Trumpp A, Salih HR, Lengerke C. Absence of NKG2D ligands defines leukaemia stem cells and mediates their immune evasion. Nature 2019; 572:254-259. [PMID: 31316209 PMCID: PMC6934414 DOI: 10.1038/s41586-019-1410-1] [Citation(s) in RCA: 252] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 06/14/2019] [Indexed: 01/09/2023]
Abstract
Patients with acute myeloid leukaemia (AML) often achieve remission after therapy, but subsequently die of relapse1 that is driven by chemotherapy-resistant leukaemic stem cells (LSCs)2,3. LSCs are defined by their capacity to initiate leukaemia in immunocompromised mice4. However, this precludes analyses of their interaction with lymphocytes as components of anti-tumour immunity5, which LSCs must escape to induce cancer. Here we demonstrate that stemness and immune evasion are closely intertwined in AML. Using xenografts of human AML as well as syngeneic mouse models of leukaemia, we show that ligands of the danger detector NKG2D-a critical mediator of anti-tumour immunity by cytotoxic lymphocytes, such as NK cells6-9-are generally expressed on bulk AML cells but not on LSCs. AML cells with LSC properties can be isolated by their lack of expression of NKG2D ligands (NKG2DLs) in both CD34-expressing and non-CD34-expressing cases of AML. AML cells that express NKG2DLs are cleared by NK cells, whereas NKG2DL-negative leukaemic cells isolated from the same individual escape cell killing by NK cells. These NKG2DL-negative AML cells show an immature morphology, display molecular and functional stemness characteristics, and can initiate serially re-transplantable leukaemia and survive chemotherapy in patient-derived xenotransplant models. Mechanistically, poly-ADP-ribose polymerase 1 (PARP1) represses expression of NKG2DLs. Genetic or pharmacologic inhibition of PARP1 induces NKG2DLs on the LSC surface but not on healthy or pre-leukaemic cells. Treatment with PARP1 inhibitors, followed by transfer of polyclonal NK cells, suppresses leukaemogenesis in patient-derived xenotransplant models. In summary, our data link the LSC concept to immune escape and provide a strong rationale for targeting therapy-resistant LSCs by PARP1 inhibition, which renders them amenable to control by NK cells in vivo.
Collapse
Affiliation(s)
- Anna M Paczulla
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Kathrin Rothfelder
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Tuebingen, Germany
- Department of Internal Medicine II, Hematology and Oncology, Eberhard-Karls University, Tuebingen, Germany
- DFG Cluster of Excellence 2180 'Image-guided and Functional Instructed Tumor Therapy' (IFIT), Eberhard-Karls University, Tuebingen, Germany
| | - Simon Raffel
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Martina Konantz
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Julia Steinbacher
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Tuebingen, Germany
- Department of Internal Medicine II, Hematology and Oncology, Eberhard-Karls University, Tuebingen, Germany
| | - Hui Wang
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Claudia Tandler
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Tuebingen, Germany
- Department of Internal Medicine II, Hematology and Oncology, Eberhard-Karls University, Tuebingen, Germany
- DFG Cluster of Excellence 2180 'Image-guided and Functional Instructed Tumor Therapy' (IFIT), Eberhard-Karls University, Tuebingen, Germany
| | - Marcelle Mbarga
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Thorsten Schaefer
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Mattia Falcone
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Eva Nievergall
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Daniela Dörfel
- Department of Internal Medicine II, Hematology and Oncology, Eberhard-Karls University, Tuebingen, Germany
| | - Pauline Hanns
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Jakob R Passweg
- Division of Clinical Hematology, University Hospital Basel, Basel, Switzerland
| | - Christoph Lutz
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Juerg Schwaller
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
- University Children's Hospital Basel, Basel, Switzerland
| | - Robert Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- Center for Biological Signaling Studies (BIOSS), University of Freiburg, Freiburg, Germany
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Michael A Caligiuri
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, USA
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
- Beckman Research Institute, Duarte, CA, USA
| | - Stephan Dirnhofer
- Institute for Pathology & Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Pontus Lundberg
- Diagnostic Hematology, Department of Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | - Lothar Kanz
- Department of Internal Medicine II, Hematology and Oncology, Eberhard-Karls University, Tuebingen, Germany
| | | | - Alexander Steinle
- Institute for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Helmut R Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Tuebingen, Germany.
- Department of Internal Medicine II, Hematology and Oncology, Eberhard-Karls University, Tuebingen, Germany.
- DFG Cluster of Excellence 2180 'Image-guided and Functional Instructed Tumor Therapy' (IFIT), Eberhard-Karls University, Tuebingen, Germany.
| | - Claudia Lengerke
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
- Division of Clinical Hematology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
16
|
Verovskaya EV, Dellorusso PV, Passegué E. Losing Sense of Self and Surroundings: Hematopoietic Stem Cell Aging and Leukemic Transformation. Trends Mol Med 2019; 25:494-515. [PMID: 31109796 DOI: 10.1016/j.molmed.2019.04.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/29/2019] [Accepted: 04/11/2019] [Indexed: 02/07/2023]
Abstract
Aging leads to functional decline of the hematopoietic system, manifested by an increased incidence of hematological disease in the elderly. Deterioration of hematopoietic integrity with age originates in part from the degraded functionality of hematopoietic stem cells (HSCs). Here, we review recent findings identifying changes in metabolic programs and loss of epigenetic identity as major drivers of old HSC dysfunction and their role in promoting leukemia onset in the context of age-related clonal hematopoiesis (ARCH). We discuss how inflammatory and growth signals from the aged bone marrow (BM) microenvironment contribute to cell-intrinsic HSC aging phenotypes and favor leukemia development. Finally, we address how metabolic, epigenetic, and inflammatory pathways could be targeted to enhance old HSC fitness and prevent leukemic transformation.
Collapse
Affiliation(s)
- Evgenia V Verovskaya
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Paul V Dellorusso
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
17
|
Bai L, Shi G, Yang Y, Chen W, Zhang L, Qin C. Rehmannia glutinosa exhibits anti-aging effect through maintaining the quiescence and decreasing the senescence of hematopoietic stem cells. Animal Model Exp Med 2018; 1:194-202. [PMID: 30891565 PMCID: PMC6388079 DOI: 10.1002/ame2.12034] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/29/2018] [Accepted: 09/04/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The time-related decline in regenerative capacity and organ homeostasis is a major feature of aging. Rehmannia glutinosa and Astragalus membranaceus have been used as traditional Chinese herbal medicines for enhanced immunity and prolonged life. However, the mechanism by which this herbal medicine slows aging is unknown. In this study, we investigated the mechanism of the herbal anti-aging effect. METHODS Mice were fed diets supplemented with R. glutinosa or A. membranaceus for 10 months; the control group was fed a standard diet. The phenotypes were evaluated using a grading score system and survival analysis. The percentages of the senescence phenotypes of hematopoietic stem cells (HSCs) were determined by fluorescence-activated cell sorting analysis. The function and the mechanism of HSCs were analyzed by clonogenic assay and the real-time polymerase chain reaction. RESULTS The anti-aging effect of R. glutinosa is due to the enhanced function of HSCs. Mice fed with R. glutinosa displayed characteristics of a slowed aging process, including decreased senescence and increased rate of survival. Flow cytometry analysis showed decreased numbers of Lin-Sca1+c-kit- (LSK) cells, long-term HSCs (LT-HSCs) and short-term HSCs (ST-HSCs) in the R. glutinosa group. In vitro, clonogenic assays showed increased self-renewal ability of LT-HSCs from the R. glutinosa group as well as maintaining LSK quiescence through upregulated p18 expression. The R. glutinosa group also showed decreased reactive oxygen species levels and the percentage of β-gal+ cells through downregulation of the cellular senescence-associated protein p53 and p16. CONCLUSION Rehmannia glutinosa exerts anti-aging effects by maintaining the quiescence and decreasing the senescence of HSCs.
Collapse
Affiliation(s)
- Lin Bai
- Key Laboratory of Human Disease Comparative MedicineMinistry of HealthInstitute of Laboratory Animal ScienceChinese Academy of Medical Sciences and Comparative Medical CenterPeking Union Medical CollegeBeijingChina
| | - Gui‐ying Shi
- Key Laboratory of Human Disease Comparative MedicineMinistry of HealthInstitute of Laboratory Animal ScienceChinese Academy of Medical Sciences and Comparative Medical CenterPeking Union Medical CollegeBeijingChina
| | - Ya‐jun Yang
- Key Laboratory of Human Disease Comparative MedicineMinistry of HealthInstitute of Laboratory Animal ScienceChinese Academy of Medical Sciences and Comparative Medical CenterPeking Union Medical CollegeBeijingChina
| | - Wei Chen
- Key Laboratory of Human Disease Comparative MedicineMinistry of HealthInstitute of Laboratory Animal ScienceChinese Academy of Medical Sciences and Comparative Medical CenterPeking Union Medical CollegeBeijingChina
| | - Lian‐feng Zhang
- Key Laboratory of Human Disease Comparative MedicineMinistry of HealthInstitute of Laboratory Animal ScienceChinese Academy of Medical Sciences and Comparative Medical CenterPeking Union Medical CollegeBeijingChina
| | - Chuan Qin
- Key Laboratory of Human Disease Comparative MedicineMinistry of HealthInstitute of Laboratory Animal ScienceChinese Academy of Medical Sciences and Comparative Medical CenterPeking Union Medical CollegeBeijingChina
| |
Collapse
|
18
|
Thioredoxin mitigates radiation-induced hematopoietic stem cell injury in mice. Stem Cell Res Ther 2017; 8:263. [PMID: 29141658 PMCID: PMC5688691 DOI: 10.1186/s13287-017-0711-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/03/2017] [Accepted: 10/24/2017] [Indexed: 12/17/2022] Open
Abstract
Background Radiation exposure poses a significant threat to public health. Hematopoietic injury is one of the major manifestations of acute radiation sickness. Protection and/or mitigation of hematopoietic stem cells (HSCs) from radiation injury is an important goal in the development of medical countermeasure agents (MCM). We recently identified thioredoxin (TXN) as a novel molecule that has marked protective and proliferative effects on HSCs. In the current study, we investigated the effectiveness of TXN in rescuing mice from a lethal dose of total body radiation (TBI) and in enhancing hematopoietic reconstitution following a lethal dose of irradiation. Methods We used in-vivo and in-vitro methods to understand the biological and molecular mechanisms of TXN on radiation mitigation. BABL/c mice were used for the survival study and a flow cytometer was used to quantify the HSC population and cell senescence. A hematology analyzer was used for the peripheral blood cell count, including white blood cells (WBCs), red blood cells (RBCs), hemoglobin, and platelets. Colony forming unit (CFU) assay was used to study the colongenic function of HSCs. Hematoxylin and eosin staining was used to determine the bone marrow cellularity. Senescence-associated β-galactosidase assay was used for cell senescence. Western blot analysis was used to evaluate the DNA damage and senescence protein expression. Immunofluorescence staining was used to measure the expression of γ-H2AX foci for DNA damage. Results We found that administration of TXN 24 h following irradiation significantly mitigates BALB/c mice from TBI-induced death: 70% of TXN-treated mice survived, whereas only 25% of saline-treated mice survived. TXN administration led to enhanced recovery of peripheral blood cell counts, bone marrow cellularity, and HSC population as measured by c-Kit+Sca-1+Lin– (KSL) cells, SLAM + KSL cells and CFUs. TXN treatment reduced cell senescence and radiation-induced double-strand DNA breaks in both murine bone marrow lineage-negative (Lin–) cells and primary fibroblasts. Furthermore, TXN decreased the expression of p16 and phosphorylated p38. Our data suggest that TXN modulates diverse cellular processes of HSCs. Conclusions Administration of TXN 24 h following irradiation mitigates radiation-induced lethality. To the best of our knowledge, this is the first report demonstrating that TXN reduces radiation-induced lethality. TXN shows potential utility in the mitigation of radiation-induced hematopoietic injury.
Collapse
|
19
|
Han X, Liu J. Cell cycle-independent roles of p19 INK4d in human terminal erythropoiesis. CHINESE JOURNAL OF CANCER 2017; 36:22. [PMID: 28228163 PMCID: PMC5322582 DOI: 10.1186/s40880-017-0189-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 01/22/2017] [Indexed: 11/18/2022]
Affiliation(s)
- Xu Han
- The State Key Laboratory of Medical Genetics & School of Life Sciences, Central South University, Changsha, 410078, Hunan, P. R. China
| | - Jing Liu
- The State Key Laboratory of Medical Genetics & School of Life Sciences, Central South University, Changsha, 410078, Hunan, P. R. China.
| |
Collapse
|
20
|
Unexpected role for p19INK4d in posttranscriptional regulation of GATA1 and modulation of human terminal erythropoiesis. Blood 2016; 129:226-237. [PMID: 27879259 DOI: 10.1182/blood-2016-09-739268] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022] Open
Abstract
Terminal erythroid differentiation is tightly coordinated with cell-cycle exit, which is regulated by cyclins, cyclin-dependent kinases, and cyclin-dependent kinase inhibitors (CDKI), yet their roles in erythropoiesis remain to be fully defined. We show here that p19INK4d, a member of CDKI family, is abundantly expressed in erythroblasts and that p19INK4d knockdown delayed erythroid differentiation, inhibited cell growth, and led to increased apoptosis and generation of abnormally nucleated late-stage erythroblasts. Unexpectedly, p19INK4d knockdown did not affect cell cycle. Rather, it led to decreased expression of GATA1 protein. Importantly, the differentiation and nuclear defects were rescued by ectopic expression of GATA1. Because the GATA1 protein is protected by nuclear heat shock protein family (HSP) member HSP70, we examined the effects of p19INK4d knockdown on HSP70 and found that p19INK4d knockdown led to decreased expression of HSP70 and its nuclear localization. The reduced levels of HSP70 are the result of reduced extracellular signal-regulated kinase (ERK) activation. Further biochemical analysis revealed that p19INK4d directly binds to Raf kinase inhibitor PEBP1 and that p19INK4d knockdown increased the expression of PEBP1, which in turn led to reduced ERK activation. Thus we have identified an unexpected role for p19INK4d via a novel PEBP1-p-ERK-HSP70-GATA1 pathway. These findings are likely to have implications for improved understanding of disordered erythropoiesis.
Collapse
|
21
|
Traumatic Brain Injury Stimulates Neural Stem Cell Proliferation via Mammalian Target of Rapamycin Signaling Pathway Activation. eNeuro 2016; 3:eN-NWR-0162-16. [PMID: 27822507 PMCID: PMC5089538 DOI: 10.1523/eneuro.0162-16.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/07/2016] [Accepted: 09/23/2016] [Indexed: 12/31/2022] Open
Abstract
Neural stem cells in the adult brain possess the ability to remain quiescent until needed in tissue homeostasis or repair. It was previously shown that traumatic brain injury (TBI) stimulated neural stem cell (NSC) proliferation in the adult hippocampus, indicating an innate repair mechanism, but it is unknown how TBI promotes NSC proliferation. In the present study, we observed dramatic activation of mammalian target of rapamycin complex 1 (mTORC1) in the hippocampus of mice with TBI from controlled cortical impact (CCI). The peak of mTORC1 activation in the hippocampal subgranular zone, where NSCs reside, is 24-48 h after trauma, correlating with the peak of TBI-enhanced NSC proliferation. By use of a Nestin-GFP transgenic mouse, in which GFP is ectopically expressed in the NSCs, we found that TBI activated mTORC1 in NSCs. With 5-bromo-2'-deoxyuridine labeling, we observed that TBI increased mTORC1 activation in proliferating NSCs. Furthermore, administration of rapamycin abolished TBI-promoted NSC proliferation. Taken together, these data indicate that mTORC1 activation is required for NSC proliferation postinjury, and thus might serve as a therapeutic target for interventions to augment neurogenesis for brain repair after TBI.
Collapse
|
22
|
Dou DR, Calvanese V, Sierra MI, Nguyen AT, Minasian A, Saarikoski P, Sasidharan R, Ramirez CM, Zack JA, Crooks GM, Galic Z, Mikkola HKA. Medial HOXA genes demarcate haematopoietic stem cell fate during human development. Nat Cell Biol 2016; 18:595-606. [PMID: 27183470 PMCID: PMC4981340 DOI: 10.1038/ncb3354] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 04/08/2016] [Indexed: 12/18/2022]
Abstract
Pluripotent stem cells (PSC) may provide a potential source of haematopoietic stem/progenitor cells (HSPCs) for transplantation; however, unknown molecular barriers prevent the self-renewal of PSC-HSPCs. Using two-step differentiation, human embryonic stem cells (hESCs) differentiated in vitro into multipotent haematopoietic cells that had CD34+CD38−/loCD90+CD45+GPI-80+ foetal liver (FL) HSC immunophenotype, but displayed poor expansion potential and engraftment ability. Transcriptome analysis of immunophenotypic hESC-HSPCs revealed that, despite their molecular resemblance to FL-HSPCs, medial HOXA genes remained suppressed. Knockdown of HOXA7 disrupted FL-HSPC function and caused transcriptome dysregulation that resembled hESC-derived progenitors. Overexpression of medial HOXA genes prolonged FL-HSPC maintenance but was insufficient to confer self-renewal to hESC-HSPCs. Stimulation of retinoic acid signalling during endothelial-to-haematopoietic transition induced the HOXA cluster and other HSC/definitive haemogenic endothelium genes, and prolonged HSPC maintenance in culture. Thus, retinoic acid signalling-induced medial HOXA gene expression marks the establishment of the definitive HSC fate and controls HSC identity and function.
Collapse
Affiliation(s)
- Diana R Dou
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California 90095, USA.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California 90095, USA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Vincenzo Calvanese
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California 90095, USA.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Maria I Sierra
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California 90095, USA.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Andrew T Nguyen
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Arazin Minasian
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California 90095, USA.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Pamela Saarikoski
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California 90095, USA.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Rajkumar Sasidharan
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California 90095, USA.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Christina M Ramirez
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Jerome A Zack
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California 90095, USA.,Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, California 90095, USA.,Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Gay M Crooks
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California 90095, USA.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California 90095, USA.,Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Zoran Galic
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California 90095, USA.,Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Hanna K A Mikkola
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California 90095, USA.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California 90095, USA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
23
|
Hao S, Chen C, Cheng T. Cell cycle regulation of hematopoietic stem or progenitor cells. Int J Hematol 2016; 103:487-97. [DOI: 10.1007/s12185-016-1984-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/07/2016] [Accepted: 03/07/2016] [Indexed: 11/24/2022]
|
24
|
Genetic and Epigenetic Mechanisms That Maintain Hematopoietic Stem Cell Function. Stem Cells Int 2015; 2016:5178965. [PMID: 26798358 PMCID: PMC4699043 DOI: 10.1155/2016/5178965] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/03/2015] [Accepted: 09/09/2015] [Indexed: 01/15/2023] Open
Abstract
All hematopoiesis cells develop from multipotent progenitor cells. Hematopoietic stem cells (HSC) have the ability to develop into all blood lineages but also maintain their stemness. Different molecular mechanisms have been identified that are crucial for regulating quiescence and self-renewal to maintain the stem cell pool and for inducing proliferation and lineage differentiation. The stem cell niche provides the microenvironment to keep HSC in a quiescent state. Furthermore, several transcription factors and epigenetic modifiers are involved in this process. These create modifications that regulate the cell fate in a more or less reversible and dynamic way and contribute to HSC homeostasis. In addition, HSC respond in a unique way to DNA damage. These mechanisms also contribute to the regulation of HSC function and are essential to ensure viability after DNA damage. How HSC maintain their quiescent stage during the entire life is still matter of ongoing research. Here we will focus on the molecular mechanisms that regulate HSC function.
Collapse
|
25
|
Bennett JA, Singh KP, Unnisa Z, Welle SL, Gasiewicz TA. Deficiency in Aryl Hydrocarbon Receptor (AHR) Expression throughout Aging Alters Gene Expression Profiles in Murine Long-Term Hematopoietic Stem Cells. PLoS One 2015. [PMID: 26208102 PMCID: PMC4514744 DOI: 10.1371/journal.pone.0133791] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Dysregulation of hematopoietic stem cell (HSC) signaling can contribute to the development of diseases of the blood system. Lack of aryl hydrocarbon receptor (AhR) has been associated with alterations in gene expression related to HSC function and the subsequent development of a myeloproliferative disorder in aging female mice. We sorted the most primitive population of HSCs with the highest stem cell potential (Long-term, or LT-HSCs) from 18-month-old AhR-null-allele (AhR-KO) and WT mice and analyzed gene expression using microarray to determine alterations in gene expression and cell signaling networks in HSCs that could potentially contribute to the aging phenotype of AhR-KO mice. Comparisons with previous array data from 8-week old mice indicated that aging alone is sufficient to alter gene expression. In addition, a significant number of gene expression differences were observed in aged LT-HSCs that are dependent on both aging and lack of AhR. Pathway analysis of these genes revealed networks related to hematopoietic stem cell activity or function. qPCR was used to confirm the differential expression of a subset of these genes, focusing on genes that may represent novel AhR targets due to the presence of a putative AhR binding site in their upstream regulatory region. We verified differential expression of PDGF-D, Smo, Wdfy1, Zbtb37 and Zfp382. Pathway analysis of this subset of genes revealed overlap between cellular functions of the novel AhR targets and AhR itself. Lentiviral-mediated knockdown of AhR in lineage-negative hematopoietic cells was sufficient to induce changes in all five of the candidate AhR targets identified. Taken together, these data suggest a role for AhR in HSC functional regulation, and identify novel HSC AhR target genes that may contribute to the phenotypes observed in AhR-KO mice.
Collapse
Affiliation(s)
- John A. Bennett
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Kameshwar P. Singh
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Zeenath Unnisa
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Stephen L. Welle
- Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Thomas A. Gasiewicz
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
26
|
Mende N, Kuchen EE, Lesche M, Grinenko T, Kokkaliaris KD, Hanenberg H, Lindemann D, Dahl A, Platz A, Höfer T, Calegari F, Waskow C. CCND1-CDK4-mediated cell cycle progression provides a competitive advantage for human hematopoietic stem cells in vivo. ACTA ACUST UNITED AC 2015; 212:1171-83. [PMID: 26150472 PMCID: PMC4516798 DOI: 10.1084/jem.20150308] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 06/22/2015] [Indexed: 12/26/2022]
Abstract
Maintenance of stem cell properties is associated with reduced proliferation but it is unknown whether the transition kinetics through distinct cell cycle phases influences the function of HSCs. Mende et al examine the effects of increasing two cell cycle complexes CCND1–CDK4 and CCNE1–CDK2 on the transition kinetics of human HSCs and their maintenance and functional alterations in vivo. Maintenance of stem cell properties is associated with reduced proliferation. However, in mouse hematopoietic stem cells (HSCs), loss of quiescence results in a wide range of phenotypes, ranging from functional failure to extensive self-renewal. It remains unknown whether the function of human HSCs is controlled by the kinetics of cell cycle progression. Using human HSCs and human progenitor cells (HSPCs), we report here that elevated levels of CCND1–CDK4 complexes promoted the transit from G0 to G1 and shortened the G1 cell cycle phase, resulting in protection from differentiation-inducing signals in vitro and increasing human leukocyte engraftment in vivo. Further, CCND1–CDK4 overexpression conferred a competitive advantage without impacting HSPC numbers. In contrast, accelerated cell cycle progression mediated by elevated levels of CCNE1–CDK2 led to the loss of functional HSPCs in vivo. Collectively, these data suggest that the transition kinetics through the early cell cycle phases are key regulators of human HSPC function and important for lifelong hematopoiesis.
Collapse
Affiliation(s)
- Nicole Mende
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Institute of Virology, Center for Regenerative Therapies, Faculty of Medicine; Deep Sequencing Group SFB655, Biotechnology Center, TU Dresden, 01307 Dresden, Germany
| | - Erika E Kuchen
- Division of Theoretical Systems Biology, German Cancer Research Center, 69120 Heidelberg, Germany Bioquant Center, University of Heidelberg, 69120 Heidelberg, Germany
| | - Mathias Lesche
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Institute of Virology, Center for Regenerative Therapies, Faculty of Medicine; Deep Sequencing Group SFB655, Biotechnology Center, TU Dresden, 01307 Dresden, Germany
| | - Tatyana Grinenko
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Institute of Virology, Center for Regenerative Therapies, Faculty of Medicine; Deep Sequencing Group SFB655, Biotechnology Center, TU Dresden, 01307 Dresden, Germany
| | | | - Helmut Hanenberg
- Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Dirk Lindemann
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Institute of Virology, Center for Regenerative Therapies, Faculty of Medicine; Deep Sequencing Group SFB655, Biotechnology Center, TU Dresden, 01307 Dresden, Germany
| | - Andreas Dahl
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Institute of Virology, Center for Regenerative Therapies, Faculty of Medicine; Deep Sequencing Group SFB655, Biotechnology Center, TU Dresden, 01307 Dresden, Germany
| | | | - Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center, 69120 Heidelberg, Germany Bioquant Center, University of Heidelberg, 69120 Heidelberg, Germany
| | - Federico Calegari
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Institute of Virology, Center for Regenerative Therapies, Faculty of Medicine; Deep Sequencing Group SFB655, Biotechnology Center, TU Dresden, 01307 Dresden, Germany
| | - Claudia Waskow
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Institute of Virology, Center for Regenerative Therapies, Faculty of Medicine; Deep Sequencing Group SFB655, Biotechnology Center, TU Dresden, 01307 Dresden, Germany
| |
Collapse
|