1
|
Minervini A, Coccaro N, Anelli L, Zagaria A, Specchia G, Albano F. HMGA Proteins in Hematological Malignancies. Cancers (Basel) 2020; 12:E1456. [PMID: 32503270 PMCID: PMC7353061 DOI: 10.3390/cancers12061456] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/25/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023] Open
Abstract
The high mobility group AT-Hook (HMGA) proteins are a family of nonhistone chromatin remodeling proteins known as "architectural transcriptional factors". By binding the minor groove of AT-rich DNA sequences, they interact with the transcription apparatus, altering the chromatin modeling and regulating gene expression by either enhancing or suppressing the binding of the more usual transcriptional activators and repressors, although they do not themselves have any transcriptional activity. Their involvement in both benign and malignant neoplasias is well-known and supported by a large volume of studies. In this review, we focus on the role of the HMGA proteins in hematological malignancies, exploring the mechanisms through which they enhance neoplastic transformation and how this knowledge could be exploited to devise tailored therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | - Francesco Albano
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy; (A.M.); (N.C.); (L.A.); (A.Z.); (G.S.)
| |
Collapse
|
2
|
Scheffer-Mendoza S, Espinosa-Padilla SE, López-Herrera G, Mujica-Guzmán F, López-Padilla MG, Berrón-Ruiz L. Reference values of leukocyte and lymphocytes populations in umbilical cord and capillary blood in healthy Mexican newborns. Allergol Immunopathol (Madr) 2020; 48:295-305. [PMID: 32312563 DOI: 10.1016/j.aller.2019.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/19/2019] [Accepted: 12/17/2019] [Indexed: 11/19/2022]
Abstract
INTRODUCTION In newborns, dramatic changes occur in the blood and bone marrow during the first hours; there are rapid fluctuations in the quantities of leukocytes populations. In this work, we investigated leukocytes subsets counts in two types of blood samples (cord blood and capillary blood) extracted from healthy newborns. METHODS Blood samples from Mexican neonates were collected by Instituto Nacional de Pediatría with written informed consent. For all samples we determined leukocytes populations; neutrophils, monocytes, total lymphocytes, and populations: T CD3+ cells, TCD4+ cells, T CD8+ cells, B CD19+ cells and NK CD16+56 cells by flow cytometry. We used the Mann-Whitney U test to compare leukocytes of cord and capillary blood; also to analyze the differences between gender and we obtained reference values of the cord and capillary blood in neonates. RESULTS We observed higher absolute counts and frequencies of total lymphocyte in capillary blood compared with cord blood. In absolute numbers, the capillary blood showed significant differences in neutrophils, monocytes, lymphocytes, T CD3+ cells, T CD4+ cells, T CD8+ cells, B CD19+ cells, and NK cells; no significant differences were observed between genders. DISCUSSION Our data contribute to newborn Mexican reference values for all these populations of leukocytes. We found that the dispersal range differs between the two types of blood, suggesting a different fate in the immune response. Immunophenotyping of the blood cell population to identify these cells is an essential tool in the diagnosis and follow-up of neonates with immunodeficiencies and other immune disorders.
Collapse
Affiliation(s)
- S Scheffer-Mendoza
- Servicio de Inmunología y Alergia, Instituto Nacional de Pediatría, SSa, Ciudad de México, Mexico
| | - S E Espinosa-Padilla
- Unidad de Investigación en Inmunodeficiencias, Instituto Nacional de Pediatría, SSa, Ciudad de México, Mexico
| | - G López-Herrera
- Laboratorio de Hematología, Instituto Nacional de Pediatría, SSa, Ciudad de México, Mexico
| | - F Mujica-Guzmán
- Laboratorio de Hematología, Instituto Nacional de Pediatría, SSa, Ciudad de México, Mexico
| | - M G López-Padilla
- Unidad Tocoquirúgica, Instituto Nacional de Enfermedades Respiratorias "Dr. Manuel Gea González", SSa, Ciudad de México, Mexico
| | - L Berrón-Ruiz
- Unidad de Investigación en Inmunodeficiencias, Instituto Nacional de Pediatría, SSa, Ciudad de México, Mexico.
| |
Collapse
|
3
|
Sudo S, Kajiya H, Okano S, Sasaki M, Katsumata Y, Ohno J, Ikebe T, Hiraki A, Okabe K. Cisplatin-induced programmed cell death ligand-2 expression is associated with metastasis ability in oral squamous cell carcinoma. Cancer Sci 2020; 111:1113-1123. [PMID: 32012401 PMCID: PMC7156784 DOI: 10.1111/cas.14336] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/20/2020] [Accepted: 01/24/2020] [Indexed: 12/12/2022] Open
Abstract
Programmed cell death ligands (PD‐Ls) are expressed in tumor cells where they bind to programmed cell death‐1, an immunocyte co–receptor, resulting in tumor cell evasion from the immune system. Chemotherapeutic drugs have been recently reported to induce the expression of PD‐L, such as PD‐L1, in some cancer cells. However, little is known regarding PD‐L2 expression and its role in oral squamous cell carcinoma (OSCC). In this study, we examined the effect of cisplatin on the expression and regulation of PD‐L2 in OSCC cell lines and analyzed malignant behavior in PD‐L2‐expressing cells using colony, transwell and transformation assays. In addition, we examined PD‐L2 expression in the tumor tissues of OSCC patients using cytology and tissue microarray methods. In OSCC cell lines, cisplatin treatment upregulated PD‐L2 expression, along with that of the drug efflux transporter ABCG2, via signal transducers and activator of transcription (STAT) 1/3 activation. Moreover, PD‐L2‐positive or PD‐L2‐overexpressing cells demonstrated upregulation in both invasion and transformation ability but not in proliferation compared with PD‐L2‐negative or PD‐L2‐silencing cells. PD‐L2 expression was also observed in OSCC cells of cytology samples and tissue from OSCC patients. The intensity of PD‐L2 expression was correlated with more malignant morphological features in the histological appearance and an invasive pattern. Our findings indicate that cisplatin‐upregulated PD‐L2 expression in OSCC via STAT1/3 activation and the expression of PD‐L2 are likely to be associated with malignancy in OSCC. The PD‐L2 expression in cisplatin‐resistant OSCC cells may be a critical factor in prognosis of advanced OSCC patients.
Collapse
Affiliation(s)
- Shunichi Sudo
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan.,Department of Oral Maxillofacial Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - Hiroshi Kajiya
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan.,Research Center for Regenerative Medicine, Fukuoka Dental College, Fukuoka, Japan
| | - Shinji Okano
- Department of Morphological Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Mina Sasaki
- Department of Oral Maxillofacial Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - Yuri Katsumata
- Department of Oral Maxillofacial Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - Jun Ohno
- Research Center for Regenerative Medicine, Fukuoka Dental College, Fukuoka, Japan
| | - Tetsuro Ikebe
- Department of Oral Maxillofacial Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - Akimitsu Hiraki
- Department of Oral Maxillofacial Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - Koji Okabe
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan
| |
Collapse
|
4
|
Kasimanickam V, Kasimanickam R. A Method to Isolate CD34+ Mononuclear Cells from Canine Peripheral Blood. ACTA ACUST UNITED AC 2019; 49:e84. [PMID: 30901513 DOI: 10.1002/cpsc.84] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Embryonic stem cells are pluripotent whereas adult stem cells are multipotent in nature. In recent years, evidence suggests that adult stem cells not only differentiate into specific cell lineages but also transdifferentiate into multiple cell lineages. Progenitor cells are found in adult bone marrow, blood, and other organs and differentiate into numerous cell lineages regardless of origin. Identifying a subset that can differentiate into mature endothelial cells is essential. This article describes peripheral blood collection in adult beagle dogs, isolation of peripheral blood mononuclear cells (PBMNCs) from the cell fraction, separation of a subset of CD34+ cells using immunomagnetic principles, characterization of PBMNCs and CD34+ cells using flow cytometry, and evaluation of gene expression of CD34, KDR, and CD133 in CD34+ fractions. Efficient methods of isolation of endothelial progenitor cells (EPCs) will promote the ex vivo expansion and transplantation of EPCs in ischemic injury to enable neovascularization. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Vanmathy Kasimanickam
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington
| | - Ramanathan Kasimanickam
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington
| |
Collapse
|
5
|
Kovilakath A, Mohamad S, Hermes F, Wang SZ, Ginder GD, Lloyd JA. In Vitro Erythroid Differentiation and Lentiviral Knockdown in Human CD34+ Cells from Umbilical Cord Blood. Methods Mol Biol 2018; 1698:259-274. [PMID: 29076096 DOI: 10.1007/978-1-4939-7428-3_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Human umbilical cord blood is a rich source of hematopoietic stem and progenitor cells. CD34+ cells in umbilical cord blood are more primitive than those in peripheral blood or bone marrow, and can proliferate at a high rate and differentiate into multiple cell types. In this protocol, a dependable method is described for the isolation of fetal CD34+ cells from umbilical cord blood and expanding these cells in culture. The cells can then be in vitro differentiated along an erythroid pathway, while simultaneously performing knockdown of a gene of choice. The use of lentiviral vectors that express small hairpin RNA (shRNA) is an efficient method to downregulate genes. Flow cytometric analyses are used to enrich for erythroid cells. Using these methods, one can generate in vitro differentiated cells to use for quantitative reverse transcriptase PCR and other purposes.
Collapse
Affiliation(s)
- Anna Kovilakath
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Safa Mohamad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Farrah Hermes
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Shou Zhen Wang
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Gordon D Ginder
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Joyce A Lloyd
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA.
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
6
|
Xian L, Georgess D, Huso T, Cope L, Belton A, Chang YT, Kuang W, Gu Q, Zhang X, Senger S, Fasano A, Huso DL, Ewald AJ, Resar LMS. HMGA1 amplifies Wnt signalling and expands the intestinal stem cell compartment and Paneth cell niche. Nat Commun 2017; 8:15008. [PMID: 28452345 PMCID: PMC5414379 DOI: 10.1038/ncomms15008] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/21/2017] [Indexed: 12/15/2022] Open
Abstract
High-mobility group A1 (Hmga1) chromatin remodelling proteins are enriched in intestinal stem cells (ISCs), although their function in this setting was unknown. Prior studies showed that Hmga1 drives hyperproliferation, aberrant crypt formation and polyposis in transgenic mice. Here we demonstrate that Hmga1 amplifies Wnt/β-catenin signalling to enhance self-renewal and expand the ISC compartment. Hmga1 upregulates genes encoding both Wnt agonist receptors and downstream Wnt effectors. Hmga1 also helps to 'build' an ISC niche by expanding the Paneth cell compartment and directly inducing Sox9, which is required for Paneth cell differentiation. In human intestine, HMGA1 and SOX9 are positively correlated, and both become upregulated in colorectal cancer. Our results define a unique role for Hmga1 in intestinal homeostasis by maintaining the stem cell pool and fostering terminal differentiation to establish an epithelial stem cell niche. This work also suggests that deregulated Hmga1 perturbs this equilibrium during intestinal carcinogenesis.
Collapse
Affiliation(s)
- Lingling Xian
- Division of Hematology, Department of Medicine, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Research Building, Room 1025, Baltimore, Maryland 21205, USA
| | - Dan Georgess
- Department of Cell Biology, The Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, Maryland 21205, USA
| | - Tait Huso
- Division of Hematology, Department of Medicine, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Research Building, Room 1025, Baltimore, Maryland 21205, USA
| | - Leslie Cope
- Division of Biostatistics, Department of Oncology, The Johns Hopkins University School of Medicine, 550 North Broadway, Baltimore, Maryland 21205, USA
| | - Amy Belton
- Division of Hematology, Department of Medicine, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Research Building, Room 1025, Baltimore, Maryland 21205, USA
| | - Yu-Ting Chang
- Department of Pathology, Pathobiology Graduate Program, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Research Building, Room 1025, Baltimore, Maryland 21205, USA
| | - Wenyong Kuang
- Division of Hematology, Department of Medicine, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Research Building, Room 1025, Baltimore, Maryland 21205, USA
| | - Qihua Gu
- Division of Hematology, Department of Medicine, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Research Building, Room 1025, Baltimore, Maryland 21205, USA
| | - Xiaoyan Zhang
- Division of Hematology, Department of Medicine, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Research Building, Room 1025, Baltimore, Maryland 21205, USA
| | - Stefania Senger
- Department of Pediatrics, Mucosal Immunology and Biology Research Center, Harvard Medical School, Massachusetts General Hospital East, 16th Street, Building 114, Charlestown, Massachusetts 02114, USA
| | - Alessio Fasano
- Department of Pediatrics, Mucosal Immunology and Biology Research Center, Harvard Medical School, Massachusetts General Hospital East, 16th Street, Building 114, Charlestown, Massachusetts 02114, USA
| | - David L Huso
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Andrew J Ewald
- Department of Cell Biology, The Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, Maryland 21205, USA.,Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Linda M S Resar
- Division of Hematology, Department of Medicine, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Research Building, Room 1025, Baltimore, Maryland 21205, USA.,Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.,Department of Pathology and Institute for Cellular Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
7
|
Potential Role for a β-Catenin Coactivator (High-Mobility Group AT-Hook 1 Protein) during the Latency-Reactivation Cycle of Bovine Herpesvirus 1. J Virol 2017; 91:JVI.02132-16. [PMID: 28003484 DOI: 10.1128/jvi.02132-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 12/12/2016] [Indexed: 12/25/2022] Open
Abstract
The latency-related (LR) RNA encoded by bovine herpesvirus 1 (BoHV-1) is abundantly expressed in latently infected sensory neurons. Although the LR gene encodes several products, ORF2 appears to mediate important steps during the latency-reactivation cycle because a mutant virus containing stop codons at the amino terminus of ORF2 does not reactivate from latency in calves. We recently found that the Wnt/β-catenin signaling pathway is regulated during the BoHV-1 latency-reactivation cycle (Y. Liu, M. Hancock, A. Workman, A. Doster, and C. Jones, J Virol 90:3148-3159, 2016). In the present study, a β-catenin coactivator, high-mobility group AT-hook 1 protein (HMGA1), was detected in significantly more neurons in the trigeminal ganglia of latently infected calves than in those of uninfected calves. Consequently, we hypothesized that HMGA1 cooperates with ORF2 and β-catenin to maintain latency. In support of this hypothesis, coimmunoprecipitation studies demonstrated that ORF2 stably interacts with a complex containing β-catenin and/or HMGA1 in transfected mouse neuroblastoma (Neuro-2A) cells. Confocal microscopy provided evidence that ORF2 was relocalized by HMGA1 and β-catenin in Neuro-2A cells. ORF2 consistently enhanced the ability of HMGA1 to stimulate β-catenin-dependent transcription, suggesting that interactions between ORF2 and a complex containing β-catenin and HMGA1 have functional significance. An ORF2 stop codon mutant, an ORF2 nuclear localization mutant, or a mutant lacking the 5 protein kinase A or C phosphorylation sites interfered with its ability to stimulate β-catenin-dependent transcription. Since the canonical Wnt/β-catenin signaling pathway promotes neurogenesis (synapse formation and remodeling) and inhibits neurodegeneration, interactions between ORF2, HMGA1, and β-catenin may be important for certain aspects of the latency-reactivation cycle.IMPORTANCE The lifelong latency of bovine herpesvirus 1 (BoHV-1) requires that significant numbers of infected sensory neurons survive infection and maintain normal functions. Consequently, we hypothesize that viral products expressed during latency cooperate with neuronal factors to maintain latency. Our studies revealed that a β-catenin coactivator, high-mobility group AT-hook 1 protein (HMGA1), was readily detected in a subset of trigeminal ganglion neurons in latently infected calves but not in uninfected calves. A viral protein (ORF2) expressed in latently infected neurons interacted with β-catenin and HMGA1 in transfected cells, which resulted in the nuclear localization of β-catenin. This interaction correlated with the ability of ORF2 to stimulate the coactivator functions of HMGA1. These findings are significant because the canonical Wnt/β-catenin signaling pathway promotes neurogenesis and inhibits neurodegeneration.
Collapse
|
8
|
Sumter TF, Xian L, Huso T, Koo M, Chang YT, Almasri TN, Chia L, Inglis C, Reid D, Resar LMS. The High Mobility Group A1 (HMGA1) Transcriptome in Cancer and Development. Curr Mol Med 2016; 16:353-93. [PMID: 26980699 DOI: 10.2174/1566524016666160316152147] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 02/15/2016] [Accepted: 03/10/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND & OBJECTIVES Chromatin structure is the single most important feature that distinguishes a cancer cell from a normal cell histologically. Chromatin remodeling proteins regulate chromatin structure and high mobility group A (HMGA1) proteins are among the most abundant, nonhistone chromatin remodeling proteins found in cancer cells. These proteins include HMGA1a/HMGA1b isoforms, which result from alternatively spliced mRNA. The HMGA1 gene is overexpressed in cancer and high levels portend a poor prognosis in diverse tumors. HMGA1 is also highly expressed during embryogenesis and postnatally in adult stem cells. Overexpression of HMGA1 drives neoplastic transformation in cultured cells, while inhibiting HMGA1 blocks oncogenic and cancer stem cell properties. Hmga1 transgenic mice succumb to aggressive tumors, demonstrating that dysregulated expression of HMGA1 causes cancer in vivo. HMGA1 is also required for reprogramming somatic cells into induced pluripotent stem cells. HMGA1 proteins function as ancillary transcription factors that bend chromatin and recruit other transcription factors to DNA. They induce oncogenic transformation by activating or repressing specific genes involved in this process and an HMGA1 "transcriptome" is emerging. Although prior studies reveal potent oncogenic properties of HMGA1, we are only beginning to understand the molecular mechanisms through which HMGA1 functions. In this review, we summarize the list of putative downstream transcriptional targets regulated by HMGA1. We also briefly discuss studies linking HMGA1 to Alzheimer's disease and type-2 diabetes. CONCLUSION Further elucidation of HMGA1 function should lead to novel therapeutic strategies for cancer and possibly for other diseases associated with aberrant HMGA1 expression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - L M S Resar
- Department of Medicine, Faculty of the Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Research Building, Room 1025, Baltimore, MD 21205-2109, USA.
| |
Collapse
|
9
|
Andreozzi M, Quintavalle C, Benz D, Quagliata L, Matter M, Calabrese D, Tosti N, Ruiz C, Trapani F, Tornillo L, Fusco A, Heim MH, Ng CK, Pallante P, Terracciano LM, Piscuoglio S. HMGA1 Expression in Human Hepatocellular Carcinoma Correlates with Poor Prognosis and Promotes Tumor Growth and Migration in in vitro Models. Neoplasia 2016; 18:724-731. [PMID: 27855356 PMCID: PMC5110473 DOI: 10.1016/j.neo.2016.10.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 10/17/2016] [Accepted: 10/17/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND HMGA1 is a non-histone nuclear protein that regulates cellular proliferation, invasion and apoptosis and is overexpressed in many carcinomas. In this study we sought to explore the expression of HMGA1 in HCCs and cirrhotic tissues, and its effect in in vitro models. METHODS We evaluated HMGA1 expression using gene expression microarrays (59 HCCs, of which 37 were matched with their corresponding cirrhotic tissue and 5 normal liver donors) and tissue microarray (192 HCCs, 108 cirrhotic tissues and 79 normal liver samples). HMGA1 expression was correlated with clinicopathologic features and patient outcome. Four liver cancer cell lines with stable induced or knockdown expression of HMGA1 were characterized using in vitro assays, including proliferation, migration and anchorage-independent growth. RESULTS HMGA1 expression increased monotonically from normal liver tissues to cirrhotic tissue to HCC (P<.01) and was associated with Edmondson grade (P<.01). Overall, 51% and 42% of HCCs and cirrhotic tissues expressed HMGA1, respectively. Patients with HMGA1-positive HCCs had earlier disease progression and worse overall survival. Forced expression of HMGA1 in liver cancer models resulted in increased cell growth and migration, and vice versa. Soft agar assay showed that forced expression of HMGA1 led to increased foci formation, suggesting an oncogenic role of HMGA1 in hepatocarcinogenesis. CONCLUSIONS HMGA1 is frequently expressed in cirrhotic tissues and HCCs and its expression is associated with high Edmondson grade and worse prognosis in HCC. Our results suggest that HMGA1 may act as oncogenic driver of progression, implicating it in tumor growth and migration potential in liver carcinogenesis.
Collapse
Affiliation(s)
| | | | - David Benz
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Luca Quagliata
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Matthias Matter
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Diego Calabrese
- Department of Biomedicine, Hepatology Laboratory, University of Basel, Basel, Switzerland
| | - Nadia Tosti
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Christian Ruiz
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Francesca Trapani
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Luigi Tornillo
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Alfredo Fusco
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), and Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples "Federico II, Naples, Italy; National Cancer Institute-INCA, Rua Andrè Cavalcanti, 37-Centro, Rio de Janeiro, Brazil
| | - Markus H Heim
- Department of Biomedicine, Hepatology Laboratory, University of Basel, Basel, Switzerland
| | - Charlotte Ky Ng
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Pierlorenzo Pallante
- Institute of Pathology, University Hospital Basel, Basel, Switzerland; Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), and Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples "Federico II, Naples, Italy
| | | | | |
Collapse
|
10
|
Li J, Wei Y, Yan L, Wang R, Zhang Y, Su Y, Yang Z, Hu M, Qi R, Tan H, Wu Q, Yin X, Pan X. Multiplacenta derived stem cell/cytokine treatment increases survival time in a mouse model with radiation-induced bone marrow damage. Cytotechnology 2016; 68:2677-2686. [PMID: 27318496 DOI: 10.1007/s10616-016-9993-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/06/2016] [Indexed: 02/02/2023] Open
Abstract
Nuclear Warfare and nuclear leakage can result in a large number of patients with radiation-induced bone marrow damage. Based on the fact that hematopoietic stem cells and hematopoietic growth factors are characterized as a novel strategy for therapy, the aim of this study was to explore a safe and routine stem cell/cytokine therapeutic strategy. Allogeneic multiplacenta derived hematopoietic and mesenchymal stem cells/cytokines were intraperitoneally injected into a moderate dose of total body irradiation-induced mouse bone marrow damage model a single time. Then, the mouse posttransplantation survival time, peripheral blood hemoglobin count, bone marrow architecture, and donor cell engraftment were assessed. Each mouse that received placenta-derived stem cells exhibited positive donor hematopoietic and mesenchymal stem cell engraftment both in the bone marrow and peripheral blood after transplantation. The peripheral blood hemoglobin count and survival time were greater in the group with the combined treatment of multiplacenta-derived stem cells and cytokines, compared with model-only controls (both P < 0.001). The blood smear mesenchymal/hematopoietic stem cell count was significantly higher in the combined treatment group than in the mice treated only with placenta-derived cells (28.08 ± 5.824 vs. 20.40 ± 5.989, P < 0.001; 7.74 ± 2.153 vs. 4.23 ± 1.608, P < 0.001, respectively). However, there was no marked change on the bone marrow pathology of any of the experimental mice after the transplantation. These results indicate that for radiation-induced bone marrow damage treatment, multiplacenta-derived stem cells and cytokines can increase the life span of model mice and delay but not abrogate the disease progression. Intraperitoneally transplanted stem cells can survive and engraft into the host body through the blood circulation. Improvement of peripheral blood hemoglobin levels, but not the bone marrow architecture response, probably explains the increase in survival time observed in this study.
Collapse
Affiliation(s)
- Jun Li
- Medical School of Kunming University, Kunming, 650214, China
- State Local Joint Engineering Laboratory of Stem Cell and Immunocyte Biomedical Technology, Kunming General Hospital of Chengdu Military Command, Kunming, 650032, China
| | - Yunfang Wei
- Medical School of Kunming University, Kunming, 650214, China
| | - Lei Yan
- Anesthesiology Department, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
| | - Rui Wang
- Medical School of Kunming University, Kunming, 650214, China
| | - Ying Zhang
- Medical School of Kunming University, Kunming, 650214, China
| | - Yingzhen Su
- Medical School of Kunming University, Kunming, 650214, China
| | - Zhaoyu Yang
- Medical School of Kunming University, Kunming, 650214, China
| | - Min Hu
- Research Center for Molecular Medicine, Kunming University, Kunming, 650214, China
| | - Rui Qi
- Medical School of Kunming University, Kunming, 650214, China
| | - Hongbo Tan
- Orthopedics Department, Kunming General Hospital of Chengdu Military Command, Kunming, 650032, China
| | - Qiong Wu
- Department of Chemical Science and Technology, Kunming University, Kunming, 650214, China
| | - Xudong Yin
- Department of Life Science and Technology, Kunming University, Kunming, 650214, China
| | - Xinghua Pan
- State Local Joint Engineering Laboratory of Stem Cell and Immunocyte Biomedical Technology, Kunming General Hospital of Chengdu Military Command, Kunming, 650032, China.
| |
Collapse
|
11
|
Hordyjewska A, Popiołek Ł, Horecka A. Characteristics of hematopoietic stem cells of umbilical cord blood. Cytotechnology 2014; 67:387-96. [PMID: 25373337 PMCID: PMC4371573 DOI: 10.1007/s10616-014-9796-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 10/10/2014] [Indexed: 11/30/2022] Open
Abstract
Umbilical cord blood collected from the postpartum placenta and cord is a rich source of hematopoietic stem cells (HSCs) and is an alternative to bone marrow transplantation. In this review we wanted to describe the differences (in phenotype, cytokine production, quantity and quality of cells) between stem cells from umbilical cord blood, bone marrow and peripheral blood. HSCs present in cord blood are more primitive than their counterparts in bone marrow or peripheral blood, and have several advantages including high proliferation. With using proper cytokine combination, HSCs can be effectively developed into different cell lines. This process is used in medicine, especially in hematology.
Collapse
Affiliation(s)
- Anna Hordyjewska
- Department of Medical Chemistry, Medical Univeristy of Lublin, Lublin, Poland
| | | | | |
Collapse
|
12
|
Yanagisawa BL, Resar LMS. Hitting the bull's eye: targeting HMGA1 in cancer stem cells. Expert Rev Anticancer Ther 2014; 14:23-30. [PMID: 24410339 DOI: 10.1586/14737140.2013.859988] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Emerging evidence suggests that when cancer cells hijack normal stem cell properties, they acquire the ability to invade, metastasize to distant sites and evade therapy. Thus, eliminating cancer cells with stem cell properties, or cancer stem cells, is of prime importance for the successful treatment of cancer, regardless of the tissue of origin. Previous efforts to target cancer stem cells (CSCs), however, have been largely unsuccessful. Recent studies led to the discovery of a novel role for the high mobility group A1 (HMGA1) protein as a master regulator in both CSCs and normal embryonic stem cells. Here, we present exciting new work unveiling HMGA1 as a promising target for therapies directed at eradicating CSCs.
Collapse
Affiliation(s)
- Breann L Yanagisawa
- Department of Medicine, Pathobiology Graduate Program, Hematology Division, Oncology, the Institute for Cellular Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
13
|
Huso TH, Resar LMS. The high mobility group A1 molecular switch: turning on cancer - can we turn it off? Expert Opin Ther Targets 2014; 18:541-53. [PMID: 24684280 DOI: 10.1517/14728222.2014.900045] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Emerging evidence demonstrates that the high mobility group A1 (HMGA1) chromatin remodeling protein is a key molecular switch required by cancer cells for tumor progression and a poorly differentiated, stem-like state. Because the HMGA1 gene and proteins are expressed at high levels in all aggressive tumors studied to date, research is needed to determine how to 'turn off' this master regulatory switch in cancer. AREAS COVERED In this review, we describe prior studies that underscore the central role of HMGA1 in refractory cancers and we discuss approaches to target HMGA1 in cancer therapy. EXPERT OPINION Given the widespread overexpression of HMGA1 in diverse, aggressive tumors, further research to develop technology to target HMGA1 holds immense promise as potent anticancer therapy. Previous work in preclinical models indicates that delivery of short hairpin RNA or interfering RNA molecules to 'switch off' HMGA1 expression dramatically impairs cancer cell growth and tumor progression. The advent of nanoparticle technology to systemically deliver DNA or RNA molecules to tumors brings this approach even closer to clinical applications, although further efforts are needed to translate these advances into therapies for cancer patients.
Collapse
Affiliation(s)
- Tait H Huso
- The Johns Hopkins University School of Medicine, Hematology Division , Ross Research Building, Room 1015, 720 Rutland Avenue, Baltimore MD 21205 , USA
| | | |
Collapse
|
14
|
Yanagisawa BL, Resar LMS. Hitting the bull’s eye: targeting HMGA1 in cancer stem cells. Expert Rev Anticancer Ther 2013. [DOI: 10.1586/14737140.2014.859988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
15
|
Shah SN, Cope L, Poh W, Belton A, Roy S, Talbot CC, Sukumar S, Huso DL, Resar LMS. HMGA1: a master regulator of tumor progression in triple-negative breast cancer cells. PLoS One 2013; 8:e63419. [PMID: 23658826 PMCID: PMC3642138 DOI: 10.1371/journal.pone.0063419] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 04/04/2013] [Indexed: 12/31/2022] Open
Abstract
Emerging evidence suggests that tumor cells metastasize by co-opting stem cell transcriptional networks, although the molecular underpinnings of this process are poorly understood. Here, we show for the first time that the high mobility group A1 (HMGA1) gene drives metastatic progression in triple negative breast cancer cells (MDA-MB-231, Hs578T) by reprogramming cancer cells to a stem-like state. Silencing HMGA1 expression in invasive, aggressive breast cancer cells dramatically halts cell growth and results in striking morphologic changes from mesenchymal-like, spindle-shaped cells to cuboidal, epithelial-like cells. Mesenchymal genes (Vimentin, Snail) are repressed, while E-cadherin is induced in the knock-down cells. Silencing HMGA1 also blocks oncogenic properties, including proliferation, migration, invasion, and orthotopic tumorigenesis. Metastatic progression following mammary implantation is almost completely abrogated in the HMGA1 knock-down cells. Moreover, silencing HMGA1 inhibits the stem cell property of three-dimensional mammosphere formation, including primary, secondary, and tertiary spheres. In addition, knock-down of HMGA1 depletes cancer initiator/cancer stem cells and prevents tumorigenesis at limiting dilutions. We also discovered an HMGA1 signature in triple negative breast cancer cells that is highly enriched in embryonic stem cells. Together, these findings indicate that HMGA1 is a master regulator of tumor progression in breast cancer by reprogramming cancer cells through stem cell transcriptional networks. Future studies are needed to determine how to target HMGA1 in therapy.
Collapse
Affiliation(s)
- Sandeep N Shah
- Hematology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Di Cello F, Dhara S, Hristov AC, Kowalski J, Elbahloul O, Hillion J, Roy S, Meijerink JPP, Winter SS, Larson RS, Huso DL, Resar L. Inactivation of the Cdkn2a locus cooperates with HMGA1 to drive T-cell leukemogenesis. Leuk Lymphoma 2013; 54:1762-8. [PMID: 23311322 DOI: 10.3109/10428194.2013.764422] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive leukemia with high relapse rates compared to B-lineage ALL. We previously showed that HMGA1a transgenic mice develop aggressive T-ALL, indicating that HMGA1 causes leukemic transformation in vivo. HMGA1 is also highly expressed in embryonic stem cells, hematopoietic stem cells and diverse, refractory human cancers. Disruption of the CDKN2A tumor suppressor locus occurs in most cases of T-ALL and is thought to contribute to leukemic transformation. To determine whether loss of function of CDKN2A cooperates with HMGA1 in T-ALL, we crossed HMGA1a transgenics onto a Cdkn2a null background. We discovered that T-ALL is markedly accelerated in HMGA1a transgenic Cdkn2a null mice. In addition, these mice recapitulate salient clinical and pathologic features of human T-ALL. HMGA1 is also highly overexpressed in human T-ALL. These findings suggest that HMGA1 plays a causative role in T-ALL and could represent a rational therapeutic target.
Collapse
Affiliation(s)
- Francescopaolo Di Cello
- Hematology Division, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Shah SN, Kerr C, Cope L, Zambidis E, Liu C, Hillion J, Belton A, Huso DL, Resar LMS. HMGA1 reprograms somatic cells into pluripotent stem cells by inducing stem cell transcriptional networks. PLoS One 2012; 7:e48533. [PMID: 23166588 PMCID: PMC3499526 DOI: 10.1371/journal.pone.0048533] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 09/26/2012] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Although recent studies have identified genes expressed in human embryonic stem cells (hESCs) that induce pluripotency, the molecular underpinnings of normal stem cell function remain poorly understood. The high mobility group A1 (HMGA1) gene is highly expressed in hESCs and poorly differentiated, stem-like cancers; however, its role in these settings has been unclear. METHODS/PRINCIPAL FINDINGS We show that HMGA1 is highly expressed in fully reprogrammed iPSCs and hESCs, with intermediate levels in ECCs and low levels in fibroblasts. When hESCs are induced to differentiate, HMGA1 decreases and parallels that of other pluripotency factors. Conversely, forced expression of HMGA1 blocks differentiation of hESCs. We also discovered that HMGA1 enhances cellular reprogramming of somatic cells to iPSCs together with the Yamanaka factors (OCT4, SOX2, KLF4, cMYC - OSKM). HMGA1 increases the number and size of iPSC colonies compared to OSKM controls. Surprisingly, there was normal differentiation in vitro and benign teratoma formation in vivo of the HMGA1-derived iPSCs. During the reprogramming process, HMGA1 induces the expression of pluripotency genes, including SOX2, LIN28, and cMYC, while knockdown of HMGA1 in hESCs results in the repression of these genes. Chromatin immunoprecipitation shows that HMGA1 binds to the promoters of these pluripotency genes in vivo. In addition, interfering with HMGA1 function using a short hairpin RNA or a dominant-negative construct blocks cellular reprogramming to a pluripotent state. CONCLUSIONS Our findings demonstrate for the first time that HMGA1 enhances cellular reprogramming from a somatic cell to a fully pluripotent stem cell. These findings identify a novel role for HMGA1 as a key regulator of the stem cell state by inducing transcriptional networks that drive pluripotency. Although further studies are needed, these HMGA1 pathways could be exploited in regenerative medicine or as novel therapeutic targets for poorly differentiated, stem-like cancers.
Collapse
Affiliation(s)
- Sandeep N. Shah
- Hematology Division, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Candace Kerr
- Obstetrics & Gynecology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Leslie Cope
- Oncology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Biostatistics, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Elias Zambidis
- Oncology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Comparative Molecular & Pathobiology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Cyndi Liu
- Obstetrics & Gynecology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Joelle Hillion
- Hematology Division, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Amy Belton
- Hematology Division, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - David L. Huso
- Oncology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Comparative Molecular & Pathobiology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Pathology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Linda M. S. Resar
- Hematology Division, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Oncology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Pediatrics, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
18
|
Belton A, Gabrovsky A, Bae YK, Reeves R, Iacobuzio-Donahue C, Huso DL, Resar LMS. HMGA1 induces intestinal polyposis in transgenic mice and drives tumor progression and stem cell properties in colon cancer cells. PLoS One 2012; 7:e30034. [PMID: 22276142 PMCID: PMC3262796 DOI: 10.1371/journal.pone.0030034] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 12/12/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Although metastatic colon cancer is a leading cause of cancer death worldwide, the molecular mechanisms that enable colon cancer cells to metastasize remain unclear. Emerging evidence suggests that metastatic cells develop by usurping transcriptional networks from embryonic stem (ES) cells to facilitate an epithelial-mesenchymal transition (EMT), invasion, and metastatic progression. Previous studies identified HMGA1 as a key transcription factor enriched in ES cells, colon cancer, and other aggressive tumors, although its role in these settings is poorly understood. METHODS/PRINCIPAL FINDINGS To determine how HMGA1 functions in metastatic colon cancer, we manipulated HMGA1 expression in transgenic mice and colon cancer cells. We discovered that HMGA1 drives proliferative changes, aberrant crypt formation, and intestinal polyposis in transgenic mice. In colon cancer cell lines from poorly differentiated, metastatic tumors, knock-down of HMGA1 blocks anchorage-independent cell growth, migration, invasion, xenograft tumorigenesis and three-dimensional colonosphere formation. Inhibiting HMGA1 expression blocks tumorigenesis at limiting dilutions, consistent with depletion of tumor-initiator cells in the knock-down cells. Knock-down of HMGA1 also inhibits metastatic progression to the liver in vivo. In metastatic colon cancer cells, HMGA1 induces expression of Twist1, a gene involved in embryogenesis, EMT, and tumor progression, while HMGA1 represses E-cadherin, a gene that is down-regulated during EMT and metastatic progression. In addition, HMGA1 is among the most enriched genes in colon cancer compared to normal mucosa. CONCLUSIONS Our findings demonstrate for the first time that HMGA1 drives proliferative changes and polyp formation in the intestines of transgenic mice and induces metastatic progression and stem-like properties in colon cancer cells. These findings indicate that HMGA1 is a key regulator, both in metastatic progression and in the maintenance of a stem-like state. Our results also suggest that HMGA1 or downstream pathways could be rational therapeutic targets in metastatic, poorly differentiated colon cancer.
Collapse
Affiliation(s)
- Amy Belton
- Hematology Division, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Alexander Gabrovsky
- Hematology Division, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Young Kyung Bae
- Department of Pathology, Yeungnam University College of Medicine, Daegu, South Korea
| | - Ray Reeves
- School of Molecular Biosciences, Washington State University, Pullman, Washington, United States of America
| | - Christine Iacobuzio-Donahue
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - David L. Huso
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Linda M. S. Resar
- Hematology Division, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
19
|
Nelson DM, Joseph B, Hillion J, Segal J, Karp JE, Resar LMS. Flavopiridol induces BCL-2 expression and represses oncogenic transcription factors in leukemic blasts from adults with refractory acute myeloid leukemia. Leuk Lymphoma 2011; 52:1999-2006. [PMID: 21728742 PMCID: PMC3214625 DOI: 10.3109/10428194.2011.591012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Flavopiridol is a cyclin-dependent kinase inhibitor that induces cell cycle arrest, apoptosis, and clinical responses in selected patients with acute myeloid leukemia (AML). A better understanding of the molecular pathways targeted by flavopiridol is needed to design optimal combinatorial therapy. Here, we report that in vivo administration of flavopiridol induced expression of the BCL-2 anti-apoptotic gene in leukemic blasts from adult patients with refractory AML. Moreover, flavopiridol repressed the expression of genes encoding oncogenic transcription factors (HMGA1, STAT3, E2F1) and the major subunit of RNA Polymerase II. Our results provide mechanistic insight into the cellular pathways targeted by flavopiridol. Although further studies are needed, our findings also suggest that blocking anti-apoptotic pathways could enhance cytotoxicity with flavopiridol.
Collapse
Affiliation(s)
- Dwella M. Nelson
- Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Biju Joseph
- Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Joelle Hillion
- Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Jodi Segal
- Division of General Internal Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Judith E. Karp
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Division of General Internal Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Linda M. S. Resar
- Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Division of General Internal Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Division of Hematologic Malignancy; The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
20
|
Surgical Therapy of End-Stage Heart Failure: Understanding Cell-Mediated Mechanisms Interacting with Myocardial Damage. Int J Artif Organs 2011; 34:529-45. [DOI: 10.5301/ijao.5000004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2011] [Indexed: 01/19/2023]
Abstract
Worldwide, cardiovascular disease results in an estimated 14.3 million deaths per year, giving rise to an increased demand for alternative and advanced treatment. Current approaches include medical management, cardiac transplantation, device therapy, and, most recently, stem cell therapy. Research into cell-based therapies has shown this option to be a promising alternative to the conventional methods. In contrast to early trials, modern approaches now attempt to isolate specific stem cells, as well as increase their numbers by means of amplifying in a culture environment. The method of delivery has also been improved to minimize the risk of micro-infarcts and embolization, which were often observed after the use of coronary catheterization. The latest approach entails direct, surgical, transepicardial injection of the stem cell mixture, as well as the use of tissue-engineered meshes consisting of embedded progenitor cells.
Collapse
|
21
|
Efficient human iPS cell derivation by a non-integrating plasmid from blood cells with unique epigenetic and gene expression signatures. Cell Res 2011; 21:518-29. [PMID: 21243013 PMCID: PMC3193421 DOI: 10.1038/cr.2011.12] [Citation(s) in RCA: 345] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
To identify accessible and permissive human cell types for efficient derivation of induced pluripotent stem cells (iPSCs), we investigated epigenetic and gene expression signatures of multiple postnatal cell types such as fibroblasts and blood cells. Our analysis suggested that newborn cord blood (CB) and adult peripheral blood (PB) mononuclear cells (MNCs) display unique signatures that are closer to iPSCs and human embryonic stem cells (ESCs) than age-matched fibroblasts to iPSCs/ESCs, thus making blood MNCs an attractive cell choice for the generation of integration-free iPSCs. Using an improved EBNA1/OriP plasmid expressing 5 reprogramming factors, we demonstrated highly efficient reprogramming of briefly cultured blood MNCs. Within 14 days of one-time transfection by one plasmid, up to 1000 iPSC-like colonies per 2 million transfected CB MNCs were generated. The efficiency of deriving iPSCs from adult PB MNCs was approximately 50-fold lower, but could be enhanced by inclusion of a second EBNA1/OriP plasmid for transient expression of additional genes such as SV40 T antigen. The duration of obtaining bona fide iPSC colonies from adult PB MNCs was reduced to half (∼14 days) as compared to adult fibroblastic cells (28–30 days). More than 9 human iPSC lines derived from PB or CB blood cells are extensively characterized, including those from PB MNCs of an adult patient with sickle cell disease. They lack V(D)J DNA rearrangements and vector DNA after expansion for 10–12 passages. This facile method of generating integration-free human iPSCs from blood MNCs will accelerate their use in both research and future clinical applications.
Collapse
|
22
|
Phase 1 and pharmacokinetic study of bolus-infusion flavopiridol followed by cytosine arabinoside and mitoxantrone for acute leukemias. Blood 2011; 117:3302-10. [PMID: 21239698 DOI: 10.1182/blood-2010-09-310862] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Flavopiridol is a protein bound, cytotoxic, cyclin-dependent kinase inhibitor. Flavopiridol given by 1-hour bolus at 50 mg/m(2) daily 3 times followed by cytosine arabinoside and mitoxantrone (FLAM) is active in adults with poor-risk acute leukemias. A pharmacologically derived "hybrid" schedule (30-minute bolus followed by 4-hour infusion) of flavopiridol was more effective than bolus administration in refractory chronic lymphocytic leukemia. Our phase 1 trial "hybrid FLAM" in 55 adults with relapsed/refractory acute leukemias began at a total flavopiridol dose of 50 mg/m(2) per day 3 times (20-mg/m(2) bolus, 30-mg/m(2) infusion). Dose-limiting toxicity occurred at level 6 (30-mg/m(2) bolus, 70-mg/m(2) infusion) with tumor lysis, hyperbilirubinemia, and mucositis. Death occurred in 5 patients (9%). Complete remission occurred in 22 (40%) across all doses. Overall and disease-free survivals for complete remission patients are more than 60% at more than 2 years. Pharmacokinetics demonstrated a dose-response for total and unbound plasma flavopiridol unrelated to total protein, albumin, peripheral blast count, or toxicity. Pharmacodynamically, flavopiridol inhibited mRNAs of multiple cell cycle regulators, but with uniform increases in bcl-2. "Hybrid FLAM" is active in relapsed/refractory acute leukemias, with a recommended "hybrid" dose of bolus 30 mg/m(2) followed by infusion of 60 mg/m(2) daily for 3 days. This clinical trial is registered at www.clinicaltrials.gov as #NCT00470197.
Collapse
|
23
|
Segawa T, Itou T, Suzuki M, Moritomo T, Nakanishi T, Sakai T. Hematopoietic cell populations in dolphin bone marrow: Analysis of colony formation and differentiation. RESULTS IN IMMUNOLOGY 2011; 1:1-5. [PMID: 24371545 DOI: 10.1016/j.rinim.2011.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 05/12/2011] [Indexed: 11/20/2022]
Abstract
Bone marrow biopsy is useful for diagnosis of hematopoietic diseases. We have recently reported that bone marrow biopsy from the flipper might be useful for diagnosis of hematopoietic diseases in dolphins. In this study, to demonstrate whether biopsy from the flipper is useful for clinical diagnosis, we investigated the gene expression profiles and proliferation and differentiation of bone marrow mononuclear cell (BMMC) isolated from the humeral bone marrow of bottlenose dolphins. BMMC exhibited gene expression profiles considered to be characteristic of hematopoietic cells. Similarly, a colony forming unit assay showed that dolphin BMMC possessed vigorous colony forming ability. The proliferation of hematopoietic progenitor cells resulted in the formation of three types of colonies, containing neutrophils, monocytes/macrophages and eosinophils with or without megakaryocytes, all of which could be identified based on the morphological characteristics and gene expression profiles typically associated with hematopoietic markers. Thus, dolphin BMMCs from humeral bone marrow contain many hematopoietic progenitor cells, and bone marrow biopsy from the flipper is suggested useful for clinical diagnosis for the dolphins.
Collapse
Key Words
- BMMC, bone marrow mononuclear cell
- Bone marrow
- CFU assay, colony forming unit assay
- Colony forming unit assay
- Dolphin
- EBF, early B-cell factor
- EPOR, erythropoietin receptor
- Epx, eosinophil peroxidase
- FBS, fetal bovine serum
- G-CSFR, granulocyte colony-stimulating factor receptor
- Gene expression
- Hematopoiesis
- Humeral bone
- IMDM, Iscove's Modified Dulbecco's Medium
- LCM, lymphocyte conditioned medium
- M-CSFR, macrophage colony-stimulating factor receptor
- M-MuLV, Moloney murine leukemia virus
- MPO, myeloperoxidase
- MSR, macrophage scavenger receptor
- NE, neutrophil elastase
- PBMC, peripheral blood mononuclear cell
- PBS, phosphate buffered saline
- PHA, phytohemagglutinin
- PMN, polymorphonuclear leukocyte
- Pax5, paired box gene 5
- RT-PCR, reverse transcription-polymerase chain reaction
- SCL, stem cell leukemia
- TCRβ, T cell receptor β
Collapse
Affiliation(s)
- Takao Segawa
- Nihon University Veterinary Research Center, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Takuya Itou
- Nihon University Veterinary Research Center, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Miwa Suzuki
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Tadaaki Moritomo
- Nihon University Veterinary Research Center, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Teruyuki Nakanishi
- Nihon University Veterinary Research Center, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Takeo Sakai
- Nihon University Veterinary Research Center, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| |
Collapse
|
24
|
da Cunha AF, Brugnerotto AF, Duarte AS, Lanaro C, Costa GGL, Saad STO, Costa FF. Global gene expression reveals a set of new genes involved in the modification of cells during erythroid differentiation. Cell Prolif 2010; 43:297-309. [PMID: 20546246 DOI: 10.1111/j.1365-2184.2010.00679.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVES Erythroid differentiation is a dynamic process in which a pluripotent stem cell undergoes a series of developmental changes that commit it to a specific lineage. These alterations involve changes in gene expression profiles. In this study, gene expression profiles during differentiation of human erythroid cells of a normal blood donor were evaluated using SAGE. MATERIALS AND METHODS Global gene expression was evaluated in cells collected immediately before addition of erythropoietin (0 h) and 192 and 336 h after addition of this hormone. Real-time PCR was used to evaluate activation of differentially expressed genes. RESULTS The data indicate that global aspects of the transcriptome were similar during differentiation of the majority of the genes and that a relatively small set of genes is probably involved in modification of erythroid cells during differentiation. We have identified 93 differentially expressed genes during erythroid development, and expression of some of these was confirmed by qPCR. Various genes including EYA3, ERH, HES6, TIMELESS and TRIB3 were found to be homologous to those of Drosophila melanogaster and here are described for the first time during erythroid development. An important and unique carboxypeptidase inhibitor described in mammalians, LXN, was also identified. CONCLUSIONS The results of this study amplify previously published data and may contribute to comprehension of erythroid differentiation and identification of new target genes involved in some erythroid concerning diseases.
Collapse
Affiliation(s)
- A F da Cunha
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil.
| | | | | | | | | | | | | |
Collapse
|
25
|
Wang SM, Zhang MQ. Transcriptome study for early hematopoiesis--achievement, challenge and new opportunity. J Cell Physiol 2010; 223:549-52. [PMID: 20143329 DOI: 10.1002/jcp.22065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Hematopoietic stem progenitor cells are the source for the entire hematopoietic system. Studying gene expression in hematopoietic stem progenitor cells will provide information to understand the genetic programs controlling early hematopoiesis, and to identify the gene targets to interfere hematopoietic disorders. Extensive efforts using cell biology, molecular biology, and genomics approaches have generated rich knowledge for the genes and functional pathways involving in early hematopoiesis. Challenges remain, however, including the rarity of the hematopoietic stem progenitor cells that set physical limitation for the study, the difficulty for reaching comprehensive transcriptome detection under the conventional genomics technologies, and the difficulty for using conventional biological methods to identify the key genes among large number of expressed genes controlling stem cell self-renewal and differentiation. The newly developed single-cell transcriptome method and the next-generation DNA sequencing technology provide new opportunities for transcriptome study for early hematopoietic. Using systems biology approach may reveal the insight of the genetic mechanisms controlling early hematopoiesis.
Collapse
Affiliation(s)
- San Ming Wang
- Northshore University HealthSystem Research Institute, University of Chicago Pritzker School of Medicine, Evanston, Illinois, USA.
| | | |
Collapse
|
26
|
D'Alessandro A, Liumbruno G, Grazzini G, Pupella S, Lombardini L, Zolla L. Umbilical cord blood stem cells: Towards a proteomic approach. J Proteomics 2010; 73:468-82. [DOI: 10.1016/j.jprot.2009.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 06/04/2009] [Accepted: 06/16/2009] [Indexed: 02/07/2023]
|
27
|
The transcriptome of human CD34+ hematopoietic stem-progenitor cells. Proc Natl Acad Sci U S A 2009; 106:8278-83. [PMID: 19416867 DOI: 10.1073/pnas.0903390106] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Studying gene expression at different hematopoietic stages provides insights for understanding the genetic basis of hematopoiesis. We analyzed gene expression in human CD34(+) hematopoietic cells that represent the stem-progenitor population (CD34(+) cells). We collected >459,000 transcript signatures from CD34(+) cells, including the de novo-generated 3' ESTs and the existing sequences of full-length cDNAs, ESTs, and serial analysis of gene expression (SAGE) tags, and performed an extensive annotation on this large set of CD34(+) transcript sequences. We determined the genes expressed in CD34(+) cells, verified the known genes and identified the new genes of different functional categories involved in hematopoiesis, dissected the alternative gene expression including alternative transcription initiation, splicing, and adenylation, identified the antisense and noncoding transcripts, determined the CD34(+) cell-specific gene expression signature, and developed the CD34(+) cell-transcription map in the human genome. Our study provides a current view on gene expression in human CD34(+) cells and reveals that early hematopoiesis is an orchestrated process with the involvement of over half of the human genes distributed in various functions. The data generated from our study provide a comprehensive and uniform resource for studying hematopoiesis and stem cell biology.
Collapse
|
28
|
Hu YF, Caron MG, Sieber-Blum M. Norepinephrine transport-mediated gene expression in noradrenergic neurogenesis. BMC Genomics 2009; 10:151. [PMID: 19356247 PMCID: PMC2679758 DOI: 10.1186/1471-2164-10-151] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2008] [Accepted: 04/08/2009] [Indexed: 11/10/2022] Open
Abstract
Background We have identified a differential gene expression profile in neural crest stem cells that is due to deletion of the norepinephrine transporter (NET) gene. NET is the target of psychotropic substances, such as tricyclic antidepressants and the drug of abuse, cocaine. NET mutations have been implicated in depression, anxiety, orthostatic intolerance and attention deficit hyperactivity disorder (ADHD). NET function in adult noradrenergic neurons of the peripheral and central nervous systems is to internalize norepinephrine from the synaptic cleft. By contrast, during embryogenesis norepinephrine (NE) transport promotes differentiation of neural crest stem cells and locus ceruleus progenitors into noradrenergic neurons, whereas NET inhibitors block noradrenergic differentiation. While the structure of NET und the regulation of NET function are well described, little is known about downstream target genes of norepinephrine (NE) transport. Results We have prepared gene expression profiles of in vitro differentiating wild type and norepinephrine transporter-deficient (NETKO) mouse neural crest cells using long serial analysis of gene expression (LongSAGE). Comparison analyses have identified a number of important differentially expressed genes, including genes relevant to neural crest formation, noradrenergic neuron differentiation and the phenotype of NETKO mice. Examples of differentially expressed genes that affect noradrenergic cell differentiation include genes in the bone morphogenetic protein (BMP) signaling pathway, the Phox2b binding partner Tlx2, the ubiquitin ligase Praja2, and the inhibitor of Notch signaling, Numbl. Differentially expressed genes that are likely to contribute to the NETKO phenotype include dopamine-β-hydroxylase (Dbh), tyrosine hydroxylase (Th), the peptide transmitter 'cocaine and amphetamine regulated transcript' (Cart), and the serotonin receptor subunit Htr3a. Real-time PCR confirmed differential expression of key genes not only in neural crest cells, but also in the adult superior cervical ganglion and locus ceruleus. In addition to known genes we have identified novel differentially expressed genes and thus provide a valuable database for future studies. Conclusion Loss of NET function during embryonic development in the mouse deregulates signaling pathways that are critically involved in neural crest formation and noradrenergic cell differentiation. The data further suggest deregulation of signaling pathways in the development and/or function of the NET-deficient peripheral, central and enteric nervous systems.
Collapse
Affiliation(s)
- Yao Fei Hu
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | | | | |
Collapse
|
29
|
Xu WJ, Li QL, Yao CJ, Wang ZX, Zhao YX, Qiao ZD. Semi-nested PCR analysis of unknown tags on serial analysis of gene expression. FEBS J 2008; 275:5422-8. [PMID: 18959766 DOI: 10.1111/j.1742-4658.2008.06671.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Serial analysis of gene expression (SAGE) is a powerful technique for studying gene expression at the genome level. However, short SAGE tags limit the further study of related data. In this study, in order to identify a gene, we developed a semi-nested PCR-based method called the two-step analysis of unknown SAGE tags (TSAT-PCR) to generate longer 3'-end cDNA fragments from unknown SAGE tags. In the procedure, a modified lock-docking oligo(dT) with two degenerate nucleotide positions at the 3'-end was used as a reverse primer to synthesize cDNAs. Afterwards, the full-length cDNAs were amplified by PCR based on 5'-RACE and 3'-RACE. The amplified cDNAs were then used for the subsequent two-step PCR of the TSAT-PCR process. The first-step PCR was carried out at an appropriately low annealing temperature; a SAGE tag-specific primer was used as the sense primer, and an 18 bp sequence (universal primer I) located at the 5'-reverse primer end was used as the antisense primer. After 15-20 PCR cycles, the 3'-end cDNA fragments containing the tag could be enriched, and the PCR products could be used as templates for the second-step PCR to obtain the specific products. The second-step PCR was performed with a SAGE tag-specific primer and a 22-bp sequence (universal primer II) upstream of universal primer I at the 5'-reverse primer with a high annealing temperature. With our innovative TSAT-PCR method, we could easily obtain specific PCR products covering SAGE from those transcripts, especially low-abundance transcripts. It can be used as a method to identify genes expressed in different cell types.
Collapse
Affiliation(s)
- Wang-Jie Xu
- College of Life Science and Technology, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
30
|
Lymphoid-affiliated genes are associated with active histone modifications in human hematopoietic stem cells. Blood 2008; 112:2722-9. [PMID: 18625888 DOI: 10.1182/blood-2008-02-140806] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To address the role of chromatin structure in the establishment of hematopoietic stem cell (HSC) multilineage potential and commitment to the lymphoid lineage, we have analyzed histone modifications at a panel of lymphoid- and myeloid-affiliated genes in multipotent and lineage-committed hematopoietic cells isolated from human cord blood. Our results show that many B- and T-lymphoid genes, although silent in HSCs, are associated with acetylated histones H3 and H4. We also detected histone H3 lysine 4 methylation but not repressive lysine 9 or 27 methylation marks at these loci, indicative of an open chromatin structure. Interestingly, the relative level of H3 lysine 4 dimethylation to trimethylation at B-specific loci was high in multipotent CD34(+)CD38(lo) progenitors and decreased as they become actively transcribed in B-lineage cells. In vitro differentiation of CD34(+) cells toward the erythroid, granulocyte, and T-cell lineages resulted in a loss of histone acetylation at nonlineage-associated genes. This study provides evidence that histone modifications involved in chromatin decondensation are already in place at lymphoid-specific genes in primary human HSCs, supporting the idea that these genes are "primed" for expression before lineage commitment. This permissive chromatin structure is progressively lost as the stem cell differentiates.
Collapse
|
31
|
Covas DT, Panepucci RA, Fontes AM, Silva WA, Orellana MD, Freitas MCC, Neder L, Santos ARD, Peres LC, Jamur MC, Zago MA. Multipotent mesenchymal stromal cells obtained from diverse human tissues share functional properties and gene-expression profile with CD146+ perivascular cells and fibroblasts. Exp Hematol 2008; 36:642-54. [PMID: 18295964 DOI: 10.1016/j.exphem.2007.12.015] [Citation(s) in RCA: 432] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2007] [Revised: 12/17/2007] [Accepted: 12/21/2007] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The relationship of multipotent mesenchymal stromal cells (MSC) with pericytes and fibroblasts has not been established thus far, although they share many markers of primitive marrow stromal cells and the osteogenic, adipogenic, and chondrogenic differentiation potentials. MATERIALS AND METHODS We compared MSCs from adult or fetal tissues, MSC differentiated in vitro, fibroblasts and cultures of retinal pericytes obtained either by separation with anti-CD146 or adhesion. The characterizations included morphological, immunophenotypic, gene-expression profile, and differentiation potential. RESULTS Osteogenic, adipocytic, and chondrocytic differentiation was demonstrated for MSC, retinal perivascular cells, and fibroblasts. Cell morphology and the phenotypes defined by 22 markers were very similar. Analysis of the global gene expression obtained by serial analysis of gene expression for 17 libraries and by reverse transcription polymerase chain reaction of 39 selected genes from 31 different cell cultures, revealed similarities among MSC, retinal perivascular cells, and hepatic stellate cells. Despite this overall similarity, there was a heterogeneous expression of genes related to angiogenesis, in MSC derived from veins, artery, perivascular cells, and fibroblasts. Evaluation of typical pericyte and MSC transcripts, such as NG2, CD146, CD271, and CD140B on CD146 selected perivascular cells and MSC by real-time polymerase chain reaction confirm the relationship between these two cell types. Furthermore, the inverse correlation between fibroblast-specific protein-1 and CD146 transcripts observed on pericytes, MSC, and fibroblasts highlight their potential use as markers of this differentiation pathway. CONCLUSION Our results indicate that human MSC and pericytes are similar cells located in the wall of the vasculature, where they function as cell sources for repair and tissue maintenance, whereas fibroblasts are more differentiated cells with more restricted differentiation potential.
Collapse
Affiliation(s)
- Dimas T Covas
- Department of Clinical Medicine, Center for Cell Therapy and Regional Blood Center, University of São Paulo, Ribeirão Preto, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Characterization of NKIP: a novel, Na+/K+-ATPase interacting protein mediates neural differentiation and apoptosis. Exp Cell Res 2007; 314:463-77. [PMID: 18096156 DOI: 10.1016/j.yexcr.2007.11.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2007] [Revised: 11/05/2007] [Accepted: 11/08/2007] [Indexed: 01/17/2023]
Abstract
Cellular differentiation and programmed cell death are tightly controlled to maintain tissue homeostasis and proper organ function. In a screen for apoptosis specific gene products, we isolated an immediate early apoptosis response gene from myelomonocytic stem cells that appears to play a key regulatory role in a number of cell types and may be of particular importance in cells of the central nervous system. The gene's 28 kDa protein product interacts with the C-terminal ectodomain of the Na+/K+-ATPase (NKA) beta 1 subunit and was therefore named NKIP (NKA Interacting Protein). NKIP is coexpressed with NKA, localizes to lysosomes and the endoplasmic reticulum and is predominantly expressed in excitable tissues including polarized epithelia and the central nervous system. NKIP has been characterized as an endogenous suppressor of the NKA as reduction of NKIP in PC12 cells significantly increases NKA activity. In pluripotent NT2 progenitor cells, NKIP induced rapidly K+-level-dependent cell death. NKIP overexpression induced growth factor-independent neurite outgrowth, which was associated with MEK-independent phosphorylation of the transcription factor ERK1/2. Thus, we have identified NKIP as an important novel protein that interacts to the NKA complex, influencing cellular ion balance, induction of apoptosis and neuronal differentiation.
Collapse
|
33
|
Li S, Jiang W, Huang R, Wang X, Liu W, Shen S. The gene expression patterns of peripheral blood mononuclear cells in patients with systemic lupus erythematosus. JOURNAL OF HUAZHONG UNIVERSITY OF SCIENCE AND TECHNOLOGY. MEDICAL SCIENCES = HUA ZHONG KE JI DA XUE XUE BAO. YI XUE YING DE WEN BAN = HUAZHONG KEJI DAXUE XUEBAO. YIXUE YINGDEWEN BAN 2007; 27:367-71. [PMID: 17828488 DOI: 10.1007/s11596-007-0405-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2007] [Indexed: 10/22/2022]
Abstract
This study examined the gene expression patterns of peripheral blood mononuclear cells (PBMCs) in patients with systemic lupus erythematosus (SLE) by using serial analysis of gene expression (SAGE) technology. Following the construction of serial analysis of gene expression (SAGE) library of PBMCs collected from 3 cases of familial SLE patients, a large scale of tag sequencing was performed. The data extracted from sequencing files was analyzed with SAGE 2000 V 4.5 software. The top 30 expressed genes of SLE patients were uploaded to http://david.niaid.nih.gov/david/ease.htm and the functional classification of genes was obtained. The differences among those expressed gene were analyzed by Chi-square tests. The results showed that a total of 1286 unique SAGE tags were identified from 1814 individual SAGE tags. Among the 1286 unique tags, 86.8% had single copy, and only 0.2% tags had more than 20 copies. And 68.4% of the tags matched known expressed sequences, 41.1% of which matched more than one known expressed sequence. About 31.6% of the tags had no match and could represent potentially novel genes. Approximately one third of the top 30 genes were ribosomal protein, and the rest were genes related to metabolism or with unknown functions. Eight tags were found to express differentially in SAGE library of SLE patients. This study draws a profile of gene expression patterns of PBMCs in patients with SLE. Comparison of SAGE database from PBMCs between normal individuals and SLE patients will help us to better understand the pathogenesis of SLE.
Collapse
Affiliation(s)
- Shouxin Li
- Department of Rheumatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | | | |
Collapse
|
34
|
Zhao Y, Raouf A, Kent D, Khattra J, Delaney A, Schnerch A, Asano J, McDonald H, Chan C, Jones S, Marra MA, Eaves CJ. A Modified Polymerase Chain Reaction-Long Serial Analysis of Gene Expression Protocol Identifies Novel Transcripts in Human CD34+Bone Marrow Cells. Stem Cells 2007; 25:1681-9. [PMID: 17412892 DOI: 10.1634/stemcells.2006-0794] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Transcriptome profiling offers a powerful approach to investigating developmental processes. Long serial analysis of gene expression (LongSAGE) is particularly attractive for this purpose because of its inherent quantitative features and independence of both hybridization variables and prior knowledge of transcript identity. Here, we describe the validation and initial application of a modified protocol for amplifying cDNA preparations from <10 ng of RNA (<10(3) cells) to allow representative LongSAGE libraries to be constructed from rare stem cell-enriched populations. Quantitative real-time polymerase chain reaction (Q-RT-PCR) analyses and comparison of tag frequencies in replicate LongSAGE libraries produced from amplified and nonamplified cDNA preparations demonstrated preservation of the relative levels of different transcripts originally present at widely varying levels. This PCR-LongSAGE protocol was then used to obtain a 200,000-tag library from the CD34+ subset of normal adult human bone marrow cells. Analysis of this library revealed many anticipated transcripts, as well as transcripts not previously known to be present in CD34+ hematopoietic cells. The latter included numerous novel tags that mapped to unique and conserved sites in the human genome but not previously identified as transcribed elements in human cells. Q-RT-PCR was used to demonstrate that 10 of these novel tags were expressed in cDNA pools and present in extracts of other sources of normal human CD34+ hematopoietic cells. These findings illustrate the power of LongSAGE to identify new transcripts in stem cell-enriched populations and indicate the potential of this approach to be extended to other sources of rare cells. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Yun Zhao
- Terry Fox Laboratory, Vancouver, BC, Canada V5Z 1L3
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Chateauvieux S, Ichanté JL, Delorme B, Frouin V, Piétu G, Langonné A, Gallay N, Sensebé L, Martin MT, Moore KA, Charbord P. Molecular profile of mouse stromal mesenchymal stem cells. Physiol Genomics 2006; 29:128-38. [PMID: 17179208 DOI: 10.1152/physiolgenomics.00197.2006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
We determined a transcriptional profile specific for clonal stromal mesenchymal stem cells from adult and fetal hematopoietic sites. To identify mesenchymal stem cell-like stromal cell lines, we evaluated the adipocytic, osteoblastic, chondrocytic, and vascular smooth muscle differentiation potential and also the hematopoietic supportive (stromal) capacity of six mouse stromal cell lines from adult bone marrow and day 14.5 fetal liver. We found that two lines were quadripotent and also supported hematopoiesis, BMC9 from bone marrow and AFT024 from fetal liver. We then ascertained the set of genes differentially expressed in the intersection set of AFT024 and BMC9 compared with those expressed in the union set of two negative control lines, 2018 and BFC012 (both from fetal liver); 346 genes were upregulated and 299 downregulated. Using Ingenuity software, we found two major gene networks with highly significant scores. One network contained downregulated genes that are known to be implicated in osteoblastic differentiation, proliferation, or transformation. The other network contained upregulated genes that belonged to two categories, cytoskeletal genes and genes implicated in the transcriptional machinery. The data extend the concept of stromal mesenchymal stem cells to clonal cell populations derived not only from bone marrow but also from fetal liver. The gene networks described should discriminate this cell type from other types of stem cells and help define the stem cell state.
Collapse
Affiliation(s)
- Sebastien Chateauvieux
- Institut National de la Santé et de la Recherche Médicale, Equipe-ESPRI/EA-3855, Université François Rabelais, Faculté de Médecine, Tours, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Wang SM. Applying the SAGE technique to study the effects of electromagnetic field on biological systems. Proteomics 2006; 6:4765-8. [PMID: 16897688 DOI: 10.1002/pmic.200500881] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Identification of genes alternatively expressed in electromagnetic field (EMF)-exposed cells could provide direct evidence for biological effects of EMF. As there are a few indications so far for certain genes to be influenced by EMF, genome-wide scans of the transcriptome appear to be necessary. Among the several technologies used for genome-wide gene expression analysis, serial analysis of gene expression (SAGE) is one promising method, which seems particularly applicable for EMF research. This review provides a brief description of the features of gene expression, illustrates the basic principle of SAGE, and discusses the advantages and limitations of SAGE as well as examples of application. This information should help investigators determine if the SAGE technique is an optimal method for evaluating the biological effects of EMF.
Collapse
Affiliation(s)
- San Ming Wang
- Center for Functional Genomics, ENH Research Institute, Department of Medicine, Northwestern University, Evanston, IL 60201, USA.
| |
Collapse
|
37
|
Liu F, Lu J, Fan HH, Wang ZQ, Cui SJ, Zhang GA, Chi M, Zhang X, Yang PY, Chen Z, Han ZG. Insights into human CD34+ hematopoietic stem/progenitor cells through a systematically proteomic survey coupled with transcriptome. Proteomics 2006; 6:2673-92. [PMID: 16596711 DOI: 10.1002/pmic.200500032] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Hematopoietic stem cells are capable of self-renewal and differentiation into different hematopoietic lineages. To gain a comprehensive understanding of hematopoietic stem/progenitor cells, a systematic proteomic survey of human CD34+ cells collected from human umbilical cord blood was performed, in which the proteins were separated by 1- and 2-DE, as well as by nano-LC, and subsequently identified by MS. A total of 370 distinct proteins identified from those cells provided new insights into the potential of the stem/progenitor cells because the nerve, gonad, and eye-associated proteins were reliably identified. Interestingly, the transcripts of 133 (35.9%) identified proteins were not found by the prevalent transcriptome approaches, although several selected transcripts could be detected by RT-PCR. Moreover, the heterogeneity of 33 proteins identified from 2-DE was attributable primarily to post-translational processes rather than to alternative splicing at transcriptional level. Furthermore, the biosyntheses of 15 proteins identified in this study appears not to be completely interrupted in spite of the fact that corresponding antisense RNAs were found in the existing transcriptome data. The integrated proteomic and transcriptomic analyses employed here provided a unique view of the human stem/progenitor cells.
Collapse
Affiliation(s)
- Feng Liu
- Chinese National Human Genome Center at Shanghai, Shanghai, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Richards M, Tan SP, Chan WK, Bongso A. Reverse Serial Analysis of Gene Expression (SAGE) Characterization of Orphan SAGE Tags from Human Embryonic Stem Cells Identifies the Presence of Novel Transcripts and Antisense Transcription of Key Pluripotency Genes. Stem Cells 2006; 24:1162-73. [PMID: 16456128 DOI: 10.1634/stemcells.2005-0304] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Serial analysis of gene expression (SAGE) is a powerful technique for the analysis of gene expression. A significant portion of SAGE tags, designated as orphan tags, however, cannot be reliably assigned to known transcripts. We used an improved reverse SAGE (rSAGE) strategy to convert human embryonic stem cell (hESC)-specific orphan SAGE tags into longer 3' cDNAs. We show that the systematic analysis of these 3' cDNAs permitted the discovery of hESC-specific novel transcripts and cis-natural antisense transcripts (cis-NATs) and improved the assignment of SAGE tags that resulted from splice variants, insertion/deletion, and single-nucleotide polymorphisms. More importantly, this is the first description of cis-NATs for several key pluripotency markers in hESCs and mouse embryonic stem cells, suggesting that the formation of short interfering RNA could be an important regulatory mechanism. A systematic large-scale analysis of the remaining orphan SAGE tags in the hESC SAGE libraries by rSAGE or other 3' cDNA extension strategies should unravel additional novel transcripts and cis-NATs that are specifically expressed in hESCs. Besides contributing to the complete catalog of human transcripts, many of them should prove to be a valuable resource for the elucidation of the molecular pathways involved in the self-renewal and lineage commitment of hESCs.
Collapse
Affiliation(s)
- Mark Richards
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543
| | | | | | | |
Collapse
|
39
|
Lee S, Chen J, Zhou G, Shi RZ, Bouffard GG, Kocherginsky M, Ge X, Sun M, Jayathilaka N, Kim YC, Emmanuel N, Bohlander SK, Minden M, Kline J, Ozer O, Larson RA, LeBeau MM, Green ED, Trent J, Karrison T, Liu PP, Wang SM, Rowley JD. Gene expression profiles in acute myeloid leukemia with common translocations using SAGE. Proc Natl Acad Sci U S A 2006; 103:1030-5. [PMID: 16418266 PMCID: PMC1347995 DOI: 10.1073/pnas.0509878103] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Identification of the specific cytogenetic abnormality is one of the critical steps for classification of acute myeloblastic leukemia (AML) which influences the selection of appropriate therapy and provides information about disease prognosis. However at present, the genetic complexity of AML is only partially understood. To obtain a comprehensive, unbiased, quantitative measure, we performed serial analysis of gene expression (SAGE) on CD15(+) myeloid progenitor cells from 22 AML patients who had four of the most common translocations, namely t(8;21), t(15;17), t(9;11), and inv(16). The quantitative data provide clear evidence that the major change in all these translocation-carrying leukemias is a decrease in expression of the majority of transcripts compared with normal CD15(+) cells. From a total of 1,247,535 SAGE tags, we identified 2,604 transcripts whose expression was significantly altered in these leukemias compared with normal myeloid progenitor cells. The gene ontology of the 1,110 transcripts that matched known genes revealed that each translocation had a uniquely altered profile in various functional categories including regulation of transcription, cell cycle, protein synthesis, and apoptosis. Our global analysis of gene expression of common translocations in AML can focus attention on the function of the genes with altered expression for future biological studies as well as highlight genes/pathways for more specifically targeted therapy.
Collapse
MESH Headings
- Apoptosis
- Cell Differentiation
- Chromosomes, Human, Pair 11/genetics
- Chromosomes, Human, Pair 9/genetics
- Computational Biology
- DNA, Complementary/metabolism
- Expressed Sequence Tags
- Gene Expression Profiling/methods
- Gene Expression Regulation
- Gene Expression Regulation, Neoplastic
- Gene Library
- Humans
- Leukemia/genetics
- Leukemia, Myeloid, Acute/genetics
- Leukocytes, Mononuclear/cytology
- Lewis X Antigen/biosynthesis
- Myeloid Progenitor Cells/cytology
- Oligonucleotide Array Sequence Analysis
- RNA/chemistry
- RNA, Messenger/metabolism
- Time Factors
- Translocation, Genetic
Collapse
Affiliation(s)
- Sanggyu Lee
- Department of Medicine, University of Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Mariappan D, Winkler J, Hescheler J, Sachinidis A. Cardiovascular genomics: a current overview of in vivo and in vitro studies. STEM CELL REVIEWS 2006; 2:59-66. [PMID: 17142888 PMCID: PMC7102225 DOI: 10.1007/s12015-006-0010-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/02/2022]
Abstract
The cardiovascular system is the first system that is developed in the embryo. The cardiovascular development is a complex process involving the coordination, differentiation, and interaction of distinct cell lineages to form the heart and the diverse array of arteries, veins, and capillaries required to supply oxygen and nutrients to all tissues. Embryonic stem cells have been proposed as an interesting model system to investigate molecular and cellular mechanisms involved in mammalian development. The present review is focused on extrinsic soluble factors, intrinsic transcription factors, receptors, signal transduction pathways, and genes regulating the development of cardiovascular system in vivo and in vitro. Special emphasis has been given to cardiovascular genomics including gene expression studies on the cardiovascular system under developmental and pathophysiological conditions.
Collapse
Affiliation(s)
- Devi Mariappan
- Center of Physiology and Pathophysiology Institute of Neurophysiology, University of Cologne, Robert Koch Strasse 39, Cologne, Germany
| | - Johannes Winkler
- Center of Physiology and Pathophysiology Institute of Neurophysiology, University of Cologne, Robert Koch Strasse 39, Cologne, Germany
| | - Jürgen Hescheler
- Center of Physiology and Pathophysiology Institute of Neurophysiology, University of Cologne, Robert Koch Strasse 39, Cologne, Germany
| | - Agapios Sachinidis
- Center of Physiology and Pathophysiology Institute of Neurophysiology, University of Cologne, Robert Koch Strasse 39, Cologne, Germany
| |
Collapse
|
41
|
Ge X, Jung YC, Wu Q, Kibbe WA, Wang SM. Annotating nonspecific SAGE tags with microarray data. Genomics 2006; 87:173-80. [PMID: 16314072 DOI: 10.1016/j.ygeno.2005.08.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2005] [Revised: 08/09/2005] [Accepted: 08/27/2005] [Indexed: 11/26/2022]
Abstract
SAGE (serial analysis of gene expression) detects transcripts by extracting short tags from the transcripts. Because of the limited length, many SAGE tags are shared by transcripts from different genes. Relying on sequence information in the general gene expression database has limited power to solve this problem due to the highly heterogeneous nature of the deposited sequences. Considering that the complexity of gene expression at a single tissue level should be much simpler than that in the general expression database, we reasoned that by restricting gene expression to tissue level, the accuracy of gene annotation for the nonspecific SAGE tags should be significantly improved. To test the idea, we developed a tissue-specific SAGE annotation database based on microarray data (). This database contains microarray expression information represented as UniGene clusters for 73 normal human tissues and 18 cancer tissues and cell lines. The nonspecific SAGE tag is first matched to the database by the same tissue type used by both SAGE and microarray analysis; then the multiple UniGene clusters assigned to the nonspecific SAGE tag are searched in the database under the matched tissue type. The UniGene cluster presented solely or at higher expression levels in the database is annotated to represent the specific gene for the nonspecific SAGE tags. The accuracy of gene annotation by this database was largely confirmed by experimental data. Our study shows that microarray data provide a useful source for annotating the nonspecific SAGE tags.
Collapse
Affiliation(s)
- Xijin Ge
- Center for Functional Genomics, ENH Research Institute, Northwestern University, Chicago, IL 60611, USA
| | | | | | | | | |
Collapse
|
42
|
Jaatinen T, Hemmoranta H, Hautaniemi S, Niemi J, Nicorici D, Laine J, Yli-Harja O, Partanen J. Global gene expression profile of human cord blood-derived CD133+ cells. Stem Cells 2005; 24:631-41. [PMID: 16210406 DOI: 10.1634/stemcells.2005-0185] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Human cord blood (CB)-derived CD133+ cells carry characteristics of primitive hematopoietic cells and proffer an alternative for CD34+ cells in hematopoietic stem cell (HSC) transplantation. To characterize the CD133+ cell population on a genetic level, a global expression analysis of CD133+ cells was performed using oligonucleotide microarrays. CD133+ cells were purified from four fresh CB units by immunomagnetic selection. All four CD133+ samples showed significant similarity in their gene expression pattern, whereas they differed clearly from the CD133- control samples. In all, 690 transcripts were differentially expressed between CD133+ and CD133- cells. Of these, 393 were increased and 297 were decreased in CD133+ cells. The highest overexpression was noted in genes associated with metabolism, cellular physiological processes, cell communication, and development. A set of 257 transcripts expressed solely in the CD133+ cell population was identified. Colony-forming unit (CFU) assay was used to detect the clonal progeny of precursors present in the studied cell populations. The results demonstrate that CD133+ cells express primitive markers and possess clonogenic progenitor capacity. This study provides a gene expression profile for human CD133+ cells. It presents a set of genes that may be used to unravel the properties of the CD133+ cell population, assumed to be highly enriched in HSCs.
Collapse
Affiliation(s)
- Taina Jaatinen
- Finnish Red Cross Blood Service, Kivihaantie 7, 00310 Helsinki, Finland
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Lee S, Johnson D, Dunbar K, Dong H, Ge X, Kim YC, Wing C, Jayathilaka N, Emmanuel N, Zhou CQ, Gerber HL, Tseng CC, Wang SM. 2.45 GHz radiofrequency fields alter gene expression in cultured human cells. FEBS Lett 2005; 579:4829-36. [PMID: 16107253 DOI: 10.1016/j.febslet.2005.07.063] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2005] [Revised: 07/27/2005] [Accepted: 07/27/2005] [Indexed: 11/27/2022]
Abstract
The biological effect of radiofrequency (RF) fields remains controversial. We address this issue by examining whether RF fields can cause changes in gene expression. We used the pulsed RF fields at a frequency of 2.45 GHz that is commonly used in telecommunication to expose cultured human HL-60 cells. We used the serial analysis of gene expression (SAGE) method to measure the RF effect on gene expression at the genome level. We observed that 221 genes altered their expression after a 2-h exposure. The number of affected genes increased to 759 after a 6-h exposure. Functional classification of the affected genes reveals that apoptosis-related genes were among the upregulated ones and the cell cycle genes among the downregulated ones. We observed no significant increase in the expression of heat shock genes. These results indicate that the RF fields at 2.45 GHz can alter gene expression in cultured human cells through non-thermal mechanism.
Collapse
Affiliation(s)
- Sanggyu Lee
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Lucas T, Pratscher B, Fink D, Wolschek M, Samorapoompichit P, Schöfer C, Pehamberger H, Müller M, Sorensen P, Jansen B. The Human Orthologue of a Novel Apoptosis Response Gene Induced During Rat Myelomonocytic Stem Cell Apoptosis Maps to 20q13.12. Stem Cells Dev 2005; 14:556-63. [PMID: 16305340 DOI: 10.1089/scd.2005.14.556] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Stem cell factor (SCF) stimulation of the receptor tyrosine kinase c-kit has effects on the proliferation, differentiation, and apoptotic regulation of hematopoietic progenitor cell populations. Rat bone marrow myelomonocytic stem cells (MSC) isolated in vitro by wheat germ agglutinin culture exclusively undergo self-renewal divisions when stimulated by SCF but bipotentially differentiate in the presence of dexamethasone or 1alpha,25-dihydroxyvitamin D(3) to granulocytes and macrophages, respectively. We show here that withdrawal of SCF from MSC induces rapid apoptosis in all stages of the cell cycle accompanied by development of an ultrastructural apoptotic morphology. To investigate immediate-early gene induction during MSC apoptosis, a differential display polymerase chain reaction (DD-PCR) screen coupled with rapid amplification of cDNA ends (RACE) PCR was performed. An immediate-early apoptosis response gene was isolated from growth factor-deprived MSC that was not expressed during self-renewal or differentiation induction cultures containing SCF. The protein contains a PEST region enriched in proline, glutamic acid, serine, and threonine residues common to proteins with a high turnover and has a cytoplasmic, vesicular localization in apoptotic MSC shown by immunohistochemistry. The human orthologous gene, isolated by RACE PCR, shows 86% homology to the rat protein and high similarity with a human uncharacterized hypothalamus predicted protein (HSMNP1) localized to the long arm of chromosome 20. Because deletions in this region are a common occurrence in a wide range of myeloproliferative disorders characterized by treatment resistance to apoptosis, HSMNP1 expression may play a role in normal and pathological myeloid development.
Collapse
Affiliation(s)
- Trevor Lucas
- Department of Clinical Pharmacology, Section of Experimental Oncology/Molecular Pharmacology, Medical University of Vienna, Vienna, Austria.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Deng YJ, Huang ZX, Zhou CJ, Wang JW, You Y, Song ZQ, Xiang MM, Zhong BY, Hao F. Gene profiling involved in immature CD4+ T lymphocyte responsible for systemic lupus erythematosus. Mol Immunol 2005; 43:1497-507. [PMID: 16143398 DOI: 10.1016/j.molimm.2005.07.039] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2005] [Indexed: 01/25/2023]
Abstract
We attempted to characterize the genes expression of CD4+ T lymphocytes for the pathogenesis of systemic lupus erythematosus (SLE). Genomewide gene expression profiles of CD4+ T cells, which were isolated from the disease severe activity (T4-1s) and nonactivity (T4-2s) with an SLE patient by using long serial analysis of gene expression (LongSAGE). We picked out 289 genes matching to Unigene cluster with different expression more than four copies between T4-1s and T4-2s libraries and analyzed their roles from the collectedly published articles of PubMed by genes functional clustering. The genes functions were related to a diverse cellular process including: (1) most of these genes were associated with CD4+ T cells functions, particularly related to cellular developments; (2) Ras pathway genes as RANBP10, GMIP, RASGRP2 and ARL5 might be responsible for the abnormal development of CD4+ T cells of SLE; (3) HIG2, TCF7, KHSRP, WWP1, SMAD3, TLK2, AES, CCNI and PIM2 belong to Wnt/beta-catenin way, they could play roles in modulating proliferation and differentiation of T lymphocytes; (4) uncertain viral infections may initiate autoimmunity because high levels expression genes were detected in T4-1s such as TRIM22, IER2, ABCE1, DUT, G1P2, G1P3, HNRPUL1, EVER2, IFNAR1, TNFSF14, TMP21 and PVRL2; and (5) apoptosis relating genes as EIF3S8, SH3BGRL3, GPX4, TOSO, PFDN5, BIN1, XIAPAF1, TEGT and CUGBP2 may contribute to over uploading of selfantigens in SLE cells. Abnormalities findings of multiple genes expression involving with a variety of CD4+ T cells process might be meaningful to understanding the pathogenesis of SLE, and immature CD4+ T cells may be responsible for SLE.
Collapse
Affiliation(s)
- Y J Deng
- Department of Dermatology, Southwest Hospital, The Third Military Medical University, ChongQing 400038, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Komor M, Güller S, Baldus CD, de Vos S, Hoelzer D, Ottmann OG, Hofmann WK. Transcriptional profiling of human hematopoiesis during in vitro lineage-specific differentiation. Stem Cells 2005; 23:1154-69. [PMID: 15955831 DOI: 10.1634/stemcells.2004-0171] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
To better understand the transcriptional program that a ccompanies orderly lineage-specific hematopoietic differentiation, we performed serial oligonucleotide microarray analysis of human normal CD34+ bone marrow cells during lineage-specific differentiation. CD34+ bone marrow cells isolated from healthy individuals were selectively stimulated in vitro with the cytokines erythropoietin (EPO), thrombopoietin (TPO), granulocyte colony-stimulating factor (G-CSF), and granulocyte macrophage colony-stimulating factor (GM-CSF). Cells from each of the lineages were harvested after 4, 7, and 11 days of culture for expression profiling. Gene expression was analyzed by oligonucleotide microarrays (HG-U133A; Affymetrix, Santa Clara, CA). Experiments were done in triplicates. We identified 258 genes that are consistently upregulated or downregulated during the course of lineage-specific differentiation within each specific lineage (horizontal change). In addition, we identified 52 genes that contributed to a specific expression profile, yielding a genetic signature specific for successive stages of differentiation within each of the three lineages. Analysis of horizontal changes selected 21 continuously upregulated genes for EPO-induced differentiation (including GTPase activator proteins RAP1GA1 and ARHGAP8, which regulate small Rho GTPases), 21 for G-CSF-induced/GM-CSF-induced differentiation, and 91 for TPO-induced differentiation (including DLK1, of which the role in normal hematopoiesis is not defined). During the lineage-specific differentiation, 58 (erythropoiesis), 30 (granulopoiesis), and 37 (thrombopoiesis) genes were significantly downregulated, respectively. The expression of selected genes was confirmed by real-time polymerase chain reaction. Our data encompass the first extensive transcriptional profile of human hematopoiesis during in vitro lineage-specific differentiation.
Collapse
Affiliation(s)
- Martina Komor
- Department of Hematology, Oncology and Transfusion Medicine, University Hospital Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
47
|
Lee S, Bao J, Zhou G, Shapiro J, Xu J, Shi RZ, Lu X, Clark T, Johnson D, Kim YC, Wing C, Tseng C, Sun M, Lin W, Wang J, Yang H, Wang J, Du W, Wu CI, Zhang X, Wang SM. Detecting novel low-abundant transcripts in Drosophila. RNA (NEW YORK, N.Y.) 2005; 11:939-46. [PMID: 15923377 PMCID: PMC1370778 DOI: 10.1261/rna.7239605] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Increasing evidence suggests that low-abundant transcripts may play fundamental roles in biological processes. In an attempt to estimate the prevalence of low-abundant transcripts in eukaryotic genomes, we performed a transcriptome analysis in Drosophila using the SAGE technique. We collected 244,313 SAGE tags from transcripts expressed in Drosophila embryonic, larval, pupae, adult, and testicular tissue. From these SAGE tags, we identified 40,823 unique SAGE tags. Our analysis showed that 55% of the 40,823 unique SAGE tags are novel without matches in currently known Drosophila transcripts, and most of the novel SAGE tags have low copy numbers. Further analysis indicated that these novel SAGE tags represent novel low-abundant transcripts expressed from loci outside of currently annotated exons including the intergenic and intronic regions, and antisense of the currently annotated exons in the Drosophila genome. Our study reveals the presence of a significant number of novel low-abundant transcripts in Drosophila, and highlights the need to isolate these novel low-abundant transcripts for further biological studies.
Collapse
Affiliation(s)
- Sanggyu Lee
- Department of Medicine, University of Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Panepucci RA, Siufi JLC, Silva WA, Proto-Siquiera R, Neder L, Orellana M, Rocha V, Covas DT, Zago MA. Comparison of gene expression of umbilical cord vein and bone marrow-derived mesenchymal stem cells. Stem Cells 2005; 22:1263-78. [PMID: 15579645 DOI: 10.1634/stemcells.2004-0024] [Citation(s) in RCA: 202] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) give origin to the marrow stromal environment that supports hematopoiesis. These cells present a wide range of differentiation potentials and a complex relationship with hematopoietic stem cells (HSCs) and endothelial cells. In addition to bone marrow (BM), MSCs can be obtained from other sites in the adult or the fetus. We isolate MSCs from the umbilical cord (UC) veins that are morphologically and immunophenotpically similar to MSCs obtained from the BM. In culture, these cells are capable of differentiating in vitro into adipocytes, osteoblasts, and condrocytes. The gene expression profiles of BM-MSCs and of UC-MSCs were compared by serial analysis of gene expression, then validated by reverse transcription polymerase chain reaction of selected genes. The two lineages shared almost all of the first thousand most expressed transcripts, including vimentin, galectin 1, osteonectin, collagens, transgelins, annexin A2, and MMP2. Nevertheless, a set of genes related to antimicrobial activity and to osteogenesis was more expressed in BM-MSCs, whereas higher expression in UC-MSCs was observed for genes that participate in pathways related to matrix remodeling via metalloproteinases and angiogenesis. Finally, cultured endothelial cells, CD34+ HSCs, MSCs, blood leukocytes, and bulk BM clustered together, separated from seven other normal nonhematopoietic tissues, on the basis of shared expressed genes. MSCs isolated from UC veins are functionally similar to BM-MSCs, but differentially expressed genes may reflect differences related to their sites of origin: BM-MSCs would be more committed to osteogenesis, whereas UC-MSCs would be more committed to angiogenesis.
Collapse
Affiliation(s)
- Rodrigo A Panepucci
- Center for Cell Therapy and Regional Blood Center, Department of Clinical Medicine, Faculty of Medicine, Ribeirão Preto, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Cai J, Ash D, Kotch LE, Jabs EW, Attie-Bitach T, Auge J, Mattei G, Etchevers H, Vekemans M, Korshunova Y, Tidwell R, Messina DN, Winston JB, Lovett M. Gene expression in pharyngeal arch 1 during human embryonic development. Hum Mol Genet 2005; 14:903-12. [PMID: 15703188 DOI: 10.1093/hmg/ddi083] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Craniofacial abnormalities are one of the most common birth defects in humans, but little is known about the human genes that control these important developmental processes. To identify relevant genes, we analyzed transcription profiles of human pharyngeal arch 1 (PA1), a conserved embryonic structure that develops into the palate and jaw. Using microdissected, normal human craniofacial structures, we constructed 12 SAGE (serial analysis of gene expression) libraries and sequenced 606 532 tags. We also performed Affymetrix microarray analysis on 25 craniofacial targets. Our data revealed not only genes "enriched" or differentially expressed in PA1 during fourth and fifth week of human development, but also 6927 genes newly identified to be expressed in human PA1. Many of these genes are involved in biosynthetic processes and have binding function and catalytic activity. We compared expression profiles of human genes with those of mouse homologs to look for genes more specific to human craniofacial development and found 766 genes expressed in human PA1, but not in mouse PA1. We also identified 1408 genes that were expressed in mouse as well as human PA1 and could be useful in creating mouse models for human conditions. We confirmed conservation of some human PA1 expression patterns in mouse embryonic samples with whole mount in situ hybridization and real-time RT-PCR. This comprehensive approach to expression profiling gives insights into the early development of the craniofacial region and provides markers for developmental structures and candidate genes, including SET and CCT3, for diseases such as orofacial clefting and micrognathia.
Collapse
Affiliation(s)
- Juanliang Cai
- Institute of Genetic Medicine, Johns Hopkins University, 733 N. Broadway, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Damiola F, Keime C, Gonin-Giraud S, Dazy S, Gandrillon O. Global transcription analysis of immature avian erythrocytic progenitors: from self-renewal to differentiation. Oncogene 2004; 23:7628-43. [PMID: 15378009 DOI: 10.1038/sj.onc.1208061] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The molecular mechanisms regulating the cell fate decision between self-renewal and differentiation/apoptosis in stem and progenitor cells are poorly understood. Here, we report the first comprehensive identification of genes potentially involved in the switch from self-renewal toward differentiation of primary, non-immortalized erythroid avian progenitor cells (T2EC cells). We used the Serial Analysis of Gene Expression (SAGE) technique in order to identify and quantify the genome fraction functionally active in a self-renewing versus a differentiating cell population. We generated two SAGE libraries and sequenced a total of 37,589 tags, thereby obtaining the first transcriptional profile characterization of a chicken cell. Tag identification was performed using a new relational database (Identitag) developed in the laboratory, which allowed a highly satisfactory level of identification. Among 123 differentially expressed genes, 11 were investigated further and for nine of them the differential expression was subsequently confirmed by real-time PCR. The comparison of tag abundance between the two libraries revealed that only a small fraction of transcripts was differentially expressed. The analysis of their functions argue against a prominent role for a master switch in T2EC cells decision-making, but are in favor of a critical role for coordinated small variations in a relatively small number of genes that can lead to essential cellular identity changes.
Collapse
Affiliation(s)
- Francesca Damiola
- Equipe 'Signalisations et identités cellulaires', Centre de Génétique Moléculaire et Cellulaire CNRS UMR 5534, France.
| | | | | | | | | |
Collapse
|