1
|
Davila KMS, Nelli RK, Mora-Díaz JC, Sang Y, Miller LC, Giménez-Lirola LG. Transcriptome Analysis in Air-Liquid Interface Porcine Respiratory Epithelial Cell Cultures Reveals That the Betacoronavirus Porcine Encephalomyelitis Hemagglutinating Virus Induces a Robust Interferon Response to Infection. Viruses 2024; 16:939. [PMID: 38932231 PMCID: PMC11209522 DOI: 10.3390/v16060939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Porcine hemagglutinating encephalomyelitis virus (PHEV) replicates in the upper respiratory tract and tonsils of pigs. Using an air-liquid interface porcine respiratory epithelial cells (ALI-PRECs) culture system, we demonstrated that PHEV disrupts respiratory epithelia homeostasis by impairing ciliary function and inducing antiviral, pro-inflammatory cytokine, and chemokine responses. This study explores the mechanisms driving early innate immune responses during PHEV infection through host transcriptome analysis. Total RNA was collected from ALI-PRECs at 24, 36, and 48 h post inoculation (hpi). RNA-seq analysis was performed using an Illumina Hiseq 600 to generate 100 bp paired-end reads. Differential gene expression was analyzed using DeSeq2. PHEV replicated actively in ALI-PRECs, causing cytopathic changes and progressive mucociliary disruption. Transcriptome analysis revealed downregulation of cilia-associated genes such as CILK1, DNAH11, LRRC-23, -49, and -51, and acidic sialomucin CD164L2. PHEV also activated antiviral signaling pathways, significantly increasing the expression of interferon-stimulated genes (RSAD2, MX1, IFIT, and ISG15) and chemokine genes (CCL5 and CXCL10), highlighting inflammatory regulation. This study contributes to elucidating the molecular mechanisms of the innate immune response to PHEV infection of the airway epithelium, emphasizing the critical roles of the mucociliary, interferon, and chemokine responses.
Collapse
Affiliation(s)
- Kaitlyn M. Sarlo Davila
- Infectious Bacterial Disease Research Unit, National Animal Disease Center, United States Department of Agriculture, Agricultural Research Service, Ames, IA 50010, USA;
| | - Rahul K. Nelli
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (R.K.N.); (J.C.M.-D.)
| | - Juan C. Mora-Díaz
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (R.K.N.); (J.C.M.-D.)
| | - Yongming Sang
- Department of Agricultural and Environmental Sciences, College of Agriculture, Tennessee State University, Nashville, TN 37209, USA;
| | - Laura C. Miller
- Virus and Prion Research Unit, National Animal Disease Center, United States Department of Agriculture, Agricultural Research Service, Ames, IA 50010, USA
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Luis G. Giménez-Lirola
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (R.K.N.); (J.C.M.-D.)
| |
Collapse
|
2
|
Genome-Wide Knockout Screen Identifies Human Sialomucin CD164 as an Essential Entry Factor for Lymphocytic Choriomeningitis Virus. mBio 2022; 13:e0020522. [PMID: 35502904 PMCID: PMC9239079 DOI: 10.1128/mbio.00205-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Lymphocytic choriomeningitis virus (LCMV) is a well-studied mammarenavirus that can be fatal in congenital infections. However, our understanding of LCMV and its interactions with human host factors remains incomplete. Here, host determinants affecting LCMV infection were investigated through a genome-wide CRISPR knockout screen in A549 cells, a human lung adenocarcinoma line. We identified and validated a variety of novel host factors that play a functional role in LCMV infection. Among these, knockout of the sialomucin CD164, a heavily glycosylated transmembrane protein, was found to ablate infection with multiple LCMV strains but not other hemorrhagic mammarenaviruses in several cell types. Further characterization revealed a dependency of LCMV entry on the cysteine-rich domain of CD164, including an N-linked glycosylation site at residue 104 in that region. Given the documented role of LCMV with respect to transplacental human infections, CD164 expression was investigated in human placental tissue and placental cell lines. CD164 was found to be highly expressed in the cytotrophoblast cells, an initial contact site for pathogens within the placenta, and LCMV infection in placental cells was effectively blocked using a monoclonal antibody specific to the cysteine-rich domain of CD164. Together, this study identifies novel factors associated with LCMV infection of human tissues and highlights the importance of CD164, a sialomucin that previously had not been associated with viral infection. IMPORTANCE Lymphocytic choriomeningitis virus (LCMV) is a human-pathogenic mammarenavirus that can be fatal in congenital infections. Although frequently used in the study of persistent infections in the field of immunology, aspects of this virus's life cycle remain incomplete. For example, while viral entry has been shown to depend on a cell adhesion molecule, DAG1, genetic knockout of this gene allows for residual viral infection, implying that additional receptors can mediate cell entry. The significance of our study is the identification of host factors important for successful infection, including the sialomucin CD164, which had not been previously associated with viral infection. We demonstrated that CD164 is essential for LCMV entry into human cells and can serve as a possible therapeutic target for treatment of congenital infection.
Collapse
|
3
|
Abstract
Lymphocytic choriomeningitis virus (LCMV) is the prototypic arenavirus and has been utilized for decades as a model to understand the host immune response against viral infection. LCMV infection can lead to fatal meningitis in immunocompromised people and can lead to congenital birth defects and spontaneous abortion if acquired during pregnancy. Using a genetic screen, we uncover host factors involved in LCMV entry that were previously unknown and are candidate therapeutic targets to combat LCMV infection. This study expands our understanding of the entry pathway of LCMV, revealing that its glycoprotein switches from utilizing the known receptor α-DG and heparan sulfate at the plasma membrane to binding the lysosomal mucin CD164 at pH levels found in endolysosomal compartments, facilitating membrane fusion. Lymphocytic choriomeningitis virus (LCMV) is a rodent-borne zoonotic arenavirus that causes congenital abnormalities and can be fatal for transplant recipients. Using a genome-wide loss-of-function screen, we identify host factors required for LCMV entry into cells. We identify the lysosomal mucin CD164, glycosylation factors, the heparan sulfate biosynthesis machinery, and the known receptor alpha-dystroglycan (α-DG). Biochemical analysis revealed that the LCMV glycoprotein binds CD164 at acidic pH and requires a sialylated glycan at residue N104. We demonstrate that LCMV entry proceeds by the virus switching binding from heparan sulfate or α-DG at the plasma membrane to CD164 prior to membrane fusion, thus identifying additional potential targets for therapeutic intervention.
Collapse
|
4
|
Tapia AL, Rowland BT, Rosen JD, Preuss M, Young K, Graff M, Choquet H, Couper DJ, Buyske S, Bien SA, Jorgenson E, Kooperberg C, Loos RJF, Morrison AC, North KE, Yu B, Reiner AP, Li Y, Raffield LM. A large-scale transcriptome-wide association study (TWAS) of 10 blood cell phenotypes reveals complexities of TWAS fine-mapping. Genet Epidemiol 2022; 46:3-16. [PMID: 34779012 PMCID: PMC8887641 DOI: 10.1002/gepi.22436] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/26/2021] [Accepted: 10/18/2021] [Indexed: 02/03/2023]
Abstract
Hematological measures are important intermediate clinical phenotypes for many acute and chronic diseases and are highly heritable. Although genome-wide association studies (GWAS) have identified thousands of loci containing trait-associated variants, the causal genes underlying these associations are often uncertain. To better understand the underlying genetic regulatory mechanisms, we performed a transcriptome-wide association study (TWAS) to systematically investigate the association between genetically predicted gene expression and hematological measures in 54,542 Europeans from the Genetic Epidemiology Research on Aging cohort. We found 239 significant gene-trait associations with hematological measures; we replicated 71 associations at p < 0.05 in a TWAS meta-analysis consisting of up to 35,900 Europeans from the Women's Health Initiative, Atherosclerosis Risk in Communities Study, and BioMe Biobank. Additionally, we attempted to refine this list of candidate genes by performing conditional analyses, adjusting for individual variants previously associated with hematological measures, and performed further fine-mapping of TWAS loci. To facilitate interpretation of our findings, we designed an R Shiny application to interactively visualize our TWAS results by integrating them with additional genetic data sources (GWAS, TWAS from multiple reference panels, conditional analyses, known GWAS variants, etc.). Our results and application highlight frequently overlooked TWAS challenges and illustrate the complexity of TWAS fine-mapping.
Collapse
Affiliation(s)
- Amanda L. Tapia
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Bryce T. Rowland
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jonathan D. Rosen
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Michael Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kris Young
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Misa Graff
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Hélène Choquet
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - David J. Couper
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Steve Buyske
- Department of Statistics, Rutgers University, Piscataway, New Jersey, USA
| | - Stephanie A. Bien
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Eric Jorgenson
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alanna C. Morrison
- Department of Epidemiology, Human Genetics, and Environmental Sciences, Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Kari E. North
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Alexander P. Reiner
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Laura M. Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
5
|
The stem cell revolution: on the role of CD164 as a human stem cell marker. NPJ Regen Med 2021; 6:33. [PMID: 34103536 PMCID: PMC8187384 DOI: 10.1038/s41536-021-00143-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 05/14/2021] [Indexed: 02/05/2023] Open
Abstract
Accurately defining hierarchical relationships between human stem cells and their progeny, and using this knowledge for new cellular therapies, will undoubtedly lead to further successful treatments for life threatening and chronic diseases, which represent substantial burdens on patient quality of life and to healthcare systems globally. Clinical translation relies in part on appropriate biomarker, in vitro manipulation and transplantation strategies. CD164 has recently been cited as an important biomarker for enriching both human haematopoietic and skeletal stem cells, yet a thorough description of extant human CD164 monoclonal antibody (Mab) characteristics, which are critical for identifying and purifying these stem cells, was not discussed in these articles. Here, we highlight earlier but crucial research describing these relevant characteristics, including the differing human CD164 Mab avidities and their binding sites on the human CD164 sialomucin, which importantly may affect subsequent stem cell function and fate.
Collapse
|
6
|
A comprehensive single cell transcriptional landscape of human hematopoietic progenitors. Nat Commun 2019; 10:2395. [PMID: 31160568 PMCID: PMC6546699 DOI: 10.1038/s41467-019-10291-0] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 05/03/2019] [Indexed: 01/06/2023] Open
Abstract
Hematopoietic Stem/Progenitor cells (HSPCs) are endowed with the role of maintaining a diverse pool of blood cells throughout the human life. Despite recent efforts, the nature of the early cell fate decisions remains contentious. Using single-cell RNA-Seq, we show that existing approaches to stratify bone marrow CD34+ cells reveal a hierarchically-structured transcriptional landscape of hematopoietic differentiation. Still, this landscape misses important early fate decisions. We here provide a broader transcriptional profiling of bone marrow lineage negative hematopoietic progenitors that recovers a key missing branchpoint into basophils and expands our understanding of the underlying structure of early adult human haematopoiesis. We also show that this map has strong similarities in topology and gene expression to that found in mouse. Finally, we identify the sialomucin CD164, as a reliable marker for the earliest branches of HSPCs specification and we showed how its use can foster the design of alternative transplantation cell products. Human Hematopoietic stem and progenitor cells (HSPCs) are commonly defined by CD34 expression. Here, the authors map single-cell RNA states both inside and outside the CD34 compartment, uncovering previously unappreciated branchpoints and validating CD164 as an efficient marker for early HSPCs.
Collapse
|
7
|
Zhang XG, Zhang T, Li CY, Zhang MH, Chen FM. CD164 promotes tumor progression and predicts the poor prognosis of bladder cancer. Cancer Med 2018; 7:3763-3772. [PMID: 30022623 PMCID: PMC6089154 DOI: 10.1002/cam4.1607] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 05/13/2018] [Accepted: 05/15/2018] [Indexed: 12/14/2022] Open
Abstract
CD164 was found to play a role in many malignant diseases. But the roles of CD164 in human bladder cancer have not yet been studied. The object of our study was to investigate the functions of CD164 in urothelial bladder carcinoma. The immunohistochemistry (IHC) was performed to evaluate the associations between the expression level of CD164 and clinical‐pathological features of patients, and IHC was used to analyze the relationship between CD164 and CXCR4 in tumor tissues. Real‐time qPCR and Western blot were used to measure the expression of relevant genes. The roles of CD164 in tumor cells and tissues were investigated by in vitro and in vivo experiments. The results of immunohistochemistry found that CD164 was associated with clinical and pathological features of patients. High level of CD164 was related to the distant metastasis and vascular invasion of bladder cancer patients. In vitro, by silencing of CD164, the proliferation, migration, and invasion of tumor cells were inhibited significantly by regulating related proteins such as Ki67, proliferating cell nuclear antigen, matrix metalloproteinases‐2, and matrix metalloproteinases‐9. In vivo, knocking‐down of CD164 could reduce the growth and metastasis of tumors in mice. In addition, a co‐expression was found between CD164 and CXCR4 in tumor tissues. In conclusion, our study demonstrated that CD164 was associated with the poor clinical outcomes of BC patients. Silencing of CD164 could inhibit the progression of tumors in vivo and in vitro, which may become an effective target in the treatment of bladder cancer.
Collapse
Affiliation(s)
- Xiao-Guang Zhang
- Department of Urology, Third Central Hospital of Tianjin, Tianjin, China
| | - Tong Zhang
- Department of Urology, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | | | - Ming-Hao Zhang
- Department of Urology, Third Central Hospital of Tianjin, Tianjin, China
| | - Fang-Min Chen
- Department of Urology, Third Central Hospital of Tianjin, Tianjin, China
| |
Collapse
|
8
|
Chen WL, Huang AF, Huang SM, Ho CL, Chang YL, Chan JYH. CD164 promotes lung tumor-initiating cells with stem cell activity and determines tumor growth and drug resistance via Akt/mTOR signaling. Oncotarget 2016; 8:54115-54135. [PMID: 28903328 PMCID: PMC5589567 DOI: 10.18632/oncotarget.11132] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 07/28/2016] [Indexed: 01/12/2023] Open
Abstract
CD164 is a cell adhesion molecule that increases hematopoietic stem cell proliferation, adhesion, and migration via C-X-C chemokine receptor type 4 (CXCR4) signaling. Emerging evidence indicates that elevated CD164 expression is associated with aggressive metastasis, advanced stages, and shorter overall survival in lung cancer. However, no data are available regarding the clinical significance of CD164 expression in lung cancer. This study explores whether CD164 promotes tumor-initiation and drug resistance through the stem cell property. Using tissue microarrays, we determine that CD164 expression is correlated with clinicopathological characteristics in human lung cancer. The CD164 overexpression in normal lung epithelial cells (BEAS2B cells) leads to malignant transformation in vitro, tumorigenicity in xenografted mice, stem cell-like property, and drug resistance through ATP-binding cassette transporters. The CD164 overexpression increases CXCR4 expression and activates Akt/mTOR signaling. Rapamycin, an mTOR inhibitor, hinders cell proliferation along with sphere formation in vitro and impedes tumor growth in vivo. In conclusion, we have provided evidence that CD164 promotes the growth of lung tumor-initiating cells with stem cell properties and induces tumor growth and drug resistance through Akt/mTOR signaling. Therefore, identification of CD164 as a cancer stem cell therapeutic marker may develop an effective therapy in patients with chemoresistant lung cancer.
Collapse
Affiliation(s)
- Wei-Liang Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan, Republic of China.,Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei 114, Taiwan, Republic of China.,Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Ai-Fang Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Shih-Ming Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan, Republic of China.,Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Ching-Liang Ho
- Division of Hematology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Yung-Lung Chang
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - James Yi-Hsin Chan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan, Republic of China.,Department of Microbiology and Immunology, National Defense Medical Center, Taipei 114, Taiwan, Republic of China.,Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| |
Collapse
|
9
|
Klamer S, Voermans C. The role of novel and known extracellular matrix and adhesion molecules in the homeostatic and regenerative bone marrow microenvironment. Cell Adh Migr 2015; 8:563-77. [PMID: 25482635 PMCID: PMC4594522 DOI: 10.4161/19336918.2014.968501] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Maintenance of haematopoietic stem cells and differentiation of committed progenitors occurs in highly specialized niches. The interactions of haematopoietic stem and progenitor cells (HSPCs) with cells, growth factors and extracellular matrix (ECM) components of the bone marrow (BM) microenvironment control homeostasis of HSPCs. We only start to understand the complexity of the haematopoietic niche(s) that comprises endosteal, arterial, sinusoidal, mesenchymal and neuronal components. These distinct niches produce a broad range of soluble factors and adhesion molecules that modulate HSPC fate during normal hematopoiesis and BM regeneration. Adhesive interactions between HSPCs and the microenvironment will influence their localization and differentiation potential. In this review we highlight the current understanding of the functional role of ECM- and adhesion (regulating) molecules in the haematopoietic niche during homeostatic and regenerative hematopoiesis. This knowledge may lead to the improvement of current cellular therapies and more efficient development of future cellular products.
Collapse
Affiliation(s)
- Sofieke Klamer
- a Department of Hematopoiesis; Sanquin Research; Landsteiner Laboratory; Academic Medical Centre ; University of Amsterdam ; Amsterdam , The Netherlands
| | | |
Collapse
|
10
|
van der Pouw Kraan TCTM, Chen WJ, Bunck MCM, van Raalte DH, van der Zijl NJ, van Genugten RE, van Bloemendaal L, Baggen JM, Serné EH, Diamant M, Horrevoets AJG. Metabolic changes in type 2 diabetes are reflected in peripheral blood cells, revealing aberrant cytotoxicity, a viral signature, and hypoxia inducible factor activity. BMC Med Genomics 2015; 8:20. [PMID: 25956355 PMCID: PMC4446948 DOI: 10.1186/s12920-015-0096-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 04/30/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Metabolic syndrome (MetS) is characterized by central obesity, insulin resistance, dysglycemia, and a pro-atherogenic plasma lipid profile. MetS creates a high risk for development of type 2 diabetes (T2DM) and cardiovascular disease (CVD), presumably by altering inflammatory responses. Presently, it is unknown how the chronic metabolic disturbances in acute hyperglycemia, MetS and T2DM affect the immune activity of peripheral blood cells. METHODS We performed genome-wide expression analysis of peripheral blood cells obtained from patients with T2DM (n = 6) and age-, sex- , BMI- and blood pressure-matched obese individuals with MetS (n = 4) and lean healthy normoglycemic controls (n = 3), both under fasting conditions and after controlled induction of acute hyperglycemia during a 70 min hyperglycemic clamp. Differential gene expression during fasting conditions was confirmed by real-time PCR, for which we included additional age-, sex-, BMI-, and blood pressure-matched obese individuals with (n = 4) or without (n = 4) MetS. RESULTS Pathway and Gene ontology analysis applied to baseline expression profiles of peripheral blood cells from MetS and T2DM patients revealed metabolic changes, highly similar to a reoviral infection gene signature in T2DM patients. Transcription factor binding site analysis indicated that increased HIF-1α activity, a transcription factor induced by either hypoxia or oxidative stress, is responsible for this aberrant metabolic profile in peripheral blood cells from T2DM patients. Acute hyperglycemia in healthy controls resulted in reduced expression of cytotoxicity-related genes, representing NK- and CD8(+) cells. In obese controls, MetS and especially T2DM patients, baseline expression of genes involved in cytotoxicity was already low, compared to healthy controls and did not further decrease upon acute hyperglycemia. CONCLUSIONS The reduced activity of cytotoxic genes in T2DM is explained by chronic hyperglycemia, but its acute effects are restricted to healthy controls. Genome expression of circulating leukocytes from T2DM patients differs from MetS individuals by a specific reovirus signature. Our data thus suggest a role for suppressed anti-viral capacity in the etiology of diabetes.
Collapse
Affiliation(s)
| | - Weena J Chen
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands.
| | - Mathijs C M Bunck
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands.
| | - Daniel H van Raalte
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands.
| | - Nynke J van der Zijl
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands.
| | - Renate E van Genugten
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands.
| | - Liselotte van Bloemendaal
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands.
| | - Josefien M Baggen
- Department of Molecular Cell Biology & Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Erik H Serné
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands.
| | - Michaela Diamant
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Anton J G Horrevoets
- Department of Molecular Cell Biology & Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
CD164 and FCRL3 are highly expressed on CD4+CD26- T cells in Sézary syndrome patients. J Invest Dermatol 2013; 134:229-236. [PMID: 23792457 PMCID: PMC3869886 DOI: 10.1038/jid.2013.279] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 05/03/2013] [Accepted: 05/20/2013] [Indexed: 12/22/2022]
Abstract
Sézary syndrome (SS) cells express cell surface molecules also found on normal activated CD4 T cells. In an effort to find a more specific surface marker for malignant SS cells, a microarray analysis of gene expression was performed. Results showed significantly increased levels of mRNA for CD164, a sialomucin found on human CD34+ hematopoietic stem cells, and FCRL3, a molecule present on a subset of human natural T regulatory cells. Both markers were increased in CD4 T cells from SS patients compared to healthy donors. Flow cytometry studies confirmed the increased expression of CD164 and FCRL3 primarily on CD4+CD26− T cells of SS patients. Importantly, a statistically significant correlation was found between an elevated percentage of CD4+CD164+ T cells and an elevated percentage of CD4+CD26− T cells in all tested SS patients but not in patients with Mycosis Fungoides and atopic dermatitis or healthy donors. FCRL3 expression was significantly increased only in high tumor burden patients. CD4+CD164+ cells displayed cerebriform morphology and their loss correlated with clinical improvement in treated patients. Our results suggest that CD164 can serve as a marker for diagnosis and for monitoring progression of CTCL/SS and that FCRL3 expression correlates with a high circulating tumor burden.
Collapse
|
12
|
Chen Z, Tang H, Qayyum R, Schick UM, Nalls MA, Handsaker R, Li J, Lu Y, Yanek LR, Keating B, Meng Y, van Rooij FJ, Okada Y, Kubo M, Rasmussen-Torvik L, Keller MF, Lange L, Evans M, Bottinger EP, Linderman MD, Ruderfer DM, Hakonarson H, Papanicolaou G, Zonderman AB, Gottesman O, Thomson C, Ziv E, Singleton AB, Loos RJ, Sleiman PM, Ganesh S, McCarroll S, Becker DM, Wilson JG, Lettre G, Reiner AP. Genome-wide association analysis of red blood cell traits in African Americans: the COGENT Network. Hum Mol Genet 2013; 22:2529-38. [PMID: 23446634 PMCID: PMC3658166 DOI: 10.1093/hmg/ddt087] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 02/09/2013] [Accepted: 02/18/2013] [Indexed: 01/02/2023] Open
Abstract
Laboratory red blood cell (RBC) measurements are clinically important, heritable and differ among ethnic groups. To identify genetic variants that contribute to RBC phenotypes in African Americans (AAs), we conducted a genome-wide association study in up to ~16 500 AAs. The alpha-globin locus on chromosome 16pter [lead SNP rs13335629 in ITFG3 gene; P < 1E-13 for hemoglobin (Hgb), RBC count, mean corpuscular volume (MCV), MCH and MCHC] and the G6PD locus on Xq28 [lead SNP rs1050828; P < 1E - 13 for Hgb, hematocrit (Hct), MCV, RBC count and red cell distribution width (RDW)] were each associated with multiple RBC traits. At the alpha-globin region, both the common African 3.7 kb deletion and common single nucleotide polymorphisms (SNPs) appear to contribute independently to RBC phenotypes among AAs. In the 2p21 region, we identified a novel variant of PRKCE distinctly associated with Hct in AAs. In a genome-wide admixture mapping scan, local European ancestry at the 6p22 region containing HFE and LRRC16A was associated with higher Hgb. LRRC16A has been previously associated with the platelet count and mean platelet volume in AAs, but not with Hgb. Finally, we extended to AAs the findings of association of erythrocyte traits with several loci previously reported in Europeans and/or Asians, including CD164 and HBS1L-MYB. In summary, this large-scale genome-wide analysis in AAs has extended the importance of several RBC-associated genetic loci to AAs and identified allelic heterogeneity and pleiotropy at several previously known genetic loci associated with blood cell traits in AAs.
Collapse
Affiliation(s)
- Zhao Chen
- Division of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health and
| | - Hua Tang
- Department of Statistics and Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Rehan Qayyum
- GeneSTAR Research Program, Division of General Internal Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Ursula M. Schick
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98195, USA
| | - Michael A. Nalls
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - Robert Handsaker
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02141, USA
| | - Jin Li
- Center for Applied Genomics, Abramson Research Center and
| | - Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine, The Genetics of Obesity and Related Metabolic Traits Program
| | - Lisa R. Yanek
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Yan Meng
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Frank J.A. van Rooij
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Yukinori Okada
- Division of Rheumatology, Immunology, and Allergy and
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Medical and Population Genetics Program, Broad Institute, Cambridge, MA 02142, USA
| | - Michiaki Kubo
- Laboratory for Genotyping Development, CGM, RIKEN, Yokohama, Japan
| | - Laura Rasmussen-Torvik
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Leslie Lange
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Michele Evans
- Health Disparities Research Section, Clinical Research Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21225, USA
| | | | - Michael D. Linderman
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology
| | | | - Hakon Hakonarson
- Center for Applied Genomics, Abramson Research Center and
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - George Papanicolaou
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (NHLBI), Bethesda, MD, USA
| | - Alan B. Zonderman
- Laboratory of Personality and Cognition, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Omri Gottesman
- The Charles Bronfman Institute for Personalized Medicine
| | | | - Cynthia Thomson
- Division of Nutrition, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ 85724, USA
| | - Elad Ziv
- Department of Medicine, University of California, San Francisco, CA 94143, USA
| | | | - Ruth J.F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Institute of Child Health and Development, The Genetics of Obesity and Related Metabolic Traits Program, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Patrick M.A. Sleiman
- Center for Applied Genomics, Abramson Research Center and
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Santhi Ganesh
- Division of Cardiology, University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Steven McCarroll
- Department of Genetics, Harvard Medical School, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Diane M. Becker
- GeneSTAR Research Program, Division of General Internal Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - James G. Wilson
- Department of Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | | | - Alexander P. Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98195, USA
- Department of Epidemiology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
13
|
Roop-ngam P, Chaiyarit S, Pongsakul N, Thongboonkerd V. Isolation and characterizations of oxalate-binding proteins in the kidney. Biochem Biophys Res Commun 2012; 424:629-34. [DOI: 10.1016/j.bbrc.2012.07.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 07/06/2012] [Indexed: 01/06/2023]
|
14
|
Tang J, Zhang L, She X, Zhou G, Yu F, Xiang J, Li G. Inhibiting CD164 expression in colon cancer cell line HCT116 leads to reduced cancer cell proliferation, mobility, and metastasis in vitro and in vivo. Cancer Invest 2012; 30:380-9. [PMID: 22409183 DOI: 10.3109/07357907.2012.666692] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND CD164 (Endolyn) is a sialomucin, which has been found to play roles in regulating proliferation, adhesion, and differentiation of hematopoietic stem cells. Possible association of CD164 with solid cancer development remains unknown. METHODS AND RESULTS We first studied CD164 expression in biopsies from colorectal cancer, breast, and ovary cancer patients by semi-quantitative immunohistochemistry, and found that CD164 was strongly expressed in all the colorectal cancer samples compared to the matching normal colon tissues. The possible roles of CD164 in colon cancer development were further investigated using a well-established human colon cancer cell line HCT116. We found that knockdown of CD164 expression in HCT116 cells significantly inhibited cell proliferation, mobility, and metastasis in vitro and in vivo. The knockdown of CD164 expression was associated with decreased chemokine receptor CXCR4 expression HCT116 cell surface and immunoprecipitation studies showed that CD164 formed complexes with CXCR4. CONCLUSIONS CD164 is highly expressed in the colon cancer sites, and it promotes HCT116 colon cancer cell proliferation and metastasis both in vitro and in vivo, and the effects may act through regulating CXCR4 signaling pathway. Therefore, CD164 may be a new target for diagnosis and treatment for colon cancer.
Collapse
Affiliation(s)
- Jingqun Tang
- Department of Cardiothoracic Surgery, Xiangya Second Hospital, Central South University, Changsha, Hunan, P.R. China
| | | | | | | | | | | | | |
Collapse
|
15
|
Tang J, Luo Z, Zhou G, Song C, Yu F, Xiang J, Li G. Cis-regulatory functions of overlapping HIF-1alpha/E-box/AP-1-like sequences of CD164. BMC Mol Biol 2011; 12:44. [PMID: 21999799 PMCID: PMC3215284 DOI: 10.1186/1471-2199-12-44] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 10/14/2011] [Indexed: 11/10/2022] Open
Abstract
Background CD164 (also known as MGC-24v or endolyn) is a sialomucin which has been suggested to participate in regulating the proliferation, cell adhesion and differentiation of hematopoietic stem and progenitor cells. CD164 is also involved in the development of cancer. The functions of cis-regulatory elements of CD164 remain relatively unknown. Methods In this study, we investigated the function of cis-regulatory elements within the promoter of CD164. We fused the 5'-flanking region of CD164 to a luciferase reporter vector. The minimal promoter region was confirmed by luciferase reporter assay. Using in silico analysis, we found the presence of one HIF-1alpha (HIF-1A) motif (5_-RCGTG-3_) overlapping E-box (CACGTG) and two AP-1-like binding sites (CGCTGTCCC, GTCTGTTG), one of which is also overlapped with HIF-1alpha sequence. Dual-luciferase assay was performed to examine the transcriptional activity of AP-1 and HIF-1alpha of CD164 promoter. Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) was performed to measure CD164 expression. Chromatin Immunoprecipitation was used to confirm the binding of HIF-1alpha and CD164. Results Co-transfection of c-jun, HIF-1alpha and minimal promoter region construct demonstrated that c-jun and HIF-1alpha bound the CD164 promoter and promoted CD164 expression. Hypoxia treatment also led to the up-regulation of CD164 expression. The mutation of overlapping sequences resulted in the reduced expression of CD164 induced by HIF-1alpha. Chromatin Immunoprecipitation demonstrated that the HIF-1alpha bound the minimal promoter region. Conclusions Determination of the optimal promoter region and transcription factors governing CD164 expression is useful in understanding CD164 functions. These results suggest that cis-regulatory elements of CD164 overlapping HIF-1alpha/E-box/AP-1-like sequences may play important regulatory roles.
Collapse
Affiliation(s)
- Jingqun Tang
- Department of Cardiothoracic Surgery, Xiangya Second Hospital, Central South University, Changsha, Hunan, 410011, PR China
| | | | | | | | | | | | | |
Collapse
|
16
|
Chirumbolo S. CD164 and other recently discovered activation markers as promising tools for allergy diagnosis: what’s new? Clin Exp Med 2011; 11:255-7. [DOI: 10.1007/s10238-011-0132-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 03/01/2011] [Indexed: 12/25/2022]
|
17
|
Haematopoietic repopulating activity in human cord blood CD133+ quiescent cells. Bone Marrow Transplant 2008; 43:627-35. [DOI: 10.1038/bmt.2008.368] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
18
|
Zhou GQ, Zhang Y, Ferguson DJP, Chen S, Rasmuson-Lestander A, Campbell FC, Watt SM. The Drosophila ortholog of the endolysosomal membrane protein, endolyn, regulates cell proliferation. J Cell Biochem 2007; 99:1380-96. [PMID: 16924678 DOI: 10.1002/jcb.20965] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Endolyn (CD164) is a sialomucin that regulates the proliferation, adhesion, and migration of human haematopoietic stem and progenitor cells. This molecule is predominately localized in endocytotic compartments, where it may contribute to endolysosomal biogenesis and trafficking. In order to more closely define the function of endolyn from an evolutionary view-point, we first analyzed endolyn orthologs in species ranging from insects, fish, and birds to mammals. The predicted molecular structures of the endolyn orthologs from these species are well conserved, particularly with respect to significant O-linked glycosylation of the extracellular domain, and the high degree of amino acid similarities within their transmembrane and cytoplasmic domains, with the latter possessing the lysosomal target signal, YXXphi. Focusing on Drosophila, our studies showed that the subcellular distribution of endolyn in non-polarized Drosophila S2 cells resembles that of its human counterpart in hematopoietic cells, with its predominant localization being within intracellular vesicles, while a small fraction occurs on the cell surface. Both Y --> A and L --> A mutations in the YHTL motif perturbed the normal subcellular distribution of Drosophila endolyn. Interestingly, embryonic and early larval development was often arrested in endolyn-deficient Drosophila mutants. This may partly be due to the role of endolyn in regulating cell proliferation, since knock-down of endolyn expression in S2 cells resulted in up to 50% inhibition of cell growth, with a proportion of cells undergoing apoptosis. Taken together, these results demonstrate that endolyn is an evolutionarily conserved sialomucin fundamentally involved in cell proliferation in both the human and Drosophila melanogaster.
Collapse
Affiliation(s)
- Guang-Qian Zhou
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, UK.
| | | | | | | | | | | | | |
Collapse
|
19
|
Jung Y, Wang J, Song J, Shiozawa Y, Wang J, Havens A, Wang Z, Sun YX, Emerson SG, Krebsbach PH, Taichman RS. Annexin II expressed by osteoblasts and endothelial cells regulates stem cell adhesion, homing, and engraftment following transplantation. Blood 2007; 110:82-90. [PMID: 17360942 PMCID: PMC1896130 DOI: 10.1182/blood-2006-05-021352] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Differentiation of hematopoietic stem cells (HSCs) after birth is largely restricted to the bone marrow cavity, where HSCs are associated closely with osteoblasts (OBs). How OBs localize HSCs to the endosteal niche remains unclear. To explore adhesive interactions between HSCs and OBs, a cell blot analysis was used that revealed 2 major bands that corresponded to monomers and multimers of annexin II (Anxa2). Immunohistochemistry revealed that OBs and marrow endothelial cells express Anxa2 at high levels. Function-blocking studies confirmed that Anxa2 mediates HSC adhesion mainly via the N-terminal portion of the Anxa2 peptide. Adhesion of HSCs to OBs derived from Anxa2-deficient animals (Anxa2(-/-)) was significantly impaired compared with OBs obtained from wild-type animals (Anxa2(+/+)). Moreover, fewer HSCs were found in the marrow of Anxa2(-/-) versus Anxa2(+/+) animals. Short-term lodging, engraftment, and survival of irradiated mice with whole marrow cells were substantially inhibited by N-terminal peptide fragments of Anxa2 or anti-Anxa2 antibodies. Similar findings were noted in long-term competitive repopulation studies. Collectively, these findings reveal that Anxa2 regulates HSC homing and binding to the bone marrow microenvironment and suggest that Anxa2 is crucial for determining the bone marrow niche of HSCs.
Collapse
Affiliation(s)
- Younghun Jung
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Martin-Rendon E, Hale SJM, Ryan D, Baban D, Forde SP, Roubelakis M, Sweeney D, Moukayed M, Harris AL, Davies K, Watt SM. Transcriptional profiling of human cord blood CD133+ and cultured bone marrow mesenchymal stem cells in response to hypoxia. Stem Cells 2006; 25:1003-12. [PMID: 17185612 DOI: 10.1634/stemcells.2006-0398] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Umbilical cord blood (UCB) and bone marrow (BM)-derived stem and progenitor cells possess two characteristics required for successful tissue regeneration: extensive proliferative capacity and the ability to differentiate into multiple cell lineages. Within the normal BM and in pathological conditions, areas of hypoxia may have a role in maintaining stem cell fate or determining the fine equilibrium between their proliferation and differentiation. In this study, the transcriptional profiles and proliferation and differentiation potential of UCB CD133(+) cells and BM mesenchymal cells (BMMC) exposed to normoxia and hypoxia were analyzed and compared. Both progenitor cell populations responded to hypoxic stimuli by stabilizing the hypoxia inducible factor (HIF)-1alpha protein. Short exposures to hypoxia increased the clonogenic myeloid capacity of UCB CD133(+) cells and promoted a significant increase in BMMC number. The differentiation potential of UCB CD133(+) clonogenic myeloid cells was unaltered by short exposures to hypoxia. In contrast, the chondrogenic differentiation potential of BMMCs was enhanced by hypoxia, whereas adipogenesis and osteogenesis were unaltered. When their transcriptional profiles were compared, 183 genes in UCB CD133(+) cells and 45 genes in BMMC were differentially regulated by hypoxia. These genes included known hypoxia-responsive targets such as BNIP3, PGK1, ENO2, and VEGFA, and other genes not previously described to be regulated by hypoxia. Several of these genes, namely CDTSPL, CCL20, LSP1, NEDD9, TMEM45A, EDG-1, and EPHA3 were confirmed to be regulated by hypoxia using quantitative reverse transcriptase polymerase chain reaction. These results, therefore, provide a global view of the signaling and regulatory network that controls oxygen sensing in human adult stem/progenitor cells derived from hematopoietic tissues.
Collapse
Affiliation(s)
- Enca Martin-Rendon
- Stem Cell Research Laboratory, National Blood Service, Oxford Centre, The John Radcliffe Hospital, Headington, OX3 9BQ, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Cheung JOP, Casals-Pascual C, Roberts DJ, Watt SM. A small-scale serum-free liquid cell culture model of erythropoiesis to assess the effects of exogenous factors. J Immunol Methods 2006; 319:104-17. [PMID: 17174973 DOI: 10.1016/j.jim.2006.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2006] [Revised: 10/31/2006] [Accepted: 11/08/2006] [Indexed: 10/23/2022]
Abstract
Anaemia is an important global health problem. Therefore, it is crucial to understand its pathophysiology in various genetic or infectious diseases where dyserythropoiesis is a key pathological feature. To this effect, reproducible and reliable models of erythropoiesis in vitro are much needed as investigative tools. We have developed a serum-free liquid culture model of erythropoiesis using human umbilical cord blood CD34(+) cells cultured in the cytokine combination, interleukin-3 (IL-3), IL-6, stem cell factor (SCF) and erythropoietin (Epo), over 14 days. We found that these culture conditions favored erythroid differentiation over the expansion of the more primitive erythroid precursors. With an initiating culture density of 5x10(4) cells per ml, the nucleated cell fold expansion increased from 7.9+/-3.9 (range 4.5 to 11.1) after 4 days to 2990.2+/-1936.1 (range 626.6 to 6912.0) after 14 days in culture. Day-14 burst-forming unit-erythroid (BFU-E) frequencies peaked at day 4 (24.0+/-8.9%), with a marked decrease in BFU-E burst size as the cultures progressed. Time-course immunophenotypical profiles were characteristically erythroid with a decrease in CD34 expression (from 96.8+/-3.0% at day 0 to 0.8+/-0.8% at day 14), and a concomitant increase in the expression of erythroid-specific markers, CD36, glycophorin A (GpA) and CD71 (from 14.8+/-5.0%, 1.7+/-1.0% and 37.9+/-18.0% to 93.0+/-7.0%, 82.1+/-14.0% and 95.7+/-3.0%, respectively). Morphological studies revealed the presence of normoblasts with the complete absence of reticulocytes and mature erythrocytes after 14 days in culture. Once the culture conditions were optimized, we scaled down our culture model from 24-well plate (large-scale) to 96-well plate cultures (small-scale). We found that the small-scale cultures compared favorably with their large-scale counterpart in terms of erythroid progenitor cell proliferation and differentiation, particularly at low CD34(+) initiating cell doses. By using tumor necrosis factor-alpha (TNF-alpha), a known inhibitor of erythropoiesis, we validated our model system and showed a dose-dependent inhibition of erythroid differentiation with TNF-alpha in our cultures. Therefore, our results demonstrate a small-scale serum-free liquid culture model of erythropoiesis that is comparable with and complements our well-defined large-scale model. Our system would prove useful for screening the effects of exogenous factors on erythropoiesis in vitro.
Collapse
Affiliation(s)
- Joyce O P Cheung
- Stem Cell and Immunotherapy, National Blood Service--Oxford, NHS Blood and Transplant, John Radcliffe Hospital, Oxford OX3 9BQ, UK
| | | | | | | |
Collapse
|
22
|
Havens AM, Jung Y, Sun YX, Wang J, Shah RB, Bühring HJ, Pienta KJ, Taichman RS. The role of sialomucin CD164 (MGC-24v or endolyn) in prostate cancer metastasis. BMC Cancer 2006; 6:195. [PMID: 16859559 PMCID: PMC1557671 DOI: 10.1186/1471-2407-6-195] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2006] [Accepted: 07/21/2006] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The chemokine stromal derived factor-1 (SDF-1 or CXCL12) and its receptor CXCR4 have been demonstrated to be crucial for the homing of stem cells and prostate cancers to the marrow. While screening prostate cancers for CXCL12-responsive adhesion molecules, we identified CD164 (MGC-24) as a potential regulator of homing. CD164 is known to function as a receptor that regulates stem cell localization to the bone marrow. RESULTS Using prostate cancer cell lines, it was demonstrated that CXCL12 induced both the expression of CD164 mRNA and protein. Functional studies demonstrated that blocking CD164 on prostate cancer cell lines reduced the ability of these cells to adhere to human bone marrow endothelial cells, and invade into extracellular matrices. Human tissue microarrays stained for CD164 demonstrated a positive correlation with prostate-specific antigen levels, while its expression was negatively correlated with the expression of androgen receptor. CONCLUSION Our findings suggest that CD164 may participate in the localization of prostate cancer cells to the marrow and is further evidence that tumor metastasis and hematopoietic stem cell trafficking may involve similar processes.
Collapse
Affiliation(s)
- AM Havens
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave., Ann Arbor, Michigan 48109-1078, USA
| | - Y Jung
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave., Ann Arbor, Michigan 48109-1078, USA
| | - YX Sun
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave., Ann Arbor, Michigan 48109-1078, USA
- Department of Ophthalmology, Affiliated hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - J Wang
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave., Ann Arbor, Michigan 48109-1078, USA
| | - RB Shah
- Department of Pathology, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, Michigan 48109-0692, USA
| | - HJ Bühring
- Department of Internal Medicine II, Division for Hematology, Immunology, Oncology and Rheumatology, University Hospital, Tübingen, Germany
| | - KJ Pienta
- Department of Urology, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, Michigan 48109-0692, USA
| | - RS Taichman
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave., Ann Arbor, Michigan 48109-1078, USA
| |
Collapse
|
23
|
Hennersdorf F, Florian S, Jakob A, Baumgärtner K, Sonneck K, Nordheim A, Biedermann T, Valent P, Bühring HJ. Identification of CD13, CD107a, and CD164 as novel basophil-activation markers and dissection of two response patterns in time kinetics of IgE-dependent upregulation. Cell Res 2005; 15:325-35. [PMID: 15916720 DOI: 10.1038/sj.cr.7290301] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Using two-colour flow cytometry >200 antibodies submitted to the 8th International Workshop of Human Leukocyte Differentiation Antigens (HLDA8) have been analyzed for their reactivity with resting and activated CD203c+ basophils. Four antibodies either non-reactive or weakly reactive with resting basophils exhibited an increased reactivity with basophils activated by anti-IgE-mediated cross-linking of the high affinity IgE receptor (FceRI). These include antibodies against CD164 (WS-80160, clone N6B6 and WS-80162, clone 67D2), as well as two reagents with previously unknown specificities that were identified as CD13 (WS-80274, clone A8) and CD107a (WS-80280, clone E63-880). The activation patterns followed either the "CD203c-like" or "CD63-like" activation profile. The CD203c profile is characterized by a rapid and significant upregulation (of CD13, CD164, and CD203c), reaching maximum levels after 5-15 min of stimulation. The Phosphoinositide-3-kinase (PI3K)-specific inhibitor Wortmannin inhibited the upregulation of these markers whereas 12-O-tetradecanoyl-phorbol-13-acetate (TPA) induced a rapid and FceRI-independent upregulation within 1-2 min. In the CD63 profile, maximum upregulation (of CD63 and CD107a) was detected only after 20-40 min, and upregulation by TPA reached maximum levels after 60 min. In summary, our data identify CD13, CD107a, and CD164 as novel basophil-activation antigens. Based on time kinetics of upregulation, we hypothesize that molecules of the "CD203c group" and the "CD63 group" are linked to two different mechanisms of basophil activation.
Collapse
Affiliation(s)
- Florian Hennersdorf
- University Clinic of Tübingen, Department of Internal Medicine II, Division of Hematology and Immunology, Tübingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Jorgensen-Tye B, Levesque JP, Royle L, Doyonnas R, Chan JYH, Dwek RA, Rudd PM, Harvey DJ, Simmons PJ, Watt SM. Epitope recognition of antibodies that define the sialomucin, endolyn (CD164), a negative regulator of haematopoiesis. ACTA ACUST UNITED AC 2005; 65:220-39. [PMID: 15730515 DOI: 10.1111/j.1399-0039.2005.00358.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Endolyn (CD164) is a sialomucin that functions as an adhesion molecule and a negative regulator of CD34+ CD38- human haematopoietic precursor cell proliferation. The 105A5 and 103B2/9E10 CD164 monoclonal antibodies (mAbs), which act as surrogate ligands, recognize distinct glycosylation-dependent classes I and II epitopes located on domain I of the native and recombinant CD164 proteins. Here, we document five new CD164 mAbs, the 96 series, that rely on conformational integrity, but not glycosylation, of exons 2- and 3-encoded CD164 domains, thereby resembling the class III mAbs, N6B6 and 67D2. Although all the 96 series class III mAbs labelled both the 105A5+ and 103B2/9E10+ cells, cross-competition and immunoblotting studies allow them to be categorized into two distinct class III subgroups, i.e. the N6B6-like subgroup that only recognizes 80-100 kDa proteins and the 67D2-like subgroup that also recognizes a higher molecular weight (>220 kDa) form. To more closely define the reactivity patterns of mAbs to the classes I and II epitopes, the global glycosylation patterns of the soluble human (h) CD164 proteins were determined using lectin binding, high-performance liquid chromatography (HPLC) and mass spectrometry. hCD164 recombinant proteins bound to the lectins, Galanthus nivalis agglutinin, Datura stramonium agglutinin, Sambucus nigra agglutinin, Maackia amurensis agglutinin and peanut agglutinin, indicating the presence of high mannose and complex N-glycans, in addition to core 1 O-glycans (the Tn antigen) and alpha2-3 and alpha2-6 sialic acid moieties. Our HPLC and mass spectrometry results revealed both high mannose and complex N-glycosylation with various numbers of branches increasing the complexity of the glycosylation pattern. Most O-glycans were small, core 1 or 2 based. High levels of sialylation in alpha2-3 and alpha2-6 linkages, without sialyl-Lewis X, indicate that the majority of these hCD164 recombinant proteins are unable to bind to selectins in our assay system, but may interact with Siglec molecules.
Collapse
Affiliation(s)
- B Jorgensen-Tye
- Stem Cell Laboratory, National Blood Service and Nuffield Department of Clinical Laboratory Sciences, The John Radcliffe Hospital, Headington, Oxford, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
McGuckin CP, Forraz N, Baradez MO, Lojo-Rial C, Wertheim D, Whiting K, Watt SM, Pettengell R. Colocalization analysis of sialomucins CD34 and CD164. Stem Cells 2003; 21:162-70. [PMID: 12634412 DOI: 10.1634/stemcells.21-2-162] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Flow cytometric protocols are employed to identify and characterize hemopoietic stem/progenitor populations before transplantation. Cell surface antigens, including CD34, are employed in this process and widely used in harvest protocols, which largely ignores the potential functional role of such antigens. Transmembrane glycoprotein sialomucins, including CD34 and CD164, have been implicated in cell-to-cell interactions and activation. CD164, also expressed on early hemopoietic populations, was reported to have a possible function facilitating CD34(+) cells to adhere to bone marrow stroma. In this study, we employed high-definition laser-scanning confocal microscopy to investigate CD34 and CD164 surface co-localization patterns on bone marrow and cord blood cells and to compare the expression patterns using a three-dimensional computer-generated method developed in house. Differential interference microscopy analysis revealed bone marrow membrane activity was higher than the corresponding cord blood counterpart, perhaps indicating the marrow microenvironmental nature. Fluorescence analysis of CD34 and CD164 antigens showed both were expressed first in a halo-like pattern and second in antigen-dense pockets. Three-dimensional computer analyses further revealed that this pocketing corresponded to dense crest-like surface structures appearing to rise from the point of adherence on the slide. Further, it was found that CD34 and CD164 display strong colocalization patterns on cells expressing both antigens. The dual nature of the CD34 and CD164 antigens discovered here lends further evidence to the previous literature implicating a strong functional link between these two sialomucins, which should be considered in the transplantation arena and in the function of such sialomucins as negative regulators of cell proliferation.
Collapse
Affiliation(s)
- Colin P McGuckin
- King-George Laboratory, St. George's Hospital Medical School and Kingston University, London, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Handgretinger R, Gordon PR, Leimig T, Chen X, Buhring HJ, Niethammer D, Kuci S. Biology and plasticity of CD133+ hematopoietic stem cells. Ann N Y Acad Sci 2003; 996:141-51. [PMID: 12799292 DOI: 10.1111/j.1749-6632.2003.tb03242.x] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AC133 (CD133) is a highly conserved antigen expressed on hematopoietic stem cells with unknown function. In order to further characterize CD133(+) progenitor cells, we purified CD133(+) stem cells using the method of magnetic activated cell sorting (MACS) from healthy adult volunteers mobilized with granulocyte colony-stimulating growth factor (G-CSF) to a mean purity of 94%. The purified CD133(+) cells highly engrafted NOD/SCID mice. In addition, unseparated mononuclear cells or CD133(+) stem cells isolated from the bone marrow of transplanted NOD/SCID mice gave rise to engraftment of secondary recipients. Upon ex vivo culture of purified CD133(+) cells with FLT3/Flk2 ligand (FL) and interleukin-6 (IL-6), a plastic-adherent cell population could be observed after 6 weeks in culture. These adherent cells did not express CD34 or CD133 antigens on their surface, nor did they express markers for endothelial, mesenchymal, or dendritic cells. After incubation of these adherent cells with stem cell factor (SCF), non-adherent cells were observed which partially co-expressed CD133, but were negative for CD34. These nonadherent CD34(-) cells showed a high engraftment capacity in NOD/SCID mice. From our results, we conclude that CD133 might be a marker of early progenitors with a high NOD/SCID engraftment potential. The fact that CD133(+) hematopoietic progenitors can give rise to an adherent population which is CD133(-) and CD34(-) and that these cells can again give rise to a CD133(+)CD34(-) stem cell population with high NOD/SCID engraftment potential indicates the plasticity of hematopoietic precursors. CD133(+) stem cells might be useful for research and for clinical application.
Collapse
Affiliation(s)
- Rupert Handgretinger
- Division of Stem Cell Transplantation, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
Kuçi S, Wessels JT, Bühring HJ, Schilbach K, Schumm M, Seitz G, Löffler J, Bader P, Schlegel PG, Niethammer D, Handgretinger R. Identification of a novel class of human adherent CD34- stem cells that give rise to SCID-repopulating cells. Blood 2003; 101:869-76. [PMID: 12393715 DOI: 10.1182/blood-2002-03-0711] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Here we describe the in vitro generation of a novel adherent cell fraction derived from highly enriched, mobilized CD133(+) peripheral blood cells after their culture with Flt3/Flk2 ligand and interleukin-6 for 3 to 5 weeks. These cells lack markers of hematopoietic stem cells, endothelial cells, mesenchymal cells, dendritic cells, and stromal fibroblasts. However, all adherent cells expressed the adhesion molecules VE-cadherin, CD54, and CD44. They were also positive for CD164 and CD172a (signal regulatory protein-alpha) and for a stem cell antigen defined by the recently described antibody W7C5. Adherent cells can either spontaneously or upon stimulation with stem cell factor give rise to a transplantable, nonadherent CD133(+)CD34(-) stem cell subset. These cells do not generate in vitro hematopoietic colonies. However, their transplantation into nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice induced substantially higher long-term multilineage engraftment compared with that of freshly isolated CD34(+) cells, suggesting that these cells are highly enriched in SCID-repopulating cells. In addition to cells of the myeloid lineage, nonadherent CD34(-) cells were able to give rise to human cells with B-, T-, and natural killer-cell phenotype. Hence, these cells possess a distinct in vivo differentiation potential compared with that of CD34(+) stem cells and may therefore provide an alternative to CD34(+) progenitor cells for transplantation.
Collapse
Affiliation(s)
- Selim Kuçi
- Department of Hematology/Oncology, University Children's Hospital, Tübingen, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Connolly NP, Jones M, Watt SM. Human Siglec-5: tissue distribution, novel isoforms and domain specificities for sialic acid-dependent ligand interactions. Br J Haematol 2002; 119:221-38. [PMID: 12358929 DOI: 10.1046/j.1365-2141.2002.03808.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Human Siglec-5 is a sialic acid binding immunoglobulin (Ig)-like lectin (Siglec), comprising one N-terminal IgV-SET domain followed by three IgC2-SET domains, and a cytoplasmic domain with ITIM and SAP motifs which regulate cell signalling. We report the differential distribution of hSiglec-5 on neutrophil and macrophage subsets in tissues using monoclonal antibodies, 1A5 and 2H8, which require the first IgC2-SET domain for binding. Interestingly, hSiglec-5 was especially prominent on macrophages in reactive lymph nodes. We have identified four isoforms of hSiglec-5 possessing three (hSiglec-5-3L and -3C) or four (hSiglec-5-4L and -4S) extracellular domains linked to long (hSiglec-5-3L and -4L) or short (hSiglec-5-4S) cytoplasmic tails or existing as a soluble isoform (hSiglec-5-3C). hSiglec-5-4L has the broadest tissue distribution, being detected in adult spleen, thymus, lymph node, peripheral blood leucocytes and bone marrow, and in fetal lung and liver. A soluble Fc chimaeric protein containing the hSiglec-5-4L extracellular domain binds in a sialic acid-dependent manner to glycophorin A on human erythrocytes and to alpha2-3- and alpha2-6-sialyllactose moieties. Domain deletion mutants of hSiglec-5(D1-4)-Fc reveal that the first three IgC2-SET domains are required for optimal binding, with adhesion being abolished if the first IgC2-SET domain is deleted. This indicates that each hSiglec-5 isoform will interact with sialic acid ligands and provides the first step towards defining structure-function relationships of hSiglec-5 isoforms.
Collapse
MESH Headings
- Amino Acid Sequence
- Antibodies, Monoclonal/metabolism
- Antigens, CD/chemistry
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/chemistry
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Chimera
- Enzyme-Linked Immunosorbent Assay/methods
- Epitopes
- Erythrocytes/metabolism
- Hematopoietic Stem Cells/metabolism
- Humans
- Lectins/chemistry
- Lectins/genetics
- Lectins/metabolism
- Macrophages/metabolism
- Molecular Sequence Data
- Monocytes/metabolism
- N-Acetylneuraminic Acid/metabolism
- Neutrophils/metabolism
- Protein Isoforms/chemistry
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Nicholas P Connolly
- National Blood Service, Stem Cell Laboratory, National Blood Service Oxford Centre, John Radcliffe Hospital, Oxford, UK
| | | | | |
Collapse
|
30
|
Goldberg M, Wei M, Tycko B, Falikovich I, Warburton D. Identification and expression analysis of the human mu-protocadherin gene in fetal and adult kidneys. Am J Physiol Renal Physiol 2002; 283:F454-63. [PMID: 12167596 DOI: 10.1152/ajprenal.00012.2002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We recently cloned mu-protocadherin, a developmentally regulated cell adhesion molecule that contains an extracellular region with four cadherin-like ectodomains and a triply repeating mucin domain in its longer isoform. Expression of mu-protocadherin in L929 cells resulted in cellular aggregation, confirming its role in intercellular adhesion. We now identify the human mu-protocadherin ortholog and study its distribution in vivo and its targeting in polarized epithelia. Basic Local Alignment Search Tool searches and fluorescent in situ hybridization analysis on the basis of human-mouse synteny reveal that mu-protocadherin maps to 11p15.5, matching a previously identified gene called MUCDHL. At least three different splicing isoforms exist for MUCDHL that vary in expression in the fetal kidney. Mu-protocadherin is apically expressed along the brush border of the proximal convoluted tubule of the adult kidney. Transfection of truncated forms of mu-protocadherin into polarized Madin-Darby canine kidney cells reveals that the NH(2) terminus is essential for targeting to the apical surface. These results suggest that although human mu-protocadherin may mediate a homotypic adhesive interaction, it may have additional functions in terminally differentiated epithelia.
Collapse
Affiliation(s)
- Michael Goldberg
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA.
| | | | | | | | | |
Collapse
|
31
|
Abstract
Expression of cell surface markers on human hematopoietic cells has provided a method for characterizing subsets of cells with distinct biological functions. This is largely due to the ability to separate highly purified subpopulations of cells for comparative analysis. Relationships between the cell surface phenotype of these subpopulations and their proliferative and differentiative capacity have been instrumental in defining the hierarchical organization of cells comprising the human hematopoietic system. The identification and isolation of human hematopoietic cells expressing AC133, combined with use of in vitro and in vivo assays, has provided novel insights into the hematopoietic progenitor and stem cell compartment in the human. More recent studies have offered evidence that AC133 expression is not limited to primitive blood cells, but also defines unique cell populations in non-hematopoietic tissues. These findings will be reviewed here in the context of human hematopoiesis and the potential role and utility of AC133 expression in the human.
Collapse
Affiliation(s)
- M Bhatia
- The John P Robarts Research Institute, Developmental Stem Cell Biology, The University of Western Ontario, Ontario, Canada
| |
Collapse
|
32
|
Simmons PJ, Levesque JP, Haylock DN. Mucin-like molecules as modulators of the survival and proliferation of primitive hematopoietic cells. Ann N Y Acad Sci 2001; 938:196-206; discussion 206-7. [PMID: 11458509 DOI: 10.1111/j.1749-6632.2001.tb03590.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Current data suggest that interplay between two classes of molecules contributes to the regulation of hematopoiesis: hematopoietic growth factors, which regulate the survival, proliferation, and development of primitive hematopoietic cells and cell adhesion molecules (CAMs), which are responsible for the localization of hematopoiesis to the bone marrow (BM) and for mediating physical association between developing hematopoietic cells and marrow stromal tissue. A range of cell surface molecules representing several CAM superfamilies including integrins, selectins, the immunoglobulin gene superfamily and an emerging family of mucin-like molecules (the sialomucins) are involved in supporting cell-cell and cell-extracellular matrix (ECM) interactions between primitive hematopoietic cells and the stromal cell-mediated hematopoietic microenvironment (HM) of the bone marrow. There is abundant evidence in non-hematopoietic tissues that CAMs are signalling molecules which participate in a range of signal transduction events important not only for regulating cell adhesion and motility, but also for cell growth and survival. Although the signalling functions of CAMs have not been studied extensively in primitive hematopoietic progenitors (HPCs), extrapolation from burgeoning data in other systems is consistent with the hypothesis that hematopoiesis within the BM is regulated by interaction between signals generated locally by CAMs and those elicited by cytokines. Evidence in support of this notion was initially provided by studies on normal HPCs demonstrating cross-talk between members of the integrin superfamily and cytokine receptors. In this article we review recent reports that mucin-like molecules are also signalling molecules on primitive hematopoietic cells and that the signals they deliver potently inhibit hematopoiesis.
Collapse
Affiliation(s)
- P J Simmons
- Stem Cell Laboratory, Peter MacCallum Cancer Institute, St. Andrew's Place, East Melbourne, VIC 3002, Australia.
| | | | | |
Collapse
|
33
|
Elwood NJ, Smith CA. Current status of retroviral vector mediated gene transfer into human hematopoietic stem cells. Leuk Lymphoma 2001; 41:465-82. [PMID: 11378565 DOI: 10.3109/10428190109060338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Genetic modification of hematopoietic stem cells (HSCs) has been proposed as a treatment strategy for a variety of hematologic diseases, tracking marked cells or conferring resistance to chemotherapeutic agents. Despite early enthusiasm, the results of clinical studies involving gene transfer into HSCs has not resulted in therapeutic benefits for the vast majority of treated patients. This review describes the limitations and advances that have been made in the areas of gene transfer vectors, identification of the appropriate HSCs to target for genetic modifications and the methods used to perform gene transfer.
Collapse
Affiliation(s)
- N J Elwood
- Center for Genetic and Cellular Therapies, Department of Medicine, Box 2601, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
34
|
Elwood NJ, Smith CA. Current status of retroviral vector mediated gene transfer into human hematopoietic stem cells. Leuk Lymphoma 2001; 41:1-18. [PMID: 11342353 DOI: 10.3109/10428190109057950] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Genetic modification of hematopoietic stem cells (HSCs) has been proposed as a treatment strategy for a variety of hematologic diseases, tracking marked cells or conferring resistance to chemotherapeutic agents. Despite early enthusiasm, the results of clinical studies involving gene transfer into HSCs have not resulted in therapeutic benefits for the vast majority of treated patients. This review describes the limitations and advances that have been made in the areas of gene transfer vectors, identification of the appropriate HSCs to target for genetic modifications and the methods used to perform gene transfer.
Collapse
Affiliation(s)
- N J Elwood
- Center for Genetic and Cellular Therapies, Department of Medicine, Box 2601, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
35
|
Prosper F, Verfaillie CM. Regulation of hematopoiesis through adhesion receptors. J Leukoc Biol 2001. [DOI: 10.1189/jlb.69.3.307] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Felipe Prosper
- Department of Hematology and Medical Oncology, Hospital Clinico Universitario, University of Valencia, Spain
| | - Catherine M. Verfaillie
- Stem Institute, and Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis
| |
Collapse
|
36
|
Affiliation(s)
- J Y Chan
- The MRC Molecular Haematology Unit, Institute of Molecular Medicine, The John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | | |
Collapse
|
37
|
Chan JY, Lee-Prudhoe JE, Jorgensen B, Ihrke G, Doyonnas R, Zannettino AC, Buckle VJ, Ward CJ, Simmons PJ, Watt SM. Relationship between novel isoforms, functionally important domains, and subcellular distribution of CD164/endolyn. J Biol Chem 2001; 276:2139-52. [PMID: 11027692 DOI: 10.1074/jbc.m007965200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Functional analyses have indicated that the human CD164 sialomucin may play a key role in hematopoiesis by facilitating the adhesion of human CD34(+) cells to the stroma and by negatively regulating CD34(+)CD38(lo/-) cell proliferation. We have identified three novel human CD164 variants derived by alternative splicing of bona fide exons from a single genomic transcription unit. The predominant CD164(E1-6) isoform, encoded by six exons, is a type I transmembrane protein containing two extracellular mucin domains (I and II) interrupted by a cysteine-rich non-mucin domain. The 103B2/9E10 and 105A5 epitopes, which specify ligand binding characteristics, are located on the exon 1-encoded mucin domain I. Three human CD164(E1-6) mRNA species, exhibiting differential polyadenylation site usage, are differentially expressed in hematopoietic and non-hematopoietic tissues. This study provides additional evidence that human CD164(E1-6) represents the ortholog of murine MGC-24v and rat endolyn. Comparative analysis of murine MGC-24v/CD164(E1-6) with human CD164(E1-6) revealed two potential splice variants and a similar genomic structure. Whereas the human CD164 gene is located on chromosome 6q21, the mouse gene occurs in a syntenic region on chromosome 10B1-B2. By confocal microscopy, human CD164 in CD34(+)CD38(+) hematopoietic progenitor (KG1B) and epithelial cell lines appears to be localized primarily in endosomes and lysosomes, with low concentrations at the cell surface. However, in a minority of KG1B cells, CD164 is more prominently expressed at the plasma membrane and in the recycling endosomes, suggesting that its distribution is regulated in cells of hematopoietic origin.
Collapse
Affiliation(s)
- J Y Chan
- Medical Research Council Molecular Hematology Unit, Institute of Molecular Medicine, Oxford OX3 9DS, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Matsumoto K, Yasui K, Yamashita N, Horie Y, Yamada T, Tani Y, Shibata H, Nakano T. In vitro proliferation potential of AC133 positive cells in peripheral blood. Stem Cells 2000; 18:196-203. [PMID: 10840073 DOI: 10.1634/stemcells.18-3-196] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
AC133 antigen is a novel marker for human hematopoietic stem/progenitor cells. In this study, we examined the expression and proliferation potential of AC133(+) cells obtained from steady-state peripheral blood (PB). The proportion of AC133(+) cells in the CD34(+) subpopulation of steady-state PB was significantly lower than that of cord blood (CB), although that of cytokine-mobilized PB was higher than that of CB. The proliferation potential of AC133(+)CD34(+) and AC133(-)CD34(+) cells was examined by colony-forming analysis and analysis of long-term culture-initiating cells (LTC-IC). Although the total number of colony-forming cells was essentially the same in the AC133(+)CD34(+) fraction as in the AC133(-)CD34(+) fraction, the proportion of LTC-IC was much higher in the AC133(+)CD34(+) fraction. Virtually no LTC-IC were detected in the AC133(-)CD34(+) fraction. In addition, the features of the colonies grown from these two fractions were quite different. Approximately 70% of the colonies derived from the AC133(+)CD34(+) fraction were granulocyte-macrophage colonies, whereas more than 90% of the colonies derived from the AC133(-)CD34(+) fraction were erythroid colonies. Furthermore, an ex vivo expansion study observed expansion of colony-forming cells only in the AC133(+)CD34(+) population, and not in the AC133(-)CD34(+) population. These findings suggest that to isolate primitive hematopoietic cells from steady-state PB, selection by AC133 expression is better than selection by CD34 expression.
Collapse
|
39
|
Doyonnas R, Yi-Hsin Chan J, Butler LH, Rappold I, Lee-Prudhoe JE, Zannettino AC, Simmons PJ, Bühring HJ, Levesque JP, Watt SM. CD164 monoclonal antibodies that block hemopoietic progenitor cell adhesion and proliferation interact with the first mucin domain of the CD164 receptor. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:840-51. [PMID: 10878358 DOI: 10.4049/jimmunol.165.2.840] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The novel sialomucin, CD164, functions as both an adhesion receptor on human CD34+ cell subsets in bone marrow and as a potent negative regulator of CD34+ hemopoietic progenitor cell proliferation. These diverse effects are mediated by at least two functional epitopes defined by the mAbs, 103B2/9E10 and 105A5. We report here the precise epitope mapping of these mAbs together with that of two other CD164 mAbs, N6B6 and 67D2. Using newly defined CD164 splice variants and a set of soluble recombinant chimeric proteins encoded by exons 1-6 of the CD164 gene, we demonstrate that the 105A5 and 103B2/9E10 functional epitopes map to distinct glycosylated regions within the first mucin domain of CD164. The N6B6 and 67D2 mAbs, in contrast, recognize closely associated and complex epitopes that rely on the conformational integrity of the CD164 molecule and encompass the cysteine-rich regions encoded by exons 2 and 3. On the basis of their sensitivities to reducing agents and to sialidase, O-sialoglycoprotease, and N-glycanase treatments, we have characterized CD164 epitopes and grouped them into three classes by analogy with CD34 epitope classification. The class I 105A5 epitope is sialidase, O-glycosidase, and O-sialoglycoprotease sensitive; the class II 103B2/9E10 epitope is N-glycanase, O-glycosidase, and O-sialoglycoprotease sensitive; and the class III N6B6 and 67D2 epitopes are not removed by such enzyme treatments. Collectively, this study indicates that the previously observed differential expression of CD164 epitopes in adult tissues is linked with cell type specific post-translational modifications and suggests a role for epitope-associated carbohydrate structures in CD164 function.
Collapse
Affiliation(s)
- R Doyonnas
- Medical Research Council Molecular Haematology Unit, Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Hematopoiesis in adult bone marrow is a tightly regulated process involving interactions between cytokine and adhesion receptors on hematopoietic progenitor cells and their cognate ligands in the immediate microenvironment. These interactions control hematopoietic stem cell self-renewal, quiescence, commitment and migration. Recently, sialomucins have assumed some importance in hematopoiesis, with six of these receptors, CD34, PSGL-1, CD43, PCLP, CD45RA and CD164, having been identified on primitive hematopoietic precursor cells and/or their associated stromal/endothelial elements. This article reviews the cloning, expression and function of the recently identified sialomucin, CD164, which is highly expressed by primitive hematopoietic progenitor cells. The CD164 receptor is implicated in mediating or regulating hematopoietic precursor cell adhesion to stroma, and may serve as a potent negative regulator of hematopoietic progenitor cell proliferation.
Collapse
Affiliation(s)
- S M Watt
- The MRC Molecular Haematology Unit, Institute of Molecular Medicine, The John Radcliffe Hospital, Oxford, UK.
| | | |
Collapse
|
41
|
Opposing effects of engagement of integrins and stimulation of cytokine receptors on cell cycle progression of normal human hematopoietic progenitors. Blood 2000. [DOI: 10.1182/blood.v95.3.846.003k31_846_854] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We evaluated the effect of β1-integrin receptor engagement on the expression and activity of cell cycle regulatory proteins in CD34+ cells under conditions that mimic the steady-state marrow microenvironment and in the presence of supraphysiological concentrations of interleukin-3 (IL3) and stem cell factor (SCF). Adhesion of CD34+ progenitors to fibronectin (FN) was similar whether IL3 or SCF was present or absent. Engagement of β1-integrins blocked S-phase entry of CD34+ cells in the absence of IL3 or SCF, whereas addition of 10 ng/mL IL3 or SCF prevented such a block in S-phase entry. In the absence of IL3 or SCF, cyclin-E levels were significantly lower and p27KIP1 levels significantly higher in FN-adherent than in FN-nonadherent cells, or than in poly-L-lysine (PLL)–adherent or (PLL)–nonadherent cells. Cyclin-dependent-kinase (cdk)-2 activity was decreased and levels of cyclin-E–cdk2 complexes were lower in FN-adherent than in PLL-adherent cells. In contrast, cyclin-E and p27KIP1 protein levels and cdk2 activity in cells adherent to FN in the presence of IL3 or SCF were similar to those in PLL-adherent and FN-nonadherent or PLL-nonadherent cells. In conclusion, under physiological cytokine conditions, integrin engagement prevents S-phase entrance of CD34+ cells, which is associated with elevated levels of the contact-dependent cyclin kinase inhibitor p27KIP1. Supraphysiological concentrations of IL3 or SCF prevent p27KIP1 elevation and override the integrin-mediated inhibition of entry into S phase.
Collapse
|
42
|
Human Signal-Regulatory Protein Is Expressed on Normal, But Not on Subsets of Leukemic Myeloid Cells and Mediates Cellular Adhesion Involving Its Counterreceptor CD47. Blood 1999. [DOI: 10.1182/blood.v94.11.3633.423k01_3633_3643] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Signal-regulatory proteins (SIRPs) comprise a novel transmembrane glycoprotein family involved in the negative regulation of receptor tyrosine kinase-coupled signaling pathways. To analyze the expression and function of SIRPs, we prepared soluble recombinant fusion proteins of the extracellular regions of SIRP1 and SIRP2, as well as a variety of monoclonal antibodies (MoAbs) against these domains. The antibodies reacted predominantly with monocytes, granulocytes, dendritic cells, and their precursors, as well as with bone marrow CD34+, AC133+, CD90+hematopoietic stem/progenitor cells. In contrast, SIRP expression was absent or significantly reduced on the majority of myeloid blasts from patients with acute myeloid leukemia (AML) or chronic myeloid leukemia (CML). Functional studies showed that the extracellular domains of SIRP1 and SIRP2 support adhesion of a number of primary hematopoietic cells and cell lines. This interaction could be blocked by 4 of 7 SIRP1-reactive MoAbs. In addition, SIRP1 and SIRP2 competed for the same cell binding site, suggesting a common widely expressed SIRP ligand. In an approach to identify this molecule, MoAbs were generated against the SIRP-binding cell line CCRF-CEM, and MoAb CC2C6 was selected because of its capacity to inhibit cell binding to SIRP1. Further analysis showed that this antibody recognized CD47, a ubiquitously expressed plasma membrane protein previously implicated in integrin function, host defense action, and neutrophil migration. In this study, we identify CD47 as the extracellular ligand for human SIRP and show that these two counterreceptors are involved in cellular adhesion.
Collapse
|
43
|
Human Signal-Regulatory Protein Is Expressed on Normal, But Not on Subsets of Leukemic Myeloid Cells and Mediates Cellular Adhesion Involving Its Counterreceptor CD47. Blood 1999. [DOI: 10.1182/blood.v94.11.3633] [Citation(s) in RCA: 192] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Signal-regulatory proteins (SIRPs) comprise a novel transmembrane glycoprotein family involved in the negative regulation of receptor tyrosine kinase-coupled signaling pathways. To analyze the expression and function of SIRPs, we prepared soluble recombinant fusion proteins of the extracellular regions of SIRP1 and SIRP2, as well as a variety of monoclonal antibodies (MoAbs) against these domains. The antibodies reacted predominantly with monocytes, granulocytes, dendritic cells, and their precursors, as well as with bone marrow CD34+, AC133+, CD90+hematopoietic stem/progenitor cells. In contrast, SIRP expression was absent or significantly reduced on the majority of myeloid blasts from patients with acute myeloid leukemia (AML) or chronic myeloid leukemia (CML). Functional studies showed that the extracellular domains of SIRP1 and SIRP2 support adhesion of a number of primary hematopoietic cells and cell lines. This interaction could be blocked by 4 of 7 SIRP1-reactive MoAbs. In addition, SIRP1 and SIRP2 competed for the same cell binding site, suggesting a common widely expressed SIRP ligand. In an approach to identify this molecule, MoAbs were generated against the SIRP-binding cell line CCRF-CEM, and MoAb CC2C6 was selected because of its capacity to inhibit cell binding to SIRP1. Further analysis showed that this antibody recognized CD47, a ubiquitously expressed plasma membrane protein previously implicated in integrin function, host defense action, and neutrophil migration. In this study, we identify CD47 as the extracellular ligand for human SIRP and show that these two counterreceptors are involved in cellular adhesion.
Collapse
|
44
|
Kurosawa N, Kanemitsu Y, Matsui T, Shimada K, Ishihama H, Muramatsu T. Genomic analysis of a murine cell-surface sialomucin, MGC-24/CD164. EUROPEAN JOURNAL OF BIOCHEMISTRY 1999; 265:466-72. [PMID: 10491205 DOI: 10.1046/j.1432-1327.1999.00777.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
MGC-24 is a sialomucin originally found in human gastric carcinoma cells, and in human hematopoietic progenitor cells. In the human, soluble and transmembrane forms of MGC-24 are present, and the transmembrane form has been implicated in adhesion of hematopoietic progenitor cells to marrow stroma cells. In the mouse, we found that only the transmembrane form was expressed in many organs. Northern blotting and in situ hybridization analysis showed that MGC-24 mRNA was widely expressed in various adult and embryonic tissues. The mouse MGC-24 gene, which we isolated, spanned about 12 kb and was comprised of six exons. The transmembrane domain and the cytoplasmic domain were encoded by a single exon; the finding agrees with the absence of an alternatively spliced product of mouse MGC-24. The minimal promoter of mouse MGC-24 was embedded in GC-rich sequences, in which two Sp1 binding motifs were found, but it lacked TATA and CAAT boxes. That the promoter resembles that of house-keeping genes is consistent with the broad expression of mouse MGC-24 mRNA.
Collapse
Affiliation(s)
- N Kurosawa
- Department of Biochemistry, Nagoya University School of Medicine, Japan.
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
AbstractPhenotypic analysis of hematopoietic stem and progenitor cells has been an invaluable tool in defining the biology of stem cell populations. We use here flow cytometry to examine the expression of human erythroid-specific surface markers during the maturation of early committed erythroid cells derived from cord blood in vitro. The temporal order of the expression of erythroid specific markers was as follows: Kell glycoprotein (gp), Rh gp, Landsteiner Wiener (LW) gp, glycophorin A (GPA), Band 3, Lutheran (Lu) gp, and Duffy (Fy) gp. The time at which some of these markers appeared suggests possible roles for some of these erythroid-specific polypeptides during the differentiation of these committed progenitors. The early appearance of Kell gp raises the possibility that it may have an important role in the early stages of hematopoiesis or cell lineage determination. Kell gp may also be a useful marker for the diagnosis of erythroleukemia. The late expression of Lu gp suggests it may be involved in the migration of erythroid precursors from the marrow. Fy gp is also expressed late consistent with a role as a scavenger receptor for cytokines in the bone marrow and circulation. Rh c antigen appeared before Rh D antigen, and it is suggested that this may reflect a reorganization of the developing erythroid cell membrane involving the Rh polypeptides and other components, including GPA and Band 3.
Collapse
|
46
|
Abstract
Phenotypic analysis of hematopoietic stem and progenitor cells has been an invaluable tool in defining the biology of stem cell populations. We use here flow cytometry to examine the expression of human erythroid-specific surface markers during the maturation of early committed erythroid cells derived from cord blood in vitro. The temporal order of the expression of erythroid specific markers was as follows: Kell glycoprotein (gp), Rh gp, Landsteiner Wiener (LW) gp, glycophorin A (GPA), Band 3, Lutheran (Lu) gp, and Duffy (Fy) gp. The time at which some of these markers appeared suggests possible roles for some of these erythroid-specific polypeptides during the differentiation of these committed progenitors. The early appearance of Kell gp raises the possibility that it may have an important role in the early stages of hematopoiesis or cell lineage determination. Kell gp may also be a useful marker for the diagnosis of erythroleukemia. The late expression of Lu gp suggests it may be involved in the migration of erythroid precursors from the marrow. Fy gp is also expressed late consistent with a role as a scavenger receptor for cytokines in the bone marrow and circulation. Rh c antigen appeared before Rh D antigen, and it is suggested that this may reflect a reorganization of the developing erythroid cell membrane involving the Rh polypeptides and other components, including GPA and Band 3.
Collapse
|
47
|
Unusually Severe Heterozygous β-Thalassemia: Evidence for an Interacting Gene Affecting Globin Translation. Blood 1998. [DOI: 10.1182/blood.v92.9.3428] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractA common β-thalassemia mutation in Asian populations is the C → T substitution at position 654 of intron 2, which leads to the activation of two cryptic splicing sites and the incorporation of 73 extra nucleotides into the mutant mRNA. Like most β-thalassemia mutations, it normally exhibits recessive inheritance. We investigated the unusually severe phenotype in two heterozygotes for this mutation, father and son, who had thalassemia intermedia and an apparent dominant mode of inheritance. An increased level of aberrantly spliced transcript in the reticulocytes of the probands compared with asymptomatic β654heterozygotes led us to investigate the production and processing of β654 RNA. We showed that large amounts of the aberrant β654 transcript were detectable in erythroblasts from one of the asymptomatic cases. The translation product of this mRNA was not detectable in vivo, and we were unable to demonstrate the translation of the mutant mRNA in a cell-free translation system. Although the reticulocyte :β mRNA ratios in the two probands were within the range observed in the asymptomatic heterozygotes, globin chain biosynthesis studies showed that the probands had considerably greater :β chain imbalance. These results imply that the more severe phenotype may be due to a second defect, possibly unlinked to the β-globin cluster, that acts at the translational or posttranslational level.© 1998 by The American Society of Hematology.
Collapse
|
48
|
Abstract
A common β-thalassemia mutation in Asian populations is the C → T substitution at position 654 of intron 2, which leads to the activation of two cryptic splicing sites and the incorporation of 73 extra nucleotides into the mutant mRNA. Like most β-thalassemia mutations, it normally exhibits recessive inheritance. We investigated the unusually severe phenotype in two heterozygotes for this mutation, father and son, who had thalassemia intermedia and an apparent dominant mode of inheritance. An increased level of aberrantly spliced transcript in the reticulocytes of the probands compared with asymptomatic β654heterozygotes led us to investigate the production and processing of β654 RNA. We showed that large amounts of the aberrant β654 transcript were detectable in erythroblasts from one of the asymptomatic cases. The translation product of this mRNA was not detectable in vivo, and we were unable to demonstrate the translation of the mutant mRNA in a cell-free translation system. Although the reticulocyte :β mRNA ratios in the two probands were within the range observed in the asymptomatic heterozygotes, globin chain biosynthesis studies showed that the probands had considerably greater :β chain imbalance. These results imply that the more severe phenotype may be due to a second defect, possibly unlinked to the β-globin cluster, that acts at the translational or posttranslational level.© 1998 by The American Society of Hematology.
Collapse
|
49
|
|
50
|
|