1
|
Cuvertino S, Garner T, Martirosian E, Walusimbi B, Kimber SJ, Banka S, Stevens A. Higher order interaction analysis quantifies coordination in the epigenome revealing novel biological relationships in Kabuki syndrome. Brief Bioinform 2024; 26:bbae667. [PMID: 39701600 DOI: 10.1093/bib/bbae667] [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: 07/25/2024] [Revised: 10/25/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024] Open
Abstract
Complex direct and indirect relationships between multiple variables, termed higher order interactions (HOIs), are characteristics of all natural systems. Traditional differential and network analyses fail to account for the omic datasets richness and miss HOIs. We investigated peripheral blood DNA methylation data from Kabuki syndrome type 1 (KS1) and control individuals, identified 2,002 differentially methylated points (DMPs), and inferred 17 differentially methylated regions, which represent only 189 DMPs. We applied hypergraph models to measure HOIs on all the CpGs and revealed differences in the coordination of DMPs with lower entropy and higher coordination of the peripheral epigenome in KS1 implying reduced network complexity. Hypergraphs also capture epigenomic trans-relationships, and identify biologically relevant pathways that escape the standard analyses. These findings construct the basis of a suitable model for the analysis of organization in the epigenome in rare diseases, which can be applied to investigate mechanism in big data.
Collapse
Affiliation(s)
- Sara Cuvertino
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Terence Garner
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Evgenii Martirosian
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Bridgious Walusimbi
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University Foundation NHS Trust Health Innovation Manchester, Manchester, UK
| | - Susan J Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Siddharth Banka
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University Foundation NHS Trust Health Innovation Manchester, Manchester, UK
| | - Adam Stevens
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
2
|
Lee S, Sbihi H, MacIsaac JL, Balshaw R, Ambalavanan A, Subbarao P, Mandhane PJ, Moraes TJ, Turvey SE, Duan Q, Brauer M, Brook JR, Kobor MS, Jones MJ. Persistent DNA Methylation Changes across the First Year of Life and Prenatal NO2 Exposure in a Canadian Prospective Birth Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:47004. [PMID: 38573328 DOI: 10.1289/ehp13034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
BACKGROUND Evidence suggests that prenatal air pollution exposure alters DNA methylation (DNAm), which could go on to affect long-term health. It remains unclear whether DNAm alterations present at birth persist through early life. Identifying persistent DNAm changes would provide greater insight into the molecular mechanisms contributing to the association of prenatal air pollution exposure with atopic diseases. OBJECTIVES This study investigated DNAm differences associated with prenatal nitrogen dioxide (NO 2 ) exposure (a surrogate measure of traffic-related air pollution) at birth and 1 y of age and examined their role in atopic disease. We focused on regions showing persistent DNAm differences from birth to 1 y of age and regions uniquely associated with postnatal NO 2 exposure. METHODS Microarrays measured DNAm at birth and at 1 y of age for an atopy-enriched subset of Canadian Health Infant Longitudinal Development (CHILD) study participants. Individual and regional DNAm differences associated with prenatal NO 2 (n = 128 ) were identified, and their persistence at age 1 y were investigated using linear mixed effects models (n = 124 ). Postnatal-specific DNAm differences (n = 125 ) were isolated, and their association with NO 2 in the first year of life was examined. Causal mediation investigated whether DNAm differences mediated associations between NO 2 and age 1 y atopy or wheeze. Analyses were repeated using biological sex-stratified data. RESULTS At birth (n = 128 ), 18 regions of DNAm were associated with NO 2 , with several annotated to HOX genes. Some of these regions were specifically identified in males (n = 73 ), but not females (n = 55 ). The effect of prenatal NO 2 across CpGs within altered regions persisted at 1 y of age. No significant mediation effects were identified. Sex-stratified analyses identified postnatal-specific DNAm alterations. DISCUSSION Regional cord blood DNAm differences associated with prenatal NO 2 persisted through at least the first year of life in CHILD participants. Some differences may represent sex-specific alterations, but replication in larger cohorts is needed. The early postnatal period remained a sensitive window to DNAm perturbations. https://doi.org/10.1289/EHP13034.
Collapse
Affiliation(s)
- Samantha Lee
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
- Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Hind Sbihi
- British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Julia L MacIsaac
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Robert Balshaw
- Centre for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Padmaja Subbarao
- Department of Pediatrics & Translational Medicine, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Piushkumar J Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Medicine, USCI University, Kuala Lumpur, Malaysia
| | - Theo J Moraes
- Department of Pediatrics & Translational Medicine, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Stuart E Turvey
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Qingling Duan
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
- School of Computing, Queen's University, Kingston, Ontario, Canada
| | - Michael Brauer
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jeffrey R Brook
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Michael S Kobor
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Meaghan J Jones
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
- Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
3
|
Lin J, Zhu H, Hong L, Tang W, Wang J, Hu H, Wu X, Chen Y, Liu G, Yang Q, Li J, Wang Y, Lin Z, Xiao Y, Dai W, Huang M, Li G, Li A, Wang J, Xiang L, Liu S. Coexpression of HOXA6 and PBX2 promotes metastasis in gastric cancer. Aging (Albany NY) 2021; 13:6606-6624. [PMID: 33535170 PMCID: PMC7993744 DOI: 10.18632/aging.202426] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 11/03/2020] [Indexed: 01/23/2023]
Abstract
HOXA6 gene plays a role of the oncogene in various cancers. Nonetheless, its effect on gastric cancer (GC) occurrence and development is still unclear. We analysed whether HOXA6 interacts with the PBX2 protein using the STRING database. The molecular mechanism by which HOXA6 synergizes with PBX2 in GC metastasis is not fully understood. Here, we found that the expression of HOXA6 was increased in GC tissues and cell lines. The upregulation of HOXA6 was closely associated with differentiation, lymph node metastasis, AJCC stage, TNM stage, and poor survival outcome in GC patients based on tissue microarray (TMA) data. Moreover, the overexpression of HOXA6 promoted, whereas siRNA-mediated repression of HOXA6 inhibited, the cell proliferation, migration, and invasion of GC cells. Furthermore, HOXA6 could physically interact with and stabilize PBX2. In addition, HOXA6 and PBX2 expression was positively correlated in GC cells and tissue. HOXA6 and PBX2 suppression in GC cells also led to decreased migration and invasion potential in vitro. In vivo, HOXA6 was shown to cooperate with PBX2 to enhance cell metastasis via orthotopic implantation. These data indicate that HOXA6 promotes cell proliferation, migration, and invasion and that the HOXA6-PBX2 axis may be a useful biomarker for disease progression in GC.
Collapse
Affiliation(s)
- Jianjiao Lin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Gastroenterology, Longgang District People’s Hospital, Shenzhen 518172, China
| | - Huiqiong Zhu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Linjie Hong
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Weimei Tang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jing Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hongsong Hu
- Department of Gastroenterology, Longgang District People’s Hospital, Shenzhen 518172, China
| | - Xiaosheng Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yaying Chen
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510515, China
| | - Guangnan Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qiong Yang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- The Second Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Jiaying Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yusi Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhizhao Lin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yizhi Xiao
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Weiyu Dai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Miaojvan Huang
- Department of Gastroenterology, Longgang District People’s Hospital, Shenzhen 518172, China
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Aimin Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jide Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Gastroenterology, Longgang District People’s Hospital, Shenzhen 518172, China
| | - Li Xiang
- Department of Gastroenterology, Longgang District People’s Hospital, Shenzhen 518172, China
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Gastroenterology, Longgang District People’s Hospital, Shenzhen 518172, China
| |
Collapse
|
4
|
Lau LY, Nguyen LT, Reverter A, Moore SS, Lynn A, McBride‐Kelly L, Phillips‐Rose L, Plath M, Macfarlane R, Vasudivan V, Morton L, Ardley R, Ye Y, Fortes MRS. Gene regulation could be attributed to TCF3 and other key transcription factors in the muscle of pubertal heifers. Vet Med Sci 2020; 6:695-710. [PMID: 32432381 PMCID: PMC7738712 DOI: 10.1002/vms3.278] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 03/13/2020] [Accepted: 04/09/2020] [Indexed: 01/17/2023] Open
Abstract
Puberty is a whole-body event, driven by the hypothalamic integration of peripheral signals such as leptin or IGF-1. In the process of puberty, reproductive development is simultaneous to growth, including muscle growth. To enhance our understanding of muscle function related to puberty, we performed transcriptome analyses of muscle samples from six pre- and six post-pubertal Brahman heifers (Bos indicus). Our aims were to perform differential expression analyses and co-expression analyses to derive a regulatory gene network associate with puberty. As a result, we identified 431 differentially expressed (DEx) transcripts (genes and non-coding RNAs) when comparing pre- to post-pubertal average gene expression. The DEx transcripts were compared with all expressed transcripts in our samples (over 14,000 transcripts) for functional enrichment analyses. The DEx transcripts were associated with "extracellular region," "inflammatory response" and "hormone activity" (adjusted p < .05). Inflammatory response for muscle regeneration is a necessary aspect of muscle growth, which is accelerated during puberty. The term "hormone activity" may signal genes that respond to progesterone signalling in the muscle, as the presence of this hormone is an important difference between pre- and post-pubertal heifers in our experimental design. The DEx transcript with the highest average expression difference was a mitochondrial gene, ENSBTAG00000043574 that might be another important link between energy metabolism and puberty. In the derived co-expression gene network, we identified six hub genes: CDC5L, MYC, TCF3, RUNX2, ATF2 and CREB1. In the same network, 48 key regulators of DEx transcripts were identified, using a regulatory impact factor metric. The hub gene TCF3 was also a key regulator. The majority of the key regulators (22 genes) are members of the zinc finger family, which has been implicated in bovine puberty in other tissues. In conclusion, we described how puberty may affect muscle gene expression in cattle.
Collapse
Affiliation(s)
- Li Yieng Lau
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Loan T. Nguyen
- Queensland Alliance for Agriculture and Food InnovationThe University of QueenslandBrisbaneQLDAustralia
| | - Antonio Reverter
- CSIRO Agriculture and FoodQueensland Biosciences PrecinctBrisbaneQLDAustralia
| | - Stephen S. Moore
- Queensland Alliance for Agriculture and Food InnovationThe University of QueenslandBrisbaneQLDAustralia
| | - Aaron Lynn
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Liam McBride‐Kelly
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Louis Phillips‐Rose
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Mackenzie Plath
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Rhys Macfarlane
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Vanisha Vasudivan
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Lachlan Morton
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Ryan Ardley
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Yunan Ye
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Marina R. S. Fortes
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
- Queensland Alliance for Agriculture and Food InnovationThe University of QueenslandBrisbaneQLDAustralia
| |
Collapse
|
5
|
Identifying Cattle Breed-Specific Partner Choice of Transcription Factors during the African Trypanosomiasis Disease Progression Using Bioinformatics Analysis. Vaccines (Basel) 2020; 8:vaccines8020246. [PMID: 32456126 PMCID: PMC7350023 DOI: 10.3390/vaccines8020246] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/13/2020] [Accepted: 05/21/2020] [Indexed: 12/18/2022] Open
Abstract
African Animal Trypanosomiasis (AAT) is a disease caused by pathogenic trypanosomes which affects millions of livestock every year causing huge economic losses in agricultural production especially in sub-Saharan Africa. The disease is spread by the tsetse fly which carries the parasite in its saliva. During the disease progression, the cattle are prominently subjected to anaemia, weight loss, intermittent fever, chills, neuronal degeneration, congestive heart failure, and finally death. According to their different genetic programs governing the level of tolerance to AAT, cattle breeds are classified as either resistant or susceptible. In this study, we focus on the cattle breeds N’Dama and Boran which are known to be resistant and susceptible to trypanosomiasis, respectively. Despite the rich literature on both breeds, the gene regulatory mechanisms of the underlying biological processes for their resistance and susceptibility have not been extensively studied. To address the limited knowledge about the tissue-specific transcription factor (TF) cooperations associated with trypanosomiasis, we investigated gene expression data from these cattle breeds computationally. Consequently, we identified significant cooperative TF pairs (especially DBP−PPARA and DBP−THAP1 in N’Dama and DBP−PAX8 in Boran liver tissue) which could help understand the underlying AAT tolerance/susceptibility mechanism in both cattle breeds.
Collapse
|
6
|
Pathways, Processes, and Candidate Drugs Associated with a Hoxa Cluster-Dependency Model of Leukemia. Cancers (Basel) 2019; 11:cancers11122036. [PMID: 31861091 PMCID: PMC6966468 DOI: 10.3390/cancers11122036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/11/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023] Open
Abstract
High expression of the HOXA cluster correlates with poor clinical outcome in acute myeloid leukemias, particularly those harboring rearrangements of the mixed-lineage-leukemia gene (MLLr). Whilst decreased HOXA expression acts as a readout for candidate experimental therapies, the necessity of the HOXA cluster for leukemia maintenance has not been fully explored. Primary leukemias were generated in hematopoietic stem/progenitor cells from Cre responsive transgenic mice for conditional deletion of the Hoxa locus. Hoxa deletion resulted in reduced proliferation and colony formation in which surviving leukemic cells retained at least one copy of the Hoxa cluster, indicating dependency. Comparative transcriptome analysis of Hoxa wild type and deleted leukemic cells identified a unique gene signature associated with key pathways including transcriptional mis-regulation in cancer, the Fanconi anemia pathway and cell cycle progression. Further bioinformatics analysis of the gene signature identified a number of candidate FDA-approved drugs for potential repurposing in high HOXA expressing cancers including MLLr leukemias. Together these findings support dependency for an MLLr leukemia on Hoxa expression and identified candidate drugs for further therapeutic evaluation.
Collapse
|
7
|
Wu F, Wu S, Tong H, He W, Gou X. HOXA6 inhibits cell proliferation and induces apoptosis by suppressing the PI3K/Akt signaling pathway in clear cell renal cell carcinoma. Int J Oncol 2019; 54:2095-2105. [PMID: 31081053 PMCID: PMC6521939 DOI: 10.3892/ijo.2019.4789] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/09/2019] [Indexed: 12/13/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common type of renal cell carcinoma and the incidence of this disease is increasing. The present study aimed to investigate the role of homeobox A6 (HOXA6) in the proliferation and apoptosis of ccRCC cells. Analysis of the GSE6344 dataset and immunohistochemistry revealed that the mRNA and protein expression levels of HOXA6 were suppressed in ccRCC tissues. To evaluate the roles of HOXA6 in cell proliferation and apoptosis, ccRCC cell lines (786‑O and 769‑P) were transfected with plasmids expressing HOXA6, empty vector, short hairpin (sh)HOXA6 and non‑targeting shRNA (NC). Cell Counting Kit‑8, colony formation and 5‑ethynyl‑2'‑deoxyuridine staining assays were performed to analyze cell proliferation. In addition, Caspase‑Glo and terminal deoxynucleotidyl transferase dUTP nick end labeling assays were performed to detect apoptosis. Furthermore, the cell cycle and apoptotic rates of 786‑O and 769‑P cells were analyzed by flow cytometry. The results demonstrated that, compared with the empty vector group, the proliferation of 786‑O and 769‑P cells decreased following HOXA6 overexpression; however, compared with the NC group, cell proliferation increased in the shHOXA6 group. The rate of apoptosis of HOXA6‑overexpressing cells was increased compared with the empty vector group, while the rate of apoptosis in the shHOXA6 group was reduced compared with the NC group. In addition, flow cytometry demonstrated that upregulated HOXA6 expression levels could inhibit the cell cycle at the G0/G1 phase. Western blotting revealed that the expression levels of phosphoinositide 3‑kinase (PI3K), phosphorylated (p)‑protein kinase B (Akt), mitogen‑activated protein kinase kinase, p‑extracellular signal‑regulated kinase (ERK) and B‑cell lymphoma 2 (Bcl‑2) were suppressed in cells overexpressing HOXA6; however, the protein expression levels of phosphatase and tensin homolog, Bcl‑2‑associated X protein, cleaved caspase‑3 and cleaved‑poly (ADP‑ribose) polymerase were increased compared with the empty vector group. Opposing results were reported for the shHOXA6 group compared with the NC group. In summary, the results demonstrated that HOXA6 suppresses cell proliferation and promotes apoptosis, which may occur via inhibition of the PI3K/Akt/ERK cascade. These findings indicate the role of HOXA6 in ccRCC; however, the underlying mechanism requires further investigation.
Collapse
Affiliation(s)
- Feixiang Wu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Shasha Wu
- Department of Gastroenterology, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan 621000, P.R. China
| | - Hang Tong
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Weiyang He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xin Gou
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
8
|
Arabkari V, Amirizadeh N, Nikougoftar M, Soleimani M. microRNA expression profiles in two- and three-dimensional culture conditions of human-umbilical-cord blood-derived CD34 + cells. J Cell Physiol 2019; 234:20072-20084. [PMID: 30953369 DOI: 10.1002/jcp.28606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 03/19/2019] [Indexed: 12/16/2022]
Abstract
Human umbilical cord blood (HUCB) is a suitable source of hematopoietic stem cells (HSCs) for therapeutic transplantation. Different approaches have been used to expand the number of HSCs to increase the rate of HSC transplantation success in patients, such as using different cocktails of cytokines, feeder cell layers, and biocompatible scaffolds. microRNAs (miRNAs) are small noncoding RNAs that regulate gene expression posttranscriptionally. They play crucial roles in hematopoiesis including stem cell proliferation, differentiation, stemness, and self-renewal properties. Here, we studied the UCB-derived CD34+ cell expansion and the miRNA signatures of CD34+ cells on two- and three-dimensional (2D and 3D) culture conditions. We successfully expanded the UCB-derived CD34+ cells in both liquid culture (2D) and on aminated polyethersulfone nanofiber scaffolds (3D). Next, we identified the miRNA signature of CD34+ cells and their target genes. We found 58 dysregulated miRNAs in 3D culture condition and 34 dysregulated miRNAs in 2D culture condition when compared to the freshly isolated CD34+ cells. Various types of target genes were also predicted in both conditions using two online databases.
Collapse
Affiliation(s)
- Vahid Arabkari
- Blood Transfusion Research Center, High Institute for Education and Research in Transfusion Medicine, Iran Blood Transfusion Organization, Tehran, Iran
| | - Naser Amirizadeh
- Blood Transfusion Research Center, High Institute for Education and Research in Transfusion Medicine, Iran Blood Transfusion Organization, Tehran, Iran
| | - Mahin Nikougoftar
- Blood Transfusion Research Center, High Institute for Education and Research in Transfusion Medicine, Iran Blood Transfusion Organization, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
9
|
Increased levels of the long noncoding RNA, HOXA-AS3, promote proliferation of A549 cells. Cell Death Dis 2018; 9:707. [PMID: 29899328 PMCID: PMC5999602 DOI: 10.1038/s41419-018-0725-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 04/26/2018] [Accepted: 05/04/2018] [Indexed: 01/01/2023]
Abstract
Many long noncoding RNAs (lncRNAs) have been identified as powerful regulators of lung adenocarcinoma (LAD). However, the role of HOXA-AS3, a novel lncRNA, in LAD is largely unknown. In this study, we showed that HOXA-AS3 was significantly upregulated in LAD tissues and A549 cells. After knockdown of HOXA-AS3, cell proliferation, migration, and invasion were inhibited. Xenografts derived from A549 cells transfected with shRNA/HOXA-AS3 had significantly lower tumor weights and smaller tumor volumes. We also demonstrated that HOXA-AS3 increased HOXA6 mRNA stability by forming an RNA duplex. In addition, HOXA6 promoted cell proliferation, migration, and invasion in vitro. Using a RNA pull-down assay, we found that HOXA-AS3 bonded with NF110, which regulated the cell localization of HOXA-AS3. Moreover, histone acetylation was involved in upregulation of HOXA-AS3. These results demonstrate that HOXA-AS3 was activated in LAD and supported cancer cell progression. Therefore, inhibition of HOXA-AS3 could be an effective targeted therapy for patients with LAD.
Collapse
|
10
|
Platais C, Hakami F, Darda L, Lambert DW, Morgan R, Hunter KD. The role of HOX genes in head and neck squamous cell carcinoma. J Oral Pathol Med 2015; 45:239-47. [PMID: 26661059 DOI: 10.1111/jop.12388] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2015] [Indexed: 12/13/2022]
Abstract
Recent decades have witnessed the publication of numerous studies reporting alterations in the genome and transcriptome of head and neck squamous cell carcinoma (HNSCC). Currently, the utilisation of these alterations as biomarkers and targets for therapy is limited and new, useful molecular characteristics are being sought. Many of the published HNSCC gene expression profiles demonstrate alterations in the expression of HOX genes. These are a family of Homeobox-containing genes which are involved in developmental patterning and morphogenesis in the embryo, and which are often aberrantly expressed in cancer. The 39 HOX genes found in the human genome are arranged in four paralogous groups at different chromosomal loci. These control a wide range of cellular processes, including proliferation and migration, which are relevant in the context of cancer development. In this review article, we will outline the biology of HOX genes in relation to cancer and summarise the accumulating evidence for their role in the development of HNSCC and the possibility that they could be a therapeutic target in this malignancy. We will also identify areas where our current understanding is weak to focus future work and appraise the ongoing strategies for pharmacological intervention.
Collapse
Affiliation(s)
- Christopher Platais
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Fahad Hakami
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK.,Department of Pathology and Laboratory Medicine, King Abdulaziz Medical City-WR, Jeddah, Saudi Arabia
| | - Lav Darda
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Daniel W Lambert
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Richard Morgan
- Institute of Cancer Therapeutics, University of Bradford, Bradford, UK
| | - Keith D Hunter
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK.,Department of Oral Pathology and Biology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
11
|
Jang SH, Lee S, Chung HY. Characterization of Leukemia-Inducing Genes Using a Proto-Oncogene/Homeobox Gene Retroviral Human cDNA Library in a Mouse In Vivo Model. PLoS One 2015; 10:e0143240. [PMID: 26606454 PMCID: PMC4659616 DOI: 10.1371/journal.pone.0143240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 11/01/2015] [Indexed: 11/18/2022] Open
Abstract
The purpose of this research is to develop a method to screen a large number of potential driver mutations of acute myeloid leukemia (AML) using a retroviral cDNA library and murine bone marrow transduction-transplantation system. As a proof-of-concept, murine bone marrow (BM) cells were transduced with a retroviral cDNA library encoding well-characterized oncogenes and homeobox genes, and the virus-transduced cells were transplanted into lethally irradiated mice. The proto-oncogenes responsible for leukemia initiation were identified by PCR amplification of cDNA inserts from genomic DNA isolated from leukemic cells. In an initial screen of ten leukemic mice, the MYC proto-oncogene was detected in all the leukemic mice. Of ten leukemic mice, 3 (30%) had MYC as the only transgene, and seven mice (70%) had additional proto-oncogene inserts. We repeated the same experiment after removing MYC-related genes from the library to characterize additional leukemia-inducing gene combinations. Our second screen using the MYC-deleted proto-oncogene library confirmed MEIS1and the HOX family as cooperating oncogenes in leukemia pathogenesis. The model system we introduced in this study will be valuable in functionally screening novel combinations of genes for leukemogenic potential in vivo, and the system will help in the discovery of new targets for leukemia therapy.
Collapse
Affiliation(s)
- Su Hwa Jang
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Sohyun Lee
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Hee Yong Chung
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
12
|
Kettyle LMJ, Liberante FG, Thompson A. Rational drug repurposing using sscMap analysis in a HOX-TALE model of leukemia. Methods Mol Biol 2014; 1196:349-70. [PMID: 25151174 DOI: 10.1007/978-1-4939-1242-1_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Drug discovery and development are often hampered by lack of target identification and clinical tractability. Repurposing of approved drugs to life-threatening diseases such as leukemia is emerging as a promising alternative approach. Connectivity mapping systems link approved drugs with disease-related gene signatures. Relevant preclinical models provide essential tools for system validation and proof-of-concept studies. Herein we describe procedures aimed at generating disease-based gene signatures and applying them to established cross-referencing databases of potential candidate drugs. As a proof of principle, we present the identification of Entinostat as a candidate drug for the treatment of HOX-TALE-related leukemia.
Collapse
Affiliation(s)
- Laura M J Kettyle
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK
| | | | | |
Collapse
|
13
|
Mulgrew NM, Kettyle LMJ, Ramsey JM, Cull S, Smyth LJ, Mervyn DM, Bijl JJ, Thompson A. c-Met inhibition in a HOXA9/Meis1 model of CN-AML. Dev Dyn 2013; 243:172-81. [PMID: 24307265 DOI: 10.1002/dvdy.24070] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Revised: 09/16/2013] [Accepted: 09/26/2013] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Hematopoiesis is a paradigm for developmental processes, hierarchically organized, with stem cells at its origin. Hematopoietic stem cells (HSCs) replenish progenitor and precursor cells of multiple lineages, which normally differentiate into short-lived mature circulating cells. Hematopoiesis has provided insight into the molecular basis of tissue homeostasis and malignancy. Malignant hematopoiesis, in particular acute myeloid leukemia (AML), results from impaired development or differentiation of HSCs and progenitors. Co-overexpression of HOX and TALE genes, particularly the HOXA cluster and MEIS1, is associated with AML. Clinically relevant models of AML are required to advance drug development for an aging patient cohort. RESULTS Molecular analysis identified altered gene, microRNA, and protein expression in HOXA9/Meis1 leukemic bone marrow compared to normal controls. A candidate drug screen identified the c-Met inhibitor SU11274 for further analysis. Altered cell cycle status, apoptosis, differentiation, and impaired colony formation were shown for SU11274 in AML cell lines and primary leukemic bone marrow. CONCLUSIONS The clonal HOXA9/Meis1 AML model is amenable to drug screening analysis. The data presented indicate that human AML cells respond in a similar manner to the HOXA9/Meis1 cells, indicating pre-clinical relevance of the mouse model.
Collapse
Affiliation(s)
- Nuala M Mulgrew
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Northern Ireland, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Pediatric acute myeloid leukemia with t(8;16)(p11;p13), a distinct clinical and biological entity: a collaborative study by the International-Berlin-Frankfurt-Munster AML-study group. Blood 2013; 122:2704-13. [PMID: 23974201 DOI: 10.1182/blood-2013-02-485524] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In pediatric acute myeloid leukemia (AML), cytogenetic abnormalities are strong indicators of prognosis. Some recurrent cytogenetic abnormalities, such as t(8;16)(p11;p13), are so rare that collaborative studies are required to define their prognostic impact. We collected the clinical characteristics, morphology, and immunophenotypes of 62 pediatric AML patients with t(8;16)(p11;p13) from 18 countries participating in the International Berlin-Frankfurt-Münster (I-BFM) AML study group. We used the AML-BFM cohort diagnosed from 1995-2005 (n = 543) as a reference cohort. Median age of the pediatric t(8;16)(p11;p13) AML patients was significantly lower (1.2 years). The majority (97%) had M4-M5 French-American-British type, significantly different from the reference cohort. Erythrophagocytosis (70%), leukemia cutis (58%), and disseminated intravascular coagulation (39%) occurred frequently. Strikingly, spontaneous remissions occurred in 7 neonates with t(8;16)(p11;p13), of whom 3 remain in continuous remission. The 5-year overall survival of patients diagnosed after 1993 was 59%, similar to the reference cohort (P = .14). Gene expression profiles of t(8;16)(p11;p13) pediatric AML cases clustered close to, but distinct from, MLL-rearranged AML. Highly expressed genes included HOXA11, HOXA10, RET, PERP, and GGA2. In conclusion, pediatric t(8;16)(p11;p13) AML is a rare entity defined by a unique gene expression signature and distinct clinical features in whom spontaneous remissions occur in a subset of neonatal cases.
Collapse
|
15
|
Dickson GJ, Liberante FG, Kettyle LM, O'Hagan KA, Finnegan DPJ, Bullinger L, Geerts D, McMullin MF, Lappin TRJ, Mills KI, Thompson A. HOXA/PBX3 knockdown impairs growth and sensitizes cytogenetically normal acute myeloid leukemia cells to chemotherapy. Haematologica 2013; 98:1216-25. [PMID: 23539541 DOI: 10.3324/haematol.2012.079012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The cytogenetically normal subtype of acute myeloid leukemia is associated with an intermediate risk which complicates therapeutic options. Lower overall HOX/TALE expression appears to correlate with more favorable prognosis/better response to treatment in some leukemias and solid cancer. The functional significance of the associated gene expression and response to chemotherapy is not known. Three independent microarray datasets obtained from large cohorts of patients along with quantitative polymerase chain reaction validation were used to identify a four-gene HOXA/TALE signature capable of prognostic stratification. Biochemical analysis was used to identify interactions between the four encoded proteins and targeted knockdown used to examine the functional importance of sustained expression of the signature in leukemia maintenance and response to chemotherapy. An 11 HOXA/TALE code identified in an intermediate-risk group of patients (n=315) compared to a group with a favorable risk (n=105) was reduced to a four-gene signature of HOXA6, HOXA9, PBX3 and MEIS1 by iterative analysis of independent platforms. This signature maintained the favorable/intermediate risk partition and where applicable, correlated with overall survival in cytogenetically normal acute myeloid leukemia. We further showed that cell growth and function are dependent on maintained levels of these core genes and that direct targeting of HOXA/PBX3 sensitizes cytogenetically normal acute myeloid leukemia cells to standard chemotherapy. Together the data support a key role for HOXA/TALE in cytogenetically normal acute myeloid leukemia and demonstrate that targeting of clinically significant HOXA/PBX3 elements may provide therapeutic benefit to patients with this subtype of leukemia.
Collapse
Affiliation(s)
- Glenda J Dickson
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Northern Ireland, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Wheadon H, Ramsey JM, Dobbin E, Dickson GJ, Corrigan PM, Freeburn RW, Thompson A. Differential Hox expression in murine embryonic stem cell models of normal and malignant hematopoiesis. Stem Cells Dev 2011; 20:1465-76. [PMID: 21083428 DOI: 10.1089/scd.2010.0226] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The Hox family are master transcriptional regulators of developmental processes, including hematopoiesis. The Hox regulators, caudal homeobox factors (Cdx1-4), and Meis1, along with several individual Hox proteins, are implicated in stem cell expansion during embryonic development, with gene dosage playing a significant role in the overall function of the integrated Hox network. To investigate the role of this network in normal and aberrant, early hematopoiesis, we employed an in vitro embryonic stem cell differentiation system, which recapitulates mouse developmental hematopoiesis. Expression profiles of Hox, Pbx1, and Meis1 genes were quantified at distinct stages during the hematopoietic differentiation process and compared with the effects of expressing the leukemic oncogene Tel/PDGFRβ. During normal differentiation the Hoxa cluster, Pbx1 and Meis1 predominated, with a marked reduction in the majority of Hox genes (27/39) and Meis1 occurring during hematopoietic commitment. Only the posterior Hoxa cluster genes (a9, a10, a11, and a13) maintained or increased expression at the hematopoietic colony stage. Cdx4, Meis1, and a subset of Hox genes, including a7 and a9, were differentially expressed after short-term oncogenic (Tel/PDGFRβ) induction. Whereas Hoxa4-10, b1, b2, b4, and b9 were upregulated during oncogenic driven myelomonocytic differentiation. Heterodimers between Hoxa7/Hoxa9, Meis1, and Pbx have previously been implicated in regulating target genes involved in hematopoietic stem cell (HSC) expansion and leukemic progression. These results provide direct evidence that transcriptional flux through the Hox network occurs at very early stages during hematopoietic differentiation and validates embryonic stem cell models for gaining insights into the genetic regulation of normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Helen Wheadon
- Paul O'Gorman Leukaemia Research Centre, University of Glasgow, Glasgow, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
17
|
Ivanović Z. Ex vivo expansion of hematopoietic cells today. SCRIPTA MEDICA 2011. [DOI: 10.5937/scrimed1102092i] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
18
|
Abstract
Abstract
HOX homeobox genes are important regulators of normal and malignant hematopoiesis. Abdominal-type HOXA genes like HOXA9 are highly leukemogenic. However, little is known about transformation by anterior HOXA genes. Here we performed a comprehensive assessment of the oncogenic potential of every HOXA gene in primary hematopoietic cells. With exception of HOXA2 and HOXA5, all HOXA genes caused a block or delay of hematopoietic differentiation and cooperated with Meis1. No evidence for the alleged tumor-suppressor function of HOXA5 could be found. Whereas all active HOXA genes immortalized mixed granulocytic/monocytic populations, HOXA13 preferentially specified monocytoid development. The anterior HOXA genes HOXA1, HOXA4, and HOXA6 transformed cells, generating permanent cell lines, although they did so less potently than HOXA9. Upon transplantation these lines induced myeloproliferation and acute myeloid leukemia in recipient animals. Kinetic studies with inducible HOX derivatives demonstrated that anterior HOXA genes autonomously contributed to cellular transformation. This function was not mediated by endogenous Hoxa9, which was persistently expressed in cells transformed by anterior HOX genes. In summary our results demonstrate a hitherto unexpected role of anterior HOXA genes in hematopoietic malignancy.
Collapse
|
19
|
Di-Poï N, Koch U, Radtke F, Duboule D. Additive and global functions of HoxA cluster genes in mesoderm derivatives. Dev Biol 2010; 341:488-98. [PMID: 20303345 DOI: 10.1016/j.ydbio.2010.03.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Revised: 03/09/2010] [Accepted: 03/09/2010] [Indexed: 01/31/2023]
Abstract
Hox genes encode transcription factors that play a central role in the specification of regional identities along the anterior to posterior body axis. In the developing mouse embryo, Hox genes from all four genomic clusters are involved in range of developmental processes, including the patterning of skeletal structures and the formation of several organs. However, the functional redundancy observed either between paralogous genes, or among neighboring genes from the same cluster, has hampered functional analyses, in particular when synergistic, cluster-specific functions are considered. Here, we report that mutant mice lacking the entire HoxA cluster in mesodermal lineages display the expected spectrum of postnatal respiratory, cardiac and urogenital defects, previously reported for single gene mutations. Likewise, mild phenotypes are observed in both appendicular and axial skeleton. However, a striking effect was uncovered in the hematopoietic system, much stronger than that seen for Hoxa9 inactivation alone, which involves stem cells (HSCs) as well as the erythroid lineage, indicating that several Hoxa genes are necessary for normal hematopoiesis to occur. Finally, the combined deletions of Hoxa and Hoxd genes reveal abnormalities in axial elongation as well as skin morphogenesis that are likely the results of defects in epithelial-mesenchymal interactions.
Collapse
Affiliation(s)
- Nicolas Di-Poï
- National Research Center Frontiers in Genetics, Department of Zoology and Animal Biology, University of Geneva, Sciences III, 1211 Geneva 4, Switzerland
| | | | | | | |
Collapse
|
20
|
Ivanovic Z, Boiron JM. [Ex vivo expansion of hematopoietic stem cells: concept and clinical benefit]. Transfus Clin Biol 2009; 16:489-500. [PMID: 19932632 DOI: 10.1016/j.tracli.2009.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Accepted: 10/13/2009] [Indexed: 10/20/2022]
Abstract
A new discipline was born and grew up over the last 4 decades of 20th century: Experimental Hematology. In addition to yield the concept of Stemness, a paradigm later applied for the other tissues than hematopoietic one, it provided the results allowing a preclinical development and a therapeutic exploitation. The concept of ex vivo expansion of hematopoietic cells for transplantation is directly issued from this knowledge. It enabled us to realize that a critical quantity of different sub-populations of stem and progenitor cells are necessary to obtain a rapid and sustained hematopoietic reconstitution. These principles, transposed to human cells (originating from: bone marrow, peripheral blood, cord blood) required some important technological innovations (conception of the specific media, recombinant technology of cytokine production...), to achieve, after several attempts, the first efficient clinical trials (at the moment for cells mobilized in peripheral blood). This goal remains to be achieved for cord blood cells too. The developments in this field as well as its actual state are the subjects of this review.
Collapse
Affiliation(s)
- Z Ivanovic
- Etablissement français du Sang-Aquitaine-limousin, place Amélie-Raba-Léon, BP24, 33035 Bordeaux cedex, France
| | | |
Collapse
|