1
|
Wang Z, Xie C, Li Y, Wang B, Jian J, Lu Y, Xia L, Chang M. Characterization of CD166 from Nile tilapia (Oreochromis niloticus) displays a broad pathogen recognition spectrum and involved the immune response to microbial aggression. FISH & SHELLFISH IMMUNOLOGY 2023; 134:108516. [PMID: 36603790 DOI: 10.1016/j.fsi.2023.108516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/20/2022] [Accepted: 01/01/2023] [Indexed: 06/17/2023]
Abstract
CD166 is a member of the immunoglobulin superfamily of cell adhesion molecules, and its mediated adhesion plays a crucial role in different physiological and pathological phenomena, especially related to leukocyte extravasation, immune synapse stability, T cell activation and proliferation. In this study, CD166 was identified from Nile tilapia (Oreochromis niloticus, OnCD166). OnCD166 contains an open reading frame of 1671 bp that encodes a peptide of 556 amino acids, and contains five consecutive extracellular immunoglobulin domains. It's tissue distribution and expression patterns after S. agalactiae challenge were also investigated. OnCD166 is widely distributed in various tissues of healthy tilapia. After Streptococcus agalactiae challenge, OnCD166 expressions were significantly up-regulated in all tested immune tissues. Meanwhile, the recombinant OnCD166 (rOnCD166E) protein showed strong agglutinating activities against both Gram-negative bacteria and Gram-positive bacteria. Moreover, rOnCD166E could promote phagocytosis of macrophages. Taken together, our results illustrated that OnCD166 might as a receptor involved in the immune recognition and phagocytosis against invading pathogen, which play important roles in the immune responses of Nile tilapia against bacterial pathogens.
Collapse
Affiliation(s)
- Zhiwen Wang
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China; State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, China
| | - Caixia Xie
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Yuan Li
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Bei Wang
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Jichang Jian
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Yishan Lu
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Liqun Xia
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China.
| | - Mingxian Chang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, China.
| |
Collapse
|
2
|
Akbarzadeh M, Maroufi NF, Tazehkand AP, Akbarzadeh M, Bastani S, Safdari R, Farzane A, Fattahi A, Nejabati HR, Nouri M, Samadi N. Current approaches in identification and isolation of cancer stem cells. J Cell Physiol 2019; 234:14759-14772. [PMID: 30741412 DOI: 10.1002/jcp.28271] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/17/2019] [Accepted: 01/22/2019] [Indexed: 01/24/2023]
Abstract
Cancer stem cells (CSCs) are tumor cells with initiating ability, self-renewal potential, and intrinsic resistance to conventional therapeutics. Efficient isolation and characterization of CSCs pave the way for more comprehensive knowledge about tumorigenesis, heterogeneity, and chemoresistance. Also a better understanding of CSCs will lead to novel era of both basic and clinical cancer research, reclassification of human tumors, and development of innovative therapeutic strategies. Finding novel diagnostic and effective therapeutic strategies also enhance the success of treatment in cancer patients. There are various methods based on the characteristics of the CSCs to detect and isolate these cells, some of which have recently developed. This review summarized current techniques for effective isolation and characterization of CSCs with a focus on advantages and limitations of each method with clinical applications.
Collapse
Affiliation(s)
- Maryam Akbarzadeh
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Fathi Maroufi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Pirpour Tazehkand
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Moloud Akbarzadeh
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Cellular and Molecular Biology, Faculty of Biological Science, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Sepideh Bastani
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Safdari
- Department of Health Information Management, School of Allied Medical Science, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Farzane
- Department of Health Information Management, School of Allied Medical Science, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Fattahi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Reza Nejabati
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Samadi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Smith NR, Davies PS, Levin TG, Gallagher AC, Keene DR, Sengupta SK, Wieghard N, El Rassi E, Wong MH. Cell Adhesion Molecule CD166/ALCAM Functions Within the Crypt to Orchestrate Murine Intestinal Stem Cell Homeostasis. Cell Mol Gastroenterol Hepatol 2017; 3:389-409. [PMID: 28462380 PMCID: PMC5404029 DOI: 10.1016/j.jcmgh.2016.12.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 12/04/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS Intestinal epithelial homeostasis is maintained by active-cycling and slow-cycling stem cells confined within an instructive crypt-based niche. Exquisite regulating of these stem cell populations along the proliferation-to-differentiation axis maintains a homeostatic balance to prevent hyperproliferation and cancer. Although recent studies focus on how secreted ligands from mesenchymal and epithelial populations regulate intestinal stem cells (ISCs), it remains unclear what role cell adhesion plays in shaping the regulatory niche. Previously we have shown that the cell adhesion molecule and cancer stem cell marker, CD166/ALCAM (activated leukocyte cell adhesion molecule), is highly expressed by both active-cycling Lgr5+ ISCs and adjacent Paneth cells within the crypt base, supporting the hypothesis that CD166 functions to mediate ISC maintenance and signal coordination. METHODS Here we tested this hypothesis by analyzing a CD166-/- mouse combined with immunohistochemical, flow cytometry, gene expression, and enteroid culture. RESULTS We found that animals lacking CD166 expression harbored fewer active-cycling Lgr5+ ISCs. Homeostasis was maintained by expansion of the transit-amplifying compartment and not by slow-cycling Bmi1+ ISC stimulation. Loss of active-cycling ISCs was coupled with deregulated Paneth cell homeostasis, manifested as increased numbers of immature Paneth progenitors due to decreased terminal differentiation, linked to defective Wnt signaling. CD166-/- Paneth cells expressed reduced Wnt3 ligand expression and depleted nuclear β-catenin. CONCLUSIONS These data support a function for CD166 as an important cell adhesion molecule that shapes the signaling microenvironment by mediating ISC-niche cell interactions. Furthermore, loss of CD166 expression results in decreased ISC and Paneth cell homeostasis and an altered Wnt microenvironment.
Collapse
Key Words
- BrdU, bromodeoxyuridine
- CD166
- CLEM, correlative light and electron microscopy
- FACS, fluorescence-activated cell sorting
- FITC, fluorescein isothiocyanate
- GFP, green fluorescent protein
- HBSS, Hank’s balanced salt solution
- Homeostasis
- IHC, immunohistochemistry
- ISC, intestinal stem cell
- Intestinal Stem Cell
- Lyz, lysozyme
- Muc2, mucin 2
- Paneth Cell
- SEM, standard error of the mean
- Stem Cell Niche
- TA, transit-amplifying
- TEM, transmission electron microscopy
- WT, wild-type
- qRT-PCR, quantitative reverse transcription polymerase chain reaction
Collapse
Affiliation(s)
- Nicholas R. Smith
- Department of Cell, Developmental and Cancer Biology and Oregon Health & Science University, Portland, OR 97239, USA
| | - Paige S. Davies
- Department of Cell, Developmental and Cancer Biology and Oregon Health & Science University, Portland, OR 97239, USA
| | - Trevor G. Levin
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Alexandra C. Gallagher
- Department of Cell, Developmental and Cancer Biology and Oregon Health & Science University, Portland, OR 97239, USA
| | | | - Sidharth K. Sengupta
- Department of Cell, Developmental and Cancer Biology and Oregon Health & Science University, Portland, OR 97239, USA
| | - Nikki Wieghard
- Department of Surgery, Oregon Health & Science University, Portland, Oregon
| | - Edward El Rassi
- Department of Otolaryngology, Oregon Health & Science University, Portland, Oregon
| | - Melissa H. Wong
- Department of Cell, Developmental and Cancer Biology and Oregon Health & Science University, Portland, OR 97239, USA,OHSU Stem Cell Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon,Correspondence Address correspondence to: Melissa H. Wong, PhD, Oregon Health & Science University, Department of Cell, Developmental and Cancer Biology, 3181 SW Sam Jackson Park Road, Mail Code L215, Portland, Oregon 97239. fax: (503) 494-4253.Oregon Health & Science UniversityDepartment of CellDevelopmental and Cancer Biology3181 SW Sam Jackson Park RoadMail Code L215PortlandOregon 97239
| |
Collapse
|
4
|
Tan F, Mosunjac M, Adams AL, Adade B, Taye O, Hu Y, Rizzo M, Ofori-Acquah SF. Enhanced down-regulation of ALCAM/CD166 in African-American Breast Cancer. BMC Cancer 2014; 14:715. [PMID: 25255861 PMCID: PMC4190464 DOI: 10.1186/1471-2407-14-715] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 09/22/2014] [Indexed: 12/31/2022] Open
Abstract
Background Variation in tumor biology in African-American (AA) and Caucasian (CAU) women with breast cancer is poorly defined. Activated leukocyte cell adhesion molecule (ALCAM) is a bad prognostic factor of breast cancer yet it has never being studied in the AA population. We tested the hypothesis that ALCAM expression would be markedly lower in cases of AA breast cancer when compared to CAU. Methods Cases of breast cancer among AA (n = 78) and CAU (n = 95) women were studied. Immunohistochemical staining was used to semi-quantitatively score ALCAM expression in tumor and adjacent non-tumor breast tissues. Clinico-pathological characteristics including histological type, histological grade, tumor size, lymph node metastasis, estrogen receptor (ER), progesterone receptor (PR), and HER2-neu status were abstracted, and their association with ALCAM expression tested. Results Univariate analysis revealed that the level of ALCAM expression at intercellular junctions of primary tumors correlates with histological grade (AA; p = 0.04, CUA; p = 0.02), ER status (AA; p = 0.0004, CAU; p = 0.0015), PR status (AA; p = 0.002, CUA p = 0.034) and triple-negative tumor status (AA; p = 0.0002, CAU; p = 0.0006,) in both ethnic groups. Multivariate analysis demonstrated that ethnicity contribute significantly to ALCAM expression after accounting for basal-like subtype, age, histological grade, tumor size, and lymph node status. Compared to CAU tumors, the AA are 4 times more likely to have low ALCAM expression (p = 0.003). Conclusions Markedly low expression of ALCAM at sites of cell-cell contact in primary breast cancer tumors regardless of differentiation, size and lymph node involvement may contribute to the more aggressive phenotype of breast cancer among AA women.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Solomon F Ofori-Acquah
- Aflac Cancer Center and Blood Disorders Service, Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Drive, Atlanta, GA 30322, USA.
| |
Collapse
|
5
|
CD146, a multi-functional molecule beyond adhesion. Cancer Lett 2012; 330:150-62. [PMID: 23266426 DOI: 10.1016/j.canlet.2012.11.049] [Citation(s) in RCA: 207] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 11/13/2012] [Accepted: 11/28/2012] [Indexed: 02/08/2023]
Abstract
CD146 is a cell adhesion molecule (CAM) that is primarily expressed at the intercellular junction of endothelial cells. CD146 was originally identified as a tumor marker for melanoma (MCAM) due to its existence only in melanoma but not in the corresponding normal counterpart. However CD146 is not just a CAM for the inter-cellular and cell-matrix adhesion. Recent evidence indicates that CD146 is actively involved in miscellaneous processes, such as development, signaling transduction, cell migration, mesenchymal stem cells differentiation, angiogenesis and immune response. CD146 has increasingly become an important molecule, especially identified as a novel bio-marker for angiogenesis and for cancer. Here we have reviewed the dynamic research of CD146, particularly newly identified functions and the underlying mechanisms of CD146.
Collapse
|
6
|
Cloning of the human activated leukocyte cell adhesion molecule promoter and identification of its tissue-independent transcriptional activation by Sp1. Cell Mol Biol Lett 2012; 17:571-85. [PMID: 22941204 PMCID: PMC3683579 DOI: 10.2478/s11658-012-0028-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 08/23/2012] [Indexed: 11/20/2022] Open
Abstract
Activated leukocyte cell adhesion molecule (ALCAM) belongs to the immunoglobulin cell adhesion molecule super family. ALCAM is implicated in tumor progression, inflammation, and the differentiation of hematopoietic stem cells. Hitherto, the identity of regulatory DNA elements and cognate transcription factors responsible for ALCAM gene expression remained unknown. In this report, the human ALCAM promoter was cloned and its transcriptional mechanisms elucidated. The promoter is TATA-less and contains multiple GC-boxes. A proximal 650-bp promoter fragment conferred tissue-independent activation, whereas two contiguous regions upstream of this region negatively influenced promoter activity in a tissue-specific manner. The positive regulatory promoter region was mapped to a core 50 base pair sequence containing a conical Sp1 element. Mutation analysis revealed that this element alone or in tandem with elements immediately upstream was required for maximal promoter activity. Chromatin analysis revealed that Sp1 binds exclusively to the canonical binding sequence in vivo, but not to DNA sequence immediately upstream. Finally, we showed that over-expression of Sp1 significantly increased the basal promoter activity. Thus, Sp1 activated the ALCAM promoter in most cells. These findings have important ramifications for unraveling the roles of ALCAM in inflammation and tumorigenesis.
Collapse
|
7
|
Handharyani E, Tsukamoto M, Tsukamoto Y. Expression of SC1, a cell adhesion molecule, promotes the metastatic activities of the Gallus gallus lymphoblastoid cell line MDCC-MSB1 derived from Marek's disease. Avian Pathol 2011; 40:111-5. [PMID: 21331955 DOI: 10.1080/03079457.2010.541901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
SC1 is an immunoglobulin superfamily cell adhesion molecule purified from the Gallus gallus spinal cord. SC1 is present in embryonic tissues and plays a role in chick development through its cell adhesive property. Interestingly, increased SC1 expression is observed in some sporadic tumours of the chicken, including Marek's disease-induced lymphomas and in nephroblastomas. To elucidate the possible functions of SC1 in tumour progression in the chicken, SC1 cDNA was introduced into the endogenous SC1-negative Marek's disease-derived chicken lymphoblastoid cell line MDCC-MSB1, and subsequently the metastatic potentials of these cell lines were analysed. The in vitro analyses revealed that the SC1-transfected MDCC-MSB1 cells were enhanced in their adhesive and migratory activities in the presence of the SC1 proteins. In addition, the metastatic potential of the SC1-transfected MDCC-MSB1 cells to the lung was enhanced after intravenous implantation into chickens. These findings suggest that the expression of SC1 contributes to the malignancy and metastatic properties of chicken Marek's disease-induced lymphomas.
Collapse
Affiliation(s)
- Ekowati Handharyani
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Bogor Agriculture University, Bogor, Indonesia
| | | | | |
Collapse
|
8
|
Affiliation(s)
- Amanda G Hansen
- Pathology and Cancer Biology, Vanderbilt University, TN 37232, US
| | - Guido W Swart
- FNWI-WiNSt (Faculty of Science, Mathematics & Informatics), Radboud University Nijmegen, 6500 GL, NL
| | - Andries Zijlstra
- Pathology and Cancer Biology, Vanderbilt University, TN 37232, US
| |
Collapse
|
9
|
Tan F, Ghosh S, Mbeunkui F, Thomas R, Weiner JA, Ofori-Acquah SF. Essential role for ALCAM gene silencing in megakaryocytic differentiation of K562 cells. BMC Mol Biol 2010; 11:91. [PMID: 21126364 PMCID: PMC3003670 DOI: 10.1186/1471-2199-11-91] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 12/02/2010] [Indexed: 01/12/2023] Open
Abstract
Background Activated leukocyte cell adhesion molecule (ALCAM/CD166) is expressed by hematopoietic stem cells. However, its role in hematopoietic differentiation has not previously been defined. Results In this study, we show that ALCAM expression is silenced in erythromegakaryocytic progenitor cell lines. In agreement with this finding, the ALCAM promoter is occupied by GATA-1 in vivo, and a cognate motif at -850 inhibited promoter activity in K562 and MEG-01 cells. Gain-of-function studies showed that ALCAM clusters K562 cells in a process that requires PKC. Induction of megakaryocytic differentiation in K562 clones expressing ALCAM activated PKC-δ and triggered apoptosis. Conclusions There is a lineage-specific silencing of ALCAM in bi-potential erythromegakaryocytic progenitor cell lines. Marked apoptosis of ALCAM-expressing K562 clones treated with PMA suggests that aberrant ALCAM expression in erythromegakaryocytic progenitors may contribute to megakaryocytopenia.
Collapse
Affiliation(s)
- Fang Tan
- Department of Pediatrics, Aflac Cancer Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | |
Collapse
|
10
|
Levin TG, Powell AE, Davies PS, Silk AD, Dismuke AD, Anderson EC, Swain JR, Wong MH. Characterization of the intestinal cancer stem cell marker CD166 in the human and mouse gastrointestinal tract. Gastroenterology 2010; 139:2072-2082.e5. [PMID: 20826154 PMCID: PMC2997177 DOI: 10.1053/j.gastro.2010.08.053] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 08/20/2010] [Accepted: 08/26/2010] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS CD166 (also called activated leukocyte cell adhesion molecule [ALCAM]) is a marker of colorectal cancer (CRC) stem cells; it is expressed by aggressive tumors. Although the presence of CD166 at the tumor cell surface has been correlated with shortened survival, little is known about its function and expression in normal intestinal epithelia. METHODS We characterized the expression pattern of CD166 in normal intestinal tissue samples from humans and mice using immunohistochemisty, flow cytometry, and quantitative reverse-transcriptase polymerase chain reaction. Human and mouse intestinal tumors were also analyzed. RESULTS CD166 was expressed on the surface of epithelial cells within the stem cell niche and along the length of the intestine; expression was conserved across species. In the small intestine, CD166 was observed on crypt-based Paneth cells and intervening crypt-based columnar cells (putative stem cells). A subset of CD166-positive, crypt-based columnar cells coexpressed the stem cell markers Lgr5, Musashi-1, or Dcamkl-1. CD166 was located in the cytoplasm and at the surface of cells within human CRC tumors. CD166-positive cells were also detected in benign adenomas in mice; rare cells coexpressed CD166 and CD44 or epithelial-specific antigen. CONCLUSIONS CD166 is highly expressed within the endogenous intestinal stem cell niche. CD166-positive cells appear at multiple stages of intestinal carcinoma progression, including benign and metastatic tumors. Further studies should investigate the function of CD166 in stem cells and the stem cell niche, which might have implications for normal intestinal homeostasis. CD166 has potential as a therapeutic target for CRC.
Collapse
Affiliation(s)
- Trevor G. Levin
- Department of Cell and Developmental Biology, Oregon Health & Science University, Portland, OR
| | - Anne E. Powell
- Department of Cell and Developmental Biology, Oregon Health & Science University, Portland, OR
| | - Paige S. Davies
- Department of Dermatology; Knight Cancer Institute; Oregon Stem Cell Center, Oregon Health & Science University, Portland, OR
| | - Alain D. Silk
- Department of Dermatology; Knight Cancer Institute; Oregon Stem Cell Center, Oregon Health & Science University, Portland, OR
| | - Adria D. Dismuke
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR
| | - Eric C. Anderson
- Department of Internal Medicine, Division of Hematology and Medical Oncology; Oregon Health & Science University, Portland, OR
| | - John R. Swain
- Department of Dermatology; Knight Cancer Institute; Oregon Stem Cell Center, Oregon Health & Science University, Portland, OR
| | - Melissa H. Wong
- Department of Cell and Developmental Biology, Oregon Health & Science University, Portland, OR
,Department of Dermatology; Knight Cancer Institute; Oregon Stem Cell Center, Oregon Health & Science University, Portland, OR
| |
Collapse
|
11
|
Swart GWM. Activated leukocyte cell adhesion molecule (CD166/ALCAM): developmental and mechanistic aspects of cell clustering and cell migration. Eur J Cell Biol 2002; 81:313-21. [PMID: 12113472 DOI: 10.1078/0171-9335-00256] [Citation(s) in RCA: 189] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Activated leukocyte cell adhesion molecule (ALCAM/CD166) is a member of the immunoglobulin superfamily and belongs to a recent subgroup with five extracellular immunoglobulin-like domains (VVC2C2C2). ALCAM mediates both heterophilic (ALCAM-CD6) and homophilic (ALCAM-ALCAM) cell-cell interactions. While expressed in a wide variety of tissues, ALCAM is usually restricted to subsets of cells involved in dynamic growth and/or migration, including neural development, branching organ development, hematopoiesis, immune response and tumor progression. Recent structure-function analyses of ALCAM hint at how its cytoskeletal anchoring and the integrity of the extracellular immunoglobulin-like domains may regulate complex cellular properties in regard to cell adhesion, growth and migration. Accumulating evidence suggests that ALCAM expression may reflect the onset of a cellular program for homeostatic control of growth saturation, which induces either growth arrest or cell migration when the upper limits are exceeded.
Collapse
Affiliation(s)
- Guido W M Swart
- Department of Biochemistry, University of Nijmegen, The Netherlands.
| |
Collapse
|
12
|
Parsons SF, Lee G, Spring FA, Willig TN, Peters LL, Gimm JA, Tanner MJ, Mohandas N, Anstee DJ, Chasis JA. Lutheran blood group glycoprotein and its newly characterized mouse homologue specifically bind alpha5 chain-containing human laminin with high affinity. Blood 2001; 97:312-20. [PMID: 11133776 DOI: 10.1182/blood.v97.1.312] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Lutheran blood group glycoproteins (Lu gps) are receptors for the extracellular matrix protein, laminin. Studies suggest that Lu gps may contribute to vaso-occlusion in sickle cell disease and it has recently been shown that sickle cells adhere to laminin isoforms containing the alpha5 chain (laminin 10/11). Laminin alpha5 is present in the subendothelium and is also a constituent of bone marrow sinusoids, suggesting a role for the Lu/laminin interaction in erythropoiesis. The objectives of the current study were to define more precisely the molecular interactions of the extracellular and intracellular regions of human Lu and to clone and characterize a mouse homologue. To this end, complementary DNA and genomic clones for the mouse homologue were sequenced and the mouse Lu gene mapped to a region on chromosome 7 with conserved synteny with human 19q13.2. Mouse and human Lu gps are highly conserved (72% identity) at the amino acid sequence level and both mouse and human Lu gps specifically bind laminin 10/11 with high affinity. Furthermore, the first 3, N-terminal, immunoglobulin superfamily domains of human Lu are critical for this interaction. The results indicated that the cytoplasmic domain of BRIC 221-labeled human Lu gp is linked with the spectrin-based skeleton, affording the speculation that this interaction may be critical for signal transduction. These results further support a role for Lu gps in sickle cell disease and indicate the utility of mouse models to explore the function of Lu gp-laminin 10/11 interaction in normal erythropoiesis and in sickle cell disease.
Collapse
Affiliation(s)
- S F Parsons
- Bristol Institute for Transfusion Sciences, Bristol, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Weber WT. In vitro characterization of chB6 positive and negative cells from early avian embryos. Cell Immunol 2000; 204:77-87. [PMID: 11069715 DOI: 10.1006/cimm.2000.1702] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Density gradient-separated embryonic spleen and bone marrow cells were enriched for chB6(+) cells with positive and negative selection procedures and magnetic cell sorting. The majority of chB6(+) cells, consisting of small, dense, strongly chB6(+) cells, were prone to apoptosis, which was further accentuated after exposure to monoclonal antibodies directed against chB6 alloantigen, but was largely inhibited by PDBU, leading to maintenance and frequently numerical increases of chB6(+) cells after an initial decline. sIgM(+) cells within that population followed a very similar pattern, suggesting a PDBU-induced upregulation of sIgM expression on a proportion of chB6(+) cells. The protective effect of PDBU on anti-chB6-exposed cells was confirmed with bursal lymphocytes and shown to be entirely PDBU concentration dependent. It was calculated that each 14-day embryonic spleen contained a minimum of 250,000 chB6(+) and 3000-4000 sIgM(+) cells, respectively. Endogenous apoptosis appeared to be increased with embryonic age, reaching a peak with bursal cells in the posthatching period. A second population of larger, less dense, and weak chB6(+) cells, often with vacuoles in a more abundant cytoplasm, differed functionally, expanding numerically in unstimulated cultures and being inhibited by PDBU. No sIgM(+) cells developed within this population. It is proposed that this chB6(+) fraction may represent progenitors of a previously suggested chB6(+) subset of macrophages, in contrast to the dense chB6(+), small cells, viewed as B cell progenitors. chB6(-) cells, consisting predominantly of granulocytes, proliferated vigorously in unstimulated cultures, but were consistently inhibited by PDBU. Coculture of age-matched embryonic bursal stroma with positively and negatively enriched chB6(+) cells revealed enhanced protection from apoptosis for chB6(+) cells and a PDBU-induced upregulation of sIgM expressing cells. Bursal stroma also had a pronounced positive effect on the proliferation of chB6(-) cells.
Collapse
Affiliation(s)
- W T Weber
- Department of Pathobiology, School of Veterinary Medicine, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
14
|
Fraboulet S, Schmidt-Petri T, Dhouailly D, Pourquié O. Expression of DM-GRASP/BEN in the developing mouse spinal cord and various epithelia. Mech Dev 2000; 95:221-4. [PMID: 10906467 DOI: 10.1016/s0925-4773(00)00330-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The expression pattern of the immunoglobulin DM-GRASP/BEN gene was studied in the mouse embryo using in situ hybridization. DM-GRASP/BEN is expressed in the spinal cord in a subset of motoneurons expressing Islet-1, and non homogeneously in the dorsal root ganglia (DRG). In contrast, it's expression is homogeneous in the vestibulo-cochlear and trigemminal ganglia. DM-GRASP/BEN is also expressed in various epithelia of ectodermal or endodermal origin like the nasal, buccopharyngal and lung epithelia. In upper lip, DM-GRASP/BEN transcripts are present in the epidermal cells of the developing hair vibrissa follicles. First detected in the hair placode, DM-GRASP/BEN expression is localized in the central cells of the epithelial hair peg and then in a thin layer of cells crushed against the outer root sheath by the outgrowth of the hair shaft.
Collapse
Affiliation(s)
- S Fraboulet
- Biologie de la Différenciation Epithéliale, UMR CNRS 5538 LEDAC, Institut Albert Bonniot, Université Joseph Fourier, Grenoble, France.
| | | | | | | |
Collapse
|
15
|
Nelissen JM, Torensma R, Pluyter M, Adema GJ, Raymakers RA, van Kooyk Y, Figdor CG. Molecular analysis of the hematopoiesis supporting osteoblastic cell line U2-OS. Exp Hematol 2000; 28:422-32. [PMID: 10781900 DOI: 10.1016/s0301-472x(00)00127-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Osteoblasts play an important role in regulating hematopoiesis in the bone marrow. Here we show that U2-OS, a widely used osteoblastic cell line derived from an osteosarcoma, has the capacity to support proliferation of human hematopoietic progenitor cells in vitro. In this study, U2-OS cells are characterized at the molecular level to unravel the molecular mechanisms underlying the support of hematopoiesis. MATERIALS AND METHODS U2-OS was analyzed in great detail using RT-PCR and flow cytometry. In addition, a cDNA library was constructed and randomly sequenced to obtain insight in the repertoire of expressed molecules. RESULTS A broad panel of growth factors and cytokines is expressed by U2-OS. TGF-beta, GM-CSF, c-kit ligand, and IL-7 are produced constitutively and IL-1beta, IL-6, IL-8, TNF-alpha, IFN-gamma, and MIP1-alpha are upregulated upon stimulation. In addition to those, mRNAs of the CC chemokine LARC and leukemia inhibitory factor were identified. U2-OS cells express high levels of beta1-integrins at the cell surface: VLA-2, VLA-3, VLA-4, VLA-5, VLA-6, and the integrin alphavbeta3. Besides integrins, ALCAM and NCAM are detected on the cell surface of U2-OS. Interestingly, we show that CD34(+) progenitor cells expressing ALCAM are highly proliferative when compared with CD34(+) ALCAM(low) cells, hinting at a role for ALCAM in anchoring progenitor cells to the bone marrow stroma. Interestingly, random sequencing of an U2-OS cDNA library yielded almost 10% of novel cDNAs with a potential role in hematopoiesis. The involvement of these novel molecules in hematopoiesis is an interesting target for future investigations. CONCLUSIONS We conclude that U2-OS supports outgrowth of hematopoietic progenitor cells and accordingly expresses adhesion molecules and growth factors and a number of novel, as yet uncharacterized potentially interesting genes.
Collapse
Affiliation(s)
- J M Nelissen
- Department of Tumor Immunology, University Hospital Nijmegen St. Radboud, Nijmegen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
16
|
Parsons SF, Spring FA, Chasis JA, Anstee DJ. Erythroid cell adhesion molecules Lutheran and LW in health and disease. Best Pract Res Clin Haematol 1999; 12:729-45. [PMID: 10895261 DOI: 10.1053/beha.1999.0050] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The Lutheran and LW glycoproteins are blood group-active proteins found at the surface of human red cells. The Lutheran glycoprotein (Lu gp) is a member of the immunoglobulin superfamily (IgSF) that binds the extracellular matrix protein laminin, in particular, laminin isoforms containing the alpha 5 subunit. The LW glycoprotein (LW gp), also an IgSF member, has substantial sequence homology with the family of intercellular adhesion molecules (ICAMs). LW gp binds the integrin very late antigen-4 (VLA-4, alpha 4 beta 1) and alpha V-containing integrins. Studies on the expression of LW and Lu gps during erythropoiesis utilizing in vitro cultures of haemopoietic progenitor cells have shown that LW gp expression precedes that of Lu gp. These observations have led to the suggestion that LW gp on erythroblasts may interact with VLA-4 on macrophages to stabilize erythroblastic islands in normal bone marrow and that Lu gp may facilitate trafficking of more mature erythroid cells to the sinusoidal endothelium where alpha 5-containing laminins are known to be expressed. Levels of Lu gp and LW gp expression on sickle red cells are greater than on normal red cells and sickle red cells adhere to alpha 5-containing laminins. These data suggest that the Lu and LW molecules may contribute to the vaso-occlusive events associated with episodes of acute pain in sickle cell disease.
Collapse
Affiliation(s)
- S F Parsons
- Bristol Institute for Transfusion Sciences, UK.
| | | | | | | |
Collapse
|
17
|
Gandrillon O, Schmidt U, Beug H, Samarut J. TGF-beta cooperates with TGF-alpha to induce the self-renewal of normal erythrocytic progenitors: evidence for an autocrine mechanism. EMBO J 1999; 18:2764-81. [PMID: 10329623 PMCID: PMC1171358 DOI: 10.1093/emboj/18.10.2764] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Simultaneous addition of both TGF-alpha and TGF-beta induces the sustained, long-term outgrowth of chicken erythrocytic progenitor cells, referred to as T2ECs from both chick bone marrow and 2-day-old chicken embryos. By analysis for differentiation antigens and gene expression, these cells were shown to represent very immature haematopoietic progenitors committed to the erythrocytic lineage. T2ECs differentiate into almost pure populations of fully mature erythrocytes within 6 days, when TGF-alpha and TGF-beta are withdrawn and the cells exposed to anaemic chicken serum plus insulin. Outgrowth of these cells from various sources invariably required both TGF-alpha and TGF-beta, as well as glucocorticoids. Proliferating, established T2ECs still require TGF-alpha, but are independent of exogenous TGF-beta. Using a TGF-beta-neutralizing antibody or expressing a dominant-negative TGF-beta receptor II, we demonstrate that T2ECs generate an autocrine loop involving TGF-beta during their establishment, which is required for sustained proliferation. Using specific inhibitors, we also show that signalling via Mek-1 is specifically required for induction and maintenance of cell proliferation driven by cooperation between the TGF-alpha and -beta receptors. These results establish a novel mechanism by which self-renewal of erythrocytic progenitors is induced and establish avian T2ECs as a new, quasi-optimal model system to study erythrocytic progenitors.
Collapse
Affiliation(s)
- O Gandrillon
- Laboratoire de Biologie Moléculaire et Cellulaire, UMR 5665 CNRS/ENS, Ecole Normale Supérieure de Lyon, 46, allée d'Italie, F-69364 Lyon Cedex 7, France.
| | | | | | | |
Collapse
|
18
|
Abstract
AbstractThe fibrinogen receptor GPIIb-IIIa integrin is known to be expressed on cells of the megakaryocytic lineage, but its presence on hematopoietic progenitors has been a controversial issue. To resolve this ambiguity unequivocally, we performed clonogenic assays and intrathymic cell-transfer experiments in congenic animals. As the ontogeny of the avian hematopoietic system is well documented, we used this experimental model to trace GPIIb-IIIa expression during embryogenesis. Consequently, we now report that the GPIIb-IIIa integrin is expressed as early as embryonic day 3.5 (E3.5) to 4 in intraaortic hematopoietic clusters, the first site of intraembryonic hematopoietic progenitor emergence, and later in E6 paraaortic foci. Myeloid and erythroid progenitors were also detected within the GPIIb-IIIa+ CD45+ population isolated from the E3.5 to 4 aortic area, while in embryonic and adult bone marrow, myeloid, erythroid, and T-cell progenitors were present in the GPIIb-IIIa+ c-kit+ population. Furthermore, we also provide the first evidence, that GPIIb-IIIa+ bone marrow cells can differentiate into T cells. Hence, GPIIb-IIIa can be used as a marker for multilineage hematopoietic progenitors, permitting identification of early intraembryonic sites of hematopoiesis, as well as the isolation of embryonic and adult hematopoietic progenitors.
Collapse
|
19
|
Abstract
The fibrinogen receptor GPIIb-IIIa integrin is known to be expressed on cells of the megakaryocytic lineage, but its presence on hematopoietic progenitors has been a controversial issue. To resolve this ambiguity unequivocally, we performed clonogenic assays and intrathymic cell-transfer experiments in congenic animals. As the ontogeny of the avian hematopoietic system is well documented, we used this experimental model to trace GPIIb-IIIa expression during embryogenesis. Consequently, we now report that the GPIIb-IIIa integrin is expressed as early as embryonic day 3.5 (E3.5) to 4 in intraaortic hematopoietic clusters, the first site of intraembryonic hematopoietic progenitor emergence, and later in E6 paraaortic foci. Myeloid and erythroid progenitors were also detected within the GPIIb-IIIa+ CD45+ population isolated from the E3.5 to 4 aortic area, while in embryonic and adult bone marrow, myeloid, erythroid, and T-cell progenitors were present in the GPIIb-IIIa+ c-kit+ population. Furthermore, we also provide the first evidence, that GPIIb-IIIa+ bone marrow cells can differentiate into T cells. Hence, GPIIb-IIIa can be used as a marker for multilineage hematopoietic progenitors, permitting identification of early intraembryonic sites of hematopoiesis, as well as the isolation of embryonic and adult hematopoietic progenitors.
Collapse
|
20
|
BEN/SC1/DM-GRASP expression during neuromuscular development: a cell adhesion molecule regulated by innervation. J Neurosci 1999. [PMID: 9952415 DOI: 10.1523/jneurosci.19-04-01382.1999] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BEN/SC1/DM-GRASP is a cell adhesion molecule belonging to the Ig superfamily that is transiently expressed during avian embryogenesis in a variety of cell types, including the motoneurons of the spinal cord. We have investigated the pattern of BEN expression during neuromuscular development of the chick. We show that both motoneurons and their target myoblasts express BEN during early embryonic development and that the protein becomes restricted at neuromuscular contacts as soon as postsynaptic acetylcholine receptor clusters are observed in muscle fibers. Muscle cells grown in vitro express and maintain BEN expression even when they fuse and give rise to mature myotubes. When embryos are deprived of innervation by neural tube ablation, BEN expression is observed in muscle fibers, whereas, in control, the protein is already restricted at neuromuscular synaptic sites. These results demonstrate that all myogenic cells intrinsically express BEN and maintain the protein in the absence of innervation. Conversely, when neurons are added to myogenic cultures, BEN is rapidly downregulated in muscle cells, demonstrating that innervation controls the restricted pattern of BEN expression seen in innervated muscles. After nerve section in postnatal muscles, BEN protein becomes again widely spread over muscle fibers. When denervated muscles are allowed to be reinnervated, the protein is reexpressed in regenerating motor axons, and reinnervation of synaptic sites leads to the concentration of BEN at neuromuscular junctions. Our results suggest that BEN cell adhesion molecule acts both in the formation of neuromuscular contacts during development and in the events leading to muscle reinnervation.
Collapse
|
21
|
Eichmann A, Corbel C, Nataf V, Vaigot P, Bréant C, Le Douarin NM. Ligand-dependent development of the endothelial and hemopoietic lineages from embryonic mesodermal cells expressing vascular endothelial growth factor receptor 2. Proc Natl Acad Sci U S A 1997; 94:5141-6. [PMID: 9144204 PMCID: PMC24645 DOI: 10.1073/pnas.94.10.5141] [Citation(s) in RCA: 308] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The existence of a common precursor for endothelial and hemopoietic cells, termed the hemangioblast, has been postulated since the beginning of the century. Recently, deletion of the endothelial-specific vascular endothelial growth factor receptor 2 (VEGFR2) by gene targeting has shown that both endothelial and hemopoietic cells are absent in homozygous null mice. This observation suggested that VEGFR2 could be expressed by the hemangioblast and essential for its further differentiation along both lineages. However, it was not possible to exclude the hypothesis that hemopoietic failure was a secondary effect resulting from the absence of an endothelial cell microenvironment. To distinguish between these two hypotheses, we have produced a mAb directed against the extracellular domain of avian VEGFR2 and isolated VEGFR2+ cells from the mesoderm of chicken embryos at the gastrulation stage. We have found that in clonal cultures, a VEGFR2+ cell gives rise to either a hemopoietic or an endothelial cell colony. The developmental decision appears to be regulated by the binding of two different VEGFR2 ligands. Thus, endothelial differentiation requires VEGF, whereas hemopoietic differentiation occurs in the absence of VEGF and is significantly reduced by soluble VEGFR2, showing that this process could be mediated by a second, yet unidentified, VEGFR2 ligand. These observations thus suggest strongly that in the absence of the VEGFR2 gene product, the precursors of both hemopoietic and vascular endothelial lineages cannot survive. These cells therefore might be the initial targets of the VEGFR2 null mutation.
Collapse
Affiliation(s)
- A Eichmann
- Institut d'Embryologie Cellulaire et Moléculaire du Centre National de la Recherche Scientifique et du Collège de France, 49bis Avenue de la Belle Gabrielle, 94736 Nogent-sur-Marne, Cedex, France
| | | | | | | | | | | |
Collapse
|
22
|
The Characterization, Molecular Cloning, and Expression of a Novel Hematopoietic Cell Antigen From CD34+ Human Bone Marrow Cells. Blood 1997. [DOI: 10.1182/blood.v89.8.2706] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
The adhesion molecule BEN/SC1/DM-GRASP (BEN) is a marker in the developing chicken nervous system that is also expressed on the surface of embryonic and adult hematopoietic cells such as immature thymocytes, myeloid progenitors, and erythroid progenitors. F84.1 and KG-CAM, two monoclonal antibodies to rat neuronal glycoproteins with similarity to BEN, cross-react with an antigen on rat hematopoietic progenitors, but F84.1 only also recognizes human blood cell progenitors. We have defined the antigen recognized by F84.1 as the hematopoietic cell antigen (HCA). HCA expression was detected on 40% to 70% of CD34+ fetal and adult bone marrow cells and mobilized peripheral blood cells. Precursor cell activity for long-term in vitro bone marrow cell culture was confined to the subset of CD34+ cells that coexpress HCA. HCA is expressed by the most primitive subsets of CD34+ cells, including all rhodamine 123lo, Thy-1+, and CD38−/lo CD34+ adult bone marrow cells. HCA was also detected on myeloid progenitors but not on early B-cell progenitors. We also describe here the cloning and characterization of cDNAs encoding two variants of the human HCA antigen (huHCA-1 and huHCA-2) and of a cDNA clone encoding rat HCA (raHCA). The deduced amino acid sequences of huHCA and raHCA are homologous to that of chicken BEN. Recombinant proteins produced from either human or rat HCA cDNAs were recognized by F84.1, whereas rat HCA but not human HCA was recognized by antirat KG-CAM. Expression of either form of huHCA in CHO cells conferred homophilic adhesion that could be competed with soluble recombinant huHCA-Fc. The molecular cloning of HCA and the availability of recombinant HCA should permit further evaluation of its role in human and rodent hematopoiesis.
Collapse
|
23
|
Karagogeos D, Pourquié C, Kyriakopoulou K, Tavian M, Stallcup W, Péault B, Pourquié O. Expression of the cell adhesion proteins BEN/SC1/DM-GRASP and TAG-1 defines early steps of axonogenesis in the human spinal cord. J Comp Neurol 1997; 379:415-27. [PMID: 9067833 DOI: 10.1002/(sici)1096-9861(19970317)379:3<415::aid-cne7>3.0.co;2-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We have studied the expression pattern of two cell adhesion proteins of the immunoglobin (Ig) superfamily, BEN/SC1/DM-GRASP (BEN) and the transient axonal glycoprotein TAG-1, during the development of the human nervous system. This study was performed by immunocytochemistry on sections of human embryos ranging from 4 to 13 weeks postconception. The overall distribution of the two proteins during development is very similar to that reported in other vertebrate species, but several important differences have been observed. Both proteins exhibit a transient expression on selected neuronal populations, which include the motor and the sensory neurons. In addition, BEN was also detected on virtually all neurons derived from the neural crest as well as in nonneuronal tissues. A major difference of expression with the chick embryo is that, in the motor neurons, BEN expression was not observed at early stages of development, thus arguing against a role of this molecule in pathfinding and fasciculation. BEN was observed to be restricted to subsets of motor neurons, such as the medial column at the upper limb level. Expression was also detected in a laterodorsal population of the ventral horn cells, which are likely to correspond to migrating preganglionic neurons that originate from the motor pool at the thoracic level. TAG-1 was found on commissural neurons and weakly on the sympathetic neurons; it was also detected on restricted nonneuronal populations. In addition, we observed TAG-1 expression in fibers that could correspond either to subsets of dorsal root ganglia (DRGs) central afferences (including the Ia fibers) or to the axons of association interneurons and in scattered motoneurons likely to correspond either to preganglionic neurons, to gamma-motoneurons, or to late-born motoneurons. Therefore, our results indicate that the molecular strategies used to establish the axonal scaffolding of the nervous system in humans are extremely conserved among the different vertebrates.
Collapse
Affiliation(s)
- D Karagogeos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, University of Crete School of Medicine, Greece
| | | | | | | | | | | | | |
Collapse
|
24
|
Vainio O, Dunon D, Aïssi F, Dangy JP, McNagny KM, Imhof BA. HEMCAM, an adhesion molecule expressed by c-kit+ hemopoietic progenitors. J Cell Biol 1996; 135:1655-68. [PMID: 8978830 PMCID: PMC2133972 DOI: 10.1083/jcb.135.6.1655] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
We have characterized the adhesion molecule HEMCAM, which is expressed by hemopoietic progenitors of embryonic bone marrow. HEMCAM belongs to the immunoglobulin superfamily and consists of the V-V-C2-C2-C2 Ig domains. There are three mRNA splice variants. One has a short cytoplasmic tail; another has a long tail; while the third seems to lack transmembrane and cytoplasmic regions. Except for the NH2-terminal sequence, HEMCAM is identical to gicerin, a molecular involved in neurite outgrowth and Wilm's kidney tumor progression in the chicken and it is significantly homologous with MUC18 a molecule involved in melanoma progression and metastasis in human beings. In the bone marrow the HEMCAM+ cell population contains c-kit+ subsets. HEMCAM+ cells coexpressing the receptor tyrosine kinase c-kit give rise to T cells at a frequency of 0.17 when injected intrathymically in congenic animals. As HEMCAM+, c-kit+ cells differentiate into myeloid and erythroid CFU's the double-positive cell population seems to contain precursors for multiple lineages. HEMCAM promotes cell-cell adhesion of transfected cells. Cross-linking of murine HEMCAM leads to cell spreading of T-lymphocyte progenitors adhering to the vascular adhesion molecules, PECAM-1 and VCAM-1. Thus, HEMCAM is likely to be involved in cellular adhesion and homing processes.
Collapse
Affiliation(s)
- O Vainio
- Basel Institute for Immunology, Switzerland
| | | | | | | | | | | |
Collapse
|
25
|
Corbel C, Pourquié O, Cormier F, Vaigot P, Le Douarin NM. BEN/SC1/DM-GRASP, a homophilic adhesion molecule, is required for in vitro myeloid colony formation by avian hemopoietic progenitors. Proc Natl Acad Sci U S A 1996; 93:2844-9. [PMID: 8610129 PMCID: PMC39721 DOI: 10.1073/pnas.93.7.2844] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
BEN/SC1/DM-GRASP is a membrane glycoprotein of the immunoglobulin superfamily isolated in the chick by several groups, including ours. Its expression is strictly developmentally regulated in several cell types of the nervous and hemopoietic systems and in certain epithelia. Each of these cell types expresses isoforms of BEN which differ by their level of N-glycosylation and by the presence or absence of the HNK-1 carbohydrate epitope. In the present work, the influence of glycosylation on BEN homophilic binding properties was investigated by two in vitro assays. First, each BEN isoform was covalently coupled to microspheres carrying different fluorescent dyes and an aggregation test was performed. We found that homophilic aggregates form indifferently between the same or different BEN isoforms, showing that glycosylation does not affect BEN homophilic binding properties. This was confirmed in the second test, where the BEN-coated microspheres bound to the neurites of BEN- expressing neurons, irrespective of the isoform considered. The transient expression of the BEN antigen on hemopoietic progenitors prompted us to see whether it might play a role in their proliferation and differentiation. When added to hemopoietic progenitor cells in an in vitro colony formation assay anti-BEN immunoglobulin strongly inhibited myeloid, but not erythroid, colony formation although both types of precursors express the molecule.
Collapse
Affiliation(s)
- C Corbel
- Insitut d'Embryologic Cellulaire et Moléculaire du Centre National de Recherche Scientifique, Nogent-sur-Marne, France
| | | | | | | | | |
Collapse
|
26
|
Nicolas-Bolnet C, Johnston PA, Kemper AE, Ricks C, Petitte JN. Synergistic action of two sources of avian growth factors on proliferative differentiation of chick embryonic hematopoietic cells. Poult Sci 1995; 74:1102-16. [PMID: 7479487 DOI: 10.3382/ps.0741102] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
During embryonic development, the components of the avian immune system undergo ontogeny in several distinct organs, including the bone marrow, spleen, thymus, and bursa of Fabricius. This process is regulated and controlled by the complex interactions of various cytokines and colony-stimulating factors (CSF). The objective was to examine the action of two different sources of hematopoietic growth factors, spleen-conditioned media (SCM) and chick embryo extract (CEE), on the proliferation of hematopoietic cells from various organs and on the differentiation of progenitor cells in semi-solid culture. Spleen and bone marrow cells obtained at Day 16 of incubation responded in a dose-dependent manner to the addition of SCM and CEE alone or in combination. No proliferative effect of SCM was observed on cells obtained from embryonic thymus or bursa. Clonal analysis of bone marrow and spleen cells suggested that CEE may contain the avian equivalents of stem cell factor, interleukin-3, granulocyte-macrophage CSF, granulocyte-CSF, and macrophage-CSF. Clonal analysis of SCM cultures suggested that in addition to myelomonocytic growth factor, which affects primarily macrophage-granulocyte lineages, a thrombocyte-CSF-like activity was also apparent. The SCM alone tended to act upon committed late progenitors. The combination of CEE and SCM amplified the size and the total number of colonies obtained and appeared to act synergistically upon progenitors with a high level of proliferative potential. This response on young progenitors was confirmed when cells were cultured in CEE and SCM prior to clonal analysis. These results document the presence of thrombocyte CSF in SCM and the effect of both CEE and SCM on the proliferative differentiation of avian embryonic hematopoietic progenitors.
Collapse
Affiliation(s)
- C Nicolas-Bolnet
- Department of Poultry Science, North Carolina State University, Raleigh 27695-7608, USA
| | | | | | | | | |
Collapse
|
27
|
Zhang G, Slaughter C, Humphries EH. v-rel Induces ectopic expression of an adhesion molecule, DM-GRASP, during B-lymphoma development. Mol Cell Biol 1995; 15:1806-16. [PMID: 7862170 PMCID: PMC230405 DOI: 10.1128/mcb.15.3.1806] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
In an effort to identify aberrantly expressed genes in v-rel-induced tumors, monoclonal antibodies were developed that reacted selectively with avian B-cell tumors. One antibody, HY78, immunoprecipitated a 120-kDa glycoprotein (p120) from cells that express v-rel. N-terminal amino acid sequencing of p120 identified a 27-amino-acid sequence that is also present in DM-GRASP, an adhesion molecule belonging to the immunoglobulin superfamily. Evidence from tissue distribution, immunological cross-reaction, PCR amplification, cDNA cloning, and DNA sequence shows that p120 is indeed DM-GRASP. Northern (RNA) analysis using a probe from the DM-GRASP gene identified a 5.3-kb transcript in mRNA from bursa, thymus, and brain as well as from v-rel-induced B-cell lymphomas but not from bursal B cells. The induction of this protein by v-rel during the development of bursal B-cell lymphomas appears, therefore, to be ectopic in nature. Overexpression of v-rel or c-rel in chicken embryonic fibroblasts, B-cell lines, and spleen mononuclear cells induces the expression of DM-GRASP. The ratio of DM-GRASP to v-Rel was fivefold higher than that of DM-GRASP/c-Rel in a B-cell line, DT95. Interestingly, the presence of HY78 antibody inhibits the in vitro proliferation of v-rel-transformed cells but not cells that immortalized by myc. These data suggest that DM-GRASP is one of the genes induced during v-rel-mediated tumor development and that DM-GRASP may be involved in the growth of v-rel tumor cells.
Collapse
MESH Headings
- Activated-Leukocyte Cell Adhesion Molecule
- Amino Acid Sequence
- Animals
- Antibodies
- Antibodies, Monoclonal
- B-Lymphocytes/metabolism
- Base Sequence
- Blotting, Western
- Brain/immunology
- Brain/metabolism
- Cell Adhesion Molecules, Neuronal/biosynthesis
- Cell Adhesion Molecules, Neuronal/isolation & purification
- Cell Line
- Chick Embryo
- DNA Primers
- Epithelium/immunology
- Epithelium/metabolism
- Extracellular Matrix Proteins/biosynthesis
- Extracellular Matrix Proteins/isolation & purification
- Gene Library
- Glycoproteins/biosynthesis
- Glycoproteins/isolation & purification
- Humans
- Lymphoid Tissue/immunology
- Lymphoid Tissue/metabolism
- Lymphoma, B-Cell/immunology
- Mice
- Mice, Inbred BALB C
- Nerve Tissue Proteins/biosynthesis
- Nerve Tissue Proteins/isolation & purification
- Oncogenes
- Organ Specificity
- Polymerase Chain Reaction
- RNA, Messenger/biosynthesis
- Restriction Mapping
- Sequence Homology, Amino Acid
- Transcription, Genetic
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- G Zhang
- Mary Babb Randolph Cancer Center, West Virginia University, Morgantown 26506-9177
| | | | | |
Collapse
|
28
|
Cormier F. Avian pluripotent haemopoietic progenitor cells: detection and enrichment from the para-aortic region of the early embryo. J Cell Sci 1993; 105 ( Pt 3):661-6. [PMID: 8408294 DOI: 10.1242/jcs.105.3.661] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In the avian embryo, the wall of the aorta is a site where haemopoiesis occurs in large diffuse foci from day 3 to day 10. In contrast to haemopoiesis in other organs of the embryo, para-aortic haemopoiesis is sustained by stem cells, which emerge in situ. Previous studies have demonstrated that the para-aortic region from the day-4 chick embryo harbours committed myeloid progenitors and committed erythroid progenitors. The present paper reports the in vitro development of para-aortic progenitors with both myelomonocytic and erythroid potentialities. Three types of myelo-erythroid progenitors were observed, giving rise to erythroblasts and monocytes, to erythroblasts and granulocytes, or to erythroblasts, monocytes and granulocytes. Their frequency in the para-aortic cell suspension was 1 per 10,000 cells. In cell sorting experiments, they co-sorted with committed progenitors in the cell population that immunolabeled with the VI-A2 monoclonal antibody, which is specific for chicken haemopoietic cells. Cell sorting also demonstrated that these multipotential progenitors did not express the BEN cell surface molecule, in contrast to late myeloid progenitors. The BEN molecule belongs to the immunoglobulin superfamily and is expressed by haemopoietic progenitors from bone marrow, selective sets of neurons and epithelial cells from the bursa of Fabricius. The myelo-erythroid progenitors were enriched 4 times in the VI-A2-positive cell population, and 2 to 5 times in the BEN-negative population. These results represent the first in vitro demonstration of avian normal myelo-erythroid progneitors.
Collapse
Affiliation(s)
- F Cormier
- Institut d'Embryologie Cellulaire et Moléculaire du CNRS et du Collège de France, Nogent-sur-Marne
| |
Collapse
|