1
|
Urasawa T, Kawasaki N. Proteomic Approach Using DIA-MS Identifies Morphogenesis-Associated Proteins during Cardiac Differentiation of Human iPS Cells. ACS OMEGA 2025; 10:344-357. [PMID: 39829588 PMCID: PMC11740111 DOI: 10.1021/acsomega.4c06371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025]
Abstract
Human-induced pluripotent stem cell (hiPSC)-derived cardiomyocytes have potential applications in regenerative medicine. The quality by design (QbD) approach enables the efficiency and quality assurance in the manufacturing of hiPSC-derived products. It requires a molecular understanding of hiPSC differentiation throughout the differentiation process; however, information on cardiac differentiation remains limited. Proteins associated with the early stages of cardiac differentiation would be useful in the cardiomyocyte quality assessment. Here, we performed quantitative proteomics of hiPSC intermediate cells in the early phase of cardiac differentiation to better understand their molecular characteristics. Proteomic profiles suggested that day 5-7 cells were in the morphogenetic stage of cardiac differentiation. Trophoblast glycoprotein (TPBG) was the most up-regulated protein in the morphogenetic stage; it was previously shown to be up-regulated during differentiation into neural stem cells. Proteomics of TPBG-knockdown cells revealed that TPBG is involved in cell proliferation and is related to the cardiomyocyte yield, suggesting that it could be used as a marker in QbD development. Our approach helps us understand the molecular basis of hiPSC differentiation and could be a powerful tool in QbD-based manufacturing.
Collapse
Affiliation(s)
- Takaya Urasawa
- Biopharmaceutical and Regenerative
Sciences, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Nana Kawasaki
- Biopharmaceutical and Regenerative
Sciences, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| |
Collapse
|
2
|
Sun L, Shi M, Wang J, Han X, Wei J, Huang Z, Yang X, Ding Y, Zhang P, He A, Liu M, Yan R, Yang X, Li R, Wang G. Overexpressed Trophoblast Glycoprotein Contributes to Preeclampsia Development by Inducing Abnormal Trophoblast Migration and Invasion Toward the Uterine Spiral Artery. Hypertension 2024; 81:1524-1536. [PMID: 38716674 DOI: 10.1161/hypertensionaha.124.22923] [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: 02/21/2024] [Accepted: 04/22/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Preeclampsia is a significant pregnancy disorder with an unknown cause, mainly attributed to impaired spiral arterial remodeling. METHODS Using RNA sequencing, we identified key genes in placental tissues from healthy individuals and preeclampsia patients. Placenta and plasma samples from pregnant women were collected to detect the expression of TPBG (trophoblast glycoprotein). Pregnant rats were injected with TPBG-carrying adenovirus to detect preeclamptic features. HTR-8/SVneo cells transfected with a TPBG overexpression lentiviral vector were used in cell function experiments. The downstream molecular mechanisms of TPBG were explored using RNA sequencing and single-cell RNA sequencing data. TPBG expression was knocked down in the lipopolysaccharide-induced preeclampsia-like rat model to rescue the preeclampsia features. We also assessed TPBG's potential as an early preeclampsia predictor using clinical plasma samples. RESULTS TPBG emerged as a crucial differentially expressed gene, expressed specifically in syncytiotrophoblasts and extravillous trophoblasts. Subsequently, we established a rat model with preeclampsia-like phenotypes by intravenously injecting TPBG-expressing adenoviruses, observing impaired spiral arterial remodeling, thus indicating a causal correlation between TPBG overexpression and preeclampsia. Studies with HTR-8/SVneo cells, chorionic villous explants, and transwell assays showed TPBG overexpression disrupts trophoblast/extravillous trophoblast migration/invasion and chemotaxis. Notably, TPBG knockdown alleviated the lipopolysaccharide-induced preeclampsia-like rat model. We enhanced preeclampsia risk prediction in early gestation by combining TPBG expression with established clinical predictors. CONCLUSIONS These findings are the first to show that TPBG overexpression contributes to preeclampsia development by affecting uterine spiral artery remodeling. We propose TPBG levels in maternal blood as a predictor of preeclampsia risk. The proposed mechanism by which TPBG overexpression contributes to the occurrence of preeclampsia via its disruptive effect on trophoblast and extravillous trophoblast migration/invasion on uterine spiral artery remodeling, thereby increasing the risk of preeclampsia.
Collapse
Affiliation(s)
- Lu Sun
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Meiting Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Jian Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Xiaoxue Han
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Jiachun Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Zhengrui Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Xiaofeng Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine (P.Z., X.Y., G.W.), Jinan University, Guangzhou, China
| | - Yuzhen Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Ping Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine (P.Z., X.Y., G.W.), Jinan University, Guangzhou, China
| | - Andong He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Mengyuan Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Ruiling Yan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Xuesong Yang
- Key Laboratory for Regenerative Medicine of the Ministry of Education (X.Y., G.W.), Jinan University, Guangzhou, China
- Clinical Research Center, Clifford Hospital, Guangzhou, China (X.Y.)
| | - Ruiman Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine (P.Z., X.Y., G.W.), Jinan University, Guangzhou, China
- Key Laboratory for Regenerative Medicine of the Ministry of Education (X.Y., G.W.), Jinan University, Guangzhou, China
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, School of Medicine (G.W.), Jinan University, Guangzhou, China
| |
Collapse
|
3
|
Trophoblast glycoprotein is a new candidate gene for Parkinson’s disease. NPJ Parkinsons Dis 2021; 7:110. [PMID: 34876581 PMCID: PMC8651753 DOI: 10.1038/s41531-021-00252-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 11/04/2021] [Indexed: 11/08/2022] Open
Abstract
AbstractParkinson’s disease (PD) is a movement disorder caused by progressive degeneration of the midbrain dopaminergic (mDA) neurons in the substantia nigra pars compacta (SNc). Despite intense research efforts over the past decades, the etiology of PD remains largely unknown. Here, we discovered the involvement of trophoblast glycoprotein (Tpbg) in the development of PD-like phenotypes in mice. Tpbg expression was detected in the ventral midbrain during embryonic development and in mDA neurons in adulthood. Genetic ablation of Tpbg resulted in mild degeneration of mDA neurons in aged mice (12–14 months) with behavioral deficits reminiscent of PD symptoms. Through in silico analysis, we predicted potential TPBG-interacting partners whose functions were relevant to PD pathogenesis; this result was substantiated by transcriptomic analysis of the SNc of aged Tpbg knockout mice. These findings suggest that Tpbg is a new candidate gene associated with PD and provide a new insight into PD pathogenesis.
Collapse
|
4
|
Takahashi Y, Takahashi H, Stern PL, Kirita T, Tsuboi A. Expression of Oncofetal Antigen 5T4 in Murine Taste Papillae. Front Cell Neurosci 2019; 13:343. [PMID: 31417363 PMCID: PMC6685444 DOI: 10.3389/fncel.2019.00343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/12/2019] [Indexed: 12/26/2022] Open
Abstract
Background: Multicellular taste buds located within taste papillae on the tongue mediate taste sensation. In taste papillae, taste bud cells (TBCs), such as taste receptor cells and taste precursor cells, and the surrounding lingual epithelium including epithelial progenitors (also called taste stem/progenitor cells) are maintained by continuous cell turnover throughout life. However, it remains unknown how the cells constituting taste buds proliferate and differentiate to maintain taste bud tissue. Based on in situ hybridization (ISH) screening, we demonstrated that the oncofetal antigen 5T4 (also known as trophoblast glycoprotein: TPBG) gene is expressed in the adult mouse tongue. Results: In immunohistochemistry of coronal tongue sections, 5T4 protein was detected at a low level exclusively in the basal part of the lingual epithelium in developing and adult mice, and at a high level particularly in foliate papillae and circumvallate papillae (CVPs). Furthermore, immunohistochemistry of the basal part of CVPs indicated that the proliferation marker PCNA (proliferating cell nuclear antigen) co-localized with 5T4. 5T4 was strongly expressed in Krt5+ epithelial progenitors and Shh+ taste precursor cells, but weakly in mature taste receptor cells. The number of proliferating cells in the CVP was higher in 5T4-knockout mice than in wild-type (WT) mice, while neither cell differentiation nor the size of taste buds differed between these two groups of mice. Notably, X-ray irradiation enhanced cell proliferation more in 5T4-knockout mice than in WT mice. Conclusion: Our results suggest that 5T4, expressed in epithelial progenitors (taste stem/progenitor cells), and taste precursor cells, may influence the maintenance of taste papillae under both normal and injury conditions.
Collapse
Affiliation(s)
- Yuka Takahashi
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Hiroo Takahashi
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho, Japan
| | - Peter L. Stern
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Tadaaki Kirita
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Akio Tsuboi
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
- Laboratory for Cellular and Molecular Neurobiology, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
5
|
Wakeham CM, Wilmarth PA, Cunliffe JM, Klimek JE, Ren G, David LL, Morgans CW. Identification of PKCα-dependent phosphoproteins in mouse retina. J Proteomics 2019; 206:103423. [PMID: 31255707 DOI: 10.1016/j.jprot.2019.103423] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/11/2019] [Accepted: 06/19/2019] [Indexed: 12/11/2022]
Abstract
Adjusting to a wide range of light intensities is an essential feature of retinal rod bipolar cell (RBC) function. While persuasive evidence suggests this modulation involves phosphorylation by protein kinase C-alpha (PKCα), the targets of PKCα phosphorylation in the retina have not been identified. PKCα activity and phosphorylation in RBCs was examined by immunofluorescence confocal microscopy using a conformation-specific PKCα antibody and antibodies to phosphorylated PKC motifs. PKCα activity was dependent on light and expression of TRPM1, and RBC dendrites were the primary sites of light-dependent phosphorylation. PKCα-dependent retinal phosphoproteins were identified using a phosphoproteomics approach to compare total protein and phosphopeptide abundance between phorbol ester-treated wild type and PKCα knockout (PKCα-KO) mouse retinas. Phosphopeptide mass spectrometry identified over 1100 phosphopeptides in mouse retina, with 12 displaying significantly greater phosphorylation in WT compared to PKCα-KO samples. The differentially phosphorylated proteins fall into the following functional groups: cytoskeleton/trafficking (4 proteins), ECM/adhesion (2 proteins), signaling (2 proteins), transcriptional regulation (3 proteins), and homeostasis/metabolism (1 protein). Two strongly differentially expressed phosphoproteins, BORG4 and TPBG, were localized to the synaptic layers of the retina, and may play a role in PKCα-dependent modulation of RBC physiology. Data are available via ProteomeXchange with identifier PXD012906. SIGNIFICANCE: Retinal rod bipolar cells (RBCs), the second-order neurons of the mammalian rod visual pathway, are able to modulate their sensitivity to remain functional across a wide range of light intensities, from starlight to daylight. Evidence suggests that this modulation requires the serine/threonine kinase, PKCα, though the specific mechanism by which PKCα modulates RBC physiology is unknown. This study examined PKCα phosophorylation patterns in mouse rod bipolar cells and then used a phosphoproteomics approach to identify PKCα-dependent phosphoproteins in the mouse retina. A small number of retinal proteins showed significant PKCα-dependent phosphorylation, including BORG4 and TPBG, suggesting a potential contribution to PKCα-dependent modulation of RBC physiology.
Collapse
Affiliation(s)
- Colin M Wakeham
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Phillip A Wilmarth
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jennifer M Cunliffe
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR 97239, USA
| | - John E Klimek
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR 97239, USA
| | - Gaoying Ren
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Larry L David
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR 97239, USA; Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Catherine W Morgans
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
6
|
Role of TPBG (Trophoblast Glycoprotein) Antigen in Human Pericyte Migratory and Angiogenic Activity. Arterioscler Thromb Vasc Biol 2019; 39:1113-1124. [DOI: 10.1161/atvbaha.119.312665] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objective—
To determine the role of the oncofetal protein TPBG (trophoblast glycoprotein) in normal vascular function and reparative vascularization.
Approach and Results—
Immunohistochemistry of human veins was used to show TPBG expression in vascular smooth muscle cells and adventitial pericyte-like cells (APCs). ELISA, Western blot, immunocytochemistry, and proximity ligation assays evidenced a hypoxia-dependent upregulation of TPBG in APCs not found in vascular smooth muscle cells or endothelial cells. This involves the transcriptional modulator CITED2 (Atypical chemokine receptor 3 CBP/p300-interacting transactivator with glutamic acid (E)/aspartic acid (D)-rich tail) and downstream activation of CXCL12 (chemokine [C-X-C motif] ligand-12) signaling through the CXCR7 (C-X-C chemokine receptor type 7) receptor and ERK1/2 (extracellular signal-regulated kinases 1/2). TPBG silencing by siRNA transfection downregulated CXCL12, CXCR7, and pERK (phospho Thr202/Tyr204 ERK1/2) and reduced the APC migratory and proangiogenic capacities. TPBG forced expression induced opposite effects, which were associated with the formation of CXCR7/CXCR4 (C-X-C chemokine receptor type 4) heterodimers and could be contrasted by CXCL12 and CXCR7 neutralization. In vivo Matrigel plug assays using APCs with or without TPBG silencing evidenced TPBG is essential for angiogenesis. Finally, in immunosuppressed mice with limb ischemia, intramuscular injection of TPBG-overexpressing APCs surpassed naïve APCs in enhancing perfusion recovery and reducing the rate of toe necrosis.
Conclusions—
TPBG orchestrates the migratory and angiogenic activities of pericytes through the activation of the CXCL12/CXCR7/pERK axis. This novel mechanism could be a relevant target for therapeutic improvement of reparative angiogenesis.
Collapse
|
7
|
Harrop R, O’Neill E, Stern PL. Cancer stem cell mobilization and therapeutic targeting of the 5T4 oncofetal antigen. Ther Adv Vaccines Immunother 2019; 7:2515135518821623. [PMID: 30719508 PMCID: PMC6348545 DOI: 10.1177/2515135518821623] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/27/2018] [Indexed: 12/21/2022] Open
Abstract
Cancer stem cells (CSCs) can act as the cellular drivers of tumors harnessing stem cell properties that contribute to tumorigenesis either as founder elements or by the gain of stem cell traits by the malignant cells. Thus, CSCs can self-renew and generate the cellular heterogeneity of tumors including a hierarchical organization similar to the normal tissue. While the principle tumor growth contribution is often from the non-CSC components, it is the ability of small numbers of CSCs to avoid the effects of therapeutic strategies that can contribute to recurrence after treatment. However, identifying and characterizing CSCs for therapeutic targeting is made more challenging by their cellular potency being influenced by a particular tissue niche or by the capacity of more committed cells to regain stem cell functions. This review discusses the properties of CSCs including the limitations of the available cell surface markers, the assays that document tumor initiation and clonogenicity, the roles of epithelial mesenchymal transition and molecular pathways such as Notch, Wnt, Hippo and Hedgehog. The ability to target and eliminate CSCs is thought to be critical in the search for curative cancer treatments. The oncofetal tumor-associated antigen 5T4 (TBGP) has been linked with CSC properties in several different malignancies. 5T4 has functional attributes that are relevant to the spread of tumors including through EMT, CXCR4/CXCL12, Wnt, and Hippo pathways which may all contribute through the mobilization of CSCs. There are several different immunotherapies targeting 5T4 in development including antibody-drug conjugates, antibody-targeted bacterial super-antigens, a Modified Vaccinia Ankara-basedvaccine and 5T4-directed chimeric antigen receptor T-cells. These immune therapies would have the advantage of targeting both the bulk tumor as well as mobilized CSC populations.
Collapse
Affiliation(s)
- Richard Harrop
- Oxford BioMedica plc, Windrush Court, Transport Way, Oxford, OX4 6LT, UK
| | - Eric O’Neill
- Department of Oncology, University of Oxford, Oxford, UK
| | - Peter L. Stern
- Division of Molecular & Clinical Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| |
Collapse
|
8
|
Harris JL, Dave K, Gorman J, Khanna KK. The breast cancer antigen 5T4 interacts with Rab11, and is a target and regulator of Rab11 mediated trafficking. Int J Biochem Cell Biol 2018; 99:28-37. [DOI: 10.1016/j.biocel.2018.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 01/19/2023]
|
9
|
Stern PL, Harrop R. 5T4 oncofoetal antigen: an attractive target for immune intervention in cancer. Cancer Immunol Immunother 2017; 66:415-426. [PMID: 27757559 PMCID: PMC11029567 DOI: 10.1007/s00262-016-1917-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 10/12/2016] [Indexed: 01/18/2023]
Abstract
The natural history of a patient's cancer is often characterised by genetic diversity and sequential sweeps of clonal dominance. It is therefore not surprising that identifying the most appropriate tumour-associated antigen for targeted intervention is challenging. The 5T4 oncofoetal antigen was identified by searching for surface molecules shared between human trophoblast and cancer cells with the rationale that they may function to allow survival of the foetus as a semi-allograft in the mother or a tumour in its host. The 5T4 protein is expressed by many different cancers but rarely in normal adult tissues. 5T4 molecules are 72 kD, heavily N-glycosylated proteins with several leucine-rich repeats which are often associated with protein-protein interactions. 5T4 expression is associated with the directional movement of cells through epithelial mesenchymal transition, potentiation of CXCL12/CXCR4 chemotaxis and inhibition of canonical Wnt/beta-catenin while favouring non-canonical pathway signalling; all processes which help drive the spread of cancer cells. The selective pattern of 5T4 tumour expression, association with a tumour-initiating phenotype plus a mechanistic involvement with cancer spread have underwritten the clinical development of different immunotherapeutic strategies including a vaccine, a tumour-targeted superantigen and an antibody drug conjugate. In addition, a chimeric antigen receptor T cell approach targeting 5T4 expressing tumour cells is in pre-clinical development. A key challenge will include how best to combine each 5T4 targeted immunotherapy with the most appropriate standard of care treatment (or adjunct therapy) to maximise the recovery of immune control and ultimately eliminate the tumour.
Collapse
Affiliation(s)
- Peter L Stern
- Institute of Cancer Studies, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK
| | - Richard Harrop
- Oxford BioMedica Plc, Windrush Court, Transport Way, Oxford, OX4 6LT, UK.
| |
Collapse
|
10
|
McGinn OJ, Krishnan S, Bourquin JP, Sapra P, Dempsey C, Saha V, Stern PL. Targeting the 5T4 oncofetal glycoprotein with an antibody drug conjugate (A1mcMMAF) improves survival in patient-derived xenograft models of acute lymphoblastic leukemia. Haematologica 2017; 102:1075-1084. [PMID: 28341731 PMCID: PMC5451339 DOI: 10.3324/haematol.2016.158485] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 03/15/2017] [Indexed: 12/29/2022] Open
Abstract
Outcome in childhood acute lymphoblastic leukemia is prognosticated from levels of minimal residual disease after remission induction therapy. Higher levels of minimal residual disease are associated with inferior results even with intensification of therapy, thus suggesting that identification and targeting of minimal residual disease cells could be a therapeutic strategy. Here we identify high expression of 5T4 in subclonal populations of patient-derived xenografts from patients with high, post-induction levels of minimal residual disease. 5T4-positive cells showed preferential ability to overcome the NOD-scidIL2Rγnull mouse xenograft barrier, migrated in vitro on a CXCL12 gradient, preferentially localized to bone marrow in vivo and displayed the ability to reconstitute the original clonal composition on limited dilution engraftment. Treatment with A1mcMMAF (a 5T4-antibody drug conjugate) significantly improved survival without overt toxicity in mice engrafted with a 5T4-positive acute lymphoblastic leukemia cell line. Mice engrafted with 5T4-positive patient-derived xenograft cells were treated with combination chemotherapy or dexamethasone alone and then given A1mcMMAF in the minimal residual disease setting. Combination chemotherapy was toxic to NOD-scidIL2Rγnull mice. While dexamethasone or A1mcMMAF alone improved outcomes, the sequential administration of dexamethasone and A1mcMMAF significantly improved survival (P=0.0006) over either monotherapy. These data show that specifically targeting minimal residual disease cells improved outcomes and support further investigation of A1mcMMAF in patients with high-risk B-cell precursor acute lymphoblastic leukemia identified by 5T4 expression at diagnosis.
Collapse
Affiliation(s)
- Owen J McGinn
- Immunology, Division of Molecular & Clinical Cancer Sciences, University of Manchester, UK
| | - Shekhar Krishnan
- Paediatric Oncology, Division of Molecular & Clinical Cancer Sciences, University of Manchester, UK.,Tata Translational Cancer Research Center, Tata Medical Center, Kolkata, India
| | - Jean-Pierre Bourquin
- Division of Oncology & Children's Research Center, University Children's Hospital, University of Zurich, Switzerland
| | - Puja Sapra
- Pfizer Inc. Pearl River, NY10965-1299, USA
| | - Clare Dempsey
- Paediatric Oncology, Division of Molecular & Clinical Cancer Sciences, University of Manchester, UK
| | - Vaskar Saha
- Paediatric Oncology, Division of Molecular & Clinical Cancer Sciences, University of Manchester, UK .,Tata Translational Cancer Research Center, Tata Medical Center, Kolkata, India
| | - Peter L Stern
- Immunology, Division of Molecular & Clinical Cancer Sciences, University of Manchester, UK
| |
Collapse
|
11
|
Hawkins K, Keramari M, Soncin F, Segal JM, Mohamet L, Miazga N, Ritson S, Bobola N, Merry CLR, Ward CM. Novel cell lines isolated from mouse embryonic stem cells exhibiting de novo methylation of the E-cadherin promoter. Stem Cells 2015; 32:2869-79. [PMID: 25074424 DOI: 10.1002/stem.1790] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 06/04/2014] [Accepted: 06/07/2014] [Indexed: 12/25/2022]
Abstract
Mouse embryonic stem cells (mESCs) and epiblast stem cells represent the naïve and primed pluripotent states, respectively. These cells self-renew via distinct signaling pathways and can transition between the two states in the presence of appropriate growth factors. Manipulation of signaling pathways has therefore allowed the isolation of novel pluripotent cell types such as Fibroblast growth factor, Activin and BIO-derived stem cells and IESCs. However, the effect of cell seeding density on pluripotency remains unexplored. In this study, we have examined whether mESCs can epigenetically regulate E-cadherin to enter a primed-like state in response to low cell seeding density. We show that low density seeding in the absence of leukaemia inhibitory factor (LIF) induces decreased apoptosis and maintenance of pluripotency via Activin/Nodal, concomitant with loss of E-cadherin, Signal transducer and activator of transcription phosphorylation, and chimera-forming ability. These cells, E-cadherin negative proliferating stem cells (ENPSCs) can be reverted to a naïve phenotype by addition of LIF or forced E-cadherin expression. However, prolonged culture of ENPSCs without LIF leads to methylation of the E-cadherin promoter (ENPSC(M)), which cannot be reversed by LIF supplementation, and increased histone H3K27 and decreased H3K4 trimethylation. Transcript analysis of ENPSC(M) revealed a primed-like phenotype and their differentiation leads to enrichment of neuroectoderm cells. The generation of ENPSCs is similar to tumorigenesis as ENPSCs exhibit transcript alterations associated with neoplasia, hyperplasia, carcinoma, and metastasis. We therefore describe a novel cell model to elucidate the role of E-cadherin in pluripotency and to investigate epigenetic regulation of this gene during mESC differentiation and tumor metastasis.
Collapse
Affiliation(s)
- Kate Hawkins
- Stem Cell Research Group, The University of Manchester, Core Technology Facility, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Kim YS, Yi BR, Kim NH, Choi KC. Role of the epithelial-mesenchymal transition and its effects on embryonic stem cells. Exp Mol Med 2014; 46:e108. [PMID: 25081188 PMCID: PMC4150931 DOI: 10.1038/emm.2014.44] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 03/04/2014] [Accepted: 04/10/2014] [Indexed: 01/02/2023] Open
Abstract
The epithelial–mesenchymal transition (EMT) is important for embryonic development and the formation of various tissues or organs. However, EMT dysfunction in normal cells leads to diseases, such as cancer or fibrosis. During the EMT, epithelial cells are converted into more invasive and active mesenchymal cells. E-box-binding proteins, including Snail, ZEB and helix–loop–helix family members, serve as EMT-activating transcription factors. These transcription factors repress the expression of epithelial markers, for example, E-cadherin, rearrange the cytoskeleton and promote the expression of mesenchymal markers, such as vimentin, fibronectin and other EMT-activating transcription factors. Signaling pathways that induce EMT, including transforming growth factor-β, Wnt/glycogen synthase kinase-3β, Notch and receptor tyrosine kinase signaling pathways, interact with each other for the regulation of this process. Although the mechanism(s) underlying EMT in cancer or embryonic development have been identified, the mechanism(s) in embryonic stem cells (ESCs) remain unclear. In this review, we describe the underlying mechanisms of important EMT factors, indicating a precise role for EMT in ESCs, and characterize the relationship between EMT and ESCs.
Collapse
Affiliation(s)
- Ye-Seul Kim
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Bo-Rim Yi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Nam-Hyung Kim
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
13
|
Understanding and exploiting 5T4 oncofoetal glycoprotein expression. Semin Cancer Biol 2014; 29:13-20. [PMID: 25066861 DOI: 10.1016/j.semcancer.2014.07.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 07/17/2014] [Indexed: 01/14/2023]
Abstract
Oncofoetal antigens are present during foetal development with generally limited expression in the adult but are upregulated in cancer. These molecules can sometimes be used to diagnose or follow treatment of tumours or as a target for different immunotherapies. The 5T4 oncofoetal glycoprotein was identified by searching for shared surface molecules of human trophoblast and cancer cells with the rationale that they may function to allow survival of the foetus as a semi-allograft in the mother or a tumour in its host, potentially influencing growth, invasion or altered immune surveillance of the host. 5T4 tumour selective expression has stimulated the development of 5T4 vaccine, 5T4 antibody targeted-superantigen and 5T4 antibody-drug therapies through preclinical and into clinical studies. It is now apparent that 5T4 expression is a marker of the use (or not) of several cellular pathways relevant to tumour growth and spread. Thus 5T4 expression is mechanistically associated with the directional movement of cells through epithelial mesenchymal transition, facilitation of CXCL12/CXCR4 chemotaxis, blocking of canonical Wnt/beta-catenin while favouring non-canonical pathway signalling. These processes are highly regulated in development and in normal adult tissues but can contribute to the spread of cancer cells. Understanding the differential impact of these pathways marked by 5T4 can potentially improve existing, or aid development of novel cancer treatment strategies.
Collapse
|
14
|
Vishwanath VR, Nadig RR, Nadig R, Prasanna JS, Karthik J, Pai VS. Differentiation of isolated and characterized human dental pulp stem cells and stem cells from human exfoliated deciduous teeth: An in vitro study. J Conserv Dent 2013; 16:423-8. [PMID: 24082571 PMCID: PMC3778624 DOI: 10.4103/0972-0707.117509] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 06/07/2013] [Accepted: 07/17/2013] [Indexed: 01/09/2023] Open
Abstract
AIMS AND OBJECTIVES Isolation, characterization and differentiation of dental pulp stem cells (DPSCs) and stem cells from exfoliated human deciduous teeth (SHED). METHODS The pulp tissue was digested in collagenase and cultured in DMEM Dulbecco's Modified Eagle's Media). The stem cells were identified and isolated. Surface characterization of cells was done with flow cytometer using surface markers. An immuno cytochemistry analysis was done. Differentiation potential was analyzed using various differentiation markers. RESULTS Flow cytometry analyses for various CD markers showed similar results for both DPSCs and SHED. The cells showed positive expression for pluripotent, ectodermal and mesodermal markers. Cells differentiated into osteoblasts and adipocytes. CONCLUSION The study demonstrated that stem cells existed in deciduous and permanent pulp tissue. The stem cells present in pulp tissue can be isolated, cultivated and expanded in vitro. Both DPSCs and SHED show almost a similar expression pattern profile for variety of antigens tested.
Collapse
|
15
|
Hawkins K, Mohamet L, Ritson S, Merry CLR, Ward CM. E-cadherin and, in its absence, N-cadherin promotes Nanog expression in mouse embryonic stem cells via STAT3 phosphorylation. Stem Cells 2013; 30:1842-51. [PMID: 22696497 DOI: 10.1002/stem.1148] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We have recently shown that loss of E-cadherin in mouse embryonic stem cells (mESCs) results in significant alterations to both the transcriptome and hierarchy of pluripotency-associated signaling pathways. Here, we show that E-cadherin promotes kruppel-like factor 4 (Klf4) and Nanog transcript and protein expression in mESCs via STAT3 phosphorylation and that β-catenin, and its binding region in E-cadherin, is required for this function. To further investigate the role of E-cadherin in leukemia inhibitory factor (LIF)-dependent pluripotency, E-cadherin null (Ecad(-/-)) mESCs were cultured in LIF/bone morphogenetic protein supplemented medium. Under these conditions, Ecad(-/-) mESCs exhibited partial restoration of cell-cell contact and STAT3 phosphorylation and upregulated Klf4, Nanog, and N-cadherin transcripts and protein. Abrogation of N-cadherin using an inhibitory peptide caused loss of phospho STAT3, Klf4, and Nanog in these cells, demonstrating that N-cadherin supports LIF-dependent pluripotency in this context. We therefore identify a novel molecular mechanism linking E- and N-cadherin to the core circuitry of pluripotency in mESCs. This mechanism may explain the recently documented role of E-cadherin in efficient induced pluripotent stem cell reprogramming.
Collapse
Affiliation(s)
- Kate Hawkins
- Stem Cell Biology Group, Core Technology Facility, The University of Manchester, Manchester, United Kingdom
| | | | | | | | | |
Collapse
|
16
|
McGinn OJ, Marinov G, Sawan S, Stern PL. CXCL12 receptor preference, signal transduction, biological response and the expression of 5T4 oncofoetal glycoprotein. J Cell Sci 2012; 125:5467-78. [PMID: 22956548 DOI: 10.1242/jcs.109488] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
CXCL12 is a pleiotropic chemokine capable of eliciting multiple signal transduction cascades and functions, via interaction with either CXCR4 or CXCR7. Factors that determine CXCL12 receptor preference, intracellular signalling route and biological response are poorly understood but are of central importance in the context of therapeutic intervention of the CXCL12 axis in multiple disease states. We have recently demonstrated that 5T4 oncofoetal glycoprotein facilitates functional CXCR4 expression leading to CXCL12 mediated chemotaxis in mouse embryonic cells. Using wild type (WT) and 5T4 knockout (5T4KO) murine embryonic fibroblasts (MEFs), we now show that CXCL12 binding to CXCR4 activates both the ERK and AKT pathways within minutes, but while these pathways are intact, they are non-functional in 5T4KO cells treated with CXCL12. Importantly, in the absence of 5T4 expression, CXCR7 is upregulated and becomes the predominant receptor for CXCL12, activating a distinct signal transduction pathway with slower kinetics involving transactivation of the epidermal growth factor receptor (EGFR), eliciting proliferation rather than chemotaxis. Thus the surface expression of 5T4 marks the use of the CXCR4 rather than the CXCR7 receptor, with distinct consequences for CXCL12 exposure, relevant to the spread and growth of a tumour. Consistent with this hypothesis, we have identified human small cell lung carcinoma cells with similar 5T4/CXCR7 reciprocity that is predictive of biological response to CXCL12 and determined that 5T4 expression is required for functional chemotaxis in these cells.
Collapse
Affiliation(s)
- Owen J McGinn
- Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester M13 9PT, UK
| | | | | | | |
Collapse
|
17
|
Iwagawa T, Ohuchi SP, Watanabe S, Nakamura Y. Selection of RNA aptamers against mouse embryonic stem cells. Biochimie 2011; 94:250-7. [PMID: 22085640 DOI: 10.1016/j.biochi.2011.10.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 10/28/2011] [Indexed: 12/30/2022]
Abstract
Embryonic stem cells (ESCs) are capable of unlimited self-renewal and differentiation into multiple cell types. Recent large-scale analyses have identified various cell surface molecules on ESCs. Some of them are considered to be beneficial markers for characterization of cellular phenotypes and/or play an essential role for regulating the differentiation state. Thus, it is desired to efficiently produce affinity reagents specific to these molecules. In this study, to develop such reagents for mouse ESCs (mESCs), we selected RNA aptamers against intact, live mESCs using several selection strategies. The initial selection provided us with several anti-mESC aptamers of distinct sequences, which unexpectedly react with the same molecule on mESCs. Then, to isolate aptamers against different surface markers on mESCs, one of the selected aptamers was used as a competitor in the subsequent selections. In addition, one of the selections further employed negative selection against differentiated mouse cells. Consequently, we successfully isolated three classes of anti-mESC aptamers that do not compete with one another. The isolated aptamers were shown to distinguish mESCs from differentiated mouse cell lines and trace the differentiation process of mESCs. These aptamers could prove useful for developing molecular probes and manipulation tools for mESCs.
Collapse
Affiliation(s)
- Toshiro Iwagawa
- Division of Molecular Biology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Tokyo 108-8639, Japan
| | | | | | | |
Collapse
|
18
|
Brookman-May S, Burger M, Wieland WF, Rössler W, May M, Denzinger S. Vaccination therapy in renal cell carcinoma: current position and future options in metastatic and localized disease. Expert Rev Vaccines 2011; 10:837-52. [PMID: 21692704 DOI: 10.1586/erv.11.64] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
As renal cell carcinoma represents one of the most immune-responsive cancers, immunotherapy exhibits a suitable treatment basis. Beside nonspecific stimulation via cytokines, passive specific and active immunotherapy are also appropriate options to recognize and destroy tumor cells. For more than 30 years, research regarding vaccination therapy has been of special interest for the treatment of renal cell carcinoma. However, apart from occasional promising results in Phase I and II trials, vaccination therapy is still considered experimental in this tumor entity, especially owing to missing results from Phase III trials demonstrating clinical efficacy. In the present article, we review data from completed clinical trials of vaccination therapy and also discuss scheduled future trials, in order to assess the current position and possible future fields of application of vaccination therapy in renal cell carcinoma in the era of recently developed targeted therapies.
Collapse
Affiliation(s)
- Sabine Brookman-May
- University of Regensburg, Department of Urology, Caritas St. Josef Medical Center, Landshuter Strasse 65, 93053 Regensburg, Germany.
| | | | | | | | | | | |
Collapse
|
19
|
Soncin F, Mohamet L, Ritson S, Hawkins K, Bobola N, Zeef L, Merry CLR, Ward CM. E-cadherin acts as a regulator of transcripts associated with a wide range of cellular processes in mouse embryonic stem cells. PLoS One 2011; 6:e21463. [PMID: 21779327 PMCID: PMC3136471 DOI: 10.1371/journal.pone.0021463] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 06/01/2011] [Indexed: 11/18/2022] Open
Abstract
Background We have recently shown that expression of the cell adhesion molecule E-cadherin is required for LIF-dependent pluripotency of mouse embryonic stem (ES) cells. Methodology In this study, we have assessed global transcript expression in E-cadherin null (Ecad-/-) ES cells cultured in either the presence or absence of LIF and compared these to the parental cell line wtD3. Results We show that LIF has little effect on the transcript profile of Ecad-/- ES cells, with statistically significant transcript alterations observed only for Sp8 and Stat3. Comparison of Ecad-/- and wtD3 ES cells cultured in LIF demonstrated significant alterations in the transcript profile, with effects not only confined to cell adhesion and motility but also affecting, for example, primary metabolic processes, catabolism and genes associated with apoptosis. Ecad-/- ES cells share similar, although not identical, gene expression profiles to epiblast-derived pluripotent stem cells, suggesting that E-cadherin expression may inhibit inner cell mass to epiblast transition. We further show that Ecad-/- ES cells maintain a functional β-catenin pool that is able to induce β-catenin/TCF-mediated transactivation but, contrary to previous findings, do not display endogenous β-catenin/TCF-mediated transactivation. We conclude that loss of E-cadherin in mouse ES cells leads to significant transcript alterations independently of β-catenin/TCF transactivation.
Collapse
Affiliation(s)
- Francesca Soncin
- Core Technology Facility, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, United Kingdom
- Materials Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Lisa Mohamet
- Core Technology Facility, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, United Kingdom
| | - Sarah Ritson
- Core Technology Facility, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, United Kingdom
| | - Kate Hawkins
- Core Technology Facility, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, United Kingdom
| | - Nicoletta Bobola
- Core Technology Facility, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, United Kingdom
| | - Leo Zeef
- Microarray Facility, Faculty of Life Sciences, Michael Smith Building, The University of Manchester, Manchester, United Kingdom
| | | | - Christopher M. Ward
- Core Technology Facility, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
20
|
Holley RJ, Pickford CE, Rushton G, Lacaud G, Gallagher JT, Kouskoff V, Merry CLR. Influencing hematopoietic differentiation of mouse embryonic stem cells using soluble heparin and heparan sulfate saccharides. J Biol Chem 2011; 286:6241-52. [PMID: 21148566 PMCID: PMC3057799 DOI: 10.1074/jbc.m110.178483] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 12/10/2010] [Indexed: 12/13/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPG) encompass some of the most abundant macromolecules on the surface of almost every cell type. Heparan sulfate (HS) chains provide a key interaction surface for the binding of numerous proteins such as growth factors and morphogens, helping to define the ability of a cell to respond selectively to environmental cues. The specificity of HS-protein interactions are governed predominantly by the order and positioning of sulfate groups, with distinct cell types expressing unique sets of HS epitopes. Embryos deficient in HS-synthesis (Ext1(-/-)) exhibit pre-gastrulation lethality and lack recognizable organized mesoderm and extraembryonic tissues. Here we demonstrate that embryonic stem cells (ESCs) derived from Ext1(-/-) embryos are unable to differentiate into hematopoietic lineages, instead retaining ESC marker expression throughout embryoid body (EB) culture. However hematopoietic differentiation can be restored by the addition of soluble heparin. Consistent with specific size and composition requirements for HS:growth factor signaling, chains measuring at least 12 saccharides were required for partial rescue of hematopoiesis with longer chains (18 saccharides or more) required for complete rescue. Critically N- and 6-O-sulfate groups were essential for rescue. Heparin addition restored the activity of multiple signaling pathways including bone morphogenic protein (BMP) with activation of phospho-SMADs re-established by the addition of heparin. Heparin addition to wild-type cultures also altered the outcome of differentiation, promoting hematopoiesis at low concentrations, yet inhibiting blood formation at high concentrations. Thus altering the levels of HS and HS sulfation within differentiating ESC cultures provides an attractive and accessible mechanism for influencing cell fate.
Collapse
Affiliation(s)
- Rebecca J. Holley
- From the School of Materials, Materials Science Centre, The University of Manchester, Manchester M13 9PL, United Kingdom and
| | - Claire E. Pickford
- From the School of Materials, Materials Science Centre, The University of Manchester, Manchester M13 9PL, United Kingdom and
| | - Graham Rushton
- the Cancer Research UK, Paterson Institute for Cancer Research, The University of Manchester, Wilmslow Road, Manchester M20 4BX, United Kingdom
| | - Georges Lacaud
- the Cancer Research UK, Paterson Institute for Cancer Research, The University of Manchester, Wilmslow Road, Manchester M20 4BX, United Kingdom
| | - John T. Gallagher
- the Cancer Research UK, Paterson Institute for Cancer Research, The University of Manchester, Wilmslow Road, Manchester M20 4BX, United Kingdom
| | - Valerie Kouskoff
- the Cancer Research UK, Paterson Institute for Cancer Research, The University of Manchester, Wilmslow Road, Manchester M20 4BX, United Kingdom
| | - Catherine L. R. Merry
- From the School of Materials, Materials Science Centre, The University of Manchester, Manchester M13 9PL, United Kingdom and
| |
Collapse
|
21
|
Using cadherin expression to assess spontaneous differentiation of embryonic stem cells. Methods Mol Biol 2011; 690:81-94. [PMID: 21042986 DOI: 10.1007/978-1-60761-962-8_5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Embryonic stem cells (ESCs) are pluripotent cells derived from preimplantation embryos and can be maintained in an undifferentiated state over prolonged periods in vitro. In addition, ESCs can be induced to differentiate into cells representative of the three primary germ layers. As such, ESCs are a useful system for studying early developmental events in vitro and have the potential to provide a ubiquitous supply of somatic cells for use in regenerative medicine. However, significant differences in the expression pattern of various cell surface markers between murine and human ESCs, e.g. the SSEA series, necessitate the use of separate markers for determining the undifferentiated state of these cells. We have recently shown that an E- to N-cadherin switch occurs during spontaneous differentiation of both murine and human ESCs. Here we describe the use of E-cadherin and N-cadherin proteins and transcript expression for assessing the proportion of undifferentiated and spontaneously differentiated cells within ESC populations. In summary, loss of cell surface E-cadherin and/or gain of N-cadherin protein expression provides a useful nondestructive assay for the determination of the proportion of spontaneously differentiated cells within an ESC population. In addition, presence of N-cadherin transcripts in an ESC population is indicative of spontaneous differentiation of a proportion of the cells.
Collapse
|
22
|
Trettner S, Seeliger A, zur Nieden NI. Embryoid body formation: recent advances in automated bioreactor technology. Methods Mol Biol 2011; 690:135-149. [PMID: 21042990 DOI: 10.1007/978-1-60761-962-8_9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
While spontaneous differentiation is an undesired feature of expanding populations of embryonic stem cells, a variety of methods have been described for their intended differentiation into specialized cell types, such as the osteoblast or chondrocyte. Most commonly, differentiation initiation involves the aggregation of ESCs into a so-called embryoid body (EB), a sphere composed of approximately 15,000 differentiating cells. EB formation has been optimized through the years, for example through invention of the hanging drop protocol. Yet, it remains a highly laborious process. Here we describe the use of computer-controllable suspension bioreactors to form EBs in an automated and highly reproducible process and their subsequent differentiation along the osteoblast lineage. The development of the differentiating cells taken from bioreactor EBs to EBs formed in static control cultures through the hanging drop method will be compared.
Collapse
Affiliation(s)
- Susanne Trettner
- Department of Cell Therapy, Applied Stem Cell Technology Unit, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | | | | |
Collapse
|
23
|
Murakami T, Abe H, Nagai K, Tominaga T, Takamatsu N, Araoka T, Kishi S, Takahashi T, Mima A, Takai Y, Kopp JB, Doi T. Trophoblast glycoprotein: possible candidate mediating podocyte injuries in glomerulonephritis. Am J Nephrol 2010; 32:505-21. [PMID: 20980737 DOI: 10.1159/000321366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2010] [Accepted: 09/17/2010] [Indexed: 11/19/2022]
Abstract
BACKGROUND trophoblast glycoprotein (Tpbg), a 72-kDa transmembrane glycoprotein, is known to regulate the phenotypes of epithelial cells by modifying actin organization and cell motility. Recently, a microarray study showed that Tpbg is upregulated in Thy1 glomerulonephritis (Thy1 GN). We hypothesized that Tpbg regulates cytoskeletal rearrangement and modulates phenotypic alteration in podocytes under pathological conditions. METHODS we examined Tpbg expression in Thy1 GN and Tpbg function in mouse podocytes. RESULTS we demonstrated that Tpbg is upregulated in the injured podocytes of Thy1 GN. In vitro, immunofluorescence studies revealed that Tpbg colocalized with the focal adhesion protein, vinculin, in parallel with stress fiber formation. This colocalization was observed even when actin filaments were depolymerized with cytochalasin D. Tpbg localization at focal adhesions was induced by dominant-active RhoA and suppressed by the ROCK1 inhibitor Y-26732. In addition, transforming growth factor-β increased Tpbg expression at focal adhesions concurrently with rearrangement of stress fibers. Stress fiber formation was suppressed in differentiated podocytes transfected with full-length Tpbg. Furthermore, knockdown of Tpbg using small interfering RNA decreased podocyte motility. CONCLUSION our findings suggest a novel role of Tpbg in the phenotypic alteration of injured podocytes, and we accordingly propose a new mechanism of glomerular injury in glomerulonephritis.
Collapse
Affiliation(s)
- Taichi Murakami
- Department of Nephrology, Graduate School of Medicine, Institute of Health-Bio-Science, University of Tokushima, Tokushima University Hospital, Tokushima, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Kleger A, Seufferlein T, Malan D, Tischendorf M, Storch A, Wolheim A, Latz S, Protze S, Porzner M, Proepper C, Brunner C, Katz SF, Varma Pusapati G, Bullinger L, Franz WM, Koehntop R, Giehl K, Spyrantis A, Wittekindt O, Lin Q, Lin Q, Zenke M, Fleischmann BK, Wartenberg M, Wobus AM, Boeckers TM, Liebau S. Modulation of calcium-activated potassium channels induces cardiogenesis of pluripotent stem cells and enrichment of pacemaker-like cells. Circulation 2010; 122:1823-36. [PMID: 20956206 DOI: 10.1161/circulationaha.110.971721] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Ion channels are key determinants for the function of excitable cells, but little is known about their role and involvement during cardiac development. Earlier work identified Ca(2+)-activated potassium channels of small and intermediate conductance (SKCas) as important regulators of neural stem cell fate. Here we have investigated their impact on the differentiation of pluripotent cells toward the cardiac lineage. METHODS AND RESULTS We have applied the SKCa activator 1-ethyl-2-benzimidazolinone on embryonic stem cells and identified this particular ion channel family as a new critical target involved in the generation of cardiac pacemaker-like cells: SKCa activation led to rapid remodeling of the actin cytoskeleton, inhibition of proliferation, induction of differentiation, and diminished teratoma formation. Time-restricted SKCa activation induced cardiac mesoderm and commitment to the cardiac lineage as shown by gene regulation, protein, and functional electrophysiological studies. In addition, the differentiation into cardiomyocytes was modulated in a qualitative fashion, resulting in a strong enrichment of pacemaker-like cells. This was accompanied by induction of the sino-atrial gene program and in parallel by a loss of the chamber-specific myocardium. In addition, SKCa activity induced activation of the Ras-Mek-Erk signaling cascade, a signaling pathway involved in the 1-ethyl-2-benzimidazolinone-induced effects. CONCLUSIONS SKCa activation drives the fate of pluripotent cells toward mesoderm commitment and cardiomyocyte specification, preferentially into nodal-like cardiomyocytes. This provides a novel strategy for the enrichment of cardiomyocytes and in particular, the generation of a specific subtype of cardiomyocytes, pacemaker-like cells, without genetic modification.
Collapse
Affiliation(s)
- Alexander Kleger
- Institute for Anatomy and Cell Biology, University of Ulm, Albert-Einstein Allee 11, 89081 Ulm, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abrogation of E-cadherin-mediated cellular aggregation allows proliferation of pluripotent mouse embryonic stem cells in shake flask bioreactors. PLoS One 2010; 5:e12921. [PMID: 20886069 PMCID: PMC2944850 DOI: 10.1371/journal.pone.0012921] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Accepted: 08/16/2010] [Indexed: 12/03/2022] Open
Abstract
Background A fundamental requirement for the exploitation of embryonic stem (ES) cells in regenerative medicine is the ability to reproducibly derive sufficient numbers of cells of a consistent quality in a cost-effective manner. However, undifferentiated ES cells are not ideally suited to suspension culture due to the formation of cellular aggregates, ultimately limiting scalability. Significant advances have been made in recent years in the culture of ES cells, including automated adherent culture and suspension microcarrier or embryoid body bioreactor culture. However, each of these methods exhibits specific disadvantages, such as high cost, additional downstream processes or reduced cell doubling times. Methodology/Principal Findings Here we show that abrogation of the cell surface protein E-cadherin, using either gene knockout (Ecad-/-) or the neutralising antibody DECMA-1 (EcadAb), allows culture of mouse ES cells as a near-single cell suspension in scalable shake flask culture over prolonged periods without additional media supplements. Both Ecad-/- and EcadAb ES cells exhibited adaptation phases in suspension culture, with optimal doubling times of 7.3 h±0.9 and 15.6 h±4.7 respectively and mean-fold increase in viable cell number of 95.1±2.0 and 16±0.9-fold over 48 h. EcadAb ES cells propagated as a dispersed cell suspension for 15 d maintained expression of pluripotent markers, exhibited a normal karyotype and high viability. Subsequent differentiation of EcadAb ES cells resulted in expression of transcripts and proteins associated with the three primary germ layers. Conclusions/Significance This is the first demonstration of the culture of pluripotent ES cells as a near-single cell suspension in a manual fed-batch shake flask bioreactor and represents a significant improvement on current ES cell culture techniques. Whilst this proof-of-principle method would be useful for the culture of human ES and iPS cells, further steps are necessary to increase cell viability of hES cells in suspension.
Collapse
|
26
|
Southgate TD, McGinn OJ, Castro FV, Rutkowski AJ, Al-Muftah M, Marinov G, Smethurst GJ, Shaw D, Ward CM, Miller CJ, Stern PL. CXCR4 mediated chemotaxis is regulated by 5T4 oncofetal glycoprotein in mouse embryonic cells. PLoS One 2010; 5:e9982. [PMID: 20376365 PMCID: PMC2848608 DOI: 10.1371/journal.pone.0009982] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Accepted: 03/12/2010] [Indexed: 12/20/2022] Open
Abstract
5T4 oncofetal molecules are highly expressed during development and upregulated in cancer while showing only low levels in some adult tissues. Upregulation of 5T4 expression is a marker of loss of pluripotency in the early differentiation of embryonic stem (ES) cells and forms an integrated component of an epithelial-mesenchymal transition, a process important during embryonic development and metastatic spread of epithelial tumors. Investigation of the transcriptional changes in early ES differentiation showed upregulation of CXCL12 and down-regulation of a cell surface protease, CD26, which cleaves this chemokine. CXCL12 binds to the widely expressed CXCR4 and regulates key aspects of development, stem cell motility and tumour metastasis to tissues with high levels of CXCL12. We show that the 5T4 glycoprotein is required for optimal functional cell surface expression of the chemokine receptor CXCR4 and CXCL12 mediated chemotaxis in differentiating murine embryonic stem cells and embryo fibroblasts (MEF). Cell surface expression of 5T4 and CXCR4 molecules is co-localized in differentiating ES cells and MEF. By contrast, differentiating ES and MEF derived from 5T4 knockout (KO) mice show only intracellular CXCR4 expression but infection with adenovirus encoding mouse 5T4 restores CXCL12 chemotaxis and surface co-localization with 5T4 molecules. A series of chimeric constructs with interchanged domains of 5T4 and the glycoprotein CD44 were used to map the 5T4 sequences relevant for CXCR4 membrane expression and function in 5T4KO MEF. These data identified the 5T4 transmembrane domain as sufficient and necessary to enable CXCR4 cell surface expression and chemotaxis. Furthermore, some monoclonal antibodies against m5T4 can inhibit CXCL12 chemotaxis of differentiating ES cells and MEF which is not mediated by simple antigenic modulation. Collectively, these data support a molecular interaction of 5T4 and CXCR4 occurring at the cell surface which directly facilitates the biological response to CXCL12. The regulation of CXCR4 surface expression by 5T4 molecules is a novel means to control responses to the chemokine CXCL12 for example during embryogenesis but can also be selected to advantage the spread of a 5T4 positive tumor from its primary site.
Collapse
Affiliation(s)
- Thomas D Southgate
- Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Elkord E, Shablak A, Stern PL, Hawkins RE. 5T4 as a target for immunotherapy in renal cell carcinoma. Expert Rev Anticancer Ther 2010; 9:1705-9. [PMID: 19954280 DOI: 10.1586/era.09.152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
28
|
Soncin F, Mohamet L, Eckardt D, Ritson S, Eastham AM, Bobola N, Russell A, Davies S, Kemler R, Merry CLR, Ward CM. Abrogation of E-cadherin-mediated cell-cell contact in mouse embryonic stem cells results in reversible LIF-independent self-renewal. Stem Cells 2009; 27:2069-80. [PMID: 19544408 DOI: 10.1002/stem.134] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
We have previously demonstrated that differentiation of embryonic stem (ES) cells is associated with downregulation of cell surface E-cadherin. In this study, we assessed the function of E-cadherin in mouse ES cell pluripotency and differentiation. We show that inhibition of E-cadherin-mediated cell-cell contact in ES cells using gene knockout (Ecad(-/-)), RNA interference (EcadRNAi), or a transhomodimerization-inhibiting peptide (CHAVC) results in cellular proliferation and maintenance of an undifferentiated phenotype in fetal bovine serum-supplemented medium in the absence of leukemia inhibitory factor (LIF). Re-expression of E-cadherin in Ecad(-/-), EcadRNAi, and CHAVC-treated ES cells restores cellular dependence to LIF supplementation. Although reversal of the LIF-independent phenotype in Ecad(-/-) ES cells is dependent on the beta-catenin binding domain of E-cadherin, we show that beta-catenin null (betacat(-/-)) ES cells also remain undifferentiated in the absence of LIF. This suggests that LIF-independent self-renewal of Ecad(-/-) ES cells is unlikely to be via beta-catenin signaling. Exposure of Ecad(-/-), EcadRNAi, and CHAVC-treated ES cells to the activin receptor-like kinase inhibitor SB431542 led to differentiation of the cells, which could be prevented by re-expression of E-cadherin. To confirm the role of transforming growth factor beta family signaling in the self-renewal of Ecad(-/-) ES cells, we show that these cells maintain an undifferentiated phenotype when cultured in serum-free medium supplemented with Activin A and Nodal, with fibroblast growth factor 2 required for cellular proliferation. We conclude that transhomodimerization of E-cadherin protein is required for LIF-dependent ES cell self-renewal and that multiple self-renewal signaling networks subsist in ES cells, with activity dependent upon the cellular context.
Collapse
Affiliation(s)
- Francesca Soncin
- Core Technology Facility, Faculty of Medical and Human Sciences, and The University of Manchester, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Elkord E, Dangoor A, Burt DJ, Southgate TD, Daayana S, Harrop R, Drijfhout JW, Sherlock D, Hawkins RE, Stern PL. Immune evasion mechanisms in colorectal cancer liver metastasis patients vaccinated with TroVax (MVA-5T4). Cancer Immunol Immunother 2009; 58:1657-67. [PMID: 19221742 PMCID: PMC11029831 DOI: 10.1007/s00262-009-0674-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Accepted: 01/27/2009] [Indexed: 01/01/2023]
Abstract
We have recently reported the results of a phase II trial in which two TroVax [modified vaccinia ankara (MVA) encoding the tumour antigen 5T4] vaccinations were given to patients both pre- and post-surgical resection of liver metastases secondary to colorectal cancer (CRC). 5T4-specific cellular responses were assessed at the entry and 2 weeks after each vaccination by proliferation of fresh lymphocytes and ELISA for antibody responses; 18 from the 19 CRC patients mounted a 5T4-specific cellular and/or humoral response. Here, we present a comparison of individual and between patient responses over the course of the treatments using cryopreserved peripheral blood mononuclear cells (PBMC) samples from the baseline until after the fourth vaccination at 14 weeks. Assays used were proliferation assay with 5T4-Fc fusion protein, overlapping 32mer 5T4 peptides, MVA-LacZ and MVA-5T4 infected autologous monocytes. Responses to 5T4 protein or one or more peptide pools were pre-existing in 12/20 patients and subsequently 10 and 12 patients showed boosted and/or de novo responses, respectively. Cumulatively, 13/20 patients showed proliferative responses by week 14. We also assessed the levels of systemic T regulatory cells, plasma cytokine levels, phenotype of tumour-infiltrating lymphocytes including T regulatory cells and tumour HLA class I loss of expression. More than half of the patients showed phenotypes consistent with relative immune suppression and/or escape highlighting the complexity of positive and negative factors challenging any simple correlation with clinical outcome.
Collapse
Affiliation(s)
- Eyad Elkord
- CR UK Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
- Department of Medical Oncology, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
- Present Address: Cellular Immunology Section, Laboratory of Immunology, National Institutes of Health, Bethesda, MD 20892 USA
| | - Adam Dangoor
- CR UK Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
- Department of Medical Oncology, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
| | - Deborah J. Burt
- CR UK Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
| | - Thomas D. Southgate
- CR UK Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
| | - Sai Daayana
- CR UK Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
| | - Richard Harrop
- Oxford BioMedica, Medawar Centre, Oxford Science Park, Oxford, OX4 4GA UK
| | - Jan W. Drijfhout
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, Netherlands
| | - David Sherlock
- Department of Surgery, North Manchester Healthcare NHS Trust, Manchester, UK
| | - Robert E. Hawkins
- Department of Medical Oncology, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
| | - Peter L. Stern
- CR UK Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
| |
Collapse
|
30
|
An MVA-based vaccine targeting the oncofetal antigen 5T4 in patients undergoing surgical resection of colorectal cancer liver metastases. J Immunother 2009; 31:820-9. [PMID: 18833005 DOI: 10.1097/cji.0b013e3181876ab3] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We investigated the use of a therapeutic vaccine, TroVax in patients undergoing surgical resection of colorectal cancer liver metastases. Systemic immunity generated by vaccination before and after resection of metastases was measured in addition to assessing safety and analyzing the function and phenotype of tumor-associated lymphocytes. Twenty patients were scheduled to receive 2 TroVax vaccinations at 2-week intervals preoperatively and 2 postoperatively; if immune responses were detected, 2 further vaccinations were offered. Blood was taken at trial entry and 2 weeks after each vaccination; tumor biopsies were collected at surgery. 5T4-specific cellular responses were assessed by lymphocyte proliferation and enzyme-linked immunosorbent spot, with antibody responses by enzyme-linked immunosorbent assay. Immunohistochemistry characterized the phenotype of tumor-infiltrating lymphocytes. Seventeen of 19 colorectal cancer patients showed 5T4 expression in the liver metastases or surrounding stroma and 18 mounted a 5T4-specific cellular and/or humoral response. In patients who received at least 4 vaccinations and potentially curative surgery (n=15), those with above median 5T4-specific proliferative responses or T-cell infiltration into the resected tumor showed significantly longer survival compared with those with below median responses. Seven of 8 patients who had preexisting proliferative responses to 5T4 were longer-term survivors; these patients showed significantly higher proliferative responses after vaccination than those who subsequently died. These data suggest that the magnitude of 5T4 proliferative responses and the density of CD3 cells in colorectal cancer liver metastases are associated with longer survival. These observations warrant more studies to identify the precise underlying mechanisms.
Collapse
|
31
|
Large histone H3 lysine 9 dimethylated chromatin blocks distinguish differentiated from embryonic stem cells. Nat Genet 2009; 41:246-50. [PMID: 19151716 PMCID: PMC2632725 DOI: 10.1038/ng.297] [Citation(s) in RCA: 460] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Accepted: 10/27/2008] [Indexed: 12/13/2022]
Abstract
Higher eukaryotes must adapt a totipotent genome to specialized cell types with stable but limited functions. One potential mechanism for lineage restriction is changes in chromatin, and differentiation-related chromatin changes have been observed for individual genes. We have taken a genome-wide view of histone H3 lysine 9 dimethylation (H3K9Me2) and find that differentiated tissues show surprisingly large K9-modified regions (up to 4.9 Mb). These regions are highly conserved between human and mouse and are differentiation specific, covering only approximately 4% of the genome in undifferentiated mouse embryonic stem (ES) cells, compared to 31% in differentiated ES cells, approximately 46% in liver and approximately 10% in brain. These modifications require histone methyltransferase G9a and are inversely related to expression of genes within the regions. We term these regions large organized chromatin K9 modifications (LOCKs). LOCKs are substantially lost in cancer cell lines, and they may provide a cell type-heritable mechanism for phenotypic plasticity in development and disease.
Collapse
|
32
|
Eastham AM, Spencer H, Soncin F, Ritson S, Merry CLR, Stern PL, Ward CM. Epithelial-mesenchymal transition events during human embryonic stem cell differentiation. Cancer Res 2008; 67:11254-62. [PMID: 18056451 DOI: 10.1158/0008-5472.can-07-2253] [Citation(s) in RCA: 228] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Epithelial-mesenchymal transition (EMT) occurs during embryonic development and may also be associated with the metastatic spread of epithelial tumors. During EMT, E-cadherin is down-regulated and this correlates with increased motility and invasion of cells. We show that differentiation of human embryonic stem (ES) cells in monolayer culture is associated with an E- to N-cadherin switch, increased vimentin expression, up-regulation of E-cadherin repressor molecules (Snail and Slug proteins), and increased gelatinase (matrix metalloproteinases; MMP-2 and MMP-9) activity and cellular motility, all characteristic EMT events. The 5T4 oncofetal antigen, previously shown to be associated with early human ES cell differentiation, is also part of this process. Abrogation of E-cadherin-mediated cell-cell contact in undifferentiated ES cells using neutralizing antibody (nAb) SHE78.7 resulted in increased cellular motility, altered actin cytoskeleton arrangement and a mesenchymal phenotype together with presentation of the 5T4 antigen at the cell surface. nAb-treated ES cells remained in an undifferentiated state, as assessed by OCT-4 protein expression, and did not express EMT-associated transcripts. Removal of nAb from ES cells resulted in the restoration of cell-cell contact, absence of cell surface 5T4, decreased mesenchymal cellular morphology and motility, and enabled the differentiation of the cells to the three germ layers upon their removal from the fibroblast feeder layer. We conclude that E-cadherin functions in human ES cells to stabilize the cortical actin cyoskeletal arrangement and this prevents cell surface localization of the 5T4 antigen. Furthermore, human ES cells represent a useful model system with which to study EMT events relevant to embryonic development and tumor cell metastasis.
Collapse
Affiliation(s)
- Angela M Eastham
- Centre for Molecular Medicine, Faculty of Medical and Human Sciences, The University of Manchester, M13 9PT, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
33
|
Perez-Campo FM, Spencer HL, Elder RH, Stern PL, Ward CM. Novel vectors for homologous recombination strategies in mouse embryonic stem cells: an ES cell line expressing EGFP under control of the 5T4 promoter. Exp Cell Res 2007; 313:3604-3615. [PMID: 17765223 DOI: 10.1016/j.yexcr.2007.07.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Revised: 07/12/2007] [Accepted: 07/13/2007] [Indexed: 01/28/2023]
Abstract
The use of gene mutation/knock-out strategies in mouse embryonic stem (ES) cells has revolutionized the study of gene function in ES cells and embryonic development. However, the construction of vectors for homologous recombination strategies requires considerable expertise and time. We describe two novel vectors that can generate site specific knock-out or EGFP knock-in ES cells within 6 weeks from construct design to identification of positive ES cell clones. As proof-of-principle, we have utilized the knock-out targeting vector to modify the NEIL2 locus in ES cells. In addition, using the knock-in vector, we have inserted EGFP downstream of the 5T4 oncofetal antigen promoter in ES cells (5T4-GFP ES cells). Undifferentiated 5T4-GFP ES cells lack EGFP and maintain expression of the pluripotent markers OCT-4 and NANOG. Upon differentiation, EGFP expression is increased in 5T4-GFP ES cells and this correlates with 5T4 transcript expression of the unmodified allele, loss of Nanog and Oct-4 transcripts and upregulation of differentiation-associated transcripts. Furthermore, we demonstrate that fluorescent activated cell sorting of 5T4-GFP ES cells allows isolation of pluripotent or differentiated cells from a heterogeneous population. These vectors provide researchers with a rapid method of modifying specific ES cell genes to study cellular differentiation and embryonic development.
Collapse
Affiliation(s)
- Flor M Perez-Campo
- Stem Cell Biology Group, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Wilmslow Road, Manchester, UK
| | | | | | | | | |
Collapse
|
34
|
Spencer HL, Eastham AM, Merry CL, Southgate TD, Perez-Campo F, Soncin F, Ritson S, Kemler R, Stern PL, Ward CM. E-cadherin inhibits cell surface localization of the pro-migratory 5T4 oncofetal antigen in mouse embryonic stem cells. Mol Biol Cell 2007; 18:2838-2851. [PMID: 17507657 PMCID: PMC1949355 DOI: 10.1091/mbc.e06-09-0875] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2006] [Revised: 04/11/2006] [Accepted: 05/04/2007] [Indexed: 12/31/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) events occur during embryonic development and are important for the metastatic spread of epithelial tumors. We show here that spontaneous differentiation of mouse embryonic stem (ES) cells is associated with an E- to N-cadherin switch, up-regulation of E-cadherin repressor molecules (Snail and Slug proteins), gelatinase activity (matrix metalloproteinase [MMP]-2 and -9), and increased cellular motility, all characteristic EMT events. The 5T4 oncofetal antigen, previously shown to be associated with very early ES cell differentiation and altered motility, is also a part of this coordinated process. E- and N-cadherin and 5T4 proteins are independently regulated during ES cell differentiation and are not required for induction of EMT-associated transcripts and proteins, as judged from the study of the respective knockout ES cells. Further, abrogation of E-cadherin-mediated cell-cell contact in undifferentiated ES cells using neutralizing antibody results in a reversible mesenchymal phenotype and actin cytoskeleton rearrangement that is concomitant with translocation of the 5T4 antigen from the cytoplasm to the cell surface in an energy-dependent manner. E-cadherin null ES cells are constitutively cell surface 5T4 positive, and although forced expression of E-cadherin cDNA in these cells is sufficient to restore cell-cell contact, cell surface expression of 5T4 antigen is unchanged. 5T4 and N-cadherin knockout ES cells exhibit significantly decreased motility during EMT, demonstrating a functional role for these proteins in this process. We conclude that E-cadherin protein stabilizes cortical actin cytoskeletal arrangement in ES cells, and this can prevent cell surface localization of the promigratory 5T4 antigen.
Collapse
Affiliation(s)
- Helen L. Spencer
- *Centre for Molecular Medicine, Faculty of Medical and Human Sciences, The University of Manchester, Manchester M13 9PT, United Kingdom
- Cancer Research UK Immunology Group and
| | - Angela M. Eastham
- *Centre for Molecular Medicine, Faculty of Medical and Human Sciences, The University of Manchester, Manchester M13 9PT, United Kingdom
- Cancer Research UK Immunology Group and
| | - Catherine L.R. Merry
- Cancer Research UK Stem Cell Biology Group, Paterson Institute for Cancer Research, University of Manchester, Christie Hospital National Health Service Trust, Manchester M20 4BX, United Kingdom
| | | | - Flor Perez-Campo
- Materials Science Centre, The University of Manchester, Manchester M1 7HS, United Kingdom; and
| | - Francesca Soncin
- *Centre for Molecular Medicine, Faculty of Medical and Human Sciences, The University of Manchester, Manchester M13 9PT, United Kingdom
- Cancer Research UK Stem Cell Biology Group, Paterson Institute for Cancer Research, University of Manchester, Christie Hospital National Health Service Trust, Manchester M20 4BX, United Kingdom
| | - Sarah Ritson
- *Centre for Molecular Medicine, Faculty of Medical and Human Sciences, The University of Manchester, Manchester M13 9PT, United Kingdom
| | - Rolf Kemler
- Department of Molecular Embryology, Max-Planck Institute of Immunobiology, D-79108 Freiburg, Germany
| | | | - Christopher M. Ward
- *Centre for Molecular Medicine, Faculty of Medical and Human Sciences, The University of Manchester, Manchester M13 9PT, United Kingdom
| |
Collapse
|
35
|
Kerkis I, Kerkis A, Dozortsev D, Stukart-Parsons GC, Gomes Massironi SM, Pereira LV, Caplan AI, Cerruti HF. Isolation and characterization of a population of immature dental pulp stem cells expressing OCT-4 and other embryonic stem cell markers. Cells Tissues Organs 2007; 184:105-16. [PMID: 17409736 DOI: 10.1159/000099617] [Citation(s) in RCA: 303] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Indexed: 12/24/2022] Open
Abstract
We report the isolation of a population of immature dental pulp stem cells (IDPSC), which express embryonic stem cell markers Oct-4, Nanog, SSEA-3, SSEA-4, TRA-1-60 and TRA-1-81 as well as several other mesenchymal stem cell markers during at least 25 passages while maintaining the normal karyotype and the rate of expansion characteristic of stem cells. The expression of these markers was maintained in subclones obtained from these cells. Moreover, in vitrothese cells can be induced to undergo uniform differentiation into smooth and skeletal muscles, neurons, cartilage, and bone under chemically defined culture conditions. After in vivo transplantation of these cells into immunocompromised mice, they showed dense engraftment in various tissues. The relative ease of recovery and the expression profiles of various markers justify further exploration of IDPSC for clinical therapy.
Collapse
Affiliation(s)
- Irina Kerkis
- Laboratório de Genética, Instituto Butantan, São Paulo, Brasil.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
zur Nieden NI, Cormier JT, Rancourt DE, Kallos MS. Embryonic stem cells remain highly pluripotent following long term expansion as aggregates in suspension bioreactors. J Biotechnol 2007; 129:421-32. [PMID: 17306403 DOI: 10.1016/j.jbiotec.2007.01.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2006] [Revised: 12/08/2006] [Accepted: 01/03/2007] [Indexed: 10/23/2022]
Abstract
Increasing attention has been drawn towards pluripotent embryonic stem cells (ESCs) and their potential use as the primary material in various tissue engineering applications. Successful clinical implementation of this technology would require a quality controlled reproducible culture system for the expansion of the cells to be used in the generation of functional tissues. Recently, we showed that suspension bioreactors could be used in the regulated large-scale expansion of highly pluripotent murine ESCs. The current study illustrates that these bioreactor protocols can be adapted for long term culture and that murine ESC cultures remain highly undifferentiated, when serially passaged in suspension bioreactors for extended periods. Flow cytometry analysis and gene expression profiles of several pluripotency markers, in addition to colony and embryoid body (EB) formation tests were conducted at the start and end of the experiment and all showed that the ESC cultures remained highly undifferentiated over extended culture time in suspension. In vivo teratoma formation and in vitro differentiation into neural, cardiomyocyte, osteoblast and chondrocyte lineages, performed at the end of the long term culture, further supported the presence of functional and undifferentiated ESCs in the expanded population. Overall, this system enables the controlled expansion of highly pluripotent murine ESC populations.
Collapse
Affiliation(s)
- Nicole I zur Nieden
- Institute of Maternal & Child Health, University of Calgary, Calgary, Alberta, Canada.
| | | | | | | |
Collapse
|
37
|
Cormier JT, zur Nieden NI, Rancourt DE, Kallos MS. Expansion of Undifferentiated Murine Embryonic Stem Cells as Aggregates in Suspension Culture Bioreactors. ACTA ACUST UNITED AC 2006; 12:3233-45. [PMID: 17518637 DOI: 10.1089/ten.2006.12.3233] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Pluripotent embryonic stem cells (ESCs) have recently been considered as a primary material for regenerating tissues lost to injuries and degenerative diseases. For clinical implementation of this technology, a quality controlled, reproducible culture system is necessary for the expansion and differentiation of the cells. Used in many bioprocess applications, suspension bioreactors have gained considerable attention for the regulated large-scale expansion of cells. The current study presents a bioreactor process for the large-scale expansion of undifferentiated murine ESCs as aggregates. In this system, the level of ESC aggregation and differentiation was effectively controlled by adjusting shear forces and inoculation density, achieving a 31-fold expansion in 5 days. Pluripotency markers Oct-4, Nanog, SSEA-1, ALP, and rex-1 were assessed using flow cytometry analysis and gene expression profiles and showed that the undifferentiated nature of the cells within the ESC aggregates was maintained. Colony-forming efficiencies and embryoid body formation tests of the expanded cultures demonstrated that characteristic functional attributes of undifferentiated cells were not lost. Overcoming a major impediment in the area of ESC expansion, this study describes a successful process for the controlled and reproducible largescale expansion of ESCs using suspension culture bioreactors.
Collapse
Affiliation(s)
- Jaymi T Cormier
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | | | | | | |
Collapse
|
38
|
Ward CM, Eastham AM, Stern PL. Cell surface 5T4 antigen is transiently upregulated during early human embryonic stem cell differentiation: effect of 5T4 phenotype on neural lineage formation. Exp Cell Res 2006; 312:1713-26. [PMID: 16616918 DOI: 10.1016/j.yexcr.2006.02.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2005] [Revised: 01/05/2006] [Accepted: 02/07/2006] [Indexed: 11/19/2022]
Abstract
The 5T4 oncofoetal antigen is a cell surface glycoprotein that is transiently expressed during mouse ES cell differentiation and correlates with decreased pluripotency of such cells. We show that 5T4 antigen is transiently unregulated during HES4 and H1 human ES cell differentiation and its expression correlates with loss of the pluripotent markers OCT-4 and Tra-1-60 and upregulation of transcript markers associated with the three primary germ layers. To confirm that absence of cell surface 5T4 antigen represents a pluripotent hES cell phenotype, we performed mechanical transfer of either 5T4-ve or 5T4+ve HES4 colonies identified using live cell staining. 5T4-ve transfers maintained expression of OCT-4 in over 90% of resultant colonies, whereas 5T4+ve transfers exhibited significantly lower numbers of OCT-4-expressing colonies (92 +/- 1.4 vs. 2.9 +/- 2.0%). Interestingly, low cell density 5T4-ve colony transfers exhibited increased numbers of OCT-4-expressing colonies compared to large 5T4-ve transfers (92 +/- 1.4 vs. 63.2 +/- 1.9%). 5T4-ve and 5T4+ve HES4 and H1 ES cell lines expressed markers representative of neuroectoderm lineages, and we assessed the formation of neural lineages from these phenotypes in serum-containing medium and N2B27 medium. Expression of 5T4 was found to be inversely related to the yield of tyrosine-hydroxylase (TH+)-expressing neurons in N2B27 medium, with additional mesoderm and endoderm transcript markers detected. Homogeneous glial cell populations were derived from low cell density 5T4-ve colony transfers cultured in serum-containing medium, with TH+ neuronal formation inhibited in a cell-density-dependent manner. We conclude that the 5T4 antigen is a transient marker of hES cell differentiation and that 5T4 phenotype, colony seeding density and culture conditions significantly influence the maintenance of pluripotent hES cells and their differentiation to neural lineages.
Collapse
Affiliation(s)
- Christopher M Ward
- Cancer Research UK Immunology Group, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Wilmslow Road, Manchester M20 4BX, UK.
| | | | | |
Collapse
|
39
|
Barrow KM, Ward CM, Rutter J, Ali S, Stern PL. Embryonic expression of murine 5T4 oncofoetal antigen is associated with morphogenetic events at implantation and in developing epithelia. Dev Dyn 2005; 233:1535-45. [PMID: 15977177 DOI: 10.1002/dvdy.20482] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Overexpression of 5T4 oncofoetal antigen, an early marker of ES cell differentiation, in vitro increases cellular motility and decreases adhesion, properties relevant to development and cancer. Embryonic expression of m5T4 antigen is first detected on trophectoderm at implantation and is restricted to extra-embryonic tissues to embryonic day (E) 11.5. In the embryo, significant m5T4 expression is detected at E12.5 in hindbrain roofplate and in various epithelia derived from all germ layers. In keratin 14-expressing epithelia, there is a congruent 5T4 expression pattern with many of these cells being Ki-67 positive. In brain, expression is observed in roofplate, ependymal layers, choroid plexus, and subventricular zones of lateral ventricles at E14.5. By E17.5, expression is decreased in the subventricular zone with further restriction to choroid plexus in adult brain. Our data demonstrate a limited 5T4 expression profile during embryogenesis associated with actively cycling, undifferentiated epithelial progenitor cells that may contribute to their migration.
Collapse
Affiliation(s)
- Katie M Barrow
- CR UK Immunology Group, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Manchester, United Kingdom
| | | | | | | | | |
Collapse
|
40
|
Ward CM, Barrow KM, Stern PL. Significant variations in differentiation properties between independent mouse ES cell lines cultured under defined conditions. Exp Cell Res 2004; 293:229-38. [PMID: 14729460 DOI: 10.1016/j.yexcr.2003.10.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Mouse embryonic stem (ES) cells are isolated from the inner cell mass (ICM)/epiblast of preimplantation embryos and are widely used in cell differentiation studies. We have previously observed differences in transcript and antigen expression following differentiation of ES cells lines in vitro. We have investigated this further by comparing the differentiation characteristics of five independently derived ES cell lines cultured and differentiated under defined conditions. Undifferentiated ES cell lines exhibited similar morphology and antigen/transcript marker expression. However, upon differentiation in monolayer culture by LIF withdrawal, only two of the lines expressed similar germ layer transcript profiles, and these were significantly altered compared to differentiation in serum-supplemented media. Neurofilament-68k was the only transcript marker common to all cell lines, however, induction of neuroectoderm lineages using 1 microM all-trans retinoic acid (RA) resulted in significant variations in cell number and morphology between the lines. Furthermore, neurons were only formed from clones of the two cell lines that exhibited similar transcript profiles, although the morphology was different between the two. We conclude that the independent ES cell lines in this study differ in their response to alterations in culture conditions in vitro, and the use of an appropriate cell line enables relatively homogeneous neuronal populations to be achieved in monolayer culture under defined conditions.
Collapse
Affiliation(s)
- Christopher M Ward
- Immunology Group, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Wilmslow Road, Manchester M20 4BX, UK.
| | | | | |
Collapse
|