1
|
Grenier JMP, Testut C, Bal M, Bardin F, De Grandis M, Gelsi-Boyer V, Vernerey J, Delahaye M, Granjeaud S, Zemmour C, Spinella JF, Chavakis T, Mancini SJC, Boher JM, Hébert J, Sauvageau G, Vey N, Schwaller J, Hospital MA, Fauriat C, Aurrand-Lions M. Genetic deletion of JAM-C in preleukemic cells rewires leukemic stem cell gene expression program in AML. Blood Adv 2024; 8:4662-4678. [PMID: 38954834 PMCID: PMC11402138 DOI: 10.1182/bloodadvances.2023011747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 05/23/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024] Open
Abstract
ABSTRACT The leukemic stem cell (LSC) score LSC-17 based on a stemness-related gene expression signature is an indicator of poor disease outcome in acute myeloid leukemia (AML). However, it is not known whether "niche anchoring" of LSC affects disease evolution. To address this issue, we conditionally inactivated the adhesion molecule JAM-C (Junctional Adhesion Molecule-C) expressed by hematopoietic stem cells (HSCs) and LSCs in an inducible mixed-lineage leukemia (iMLL)-AF9-driven AML mouse model. Deletion of Jam3 (encoding JAM-C) before induction of the leukemia-initiating iMLL-AF9 fusion resulted in a shift from long-term to short-term HSC expansion, without affecting disease initiation and progression. In vitro experiments showed that JAM-C controlled leukemic cell nesting irrespective of the bone marrow stromal cells used. RNA sequencing performed on leukemic HSCs isolated from diseased mice revealed that genes upregulated in Jam3-deficient animals belonged to activation protein-1 (AP-1) and tumor necrosis factor α (TNF-α)/NF-κB pathways. Human orthologs of dysregulated genes allowed to identify a score that was distinct from, and complementary to, the LSC-17 score. Substratification of patients with AML using LSC-17 and AP-1/TNF-α genes signature defined 4 groups with median survival ranging from <1 year to a median of "not reached" after 8 years. Finally, coculture experiments showed that AP-1 activation in leukemic cells was dependent on the nature of stromal cells. Altogether, our results identify the AP-1/TNF-α gene signature as a proxy of LSC anchoring in bone marrow niches, which improves the prognostic value of the LSC-17 score. This trial was registered at www.ClinicalTrials.gov as #NCT02320656.
Collapse
Affiliation(s)
- Julien M. P. Grenier
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
- UMR 7268, Aix-Marseille Université, EFS, CNRS, GENGLOBE, Marseille, France
| | - Céline Testut
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Matthieu Bal
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
- Département de la Recherche Clinique et de l’Innovation, Institut Paoli-Calmettes, Marseille, France
| | - Florence Bardin
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Maria De Grandis
- Aix-Marseille University, CNRS, EFS, ADES, Biologie des Groupes Sanguins, Marseille, France
- UMR 7268, Aix-Marseille Université, EFS, CNRS, GENGLOBE, Marseille, France
| | - Véronique Gelsi-Boyer
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Julien Vernerey
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Marjorie Delahaye
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Samuel Granjeaud
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Christophe Zemmour
- Département de la Recherche Clinique et de l’Innovation, Institut Paoli-Calmettes, Marseille, France
| | - Jean-François Spinella
- Laboratory of Molecular Genetics of Stem Cells, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Stéphane J. C. Mancini
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Jean-Marie Boher
- Département de la Recherche Clinique et de l’Innovation, Institut Paoli-Calmettes, Marseille, France
| | - Josée Hébert
- Division of Hematology-Oncology, Department of Medicine, Maisonneuve-Rosemont Hospital, Université de Montréal, Montreal, QC, Canada
| | - Guy Sauvageau
- Laboratory of Molecular Genetics of Stem Cells, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada
| | - Norbert Vey
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Jürg Schwaller
- Department of Biomedicine, University Children’s Hospital, University of Basel, Basel, Switzerland
| | | | - Cyril Fauriat
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Michel Aurrand-Lions
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| |
Collapse
|
2
|
Ishida T, Heck AM, Varnum-Finney B, Dozono S, Nourigat-McKay C, Kraskouskas K, Wellington R, Waltner O, Root, Jackson DL, Delaney C, Rafii S, Bernstein ID, Trapnell, Hadland B. Differentiation latency and dormancy signatures define fetal liver HSCs at single cell resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.01.543314. [PMID: 37333272 PMCID: PMC10274697 DOI: 10.1101/2023.06.01.543314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Decoding the gene regulatory mechanisms mediating self-renewal of hematopoietic stem cells (HSCs) during their amplification in the fetal liver (FL) is relevant for advancing therapeutic applications aiming to expand transplantable HSCs, a long-standing challenge. Here, to explore intrinsic and extrinsic regulation of self-renewal in FL-HSCs at the single cell level, we engineered a culture platform designed to recapitulate the FL endothelial niche, which supports the amplification of serially engraftable HSCs ex vivo. Leveraging this platform in combination with single cell index flow cytometry, serial transplantation assays, and single cell RNA-sequencing, we elucidated previously unrecognized heterogeneity in immunophenotypically defined FL-HSCs and demonstrated that differentiation latency and transcriptional signatures of biosynthetic dormancy are distinguishing properties of self-renewing FL-HSCs with capacity for serial, long-term multilineage hematopoietic reconstitution. Altogether, our findings provide key insights into HSC expansion and generate a novel resource for future exploration of the intrinsic and niche-derived signaling pathways that support FL-HSC self-renewal.
Collapse
Affiliation(s)
- Takashi Ishida
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Adam M. Heck
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Barbara Varnum-Finney
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Stacey Dozono
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Cynthia Nourigat-McKay
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Katie Kraskouskas
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Rachel Wellington
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology, School of Medicine, University of Washington, Seattle, WA
| | - Olivia Waltner
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Root
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Dana L Jackson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Colleen Delaney
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Deverra Therapeutics, Seattle, WA, USA
- Division of Pediatric Hematology/Oncology, University of Washington, Seattle, WA, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Irwin D. Bernstein
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Pediatric Hematology/Oncology, University of Washington, Seattle, WA, USA
| | - Trapnell
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Brandon Hadland
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Pediatric Hematology/Oncology, University of Washington, Seattle, WA, USA
| |
Collapse
|
3
|
Grenier JMP, Testut C, Fauriat C, Mancini SJC, Aurrand-Lions M. Adhesion Molecules Involved in Stem Cell Niche Retention During Normal Haematopoiesis and in Acute Myeloid Leukaemia. Front Immunol 2021; 12:756231. [PMID: 34867994 PMCID: PMC8636127 DOI: 10.3389/fimmu.2021.756231] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/27/2021] [Indexed: 12/11/2022] Open
Abstract
In the bone marrow (BM) of adult mammals, haematopoietic stem cells (HSCs) are retained in micro-anatomical structures by adhesion molecules that regulate HSC quiescence, proliferation and commitment. During decades, researchers have used engraftment to study the function of adhesion molecules in HSC's homeostasis regulation. Since the 90's, progress in genetically engineered mouse models has allowed a better understanding of adhesion molecules involved in HSCs regulation by BM niches and raised questions about the role of adhesion mechanisms in conferring drug resistance to cancer cells nested in the BM. This has been especially studied in acute myeloid leukaemia (AML) which was the first disease in which the concept of cancer stem cell (CSC) or leukemic stem cells (LSCs) was demonstrated. In AML, it has been proposed that LSCs propagate the disease and are able to replenish the leukemic bulk after complete remission suggesting that LSC may be endowed with drug resistance properties. However, whether such properties are due to extrinsic or intrinsic molecular mechanisms, fully or partially supported by molecular crosstalk between LSCs and surrounding BM micro-environment is still matter of debate. In this review, we focus on adhesion molecules that have been involved in HSCs or LSCs anchoring to BM niches and discuss if inhibition of such mechanism may represent new therapeutic avenues to eradicate LSCs.
Collapse
Affiliation(s)
- Julien M P Grenier
- Aix-Marseille Université, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Paoli Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Labellisée Ligue Nationale Contre le Cancer 2020, Marseille, France
| | - Céline Testut
- Aix-Marseille Université, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Paoli Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Labellisée Ligue Nationale Contre le Cancer 2020, Marseille, France
| | - Cyril Fauriat
- Aix-Marseille Université, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Paoli Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Labellisée Ligue Nationale Contre le Cancer 2020, Marseille, France
| | - Stéphane J C Mancini
- Aix-Marseille Université, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Paoli Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Labellisée Ligue Nationale Contre le Cancer 2020, Marseille, France
| | - Michel Aurrand-Lions
- Aix-Marseille Université, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Paoli Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Labellisée Ligue Nationale Contre le Cancer 2020, Marseille, France
| |
Collapse
|
4
|
Gorshkova O, Cappaï J, Maillot L, Sergé A. Analyzing normal and disrupted leukemic stem cell adhesion to bone marrow stromal cells by single-molecule tracking nanoscopy. J Cell Sci 2021; 134:271951. [PMID: 34435622 DOI: 10.1242/jcs.258736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/02/2021] [Indexed: 01/02/2023] Open
Abstract
Leukemic stem cells (LSCs) adhere to bone niches through adhesion molecules. These interactions, which are deeply reorganized in tumors, contribute to LSC resistance to chemotherapy and leukemia relapse. However, LSC adhesion mechanisms and potential therapeutic disruption using blocking antibodies remain largely unknown. Junctional adhesion molecule C (JAM-C, also known as JAM3) overexpression by LSCs correlates with increased leukemia severity, and thus constitutes a putative therapeutic target. Here, we took advantage of the ability of nanoscopy to detect single molecules with nanometric accuracy to characterize junctional adhesion molecule (JAM) dynamics at leuko-stromal contacts. Videonanoscopy trajectories were reconstructed using our dedicated multi-target tracing algorithm, pipelined with dual-color analyses (MTT2col). JAM-C expressed by LSCs engaged in transient interactions with JAM-B (also known as JAM2) expressed by stromal cells. JAM recruitment and colocalization at cell contacts were proportional to JAM-C level and reduced by a blocking anti-JAM-C antibody. MTT2col revealed, at single-molecule resolution, the ability of blocking antibodies to destabilize LSC binding to their niches, opening opportunities for disrupting LSC resistance mechanisms.
Collapse
Affiliation(s)
- Oksana Gorshkova
- Centre de recherche en cancérologie de Marseille (CRCM), Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (Inserm), Institut Paoli-Calmettes (IPC), Aix-Marseille Université, F-13273 Marseille, France
| | - Jessica Cappaï
- Centre de recherche en cancérologie de Marseille (CRCM), Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (Inserm), Institut Paoli-Calmettes (IPC), Aix-Marseille Université, F-13273 Marseille, France
| | - Loriane Maillot
- Laboratoire adhésion inflammation (LAI), Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (Inserm), Aix-Marseille Université, F-13288 Marseille, France
| | - Arnauld Sergé
- Centre de recherche en cancérologie de Marseille (CRCM), Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (Inserm), Institut Paoli-Calmettes (IPC), Aix-Marseille Université, F-13273 Marseille, France.,Laboratoire adhésion inflammation (LAI), Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (Inserm), Aix-Marseille Université, F-13288 Marseille, France
| |
Collapse
|
5
|
Flow Cytometry Analysis of Mouse Hematopoietic Stem and Multipotent Progenitor Cells. Methods Mol Biol 2021. [PMID: 34057715 DOI: 10.1007/978-1-0716-1425-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Flow cytometry has been widely used to detect a single event by means of multiparametric fluorescence measurements. Here we describe a method to analyze subsets of hematopoietic stem and progenitor cells isolated from long bones of mice. We further show that this method allows for comparing JAM-C protein expression between subsets of hematopoietic stem and progenitor cells.
Collapse
|
6
|
JAM-C/ Jam-C Expression Is Primarily Expressed in Mouse Hematopoietic Stem Cells. Hemasphere 2021; 5:e594. [PMID: 34131634 PMCID: PMC8196118 DOI: 10.1097/hs9.0000000000000594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/26/2021] [Indexed: 12/02/2022] Open
|
7
|
Henry E, Arcangeli ML. How Hematopoietic Stem Cells Respond to Irradiation: Similarities and Differences between Low and High Doses of Ionizing Radiations. Exp Hematol 2020; 94:11-19. [PMID: 33290858 DOI: 10.1016/j.exphem.2020.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/12/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022]
Abstract
In this review, we will specifically address the newest insights on the effect of low doses of ionizing radiations on the hematopoietic stem cells, which are prone to long-term deleterious effects. Impact of high doses of irradiation on hematopoietic cells has been widely studied over the years, in line with the risk of accidental or terrorist exposure to irradiation and with a particular attention to the sensitivity of the hematopoietic system. Recently, more studies have focused on lower doses of irradiation on different tissues, due to the increasing exposure caused by medical imaging, radiotherapy or plane travelling for instance. Hence, we will delineate similarities and discrepancies in HSC response to high and low doses of irradiation from these studies.
Collapse
Affiliation(s)
- Elia Henry
- Team Niche and Cancer in Hematopoiesis, U1274, INSERM, 92260 Fontenay-aux-Roses, France; Laboratory of Hematopoietic Stem Cells and Leukemia/Service Stem Cells and Radiation/iRCM/JACOB/DRF, CEA, Fontenay-aux-Roses, France; UMR Stabilité Génétique Cellules Souches et Radiations, Université de Paris, CEA, Fontenay-aux-Roses, France; UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris-Saclay, CEA, Fontenay-aux-Roses, France
| | - Marie-Laure Arcangeli
- Team Niche and Cancer in Hematopoiesis, U1274, INSERM, 92260 Fontenay-aux-Roses, France; Laboratory of Hematopoietic Stem Cells and Leukemia/Service Stem Cells and Radiation/iRCM/JACOB/DRF, CEA, Fontenay-aux-Roses, France; UMR Stabilité Génétique Cellules Souches et Radiations, Université de Paris, CEA, Fontenay-aux-Roses, France; UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris-Saclay, CEA, Fontenay-aux-Roses, France.
| |
Collapse
|
8
|
In Vivo Pre-Instructed HSCs Robustly Execute Asymmetric Cell Divisions In Vitro. Int J Mol Sci 2020; 21:ijms21218225. [PMID: 33153113 PMCID: PMC7663432 DOI: 10.3390/ijms21218225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/31/2020] [Accepted: 10/31/2020] [Indexed: 01/12/2023] Open
Abstract
Hematopoietic stem cells (HSCs) are responsible for life-long production of all mature blood cells. Under homeostasis, HSCs in their native bone marrow niches are believed to undergo asymmetric cell divisions (ACDs), with one daughter cell maintaining HSC identity and the other committing to differentiate into various mature blood cell types. Due to the lack of key niche signals, in vitro HSCs differentiate rapidly, making it challenging to capture and study ACD. To overcome this bottleneck, in this study, we used interferon alpha (IFNα) treatment to "pre-instruct" HSC fate directly in their native niche, and then systematically studied the fate of dividing HSCs in vitro at the single cell level via time-lapse analysis, as well as multigene and protein expression analysis. Triggering HSCs' exit from dormancy via IFNα was found to significantly increase the frequency of asynchronous divisions in paired daughter cells (PDCs). Using single-cell gene expression analyses, we identified 12 asymmetrically expressed genes in PDCs. Subsequent immunocytochemistry analysis showed that at least three of the candidates, i.e., Glut1, JAM3 and HK2, were asymmetrically distributed in PDCs. Functional validation of these observations by colony formation assays highlighted the implication of asymmetric distribution of these markers as hallmarks of HSCs, for example, to reliably discriminate committed and self-renewing daughter cells in dividing HSCs. Our data provided evidence for the importance of in vivo instructions in guiding HSC fate, especially ACD, and shed light on putative molecular players involved in this process. Understanding the mechanisms of cell fate decision making should enable the development of improved HSC expansion protocols for therapeutic applications.
Collapse
|
9
|
Yu X, Sun P, Huang X, Chen H, Huang W, Ruan Y, Jiang W, Tan X, Liu Z. RNA-seq reveals tight junction-relevant erythropoietic fate induced by OCT4 in human hair follicle mesenchymal stem cells. Stem Cell Res Ther 2020; 11:454. [PMID: 33109258 PMCID: PMC7590701 DOI: 10.1186/s13287-020-01976-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/14/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Human hair follicle mesenchymal stem cells (hHFMSCs) isolated from hair follicles possess multilineage differentiation potential. OCT4 is a gene critically associated with pluripotency properties. The cell morphology and adhesion of hHFMSCs significantly changed after transduction of OCT4 and two subpopulations emerged, including adherent cells and floating cell. Floating cells cultured in hematopoietic induction medium and stimulated with erythropoetic growth factors could transdifferentiate into mature erythrocytes, whereas adherent cells formed negligible hematopoietic colonies. The aim of this study was to reveal the role of cell morphology and adhesion on erythropoiesis induced by OCT4 in hHFMSCs and to characterize the molecular mechanisms involved. METHODS Floating cell was separated from adherent cell by centrifugation of the upper medium during cell culture. Cell size was observed through flow cytometry and cell adhesion was tested by disassociation and adhesion assays. RNA sequencing was performed to detect genome-wide transcriptomes and identify differentially expressed genes. GO enrichment analysis and KEGG pathway analysis were performed to analysis the functions and pathways enriched by differentially expressed genes. The expression of tight junction core members was verified by qPCR and Western blot. A regulatory network was constructed to figure out the relationship between cell adhesin, cytoskeleton, pluripotency, and hematopoiesis. RESULTS The overexpression of OCT4 influenced the morphology and adhesion of hHFMSCs. Transcripts in floating cells and adherent cells are quite different. Data analysis showed that upregulated genes in floating cells were mainly related to pluripotency, germ layer development (including hematopoiesis lineage development), and downregulated genes were mainly related to cell adhesion, cell junctions, and the cytoskeleton. Most molecules of the tight junction (TJ) pathway were downregulated and molecular homeostasis of the TJ was disturbed, as CLDNs were disrupted, and JAMs and TJPs were upregulated. The dynamic expression of cell adhesion-related gene E-cadherin and cytoskeleton-related gene ACTN2 might cause different morphology and adhesion. Finally, a regulatory network centered to OCT4 was constructed, which elucidated that he TJ pathway critically bridges pluripotency and hematopoiesis in a TJP1-dependent way. CONCLUSIONS Regulations of cell morphology and adhesion via the TJ pathway conducted by OCT4 might modulate hematopoiesis in hHFMSCs, thus developing potential mechanism of erythropoiesis in vitro.
Collapse
Affiliation(s)
- Xiaozhen Yu
- Department of Pathology, College of Basic Medical Sciences, Qingdao University, 308 Ningxia Road, Qingdao, 266071, Shandong, China
| | - Pengpeng Sun
- Department of Critical Care Medicine, Qingdao Center Hospital, affiliated with Qingdao University, 127 Siliunan Road, Qingdao, 266042, Shandong, China
| | - Xingang Huang
- Department of Pathology, Qingdao Municipal Hospital, affiliated with Qingdao University, 1 Jiaozhou Road, Qingdao, 266011, Shandong, China
| | - Hua Chen
- Department of Pathology, College of Basic Medical Sciences, Qingdao University, 308 Ningxia Road, Qingdao, 266071, Shandong, China.,Department of Pathology, Qingdao Municipal Hospital, affiliated with Qingdao University, 1 Jiaozhou Road, Qingdao, 266011, Shandong, China
| | - Weiqing Huang
- Department of Pathology, Qingdao Municipal Hospital, affiliated with Qingdao University, 1 Jiaozhou Road, Qingdao, 266011, Shandong, China
| | - Yingchun Ruan
- Department of Pathology, College of Basic Medical Sciences, Qingdao University, 308 Ningxia Road, Qingdao, 266071, Shandong, China
| | - Weina Jiang
- Department of Pathology, Qingdao Municipal Hospital, affiliated with Qingdao University, 1 Jiaozhou Road, Qingdao, 266011, Shandong, China
| | - Xiaohua Tan
- Department of Pathology, College of Basic Medical Sciences, Qingdao University, 308 Ningxia Road, Qingdao, 266071, Shandong, China
| | - Zhijing Liu
- Department of Pathology, Qingdao Municipal Hospital, affiliated with Qingdao University, 1 Jiaozhou Road, Qingdao, 266011, Shandong, China.
| |
Collapse
|
10
|
Radulovic V, van der Garde M, Koide S, Sigurdsson V, Lang S, Kaneko S, Miharada K. Junctional Adhesion Molecule 2 Represents a Subset of Hematopoietic Stem Cells with Enhanced Potential for T Lymphopoiesis. Cell Rep 2020; 27:2826-2836.e5. [PMID: 31167130 DOI: 10.1016/j.celrep.2019.05.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 03/18/2019] [Accepted: 05/06/2019] [Indexed: 01/29/2023] Open
Abstract
The distinct lineage potential is a key feature of hematopoietic stem cell (HSC) heterogeneity, but a subset of HSCs specialized for a single lymphoid compartment has not been identified. Here we report that HSCs expressing junctional adhesion molecule 2 (Jam2) at a higher level (Jam2high HSCs) have a greater T cell reconstitution capacity. Jam2high HSCs are metabolically dormant but preferentially differentiate toward lymphocytes, especially T cell lineages. Jam2high HSCs uniquely express T cell-related genes, and the interaction with Jam1 facilitates the Notch/Delta signaling pathway. Frequency of Jam2high HSCs changes upon T cell depletion in vivo, potentially suggesting that Jam2 expression may reflect scarcity of T cells and requirement of T cell replenishment. Our findings highlight Jam2 as a potential marker for a subfraction of HSCs with an extensive lymphopoietic capacity, mainly in T lymphopoiesis.
Collapse
Affiliation(s)
- Visnja Radulovic
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Mark van der Garde
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Shuhei Koide
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Valgardur Sigurdsson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Stefan Lang
- StemTherapy Bioinformatics Core Facility, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Shin Kaneko
- Center of iPS Cell Research and Application, Kyoto University, 606-8507 Kyoto, Japan
| | - Kenichi Miharada
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden.
| |
Collapse
|
11
|
Post-translational modifications of tight junction transmembrane proteins and their direct effect on barrier function. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183330. [PMID: 32376223 DOI: 10.1016/j.bbamem.2020.183330] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/21/2020] [Accepted: 04/27/2020] [Indexed: 12/24/2022]
Abstract
Post-translational modifications (PTMs) such as phosphorylation, ubiquitination or glycosylation are processes affecting the conformation, stability, localization and function of proteins. There is clear evidence that PTMs can act upon tight junction (TJ) proteins, thus modulating epithelial barrier function. Compared to transcriptional or translational regulation, PTMs are rapid and more dynamic processes so in the context of barrier maintenance they might be essential for coping with changing environmental or external impacts. The aim of this review is to extract literature deciphering PTMs in TJ proteins directly contributing to epithelial barrier changes in permeability to ions and macromolecules. It is not intended to cover the entire scope of PTMs in TJ proteins and should rather be understood as a digest of TJ protein modifications directly resulting in the tightening or opening of the epithelial barrier.
Collapse
|
12
|
Graham HK, Duan X. Molecular mechanisms regulating synaptic specificity and retinal circuit formation. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e379. [PMID: 32267095 DOI: 10.1002/wdev.379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 03/13/2020] [Accepted: 03/19/2020] [Indexed: 12/28/2022]
Abstract
The central nervous system (CNS) is composed of precisely assembled circuits which support a variety of physiological functions and behaviors. These circuits include multiple subtypes of neurons with unique morphologies, electrical properties, and molecular identities. How these component parts are precisely wired-up has been a topic of great interest to the field of developmental neurobiology and has implications for our understanding of the etiology of many neurological disorders and mental illnesses. To date, many molecules involved in synaptic choice and specificity have been identified, including members of several families of cell-adhesion molecules (CAMs), which are cell-surface molecules that mediate cell-cell contacts and subsequent intracellular signaling. One favored hypothesis is that unique expression patterns of CAMs define specific neuronal subtype populations and determine compatible pre- and postsynaptic neuronal partners based on the expression of these unique CAMs. The mouse retina has served as a beautiful model for investigations into mammalian CAM interactions due to its well-defined neuronal subtypes and distinct circuits. Moreover, the retina is readily amenable to visualization of circuit organization and electrophysiological measurement of circuit function. The advent of recent genetic, genomic, and imaging technologies has opened the field up to large-scale, unbiased approaches for identification of new molecular determinants of synaptic specificity. Thus, building on the foundation of work reviewed here, we can expect rapid expansion of the field, harnessing the mouse retina as a model to understand the molecular basis for synaptic specificity and functional circuit assembly. This article is categorized under: Nervous System Development > Vertebrates: General Principles Nervous System Development > Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Hannah K Graham
- Department of Ophthalmology, University of California San Francisco, San Francisco, California, USA.,Neuroscience Graduate Program, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Xin Duan
- Department of Ophthalmology, University of California San Francisco, San Francisco, California, USA.,Neuroscience Graduate Program, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA.,Department of Physiology, University of California San Francisco, San Francisco, California, USA.,Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
13
|
Bailly AL, Grenier JMP, Cartier-Michaud A, Bardin F, Balzano M, Goubard A, Lissitzky JC, De Grandis M, Mancini SJC, Serge A, Aurrand-Lions M. GRASP55 Is Dispensable for Normal Hematopoiesis but Necessary for Myc-Dependent Leukemic Growth. THE JOURNAL OF IMMUNOLOGY 2020; 204:2685-2696. [PMID: 32229537 DOI: 10.4049/jimmunol.1901124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 03/03/2020] [Indexed: 11/19/2022]
Abstract
Grasp55 is a ubiquitous Golgi stacking protein involved in autophagy, protein trafficking, and glucose deprivation sensing. The function of Grasp55 in protein trafficking has been attributed to its PDZ-mediated interaction with the C-terminal PDZ-binding motifs of protein cargos. We have recently shown that such an interaction occurs between Grasp55 and the adhesion molecule Jam-C, which plays a central role in stemness maintenance of hematopoietic and spermatogenic cells. Accordingly, we have found that Grasp55-deficient mice suffer from spermatogenesis defects similar to Jam-C knockout mice. However, whether Grasp55 is involved in the maintenance of immunohematopoietic homeostasis through regulation of protein transport and Jam-C expression remains unknown. In this study, we show that Grasp55 deficiency does not affect hematopoietic stem cell differentiation, engraftment, or mobilization, which are known to depend on expression of Grasp55-dependent protein cargos. In contrast, using an Myc-dependent leukemic model addicted to autophagy, we show that knockdown of Grasp55 in leukemic cells reduces spleen and bone marrow tumor burden upon i.v. leukemic engraftment. This is not due to reduced homing of Grasp55-deficient cells to these organs but to increased spontaneous apoptosis of Grasp55-deficient leukemic cells correlated with increased sensitivity of the cells to glucose deprivation. These results show that Grasp55 plays a role in Myc-transformed hematopoietic cells but not in normal hematopoietic cells in vivo.
Collapse
Affiliation(s)
- Anne-Laure Bailly
- Equipe Labellisée Ligue contre le Cancer, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Cancerology Research Center of Marseille, Marseille 13009, France; and
| | - Julien M P Grenier
- Equipe Labellisée Ligue contre le Cancer, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Cancerology Research Center of Marseille, Marseille 13009, France; and
| | - Amandine Cartier-Michaud
- Equipe Labellisée Ligue contre le Cancer, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Cancerology Research Center of Marseille, Marseille 13009, France; and
| | - Florence Bardin
- Equipe Labellisée Ligue contre le Cancer, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Cancerology Research Center of Marseille, Marseille 13009, France; and
| | - Marielle Balzano
- Equipe Labellisée Ligue contre le Cancer, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Cancerology Research Center of Marseille, Marseille 13009, France; and
| | - Armelle Goubard
- Equipe Labellisée Ligue contre le Cancer, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Cancerology Research Center of Marseille, Marseille 13009, France; and
| | - Jean-Claude Lissitzky
- Equipe Labellisée Ligue contre le Cancer, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Cancerology Research Center of Marseille, Marseille 13009, France; and
| | - Maria De Grandis
- Etablissement Français du Sang PACA Corse, Biologie des Groupes Sanguins, UMR 7268, Aix Marseille Université, CNRS, Marseille 13005, France
| | - Stéphane J C Mancini
- Equipe Labellisée Ligue contre le Cancer, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Cancerology Research Center of Marseille, Marseille 13009, France; and
| | - Arnauld Serge
- Equipe Labellisée Ligue contre le Cancer, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Cancerology Research Center of Marseille, Marseille 13009, France; and
| | - Michel Aurrand-Lions
- Equipe Labellisée Ligue contre le Cancer, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Cancerology Research Center of Marseille, Marseille 13009, France; and
| |
Collapse
|
14
|
Henry E, Souissi-Sahraoui I, Deynoux M, Lefèvre A, Barroca V, Campalans A, Ménard V, Calvo J, Pflumio F, Arcangeli ML. Human hematopoietic stem/progenitor cells display reactive oxygen species-dependent long-term hematopoietic defects after exposure to low doses of ionizing radiations. Haematologica 2019; 105:2044-2055. [PMID: 31780635 PMCID: PMC7395291 DOI: 10.3324/haematol.2019.226936] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/27/2019] [Indexed: 01/16/2023] Open
Abstract
Hematopoietic stem cells are responsible for life-long blood cell production and are highly sensitive to exogenous stresses. The effects of low doses of ionizing radiations on radiosensitive tissues such as the hematopoietic tissue are still unknown despite their increasing use in medical imaging. Here, we study the consequences of low doses of ionizing radiations on differentiation and self-renewal capacities of human primary hematopoietic stem/progenitor cells (HSPC). We found that a single 20 mGy dose impairs the hematopoietic reconstitution potential of human HSPC but not their differentiation properties. In contrast to high irradiation doses, low doses of irradiation do not induce DNA double strand breaks in HSPC but, similar to high doses, induce a rapid and transient increase of reactive oxygen species (ROS) that promotes activation of the p38MAPK pathway. HSPC treatment with ROS scavengers or p38MAPK inhibitor prior exposure to 20 mGy irradiation abolishes the 20 mGy-induced defects indicating that ROS and p38MAPK pathways are transducers of low doses of radiation effects. Taken together, these results show that a 20 mGy dose of ionizing radiation reduces the reconstitution potential of HSPC suggesting an effect on the self-renewal potential of human hematopoietic stem cells and pinpointing ROS or the p38MAPK as therapeutic targets. Inhibition of ROS or the p38MAPK pathway protects human primary HSPC from low-dose irradiation toxicity.
Collapse
Affiliation(s)
- Elia Henry
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis".,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| | - Inès Souissi-Sahraoui
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis".,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| | - Margaux Deynoux
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis".,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| | - Andréas Lefèvre
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis".,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| | - Vilma Barroca
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis".,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| | - Anna Campalans
- UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay.,CEA, DRF-JACOB-IRCM-SIGRR-LRIG, UMR "Genetic stability, Stem Cells and Radiation"
| | - Véronique Ménard
- UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay.,UMR "Genetic stability, Stem Cells and Radiation", F-92265 Fontenay-aux-Roses, France
| | - Julien Calvo
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis".,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| | - Françoise Pflumio
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis".,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| | - Marie-Laure Arcangeli
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis" .,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| |
Collapse
|
15
|
Herrmann M, Jakob F. Bone Marrow Niches for Skeletal Progenitor Cells and their Inhabitants in Health and Disease. Curr Stem Cell Res Ther 2019; 14:305-319. [PMID: 30674266 DOI: 10.2174/1574888x14666190123161447] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/04/2018] [Accepted: 01/02/2019] [Indexed: 12/19/2022]
Abstract
The bone marrow hosts skeletal progenitor cells which have most widely been referred to as Mesenchymal Stem or Stromal Cells (MSCs), a heterogeneous population of adult stem cells possessing the potential for self-renewal and multilineage differentiation. A consensus agreement on minimal criteria has been suggested to define MSCs in vitro, including adhesion to plastic, expression of typical surface markers and the ability to differentiate towards the adipogenic, osteogenic and chondrogenic lineages but they are critically discussed since the differentiation capability of cells could not always be confirmed by stringent assays in vivo. However, these in vitro characteristics have led to the notion that progenitor cell populations, similar to MSCs in bone marrow, reside in various tissues. MSCs are in the focus of numerous (pre)clinical studies on tissue regeneration and repair. Recent advances in terms of genetic animal models enabled a couple of studies targeting skeletal progenitor cells in vivo. Accordingly, different skeletal progenitor cell populations could be identified by the expression of surface markers including nestin and leptin receptor. While there are still issues with the identity of, and the overlap between different cell populations, these studies suggested that specific microenvironments, referred to as niches, host and maintain skeletal progenitor cells in the bone marrow. Dynamic mutual interactions through biological and physical cues between niche constituting cells and niche inhabitants control dormancy, symmetric and asymmetric cell division and lineage commitment. Niche constituting cells, inhabitant cells and their extracellular matrix are subject to influences of aging and disease e.g. via cellular modulators. Protective niches can be hijacked and abused by metastasizing tumor cells, and may even be adapted via mutual education. Here, we summarize the current knowledge on bone marrow skeletal progenitor cell niches in physiology and pathophysiology. We discuss the plasticity and dynamics of bone marrow niches as well as future perspectives of targeting niches for therapeutic strategies.
Collapse
Affiliation(s)
- Marietta Herrmann
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Clinics Wuerzburg, Wuerzburg, Germany.,Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
16
|
Kobayashi I, Kobayashi-Sun J, Hirakawa Y, Ouchi M, Yasuda K, Kamei H, Fukuhara S, Yamaguchi M. Dual role of Jam3b in early hematopoietic and vascular development. Development 2019; 147:dev.181040. [DOI: 10.1242/dev.181040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 12/11/2019] [Indexed: 12/23/2022]
Abstract
In order to efficiently derive hematopoietic stem cells (HSCs) from pluripotent precursors, it is crucial to understand how mesodermal cells acquire hematopoietic and endothelial identities, two divergent, but closely related cell fates. Although Npas4 has been recently identified as a conserved master regulator of hemato-vascular development, the molecular mechanisms underlying cell fate divergence between hematopoietic and vascular endothelial cells are still unclear. Here, we show in zebrafish that mesodermal cell differentiation into hematopoietic and vascular endothelial cells is regulated by Junctional adhesion molecule 3b (Jam3b) via two independent signaling pathways. Mutation of jam3b led to a reduction in npas4l expression in the posterior lateral plate mesoderm and defects in both hematopoietic and vascular development. Mechanistically, we uncover that Jam3b promotes endothelial specification by regulating npas4l expression through repression of the Rap1a-Erk signaling cascade. Jam3b subsequently promotes hematopoietic development, including HSCs, by regulating lrrc15 expression in endothelial precursors through the activation of an integrin-dependent signaling cascade. Our data provide insight into the divergent mechanisms for instructing hematopoietic or vascular fates from mesodermal cells.
Collapse
Affiliation(s)
- Isao Kobayashi
- Faculty of Biological Science and Technology, Institute of Science and Engineering, Kanazawa University, Ishikawa, Japan
| | - Jingjing Kobayashi-Sun
- Division of Life Sciences, Graduate School of Natural Science and Technology, Kanazawa University, Ishikawa, Japan
| | - Yuto Hirakawa
- Division of Life Sciences, Graduate School of Natural Science and Technology, Kanazawa University, Ishikawa, Japan
| | - Madoka Ouchi
- Division of Life Sciences, Graduate School of Natural Science and Technology, Kanazawa University, Ishikawa, Japan
| | - Koyuki Yasuda
- Faculty of Natural System, Institute of Science and Engineering, Kanazawa University, Ishikawa, Japan
| | - Hiroyasu Kamei
- Faculty of Biological Science and Technology, Institute of Science and Engineering, Kanazawa University, Ishikawa, Japan
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School, Kanagawa, Japan
| | - Masaaki Yamaguchi
- Faculty of Biological Science and Technology, Institute of Science and Engineering, Kanazawa University, Ishikawa, Japan
| |
Collapse
|
17
|
Tietz S, Périnat T, Greene G, Enzmann G, Deutsch U, Adams R, Imhof B, Aurrand-Lions M, Engelhardt B. Lack of junctional adhesion molecule (JAM)-B ameliorates experimental autoimmune encephalomyelitis. Brain Behav Immun 2018; 73:3-20. [PMID: 29920328 DOI: 10.1016/j.bbi.2018.06.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/11/2018] [Accepted: 06/15/2018] [Indexed: 12/28/2022] Open
Abstract
In multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE) autoaggressive CD4+ T cells cross the blood-brain barrier (BBB) and cause neuroinflammation. Therapeutic targeting of CD4+ T-cell trafficking into the CNS by blocking α4-integrins has proven beneficial for the treatment of MS but comes with associated risks, probably due to blocking CD8+ T cell mediated CNS immune surveillance. Our recent observations show that CD8+ T cells also rely on α4β1-integrins to cross the BBB. Besides vascular cell adhesion molecule-1 (VCAM-1), we identified junctional adhesion molecule-B (JAM-B) as a novel vascular α4β1-integrin ligand involved in CD8+ T-cell migration across the BBB. This prompted us to investigate, if JAM-B also mediates CD4+ T-cell migration across the BBB. We first ensured that encephalitogenic T cells can bind to JAM-B in vitro and next compared EAE pathogenesis in JAM-B-/- C57BL/6J mice and their wild-type littermates. Following immunization with MOGaa35-55 peptide, JAM-B-/- mice developed ameliorated EAE compared to their wild-type littermates. At the same time, we isolated higher numbers of CD45+ infiltrating immune cells from the CNS of JAM-B-/- C57BL/6J mice suffering from EAE. Immunofluorescence staining revealed that the majority of CD45+ inflammatory cells accumulated in the leptomeningeal and perivascular spaces of the CNS behind the BBB but do not gain access to the CNS parenchyma. Trapping of CNS inflammatory cells was not due to increased inflammatory cell proliferation. Neither a loss of BBB integrity or BBB polarity potentially affecting local chemokine gradients nor a lack of focal gelatinase activation required for CNS parenchymal immune cell entry across the glia limitans could be detected in JAM-B-/- mice. Lack of a role for JAM-B in the effector phase of EAE was supported by the observation that we did not detect any role for JAM-B in EAE pathogenesis, when EAE was elicited by in vitro activated MOG aa35-55-specific CD4+ effector T cells. On the other hand, we also failed to demonstrate any role of JAM-B in in vivo priming, proliferation or polarization of MOGaa35-55-specific CD4+ T cells in peripheral immune organs. Finally, our study excludes expression of and thus a role for JAM-B on peripheral and CNS infiltrating myeloid cells. Taken together, although endothelial JAM-B is not required for immune cell trafficking across the BBB in EAE, in its absence accumulation of inflammatory cells mainly in CNS leptomeningeal spaces leads to amelioration of EAE.
Collapse
MESH Headings
- Animals
- Blood-Brain Barrier/metabolism
- CD8-Positive T-Lymphocytes/metabolism
- Cell Movement/physiology
- Central Nervous System/metabolism
- Central Nervous System/physiology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Endothelium, Vascular/metabolism
- Female
- Integrin alpha4beta1/metabolism
- Junctional Adhesion Molecule B/genetics
- Junctional Adhesion Molecule B/metabolism
- Junctional Adhesion Molecule B/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/physiopathology
- Myelin-Oligodendrocyte Glycoprotein/pharmacology
- Myeloid Cells/metabolism
- Myeloid Cells/physiology
- Tight Junctions/metabolism
Collapse
Affiliation(s)
- Silvia Tietz
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Therese Périnat
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Gretchen Greene
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Gaby Enzmann
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Urban Deutsch
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Ralf Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Beat Imhof
- Department of Pathology and Immunology, University of Geneva, CMU Geneva, Switzerland
| | - Michel Aurrand-Lions
- Centre de Recherche en Cancerologie de Marseille, INSERM, CNRS, Aix-Marseille University, Marseille, France
| | | |
Collapse
|
18
|
Hamidi S, Sheng G. Epithelial-mesenchymal transition in haematopoietic stem cell development and homeostasis. J Biochem 2018; 164:265-275. [PMID: 30020470 DOI: 10.1093/jb/mvy063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/14/2018] [Indexed: 01/03/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a morphogenetic process of cells that adopt an epithelial organization in their developmental ontogeny or homeostatic maintenance. Abnormalities in EMT regulation result in many malignant tumours in the human body. Tumours associated with the haematopoietic system, however, are traditionally not considered to involve EMT and haematopoietic stem cells (HSCs) are generally not associated with epithelial characteristics. In this review, we discuss the ontogeny and homeostasis of adult HSCs in the context of EMT intermediate states. We provide evidence that cell polarity regulation is critical for both HSC formation from embryonic dorsal aorta and HSC self-renewal and differentiation in adult bone marrow. HSC polarity is controlled by the same set of surface and transcriptional regulators as those described in canonical EMT processes. With an emphasis on partial EMT, we propose that the concept of EMT can be similarly applied in the study of HSC generation, maintenance and pathogenesis.
Collapse
Affiliation(s)
- Sofiane Hamidi
- Laboratory of Developmental Morphogenesis, International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Guojun Sheng
- Laboratory of Developmental Morphogenesis, International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
19
|
Heterocellular molecular contacts in the mammalian stem cell niche. Eur J Cell Biol 2018; 97:442-461. [PMID: 30025618 DOI: 10.1016/j.ejcb.2018.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 07/03/2018] [Indexed: 12/16/2022] Open
Abstract
Adult tissue homeostasis and repair relies on prompt and appropriate intervention by tissue-specific adult stem cells (SCs). SCs have the ability to self-renew; upon appropriate stimulation, they proliferate and give rise to specialized cells. An array of environmental signals is important for maintenance of the SC pool and SC survival, behavior, and fate. Within this special microenvironment, commonly known as the stem cell niche (SCN), SC behavior and fate are regulated by soluble molecules and direct molecular contacts via adhesion molecules providing connections to local supporting cells and the extracellular matrix. Besides the extensively discussed array of soluble molecules, the expression of adhesion molecules and molecular contacts is another fundamental mechanism regulating niche occupancy and SC mobilization upon activation. Some adhesion molecules are differentially expressed and have tissue-specific consequences, likely reflecting the structural differences in niche composition and design, especially the presence or absence of a stromal counterpart. However, the distribution and identity of intercellular molecular contacts for adhesion and adhesion-mediated signaling within stromal and non-stromal SCN have not been thoroughly studied. This review highlights common details or significant differences in cell-to-cell contacts within representative stromal and non-stromal niches that could unveil new standpoints for stem cell biology and therapy.
Collapse
|
20
|
Hintermann E, Bayer M, Conti CB, Fuchs S, Fausther M, Leung PS, Aurrand-Lions M, Taubert R, Pfeilschifter JM, Friedrich-Rust M, Schuppan D, Dranoff JA, Gershwin ME, Manns MP, Imhof BA, Christen U. Junctional adhesion molecules JAM-B and JAM-C promote autoimmune-mediated liver fibrosis in mice. J Autoimmun 2018; 91:83-96. [PMID: 29753567 DOI: 10.1016/j.jaut.2018.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/26/2018] [Accepted: 05/02/2018] [Indexed: 12/17/2022]
Abstract
Fibrosis remains a serious health concern in patients with chronic liver disease. We recently reported that chemically induced chronic murine liver injury triggers increased expression of junctional adhesion molecules (JAMs) JAM-B and JAM-C by endothelial cells and de novo synthesis of JAM-C by hepatic stellate cells (HSCs). Here, we demonstrate that biopsies of patients suffering from primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC) or autoimmune hepatitis (AIH) display elevated levels of JAM-C on portal fibroblasts (PFs), HSCs, endothelial cells and cholangiocytes, whereas smooth muscle cells expressed JAM-C constitutively. Therefore, localization and function of JAM-B and JAM-C were investigated in three mouse models of autoimmune-driven liver inflammation. A PBC-like disease was induced by immunization with 2-octynoic acid-BSA conjugate, which resulted in the upregulation of both JAMs in fibrotic portal triads. Analysis of a murine model of PSC revealed a role of JAM-C in PF cell-cell adhesion and contractility. In mice suffering from AIH, endothelial cells increased JAM-B level and HSCs and capsular fibroblasts became JAM-C-positive. Most importantly, AIH-mediated liver fibrosis was reduced in JAM-B-/- mice or when JAM-C was blocked by soluble recombinant JAM-C. Interestingly, loss of JAM-B/JAM-C function had no effect on leukocyte infiltration, suggesting that the well-documented function of JAMs in leukocyte recruitment to inflamed tissue was not effective in the tested chronic models. This might be different in patients and may even be complicated by the fact that human leukocytes express JAM-C. Our findings delineate JAM-C as a mediator of myofibroblast-operated contraction of the liver capsule, intrahepatic vasoconstriction and bile duct stricture. Due to its potential to interact heterophilically with endothelial JAM-B, JAM-C supports also HSC/PF mural cell function. Together, these properties allow JAM-B and JAM-C to actively participate in vascular remodeling associated with liver/biliary fibrosis and suggest them as valuable targets for anti-fibrosis therapies.
Collapse
Affiliation(s)
- Edith Hintermann
- Pharmazentrum Frankfurt, ZAFES, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany.
| | - Monika Bayer
- Pharmazentrum Frankfurt, ZAFES, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany.
| | - Clara Benedetta Conti
- Department of Internal Medicine 1, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany; Fondazione IRCCS Cà, Granda Ospedale Maggiore Policlinico, Department of Pathophysiology and Organ Transplantation, Milan, Italy.
| | - Sina Fuchs
- Pharmazentrum Frankfurt, ZAFES, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany.
| | - Michel Fausther
- Division of Gastroenterology and Hepatology, University of Arkansas, Little Rock, AR, USA.
| | - Patrick S Leung
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA, USA.
| | - Michel Aurrand-Lions
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France.
| | - Richard Taubert
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.
| | - Josef M Pfeilschifter
- Pharmazentrum Frankfurt, ZAFES, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany.
| | - Mireen Friedrich-Rust
- Department of Internal Medicine 1, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany.
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany.
| | - Jonathan A Dranoff
- Division of Gastroenterology and Hepatology, University of Arkansas, Little Rock, AR, USA.
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA, USA.
| | - Michael P Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.
| | - Beat A Imhof
- Department of Pathology and Immunology, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland.
| | - Urs Christen
- Pharmazentrum Frankfurt, ZAFES, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
21
|
Zhang Y, Xia F, Liu X, Yu Z, Xie L, Liu L, Chen C, Jiang H, Hao X, He X, Zhang F, Gu H, Zhu J, Bai H, Zhang CC, Chen GQ, Zheng J. JAM3 maintains leukemia-initiating cell self-renewal through LRP5/AKT/β-catenin/CCND1 signaling. J Clin Invest 2018; 128:1737-1751. [PMID: 29584620 DOI: 10.1172/jci93198] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 02/08/2018] [Indexed: 12/14/2022] Open
Abstract
Leukemia-initiating cells (LICs) are responsible for the initiation, development, and relapse of leukemia. The identification of novel therapeutic LIC targets is critical to curing leukemia. In this report, we reveal that junctional adhesion molecule 3 (JAM3) is highly enriched in both mouse and human LICs. Leukemogenesis is almost completely abrogated upon Jam3 deletion during serial transplantations in an MLL-AF9-induced murine acute myeloid leukemia model. In contrast, Jam3 deletion does not affect the functions of mouse hematopoietic stem cells. Moreover, knockdown of JAM3 leads to a dramatic decrease in the proliferation of both human leukemia cell lines and primary LICs. JAM3 directly associates with LRP5 to activate the downstream PDK1/AKT pathway, followed by the downregulation of GSK3β and activation of β-catenin/CCND1 signaling, to maintain the self-renewal ability and cell cycle entry of LICs. Thus, JAM3 may serve as a functional LIC marker and play an important role in the maintenance of LIC stemness through unexpected LRP5/PDK1/AKT/GSK3β/β-catenin/CCND1 signaling pathways but not via its canonical role in cell junctions and migration. JAM3 may be an ideal therapeutic target for the eradication of LICs without influencing normal hematopoiesis.
Collapse
Affiliation(s)
- Yaping Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangzhen Xia
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoye Liu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuo Yu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Xie
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ligen Liu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chiqi Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haishan Jiang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxin Hao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxiao He
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feifei Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Gu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Zhu
- Department of Hematology, First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haitao Bai
- Department of Hematology, First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng Cheng Zhang
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Guo-Qiang Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junke Zheng
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
De Grandis M, Mancini SJ, Aurrand-Lions M. In quest for leukemia initiating cells in AML. Oncoscience 2018; 5:9-10. [PMID: 29556512 PMCID: PMC5854287 DOI: 10.18632/oncoscience.394] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 02/14/2018] [Indexed: 01/22/2023] Open
Affiliation(s)
- Maria De Grandis
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Stéphane Jc Mancini
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Michel Aurrand-Lions
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| |
Collapse
|
23
|
Rot A, Massberg S, Khandoga AG, von Andrian UH. Chemokines and Hematopoietic Cell Trafficking. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00013-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
24
|
Liu J, Sanes JR. Cellular and Molecular Analysis of Dendritic Morphogenesis in a Retinal Cell Type That Senses Color Contrast and Ventral Motion. J Neurosci 2017; 37:12247-12262. [PMID: 29114073 PMCID: PMC5729193 DOI: 10.1523/jneurosci.2098-17.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/27/2017] [Accepted: 10/17/2017] [Indexed: 01/08/2023] Open
Abstract
As neuronal dendrites develop, they acquire cell-type-specific features including characteristic size, shape, arborization, location and synaptic patterns. These features, in turn, are major determinants of type-specific neuronal function. Because neuronal diversity complicates the task of relating developmental programs to adult structure and function, we analyzed dendritic morphogenesis in a single retinal ganglion cell (RGC) type in mouse called J-RGC. We documented the emergence of five dendritic features that underlie J-RGC physiology: (1) dendritic field size, which approximate receptive field size; (2) dendritic complexity, which affects how J-RGCs sample space; (3) asymmetry, which contributes to direction-selectivity; (4) restricted lamination within the inner plexiform layer (IPL), which renders J-RGCs responsive to light decrements; and (5) distribution of synaptic inputs, which generate a color-opponent receptive field. We found dendritic growth in J-RGCs is accompanied by a refinement in dendritic self-crossing. Asymmetry arises by a combination of selective pruning and elaboration, whereas laminar restriction results from biased outgrowth toward the outermost IPL. Interestingly, asymmetry develops in a protracted dorsoventral wave, whereas lamination does so in a rapid centrifugal wave. As arbors mature, they acquire excitatory and inhibitory synapses, with the latter forming first and being concentrated in proximal dendrites. Thus, distinct mechanisms operate in different spatiotemporal dimensions of J-RGC dendritic patterning to generate the substrate for specific patterns of synaptogenesis. Finally, we asked whether the defining molecular signature of J-RGCs, the adhesion molecule JAM-B, regulates morphogenesis, and showed that it promotes dendro-dendritic interactions. Our results reveal multiple mechanisms that shape a dendritic arbor.SIGNIFICANCE STATEMENT Visual perception begins in the retina, where distinct types of retinal ganglion cells (RGCs) are tuned to specific visual features such as direction of motion. The features to which each RGC type responds are determined largely by the number and type of synaptic inputs it receives, and these, in turn, are greatly influenced by the size, shape, arborization pattern, and location of its dendrites. We analyzed dendritic morphogenesis in a functionally characterized RGC type, the J-RGC, demonstrating distinct mechanisms that operate in different dimensions to generate the dendritic scaffold and synaptic patterns for feature detection. Our work elucidates cellular and molecular mechanisms that shape dendritic arbors and synaptic distribution, enabling J-RGC connectivity and thus, function.
Collapse
Affiliation(s)
- Jinyue Liu
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138, and
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts 02115
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138, and
| |
Collapse
|
25
|
Keyes BE, Fuchs E. Stem cells: Aging and transcriptional fingerprints. J Cell Biol 2017; 217:79-92. [PMID: 29070608 PMCID: PMC5748991 DOI: 10.1083/jcb.201708099] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/06/2017] [Accepted: 10/10/2017] [Indexed: 12/31/2022] Open
Abstract
Keyes and Fuchs discuss the decline in stem cell renewal and function with aging and the ensuing consequences on tissue homeostasis and regeneration. Stem cells are imbued with unique qualities. They have the capacity to propagate themselves through symmetric divisions and to divide asymmetrically to engender new cells that can progress to differentiate into tissue-specific, terminal cell types. Armed with these qualities, stem cells in adult tissues are tasked with replacing decaying cells and regenerating tissue after injury to maintain optimal tissue function. With increasing age, stem cell functional abilities decline, resulting in reduced organ function and delays in tissue repair. Here, we review the effect of aging in five well-studied adult murine stem cell populations and explore age-related declines in stem cell function and their consequences for stem cell self-renewal, tissue homeostasis, and regeneration. Finally, we examine transcriptional changes that have been documented in aged stem cell populations and discuss new questions and future directions that this collection of data has uncovered.
Collapse
Affiliation(s)
| | - Elaine Fuchs
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY
| |
Collapse
|
26
|
De Grandis M, Bardin F, Fauriat C, Zemmour C, El-Kaoutari A, Sergé A, Granjeaud S, Pouyet L, Montersino C, Chretien AS, Mozziconacci MJ, Castellano R, Bidaut G, Boher JM, Collette Y, Mancini SJC, Vey N, Aurrand-Lions M. JAM-C Identifies Src Family Kinase-Activated Leukemia-Initiating Cells and Predicts Poor Prognosis in Acute Myeloid Leukemia. Cancer Res 2017; 77:6627-6640. [PMID: 28972073 DOI: 10.1158/0008-5472.can-17-1223] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/24/2017] [Accepted: 09/25/2017] [Indexed: 11/16/2022]
Abstract
Acute myeloid leukemia (AML) originates from hematopoietic stem and progenitor cells that acquire somatic mutations, leading to disease and clonogenic evolution. AML is characterized by accumulation of immature myeloid cells in the bone marrow and phenotypic cellular heterogeneity reflective of normal hematopoietic differentiation. Here, we show that JAM-C expression defines a subset of leukemic cells endowed with leukemia-initiating cell activity (LIC). Stratification of de novo AML patients at diagnosis based on JAM-C-expressing cells frequencies in the blood served as an independent prognostic marker for disease outcome. Using publicly available leukemic stem cell (LSC) gene expression profiles and gene expression data generated from JAM-C-expressing leukemic cells, we defined a single cell core gene expression signature correlated to JAM-C expression that reveals LSC heterogeneity. Finally, we demonstrated that JAM-C controls Src family kinase (SFK) activation in LSC and that LIC with exacerbated SFK activation was uniquely found within the JAM-C-expressing LSC compartment. Cancer Res; 77(23); 6627-40. ©2017 AACR.
Collapse
Affiliation(s)
- Maria De Grandis
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Florence Bardin
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Cyril Fauriat
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Christophe Zemmour
- Unité de Biostatistique et de Méthodologie, Département de la Recherche Clinique et de l'Innovation, Institut Paoli-Calmettes, Marseille, France
| | | | - Arnauld Sergé
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Samuel Granjeaud
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Laurent Pouyet
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Camille Montersino
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Anne-Sophie Chretien
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Marie-Joelle Mozziconacci
- Département de Biopathologie, Cytogénétique et Biologie Moléculaire, Institut Paoli-Calmettes, Marseille, France
| | - Remy Castellano
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Ghislain Bidaut
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Jean-Marie Boher
- Unité de Biostatistique et de Méthodologie, Département de la Recherche Clinique et de l'Innovation, Institut Paoli-Calmettes, Marseille, France
- Aix Marseille Univ, INSERM, IRD, SESSTIM, Marseille, France
| | - Yves Collette
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Stéphane J C Mancini
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Norbert Vey
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
- Département d'Hématologie, Institut Paoli-Calmettes, Marseille, France
| | - Michel Aurrand-Lions
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France.
| |
Collapse
|
27
|
Ebnet K. Junctional Adhesion Molecules (JAMs): Cell Adhesion Receptors With Pleiotropic Functions in Cell Physiology and Development. Physiol Rev 2017; 97:1529-1554. [PMID: 28931565 DOI: 10.1152/physrev.00004.2017] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/04/2017] [Accepted: 05/11/2017] [Indexed: 02/06/2023] Open
Abstract
Junctional adhesion molecules (JAM)-A, -B and -C are cell-cell adhesion molecules of the immunoglobulin superfamily which are expressed by a variety of tissues, both during development and in the adult organism. Through their extracellular domains, they interact with other adhesion receptors on opposing cells. Through their cytoplasmic domains, they interact with PDZ domain-containing scaffolding and signaling proteins. In combination, these two properties regulate the assembly of signaling complexes at specific sites of cell-cell adhesion. The multitude of molecular interactions has enabled JAMs to adopt distinct cellular functions such as the regulation of cell-cell contact formation, cell migration, or mitotic spindle orientation. Not surprisingly, JAMs regulate diverse processes such as epithelial and endothelial barrier formation, hemostasis, angiogenesis, hematopoiesis, germ cell development, and the development of the central and peripheral nervous system. This review summarizes the recent progress in the understanding of JAMs, including their characteristic structural features, their molecular interactions, their cellular functions, and their contribution to a multitude of processes during vertebrate development and homeostasis.
Collapse
Affiliation(s)
- Klaus Ebnet
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity", Institute of Medical Biochemistry, ZMBE, Cells-In-Motion Cluster of Excellence (EXC1003-CiM), and Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany
| |
Collapse
|
28
|
Roch A, Giger S, Girotra M, Campos V, Vannini N, Naveiras O, Gobaa S, Lutolf MP. Single-cell analyses identify bioengineered niches for enhanced maintenance of hematopoietic stem cells. Nat Commun 2017; 8:221. [PMID: 28790449 PMCID: PMC5548907 DOI: 10.1038/s41467-017-00291-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 06/18/2017] [Indexed: 11/13/2022] Open
Abstract
The in vitro expansion of long-term hematopoietic stem cells (HSCs) remains a substantial challenge, largely because of our limited understanding of the mechanisms that control HSC fate choices. Using single-cell multigene expression analysis and time-lapse microscopy, here we define gene expression signatures and cell cycle hallmarks of murine HSCs and the earliest multipotent progenitors (MPPs), and analyze systematically single HSC fate choices in culture. Our analysis revealed twelve differentially expressed genes marking the quiescent HSC state, including four genes encoding cell–cell interaction signals in the niche. Under basal culture conditions, most HSCs rapidly commit to become early MPPs. In contrast, when we present ligands of the identified niche components such as JamC or Esam within artificial niches, HSC cycling is reduced and long-term multipotency in vivo is maintained. Our approach to bioengineer artificial niches should be useful in other stem cell systems. Haematopoietic stem cell (HSC) self-renewal is not sufficiently understood to recapitulate in vitro. Here, the authors generate gene signature and cell cycle hallmarks of single murine HSCs, and use identified endothelial receptors Esam and JamC as substrates to enhance HSC growth in engineered niches.
Collapse
Affiliation(s)
- Aline Roch
- Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Sonja Giger
- Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Mukul Girotra
- Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Vasco Campos
- Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Nicola Vannini
- Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Olaia Naveiras
- Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland.,Department of Medicine, Centre Hospitaler Universitaire Vaudois (CHUV), CH-1015, Lausanne, Switzerland
| | - Samy Gobaa
- Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Matthias P Lutolf
- Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland. .,Institute of Chemical Sciences and Engineering, School of Basic Sciences, EPFL, CH-1015, Lausanne, Switzerland.
| |
Collapse
|
29
|
Cartier-Michaud A, Bailly AL, Betzi S, Shi X, Lissitzky JC, Zarubica A, Sergé A, Roche P, Lugari A, Hamon V, Bardin F, Derviaux C, Lembo F, Audebert S, Marchetto S, Durand B, Borg JP, Shi N, Morelli X, Aurrand-Lions M. Genetic, structural, and chemical insights into the dual function of GRASP55 in germ cell Golgi remodeling and JAM-C polarized localization during spermatogenesis. PLoS Genet 2017; 13:e1006803. [PMID: 28617811 PMCID: PMC5472279 DOI: 10.1371/journal.pgen.1006803] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 05/05/2017] [Indexed: 01/01/2023] Open
Abstract
Spermatogenesis is a dynamic process that is regulated by adhesive interactions between germ and Sertoli cells. Germ cells express the Junctional Adhesion Molecule-C (JAM-C, encoded by Jam3), which localizes to germ/Sertoli cell contacts. JAM-C is involved in germ cell polarity and acrosome formation. Using a proteomic approach, we demonstrated that JAM-C interacted with the Golgi reassembly stacking protein of 55 kDa (GRASP55, encoded by Gorasp2) in developing germ cells. Generation and study of Gorasp2-/- mice revealed that knock-out mice suffered from spermatogenesis defects. Acrosome formation and polarized localization of JAM-C in spermatids were altered in Gorasp2-/- mice. In addition, Golgi morphology of spermatocytes was disturbed in Gorasp2-/- mice. Crystal structures of GRASP55 in complex with JAM-C or JAM-B revealed that GRASP55 interacted via PDZ-mediated interactions with JAMs and induced a conformational change in GRASP55 with respect of its free conformation. An in silico pharmacophore approach identified a chemical compound called Graspin that inhibited PDZ-mediated interactions of GRASP55 with JAMs. Treatment of mice with Graspin hampered the polarized localization of JAM-C in spermatids, induced the premature release of spermatids and affected the Golgi morphology of meiotic spermatocytes.
Collapse
Affiliation(s)
| | - Anne-Laure Bailly
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Stéphane Betzi
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Xiaoli Shi
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, China
| | | | - Ana Zarubica
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Arnauld Sergé
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Philippe Roche
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Adrien Lugari
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Véronique Hamon
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Florence Bardin
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Carine Derviaux
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Frédérique Lembo
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Stéphane Audebert
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Sylvie Marchetto
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Bénédicte Durand
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Institut NeuroMyoGène, Lyon, France
| | - Jean-Paul Borg
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Ning Shi
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, China
| | - Xavier Morelli
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Michel Aurrand-Lions
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
- * E-mail:
| |
Collapse
|
30
|
Redmond SA, Mei F, Eshed-Eisenbach Y, Osso LA, Leshkowitz D, Shen YAA, Kay JN, Aurrand-Lions M, Lyons DA, Peles E, Chan JR. Somatodendritic Expression of JAM2 Inhibits Oligodendrocyte Myelination. Neuron 2016; 91:824-836. [PMID: 27499083 DOI: 10.1016/j.neuron.2016.07.021] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 06/08/2016] [Accepted: 07/01/2016] [Indexed: 02/05/2023]
Abstract
Myelination occurs selectively around neuronal axons to increase the efficiency and velocity of action potentials. While oligodendrocytes are capable of myelinating permissive structures in the absence of molecular cues, structurally permissive neuronal somata and dendrites remain unmyelinated. Utilizing a purified spinal cord neuron-oligodendrocyte myelinating co-culture system, we demonstrate that disruption of dynamic neuron-oligodendrocyte signaling by chemical cross-linking results in aberrant myelination of the somatodendritic compartment of neurons. We hypothesize that an inhibitory somatodendritic cue is necessary to prevent non-axonal myelination. Using next-generation sequencing and candidate profiling, we identify neuronal junction adhesion molecule 2 (JAM2) as an inhibitory myelin-guidance molecule. Taken together, our results demonstrate that the somatodendritic compartment directly inhibits myelination and suggest a model in which broadly indiscriminate myelination is tailored by inhibitory signaling to meet local myelination requirements.
Collapse
Affiliation(s)
- Stephanie A Redmond
- Department of Neurology and Program in Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Feng Mei
- Department of Neurology and Program in Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yael Eshed-Eisenbach
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Lindsay A Osso
- Department of Neurology and Program in Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dena Leshkowitz
- Bioinformatics Unit, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yun-An A Shen
- Department of Neurology and Program in Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeremy N Kay
- Departments of Neurobiology and Ophthalmology, Duke University School of Medicine, Durham, NC 27703, USA
| | - Michel Aurrand-Lions
- Centre de Recherche en Cancérologie de Marseille, Inserm, CNRS, Aix-Marseille University, UMR1068, 13284 Marseille, France
| | - David A Lyons
- Centre for Neuroregeneration, Centre for Multiple Sclerosis Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Elior Peles
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Jonah R Chan
- Department of Neurology and Program in Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
31
|
Lian S, Meng L, Xing X, Yang Y, Qu L, Shou C. PRL-3 promotes cell adhesion by interacting with JAM2 in colon cancer. Oncol Lett 2016; 12:1661-1666. [PMID: 27588115 PMCID: PMC4998106 DOI: 10.3892/ol.2016.4836] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 03/08/2016] [Indexed: 01/29/2023] Open
Abstract
Phosphatase of regenerating liver-3 (PRL-3), also termed PTP4A3, is a metastasis-related protein tyrosine phosphatase. Its expression levels are significantly correlated with the progression and survival of a wide range of malignant tumors. However, the mechanism by which PRL-3 promotes tumor invasion and metastasis is not clear. In the present study, the functions of PRL-3 were systemically analyzed in the key events of metastasis including, motility and adhesion. A cell wounding assay, cell spread assay and cell-matrix adhesion assay were carried out to analyze the cell movement and cell adhesion ability of colon cancer, immunoprecipitation and immunofluorescence assay was confirmed the interaction of PRL-3 and JAM2. It was demonstrated that PRL-3 promoted the motility of Flp-In-293 and LoVo colon cancer cells and increased the distribution of cell skeleton proteins on the cell protrusions. In addition, stably expressing PRL-3 reduced the spreading speed of colon cancer cells and cell adhesion on uncoated, fibronectin-coated and collagen I-coated plates. Mechanistically, junction adhesion molecular 2 (JAM2) was identified as a novel interacting protein of PRL-3. The findings of the present study revealed the roles of PRL-3 in cancer cell motility and adhesion process, and provided information on the possibility of PRL-3 increase cell-cell adhesion by associating with JAM2.
Collapse
Affiliation(s)
- Shenyi Lian
- Department of Biochemistry and Molecular Biology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing 100142, P.R. China
| | - Lin Meng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing 100142, P.R. China
| | - Xiaofang Xing
- Department of Gastrointestinal Translational Research, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing 100142, P.R. China
| | - Yongyong Yang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing 100142, P.R. China
| | - Like Qu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing 100142, P.R. China
| | - Chengchao Shou
- Department of Biochemistry and Molecular Biology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing 100142, P.R. China
| |
Collapse
|
32
|
Sergé A. The Molecular Architecture of Cell Adhesion: Dynamic Remodeling Revealed by Videonanoscopy. Front Cell Dev Biol 2016; 4:36. [PMID: 27200348 PMCID: PMC4854873 DOI: 10.3389/fcell.2016.00036] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 04/18/2016] [Indexed: 12/20/2022] Open
Abstract
The plasma membrane delimits the cell, which is the basic unit of living organisms, and is also a privileged site for cell communication with the environment. Cell adhesion can occur through cell-cell and cell-matrix contacts. Adhesion proteins such as integrins and cadherins also constitute receptors for inside-out and outside-in signaling within proteolipidic platforms. Adhesion molecule targeting and stabilization relies on specific features such as preferential segregation by the sub-membrane cytoskeleton meshwork and within membrane proteolipidic microdomains. This review presents an overview of the recent insights brought by the latest developments in microscopy, to unravel the molecular remodeling occurring at cell contacts. The dynamic aspect of cell adhesion was recently highlighted by super-resolution videomicroscopy, also named videonanoscopy. By circumventing the diffraction limit of light, nanoscopy has allowed the monitoring of molecular localization and behavior at the single-molecule level, on fixed and living cells. Accessing molecular-resolution details such as quantitatively monitoring components entering and leaving cell contacts by lateral diffusion and reversible association has revealed an unexpected plasticity. Adhesion structures can be highly specialized, such as focal adhesion in motile cells, as well as immune and neuronal synapses. Spatiotemporal reorganization of adhesion molecules, receptors, and adaptors directly relates to structure/function modulation. Assembly of these supramolecular complexes is continuously balanced by dynamic events, remodeling adhesions on various timescales, notably by molecular conformation switches, lateral diffusion within the membrane and endo/exocytosis. Pathological alterations in cell adhesion are involved in cancer evolution, through cancer stem cell interaction with stromal niches, growth, extravasation, and metastasis.
Collapse
Affiliation(s)
- Arnauld Sergé
- Centre de Cancérologie de Marseille, Équipe "Interactions Leuco/Stromales", Institut Paoli-Calmettes, Institut National de la Santé et de la Recherche Médicale U1068, Centre National de la Recherche Scientifique UMR7258, Aix-Marseille Université UM105 Marseille, France
| |
Collapse
|
33
|
Coppin E, De Grandis M, Pandolfi PP, Arcangeli ML, Aurrand-Lions M, Nunès JA. Dok1 and Dok2 Proteins Regulate Cell Cycle in Hematopoietic Stem and Progenitor Cells. THE JOURNAL OF IMMUNOLOGY 2016; 196:4110-21. [DOI: 10.4049/jimmunol.1501037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 03/11/2016] [Indexed: 01/27/2023]
|
34
|
Lhoumeau AC, Arcangeli ML, De Grandis M, Giordano M, Orsoni JC, Lembo F, Bardin F, Marchetto S, Aurrand-Lions M, Borg JP. Ptk7-Deficient Mice Have Decreased Hematopoietic Stem Cell Pools as a Result of Deregulated Proliferation and Migration. THE JOURNAL OF IMMUNOLOGY 2016; 196:4367-77. [DOI: 10.4049/jimmunol.1500680] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 03/11/2016] [Indexed: 01/20/2023]
|
35
|
Chang CH, Hale SJ, Cox CV, Blair A, Kronsteiner B, Grabowska R, Zhang Y, Cook D, Khoo CP, Schrader JB, Kabuga SB, Martin-Rendon E, Watt SM. Junctional Adhesion Molecule-A Is Highly Expressed on Human Hematopoietic Repopulating Cells and Associates with the Key Hematopoietic Chemokine Receptor CXCR4. Stem Cells 2016; 34:1664-78. [PMID: 26866290 DOI: 10.1002/stem.2340] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 01/11/2016] [Indexed: 12/16/2022]
Abstract
Hematopoietic stem/progenitor cells (HSPCs) reside in specialized bone marrow microenvironmental niches, with vascular elements (endothelial/mesenchymal stromal cells) and CXCR4-CXCL12 interactions playing particularly important roles for HSPC entry, retention, and maintenance. The functional effects of CXCL12 are dependent on its local concentration and rely on complex HSPC-niche interactions. Two Junctional Adhesion Molecule family proteins, Junctional Adhesion Molecule-B (JAM)-B and JAM-C, are reported to mediate HSPC-stromal cell interactions, which in turn regulate CXCL12 production by mesenchymal stromal cells (MSCs). Here, we demonstrate that another JAM family member, JAM-A, is most highly expressed on human hematopoietic stem cells with in vivo repopulating activity (p < .01 for JAM-A(high) compared to JAM-A(Int or Low) cord blood CD34(+) cells). JAM-A blockade, silencing, and overexpression show that JAM-A contributes significantly (p < .05) to the adhesion of human HSPCs to IL-1β activated human bone marrow sinusoidal endothelium. Further studies highlight a novel association of JAM-A with CXCR4, with these molecules moving to the leading edge of the cell upon presentation with CXCL12 (p < .05 compared to no CXCL12). Therefore, we hypothesize that JAM family members differentially regulate CXCR4 function and CXCL12 secretion in the bone marrow niche. Stem Cells 2016;34:1664-1678.
Collapse
Affiliation(s)
- Chao-Hui Chang
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Sarah J Hale
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Charlotte V Cox
- Bristol Institute for Transfusion Sciences, NHS Blood and Transplant, Bristol, United Kingdom.,Cancer Research School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Allison Blair
- Bristol Institute for Transfusion Sciences, NHS Blood and Transplant, Bristol, United Kingdom.,Cancer Research School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Barbara Kronsteiner
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Rita Grabowska
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Youyi Zhang
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - David Cook
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Cheen P Khoo
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Jack B Schrader
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Suranahi Buglass Kabuga
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Enca Martin-Rendon
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Suzanne M Watt
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
36
|
Stelzer IA, Mori M, DeMayo F, Lydon J, Arck PC, Solano ME. Differential mouse-strain specific expression of Junctional Adhesion Molecule (JAM)-B in placental structures. Cell Adh Migr 2016; 10:2-17. [PMID: 26914234 DOI: 10.1080/19336918.2015.1118605] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The junctional adhesion molecule (JAM)-B, a member of the immunoglobulin superfamily, is involved in stabilization of interendothelial cell-cell contacts, formation of vascular tubes, homeostasis of stem cell niches and promotion of leukocyte adhesion and transmigration. In the human placenta, JAM-B protein is abundant and mRNA transcripts are enriched in first-trimester extravillous trophoblast in comparison to the villous trophoblast. We here aimed to elucidate the yet unexplored spatio-temporal expression of JAM-B in the mouse placenta. We investigated and semi-quantified JAM-B protein expression by immunohistochemistry in early post-implantation si tes and in mid- to late gestation placentae of various murine mating combinations. Surprisingly, the endothelium of the placental labyrinth was devoid of JAM-B expression. JAM-B was mainly present in spongiotrophoblast cells of the junctional zone, as well as in the fetal vessels of the chorionic plate, the umbilical cord and in maternal myometrial smooth muscle. We observed a strain-specific placental increase of JAM-B protein expression from mid- to late gestation in Balb/c-mated C57BL/6 females, which was absent in DBA/2J-mated Balb/c females. Due to the essential role of progesterone during gestation, we further assessed a possible modulation of JAM-B in mid-gestational placentae deficient in the progesterone receptor (Pgr(-/-)) and observed an increased expression of JAM-B in Pgr(-/-) placentae, compared to Pgr(+/+) tissue samples. We propose that JAM-B is an as yet underappreciated trophoblast lineage-specific protein, which is modulated via the progesterone receptor and shows unique strain-specific kinetics. Future work is needed to elucidate its possible contribution to placental processes necessary to ensuring its integrity, ultimately facilitating placental development and fetal growth.
Collapse
Affiliation(s)
- Ina Annelies Stelzer
- a Laboratory for Exp. Feto-Maternal Medicine, Department of Obstetrics and Prenatal Medicine, University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Mayumi Mori
- a Laboratory for Exp. Feto-Maternal Medicine, Department of Obstetrics and Prenatal Medicine, University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | | | - John Lydon
- b Baylor College of Medicine , Houston , TX , USA
| | - Petra Clara Arck
- a Laboratory for Exp. Feto-Maternal Medicine, Department of Obstetrics and Prenatal Medicine, University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Maria Emilia Solano
- a Laboratory for Exp. Feto-Maternal Medicine, Department of Obstetrics and Prenatal Medicine, University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| |
Collapse
|
37
|
De Grandis M, Lhoumeau AC, Mancini SJC, Aurrand-Lions M. Adhesion receptors involved in HSC and early-B cell interactions with bone marrow microenvironment. Cell Mol Life Sci 2016; 73:687-703. [PMID: 26495446 PMCID: PMC11108274 DOI: 10.1007/s00018-015-2064-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 09/16/2015] [Accepted: 10/08/2015] [Indexed: 02/06/2023]
Abstract
Hematopoiesis takes place in the bone marrow of adult mammals and is the process by which blood cells are replenished every day throughout life. Differentiation of hematopoietic cells occurs in a stepwise manner through intermediates of differentiation that could be phenotypically identified. This has allowed establishing hematopoietic cell classification with hematopoietic stem cells (HSCs) at the top of the hierarchy. HSCs are mostly quiescent and serve as a reservoir for maintenance of lifelong hematopoiesis. Over recent years, it has become increasingly clear that HSC quiescence is not only due to intrinsic properties, but is also mediated by cognate interactions between HSCs and surrounding cells within micro-anatomical sites called “niches”. This hematopoietic/stromal crosstalk model also applies to more mature progenitors such as B cell progenitors, which are thought to reside in distinct “niches”. This prompted many research teams to search for specific molecular mechanisms supporting leuko-stromal crosstalk in the bone marrow and acting at specific stage of differentiation to regulate hematopoietic homeostasis. Here, we review recent data on adhesion mechanisms involved in HSCs and B cell progenitors interactions with surrounding bone marrow stromal cells.
Collapse
Affiliation(s)
- Maria De Grandis
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm U1068, CNRS UMR7258, Aix-Marseille Université UM105, Marseille, France
| | - Anne-Catherine Lhoumeau
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm U1068, CNRS UMR7258, Aix-Marseille Université UM105, Marseille, France
| | - Stéphane J. C. Mancini
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm U1068, CNRS UMR7258, Aix-Marseille Université UM105, Marseille, France
| | - Michel Aurrand-Lions
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm U1068, CNRS UMR7258, Aix-Marseille Université UM105, Marseille, France
| |
Collapse
|
38
|
Meguenani M, Miljkovic-Licina M, Fagiani E, Ropraz P, Hammel P, Aurrand-Lions M, Adams RH, Christofori G, Imhof BA, Garrido-Urbani S. Junctional adhesion molecule B interferes with angiogenic VEGF/VEGFR2 signaling. FASEB J 2015; 29:3411-25. [PMID: 25911611 DOI: 10.1096/fj.15-270223] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/16/2015] [Indexed: 12/22/2022]
Abstract
De novo formation of blood vessels is a pivotal mechanism during cancer development. During the past few years, antiangiogenic drugs have been developed to target tumor vasculature. However, because of limitations and adverse effects observed with current therapies, there is a strong need for alternative antiangiogenic strategies. Using specific anti-junctional adhesion molecule (JAM)-B antibodies and Jam-b-deficient mice, we studied the role in antiangiogenesis of JAM-B. We found that antibodies against murine JAM-B, an endothelium-specific adhesion molecule, inhibited microvessel outgrowth from ex vivo aortic rings and in vitro endothelial network formation. In addition, anti-JAM-B antibodies blocked VEGF signaling, an essential pathway for angiogenesis. Moreover, increased aortic ring branching was observed in aortas isolated from Jam-b-deficient animals, suggesting that JAM-B negatively regulates proangiogenic pathways. In mice, JAM-B expression was detected in de novo-formed blood vessels of tumors, but anti-JAM-B antibodies unexpectedly did not reduce tumor growth. Accordingly, JAM-B deficiency in vivo had no impact on blood vessel formation, suggesting that targeting JAM-B in vivo may be offset by other proangiogenic mechanisms. In conclusion, despite the promising effects observed in vitro, targeting JAM-B during tumor progression seems to be inefficient as a stand-alone antiangiogenesis therapy.
Collapse
Affiliation(s)
- Mehdi Meguenani
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Marijana Miljkovic-Licina
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Ernesta Fagiani
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Patricia Ropraz
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Philippe Hammel
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Michel Aurrand-Lions
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Ralf H Adams
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Gerhard Christofori
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Beat A Imhof
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Sarah Garrido-Urbani
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| |
Collapse
|
39
|
Siegemund S, Shepherd J, Xiao C, Sauer K. hCD2-iCre and Vav-iCre mediated gene recombination patterns in murine hematopoietic cells. PLoS One 2015; 10:e0124661. [PMID: 25884630 PMCID: PMC4401753 DOI: 10.1371/journal.pone.0124661] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/17/2015] [Indexed: 12/13/2022] Open
Abstract
Cre-recombinase mediated conditional deletion of Lox-P site flanked ("floxed") genes is widely used for functional gene annotation in mice. Many different Cre-transgenic mouse lines have been developed for cell-type specific gene disruption. But often, the precise tissue-patterns of Cre activity remain incompletely characterized. Two widely used transgenes for conditional gene recombination in hematopoietic cells are Vav-iCre driven from the murine Vav1 promotor, and hCD2-iCre driven from the human CD2 promotor. Vav-iCre expresses active Cre in fetal and adult hematopoietic stem cells and all descendants, hCD2-iCre in immature and mature B and T lymphocytes. To better characterize which hematopoietic cells contain hCD2-iCre activity, we compared EYFP fluorescence in hCD2-iCre+/- R26-stop-EYFP+/- and Vav-iCre+/- R26-stop-EYFP+/-mice. R26-stop-EYFP ubiquitously encodes EYFP preceded by a floxed stop cassette. By removing it, Cre activity induces measurable EYFP expression. Our results confirm the known activity patterns for both Cre transgenes and unveil additional hCD2-iCre mediated reporter gene recombination in common lymphoid progenitors, in natural killer cells and their progenitors, and in plasmacytoid and conventional dendritic cells. This supports previously proposed common lymphoid origins for natural killer cells and subsets of dendritic cells, and indicates the need to consider pleiotropic effects when studying hCD2-iCre mediated conditional knockout mice. Vav-iCre+/- R26-stop-EYFP+/-mice did not show the non-hematopoietic recombination in vascular endothelial cells seen in other Vav-Cre mouse lines, but displayed an unexpected Vav-iCre mediated recombination in a bone cell subset lacking hematopoietic markers. This pinpoints the need to consider stromal cell contributions to phenotypes of Vav-iCre mediated conditional knockout mice. Altogether, our data provide the first detailed assessment of hCD2-iCre and Vav-iCre mediated deletion of floxed genes during lymphocyte development from hematopoietic stem cells and open up novel applications for either Cre-transgenic mouse line.
Collapse
Affiliation(s)
- Sabine Siegemund
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Jovan Shepherd
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Changchun Xiao
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Karsten Sauer
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, United States of America
| |
Collapse
|
40
|
Arcangeli ML, Bardin F, Frontera V, Bidaut G, Obrados E, Adams RH, Chabannon C, Aurrand-Lions M. Function of Jam-B/Jam-C interaction in homing and mobilization of human and mouse hematopoietic stem and progenitor cells. Stem Cells 2015; 32:1043-54. [PMID: 24357068 DOI: 10.1002/stem.1624] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 09/21/2013] [Accepted: 10/12/2013] [Indexed: 01/05/2023]
Abstract
The junctional adhesion molecules Jam-b and Jam-c interact together at interendothelial junctions and have been involved in the regulation of immune response, inflammation, and leukocyte migration. More recently, Jam-c has been found to be expressed by hematopoietic stem and progenitor cells (HSPC) in mouse. Conversely, we have reported that Jam-b is present on bone marrow stromal cells and that Jam-b-deficient mice have defects in the regulation of hematopoietic stem cell pool. In this study, we have addressed whether interaction between Jam-b and Jam-c participates to HSPC mobilization or hematopoietic reconstitution after irradiation. We show that a blocking monoclonal antibody directed against Jam-c inhibits hematopoietic reconstitution, progenitor homing to the bone marrow, and induces HSPC mobilization in a Jam-b dependent manner. In the latter setting, antibody treatment over a period of 3 days does not alter hematopoietic differentiation nor induce leukocytosis. Results are translated to human hematopoietic system in which a functional adhesive interaction between JAM-B and JAM-C is found between human HSPC and mesenchymal stem cells. Such an interaction does not occur between HSPC and human endothelial cells or osteoblasts. It is further shown that anti-JAM-C blocking antibody interferes with CD34(+) hematopoietic progenitor homing in mouse bone marrow suggesting that monoclonal antibodies inhibiting JAM-B/JAM-C interaction may represent valuable therapeutic tools to improve stem cell mobilization protocols.
Collapse
Affiliation(s)
- Marie-Laure Arcangeli
- Centre de Recherche en Cancérologie de Marseille, Inserm, UMR1068, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Aix-Marseille Université, Marseille, France; CNRS, UMR7258, Marseille, France
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Jam1a-Jam2a interactions regulate haematopoietic stem cell fate through Notch signalling. Nature 2014; 512:319-23. [PMID: 25119047 PMCID: PMC4237229 DOI: 10.1038/nature13623] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 06/27/2014] [Indexed: 12/20/2022]
Abstract
Notch signalling plays a key role in the generation of haematopoietic stem cells (HSCs) during vertebrate development1-3 and requires intimate contact between signal emitting and receiving cells, although little is known regarding when, where, and how these intercellular events occur. We previously reported that the somitic Notch ligands, Dlc and Dld, are essential for HSC specification4. It has remained unclear, however, how these somitic requirements are connected to the later emergence of HSCs from the dorsal aorta (DA). Here we show that Notch signalling establishes HSC fate as their shared vascular precursors migrate across the ventral face of the somite and that Junctional adhesion molecules (JAMs) mediate this required Notch signal transduction. HSC precursors express jam1a and migrate axially across the ventral somite, where Jam2a and Notch ligands Dlc and Dld are expressed. Despite no alteration in the expression of Notch ligand or receptor genes, loss of function of jam1a led to loss of Notch signalling and loss of HSCs. Enforced activation of Notch in shared vascular precursors rescued HSCs in jam1a or jam2a deficient embryos. Together, these results indicate that Jam1a – Jam2a interactions facilitate the transduction of requisite Notch signals from the somite to the precursors of HSCs, and that these events occur well before formation of the DA.
Collapse
|
42
|
Doñate C, Ody C, McKee T, Ruault-Jungblut S, Fischer N, Ropraz P, Imhof BA, Matthes T. Homing of human B cells to lymphoid organs and B-cell lymphoma engraftment are controlled by cell adhesion molecule JAM-C. Cancer Res 2012; 73:640-51. [PMID: 23221386 DOI: 10.1158/0008-5472.can-12-1756] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Junctional adhesion molecule C (JAM-C) is expressed by vascular endothelium and human but not mouse B lymphocytes. The level of JAM-C expression defines B-cell differentiation stages and allows the classification of marginal zone-derived (JAM-C-positive) and germinal center-derived (JAM-C-negative) B-cell lymphomas. In the present study, we investigated the role of JAM-C in homing of human B cells, using a xenogeneic nonobese diabetic/severe combined immunodeficient mouse model. Treatment with anti-JAM-C antibodies in short-term experiments reduced migration of normal and malignant JAM-C-expressing B cells to bone marrow, lymph nodes, and spleen. Blocking homing to the spleen is remarkable, as most other antiadhesion antibodies reduce homing of B cells only to bone marrow and lymph nodes. Long-term administration of anti-JAM-C antibodies prevented engraftment of JAM-Cpos lymphoma cells in bone marrow, spleen, and lymph nodes of mice. Plasmon resonance studies identified JAM-B as the major ligand for JAM-C, whereas homotypic JAM-C interactions remained at background levels. Accordingly, anti-JAM-C antibodies blocked adhesion of JAM-C-expressing B cells to their ligand JAM-B, and immunofluorescence analysis showed the expression of JAM-B on murine and human lymphatic endothelial cells. Targeting JAM-C could thus constitute a new therapeutic strategy to prevent lymphoma cells from reaching supportive microenvironments not only in the bone marrow and lymph nodes but also in the spleen.
Collapse
Affiliation(s)
- Carmen Doñate
- Hematology Service, University Hospital, Geneva, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Arcangeli ML, Frontera V, Bardin F, Thomassin J, Chetaille B, Adams S, Adams RH, Aurrand-Lions M. The Junctional Adhesion Molecule-B regulates JAM-C-dependent melanoma cell metastasis. FEBS Lett 2012; 586:4046-51. [PMID: 23068611 DOI: 10.1016/j.febslet.2012.10.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 10/01/2012] [Accepted: 10/02/2012] [Indexed: 01/16/2023]
Abstract
Metastasis is a major clinical issue and results in poor prognosis for most cancers. The Junctional Adhesion Molecule-C (JAM-C) expressed by B16 melanoma and endothelial cells has been involved in metastasis of tumor cells through homophilic JAM-C/JAM-C trans-interactions. Here, we show that JAM-B expressed by endothelial cells contributes to murine B16 melanoma cells metastasis through its interaction with JAM-C on tumor cells. We further show that this adhesion molecular pair mediates melanoma cell adhesion to primary Lung Microvascular Endothelial Cells and that it is functional in vivo as demonstrated by the reduced metastasis of B16 cells in Jam-b deficient mice.
Collapse
Affiliation(s)
- Marie-Laure Arcangeli
- Centre de Recherche en Cancérologie de Marseille, Unité Mixte de Recherche, Institut National de la Santé et de la Recherche Médicale 1068/Centre National de la Recherche Scientifique 7258, Marseille, France.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Function of junctional adhesion molecules (JAMs) in leukocyte migration and homeostasis. Arch Immunol Ther Exp (Warsz) 2012; 61:15-23. [PMID: 22940878 DOI: 10.1007/s00005-012-0199-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 07/23/2012] [Indexed: 01/09/2023]
Abstract
Homeostasis is a word widely used in the scientific community to refer to the property of a system to maintain its uniformity and functionality. In living organisms, the word refers to the concept enunciated 150 years ago by C. Bernard by which external variations must be compensated for in order to maintain internal conditions compatible with life. This is especially true in the case of highly dynamic system such as the hematopoietic system that requires the coordinated control of cell proliferation and death within specialized microenvironments that are anatomically distinct. As a consequence, hematopoietic cell adhesion and migration must be tightly controlled in order for hematopoietic cells to reach and to be maintained in appropriate microenvironments. The junctional adhesion molecules (JAMs) are adhesion molecules that belong to the immunoglobulin superfamily (IgSf) and that have been initially identified as important players controlling vascular permeability and leukocyte transendothelial migration. This involves the regulated localization of the JAMs at lateral endothelial cell/cell borders and their interaction with leukocyte integrins. More recently, some of the JAM family members have also been found to be expressed by stromal cells and to regulate chemokine secretion within lymphoid organs, acting not only on leukocyte transendothelial migration, but also on hematopoietic cell retention within specialized microenvironments. This review summarizes recent progress in understanding the role of the JAMs in leukocyte adhesion and migration to tentatively draw an integrated view of the homeostatic function of the JAMs within the hematopoietic system.
Collapse
|
45
|
Chomel JC, Turhan AG. Chronic myeloid leukemia stem cells in the era of targeted therapies: resistance, persistence and long-term dormancy. Oncotarget 2012; 2:713-27. [PMID: 21946665 PMCID: PMC3248215 DOI: 10.18632/oncotarget.333] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Targeted therapies of chronic myeloid leukemia (CML) using tyrosine kinase inhibitors (TKI) have profoundly changed the natural history of the disease with a major impact on survival. Molecular monitoring with BCR-ABL quantification shows that a status of undetectable molecular residual disease (UMRD) is obtained in a significant minority of patients. However, it remains unclear whether these patients are definitively cured of their leukemia. Imatinib mesylate withdrawal trials have demonstrated the rapid appearance of the malignant clone in the majority of the patients whereas some patients remain in a state of UMRD. It has clearly been demonstrated that the most primitive stem cells are refractory to all TKIs used in clinical practice. In addition, long-term dormancy is one of the most fundamental characteristics of hematopoietic stem cells. In this context, we have recently undertaken a systematic analysis of the bone marrow stem cell compartment in several patients in durable UMRD. We have demonstrated the long-term persistence of a considerable amount of BCR-ABL-expressing stem cells, even in the absence of relapse. The phenomenon of long-term leukemic stem cell dormancy is of major importance in CML and one of the key questions in cancer biology in general. We discuss, here, the potential mechanisms, including intrinsic and microenvironmental factors, that control the response of leukemic stem cells (LSCs) to targeted therapies and potential novel strategies currently in progress with a curative intent. Moreover, we propose a molecular evaluation of the residual LSC compartment in selected patients in order to develop rational TKI-cessation strategies in CML.
Collapse
Affiliation(s)
- Jean-Claude Chomel
- Service d'Hématologie et Oncologie Biologique, CHU de Poitiers, Université de Poitiers, France
| | | |
Collapse
|