1
|
Wu Y, Ma J, Chen J, Liu X, Wang Z, Luo L, Sun C. Ablation of CD44 Attenuates Adipogenesis in White Adipocytes via the Tryptophan 5-Hydroxylase 2/5-Hydroxytryptamine Axis to Protect Mice from High-Fat Diet-Induced Obesity. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:247-264. [PMID: 39476955 DOI: 10.1016/j.ajpath.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 11/11/2024]
Abstract
CD44 is a transmembrane protein that plays an essential role in transducing extracellular stimuli into intracellular signaling cascades, especially in cancer cells. Recent studies have shown that CD44 contributes to metabolic regulation. However, the effect of CD44 on adipogenesis in white adipose tissue (WAT) remains unclear. Herein, the expression of CD44 was largely increased in the inguinal and epididymal WAT of obese mice. Ablation or neutralization of CD44 inhibited adipogenesis in cultured adipocytes. CD44-deficient mice were resistant to high-fat diet-induced obesity and metabolic dysfunction. RNA-sequencing, together with functional studies, revealed that reduced expression of tryptophan 5-hydroxylase 2 (Tph2) in WAT was responsible for the repressed adipogenesis in the absence of CD44. The application of 5-hydroxytryptamine, a product of TPH2, rescued the repressed adipogenesis induced by CD44 neutralization. Moreover, the inhibition of TPH2 by p-chlorophenylalanine recapitulated the beneficial phenotypes observed in CD44-deficient mice. Taken together, these data indicate that CD44 plays a pivotal role in adipogenesis in WAT. In this regard, CD44 and its downstream target TPH2 may hold great therapeutic potential for treating excessive adiposity-related metabolic disorders, such as obesity, insulin resistance, and type 2 diabetes.
Collapse
Affiliation(s)
- Yuting Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jinyu Ma
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jing Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Xiaoyu Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Zhe Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Lan Luo
- Department of Geriatrics, Affiliated Hospital of Nantong University, Nantong, China.
| | - Cheng Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China.
| |
Collapse
|
2
|
Mehner LM, Munoz-Sagredo L, Sonnentag SJ, Treffert SM, Orian-Rousseau V. Targeting CD44 and other pleiotropic co-receptors as a means for broad inhibition of tumor growth and metastasis. Clin Exp Metastasis 2024; 41:599-611. [PMID: 38761292 PMCID: PMC11499327 DOI: 10.1007/s10585-024-10292-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 05/02/2024] [Indexed: 05/20/2024]
Abstract
Although progress has been made in the treatment of cancer, particularly for the four major types of cancers affecting the lungs, colon, breast and prostate, resistance to cancer treatment often emerges upon inhibition of major signaling pathways, which leads to the activation of additional pathways as a last-resort survival mechanism by the cancer cells. This signaling plasticity provides cancer cells with a level of operational freedom, reducing treatment efficacy. Plasticity is a characteristic of cancer cells that are not only able to switch signaling pathways but also from one cellular state (differentiated cells to stem cells or vice versa) to another. It seems implausible that the inhibition of one or a few signaling pathways of heterogeneous and plastic tumors can sustain a durable effect. We propose that inhibiting molecules with pleiotropic functions such as cell surface co-receptors can be a key to preventing therapy escape instead of targeting bona fide receptors. Therefore, we ask the question whether co-receptors often considered as "accessory molecules" are an overlooked key to control cancer cell behavior.
Collapse
Affiliation(s)
- Lisa-Marie Mehner
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Leonel Munoz-Sagredo
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany
- School of Medicine, Universidad de Valparaiso, Valparaiso, Chile
| | - Steffen Joachim Sonnentag
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Sven Máté Treffert
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Véronique Orian-Rousseau
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany.
| |
Collapse
|
3
|
Gerardo-Ramírez M, Giam V, Becker D, Groth M, Hartmann N, Morrison H, May-Simera HL, Radsak MP, Marquardt JU, Galle PR, Herrlich P, Straub BK, Hartmann M. Deletion of Cd44 Inhibits Metastasis Formation of Liver Cancer in Nf2-Mutant Mice. Cells 2023; 12:cells12091257. [PMID: 37174657 PMCID: PMC10177437 DOI: 10.3390/cells12091257] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Primary liver cancer is the third leading cause of cancer-related death worldwide. An increasing body of evidence suggests that the Hippo tumor suppressor pathway plays a critical role in restricting cell proliferation and determining cell fate during physiological and pathological processes in the liver. Merlin (Moesin-Ezrin-Radixin-like protein) encoded by the NF2 (neurofibromatosis type 2) gene is an upstream regulator of the Hippo signaling pathway. Targeting of Merlin to the plasma membrane seems to be crucial for its major tumor-suppressive functions; this is facilitated by interactions with membrane-associated proteins, including CD44 (cluster of differentiation 44). Mutations within the CD44-binding domain of Merlin have been reported in many human cancers. This study evaluated the relative contribution of CD44- and Merlin-dependent processes to the development and progression of liver tumors. To this end, mice with a liver-specific deletion of the Nf2 gene were crossed with Cd44-knockout mice and subjected to extensive histological, biochemical and molecular analyses. In addition, cells were isolated from mutant livers and analyzed by in vitro assays. Deletion of Nf2 in the liver led to substantial liver enlargement and generation of hepatocellular carcinomas (HCCs), intrahepatic cholangiocarcinomas (iCCAs), as well as mixed hepatocellular cholangiocarcinomas. Whilst deletion of Cd44 had no influence on liver size or primary liver tumor development, it significantly inhibited metastasis formation in Nf2-mutant mice. CD44 upregulates expression of integrin β2 and promotes transendothelial migration of liver cancer cells, which may facilitate metastatic spreading. Overall, our results suggest that CD44 may be a promising target for intervening with metastatic spreading of liver cancer.
Collapse
Affiliation(s)
- Monserrat Gerardo-Ramírez
- Department of Medicine I, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Vanessa Giam
- Department of Medicine I, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Diana Becker
- Department of Medicine I, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Marco Groth
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), 07745 Jena, Germany
| | - Nils Hartmann
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Helen Morrison
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), 07745 Jena, Germany
- Faculty of Biological Sciences, Friedrich-Schiller University, 07745 Jena, Germany
| | - Helen L May-Simera
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Markus P Radsak
- Department of Medicine III, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Jens U Marquardt
- Department of Medicine I, University Medical Center Schleswig-Holstein, Campus Lübeck, 23558 Lübeck, Germany
| | - Peter R Galle
- Department of Medicine I, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Peter Herrlich
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), 07745 Jena, Germany
| | - Beate K Straub
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Monika Hartmann
- Department of Medicine I, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| |
Collapse
|
4
|
Drygalski K, Lecoutre S, Clément K, Dugail I. Hyaluronan in Adipose Tissue, Metabolic Inflammation, and Diabetes: Innocent Bystander or Guilty Party? Diabetes 2023; 72:159-169. [PMID: 36668999 DOI: 10.2337/db22-0676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/03/2022] [Indexed: 01/21/2023]
Abstract
Hyaluronic acid, or hyaluronan (HA), is a nonsulfated glucosaminoglycan that has long been recognized for its hydrophilic properties and is widely used as a dermal filler. Despite much attention given to the study of other extracellular matrix (ECM) components, in the field of ECM properties and their contribution to tissue fibroinflammation, little is known of HA's potential role in the extracellular milieu. However, recent studies suggest that it is involved in inflammatory response, diet-induced insulin resistance, adipogenesis, and autoimmunity in type 1 diabetes. Based on its unique physical property as a regulator of osmotic pressure, we emphasize underestimated implications in adipose tissue function, adipogenesis, and obesity-related dysfunction.
Collapse
Affiliation(s)
- Krzysztof Drygalski
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
- Clinical Research Center, Medical University of Bialystok, Bialystok, Poland
| | - Simon Lecoutre
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
- Nutrition Department, Assistance Publique Hôpitaux de Paris, Centre de Recherche en Nutrition Humaine Ile-de-France, Pitié-Salpêtrière Hospital, Paris, France
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| |
Collapse
|
5
|
RNA-Seq Analysis Identifies Differentially Expressed Genes in the Longissimus dorsi of Wagyu and Chinese Red Steppe Cattle. Int J Mol Sci 2022; 24:ijms24010387. [PMID: 36613828 PMCID: PMC9820533 DOI: 10.3390/ijms24010387] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/08/2022] [Accepted: 12/16/2022] [Indexed: 12/29/2022] Open
Abstract
Meat quality has a close relationship with fat and connective tissue; therefore, screening and identifying functional genes related to lipid metabolism is essential for the production of high-grade beef. The transcriptomes of the Longissimus dorsi muscle in Wagyu and Chinese Red Steppe cattle, breeds with significant differences in meat quality and intramuscular fat deposition, were analyzed using RNA-seq to screen for candidate genes associated with beef quality traits. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that the 388 differentially expressed genes (DEGs) were involved in biological processes such as short-chain fatty acid metabolism, regulation of fatty acid transport and the peroxisome proliferator-activated receptor (PPAR) signaling pathway. In addition, crystallin alpha B (CRYAB), ankyrin repeat domain 2 (ANKRD2), aldehyde dehydrogenase 9 family member A1 (ALDH9A1) and enoyl-CoA hydratase and 3-hydroxyacyl CoA dehydrogenase (EHHADH) were investigated for their effects on intracellular triglyceride and fatty acid content and their regulatory effects on genes in lipogenesis and fatty acid metabolism pathways. This study generated a dataset from transcriptome profiling of two cattle breeds, with differing capacities for fat-deposition in the muscle, and revealed molecular evidence that CRYAB, ANKRD2, ALDH9A1 and EHHADH are related to fat metabolism in bovine fetal fibroblasts (BFFs). The results provide potential functional genes for maker-assisted selection and molecular breeding to improve meat quality traits in beef cattle.
Collapse
|
6
|
Ivanova EL, Costa B, Eisemann T, Lohr S, Boskovic P, Eichwald V, Meckler J, Jugold M, Orian-Rousseau V, Peterziel H, Angel P. CD44 expressed by myeloid cells promotes glioma invasion. Front Oncol 2022; 12:969787. [PMID: 35992852 PMCID: PMC9386454 DOI: 10.3389/fonc.2022.969787] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/11/2022] [Indexed: 12/07/2022] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and malignant brain tumors in adulthood with a median survival of only 15 months. This poor prognosis is related to GBM’s ability to extensively infiltrate the surrounding brain parenchyma resulting in diffuse spread of neoplastic cells in the brain, responsible for high rate of recurrence. CD44 (Cluster of Differentiation 44) is a transmembrane protein, overexpressed in multiple cancer types, including gliomas, and implicated in cell motility, proliferation and angiogenesis. Multiple studies have investigated the role of CD44 in GBM cells and have highlighted a link between tumor malignancy and CD44 expression. However up to date, little is known of the role of CD44 on cells from the tumor microenvironment (TME). Here, we have investigated a potential role of CD44 in the TME in regards to GBM invasiveness. Using an ex-vivo organotypic brain slice invasion assay, we show that absence of CD44 from the TME impairs the ability of glioma cells to invade the surrounding brain parenchyma. By deleting CD44 in the astrocytic, endothelial and myeloid compartments, we show that it is specifically CD44 expression in myeloid cells that is responsible for the observed phenotype. Combining in vivo studies in cell-specific knock-out mice and in vitro analyses on primary microglia we demonstrate that myeloid CD44 is implicated in Toll Like Receptor 2 signaling and is a major regulator of Matrix metalloproteinase 9 expression.
Collapse
Affiliation(s)
- Ekaterina L. Ivanova
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Barbara Costa
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tanja Eisemann
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabrina Lohr
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pavle Boskovic
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Viktoria Eichwald
- Core Facility Small Animal Imaging Center, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jasmin Meckler
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manfred Jugold
- Core Facility Small Animal Imaging Center, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Veronique Orian-Rousseau
- Karlsruhe Institute of Technology (KIT), Institute of Biological and Chemical Systems – Functional Molecular Systems (IBCS-FMS), Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, Germany
| | - Heike Peterziel
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Angel
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
- *Correspondence: Peter Angel,
| |
Collapse
|
7
|
Garantziotis S, Savani RC. Proteoglycans in Toll-like receptor responses and innate immunity. Am J Physiol Cell Physiol 2022; 323:C202-C214. [PMID: 35675639 DOI: 10.1152/ajpcell.00088.2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The extracellular matrix (ECM) is an active and dynamic feature of tissues that not only provides gross structure but also plays key roles in cellular responses. The ever-changing microenvironment responds dynamically to cellular and external signals, and in turn influences cell fate, tissue development, and response to environmental injury or microbial invasion. It is therefore paramount to understand how the ECM components interact with each other, the environment and cells, and how they mediate their effects. Among the ECM components that have recently garnered increased attention, proteoglycans (PGs) deserve special note. Recent evidence strongly suggests that they play a crucial role both in health maintenance and disease development. In particular, proteoglycans dictate whether homeostasis or cell death will result from a given injury, by triggering and modulating activation of the innate immune system, via a conserved array of receptors that recognize exogenous (infectious) or endogenous (tissue damage) molecular patterns. Innate immune activation by proteoglycans has important implications for the understanding of cell-matrix interactions in health and disease. In this review, we will summarize the current state of knowledge of innate immune signaling by proteoglycans, discuss the implications, and explore future directions to define progress in this area of extracellular matrix biology.
Collapse
Affiliation(s)
- Stavros Garantziotis
- Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Rashmin C Savani
- Division of Neonatal-Perinatal Medicine, Center for Pulmonary & Vascular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
8
|
Hayashida K, Aquino RS, Park PW. Coreceptor Functions of Cell Surface Heparan Sulfate Proteoglycans. Am J Physiol Cell Physiol 2022; 322:C896-C912. [PMID: 35319900 PMCID: PMC9109798 DOI: 10.1152/ajpcell.00050.2022] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Receptor-ligand interactions play an important role in many biological processes by triggering specific cellular responses. These interactions are frequently regulated by coreceptors that facilitate, alter, or inhibit signaling. Coreceptors work in parallel with other specific and accessory molecules to coordinate receptor-ligand interactions. Cell surface heparan sulfate proteoglycans (HSPGs) function as unique coreceptors because they can bind to many ligands and receptors through their HS and core protein motifs. Cell surface HSPGs are typically expressed in abundance of the signaling receptors and, thus, are capable of mediating the initial binding of ligands to the cell surface. HSPG coreceptors do not possess kinase domains or intrinsic enzyme activities and, for the most part, binding to cell surface HSPGs does not directly stimulate intracellular signaling. Because of these features, cell surface HSPGs primarily function as coreceptors for many receptor-ligand interactions. Given that cell surface HSPGs are widely conserved, they likely serve fundamental functions to preserve basic physiological processes. Indeed, cell surface HSPGs can support specific cellular interactions with growth factors, morphogens, chemokines, extracellular matrix (ECM) components, and microbial pathogens and their secreted virulence factors. Through these interactions, HSPG coreceptors regulate cell adhesion, proliferation, migration and differentiation, and impact the onset, progression, and outcome of pathophysiological processes, such as development, tissue repair, inflammation, infection, and tumorigenesis. This review seeks to provide an overview of the various mechanisms of how cell surface HSPGs function as coreceptors.
Collapse
Affiliation(s)
- Kazutaka Hayashida
- Department of Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Rafael S Aquino
- Department of Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Pyong Woo Park
- Department of Medicine, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
9
|
Mu M, Li B, Zou Y, Wang W, Cao H, Zhang Y, Sun Q, Chen H, Ge D, Tao H, Hu D, Yuan L, Tao X, Wang J. Coal dust exposure triggers heterogeneity of transcriptional profiles in mouse pneumoconiosis and Vitamin D remedies. Part Fibre Toxicol 2022; 19:7. [PMID: 35057792 PMCID: PMC8772169 DOI: 10.1186/s12989-022-00449-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/13/2022] [Indexed: 12/22/2022] Open
Abstract
Background Coal dust particles (CDP), an inevitable by-product of coal mining for the environment, mainly causes coal workers’ pneumoconiosis (CWP). Long-term exposure to coal dust leads to a complex alternation of biological processes during regeneration and repair in the healing lung. However, the cellular and complete molecular changes associated with pulmonary homeostasis caused by respiratory coal dust particles remain unclear. Methods This study mainly investigated the pulmonary toxicity of respirable-sized CDP in mice using unbiased single-cell RNA sequencing. CDP (< 5 μm) collected from the coal mine was analyzed by Scanning Electron Microscope (SEM) and Mass Spectrometer. In addition, western blotting, Elisa, QPCR was used to detect gene expression at mRNA or protein levels. Pathological analysis including HE staining, Masson staining, immunohistochemistry, and immunofluorescence staining were performed to characterize the structure and functional alternation in the pneumoconiosis mouse and verify the reliability of single-cell sequencing results. Results SEM image and Mass Spectrometer analysis showed that coal dust particles generated during coal mine production have been crushed and screened with a diameter of less than 5 µm and contained less than 10% silica. Alveolar structure and pulmonary microenvironment were destroyed, inflammatory and death (apoptosis, autophagy, and necrosis) pathways were activated, leading to pneumoconiosis in post 9 months coal dust stimulation. A distinct abnormally increased alveolar type 2 epithelial cell (AT2) were classified with a highly active state but reduced the antimicrobial-related protein expression of LYZ and Chia1 after CDP exposure. Beclin1, LC3B, LAMP2, TGF-ß, and MLPH were up-regulated induced by CDP, promoting autophagy and pulmonary fibrosis. A new subset of macrophages with M2-type polarization double expressed MLPH + /CD206 + was found in mice having pneumoconiosis but markedly decreased after the Vitamin D treatment. Activated MLPH + /CD206 + M2 macrophages secreted TGF-β1 and are sensitive to Vitamin D treatment. Conclusions This is the first study to reconstruct the pathologic progression and transcriptome pattern of coal pneumoconiosis in mice. Coal dust had obvious toxic effects on lung epithelial cells and macrophages and eventually induced pulmonary fibrosis. CDP-induced M2-type macrophages could be inhibited by VD, which may be related to the alleviation of the pulmonary fibrosis process. Supplementary Information The online version contains supplementary material available at 10.1186/s12989-022-00449-y.
Collapse
|
10
|
Abstract
Cell surface proteoglycans, such as syndecans and glypicans, regulate molecular interactions that mediate cell adhesion, migration, proliferation, and differentiation. Through these activities, surface proteoglycans modulate critical biological processes of development, inflammation, infection, tissue repair, and cancer metastasis. Proteoglycans are unique glycoproteins comprised of one or several glycosaminoglycans attached covalently to core proteins. Glycosaminoglycans mediate the majority of ligand-binding functions of proteoglycans. Accumulating evidence indicates that surface proteoglycans regulate the onset, progression, and outcome of lung diseases, including lung injury, infection, fibrosis, and cancer. This article will review key features of surface proteoglycan biology in lung health and disease.
Collapse
|
11
|
Garantziotis S. Modulation of hyaluronan signaling as a therapeutic target in human disease. Pharmacol Ther 2021; 232:107993. [PMID: 34587477 DOI: 10.1016/j.pharmthera.2021.107993] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/14/2022]
Abstract
The extracellular matrix is an active participant, modulator and mediator of the cell, tissue, organ and organismal response to injury. Recent research has highlighted the role of hyaluronan, an abundant glycosaminoglycan constituent of the extracellular matrix, in many fundamental biological processes underpinning homeostasis and disease development. From this basis, emerging studies have demonstrated the therapeutic potential of strategies which target hyaluronan synthesis, biology and signaling, with significant promise as therapeutics for a variety of inflammatory and immune diseases. This review summarizes the state of the art in this field and discusses challenges and opportunities in what could emerge as a new class of therapeutic agents, that we term "matrix biologics".
Collapse
Affiliation(s)
- Stavros Garantziotis
- Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA.
| |
Collapse
|
12
|
Sottoriva K, Pajcini KV. Notch Signaling in the Bone Marrow Lymphopoietic Niche. Front Immunol 2021; 12:723055. [PMID: 34394130 PMCID: PMC8355626 DOI: 10.3389/fimmu.2021.723055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Lifelong mammalian hematopoiesis requires continuous generation of mature blood cells that originate from Hematopoietic Stem and Progenitor Cells (HSPCs) situated in the post-natal Bone Marrow (BM). The BM microenvironment is inherently complex and extensive studies have been devoted to identifying the niche that maintains HSPC homeostasis and supports hematopoietic potential. The Notch signaling pathway is required for the emergence of the definitive Hematopoietic Stem Cell (HSC) during embryonic development, but its role in BM HSC homeostasis is convoluted. Recent work has begun to explore novel roles for the Notch signaling pathway in downstream progenitor populations. In this review, we will focus an important role for Notch signaling in the establishment of a T cell primed sub-population of Common Lymphoid Progenitors (CLPs). Given that its activation mechanism relies primarily on cell-to-cell contact, Notch signaling is an ideal means to investigate and define a novel BM lymphopoietic niche. We will discuss how new genetic model systems indicate a pre-thymic, BM-specific role for Notch activation in early T cell development and what this means to the paradigm of lymphoid lineage commitment. Lastly, we will examine how leukemic T-cell acute lymphoblastic leukemia (T-ALL) blasts take advantage of Notch and downstream lymphoid signals in the pathological BM niche.
Collapse
Affiliation(s)
- Kilian Sottoriva
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, United States
| | - Kostandin V Pajcini
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, United States
| |
Collapse
|
13
|
The Role of Tumor Microenvironment in Multiple Myeloma Development and Progression. Cancers (Basel) 2021; 13:cancers13020217. [PMID: 33435306 PMCID: PMC7827690 DOI: 10.3390/cancers13020217] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Multiple Myeloma (MM) is a hematologic malignancy caused by aberrant plasma cell proliferation in the bone marrow (BM) and constitutes the second most common hematological disease after non-Hodgkin lymphoma. The disease progression is drastically regulated by the immunosuppressive tumor microenvironment (TME) generated by soluble factors and different cells that naturally reside in the BM. This microenvironment does not remain unchanged and alterations favor cancer dissemination. Despite therapeutic advances over the past 15 years, MM remains incurable and therefore understanding the elements that control the TME in MM would allow better-targeted therapies to cure this disease. In this review, we discuss the main events and changes that occur in the BM milieu during MM development. Abstract Multiple myeloma (MM) is a hematologic cancer characterized by clonal proliferation of plasma cells in the bone marrow (BM). The progression, from the early stages of the disease as monoclonal gammopathy of undetermined significance (MGUS) and smoldering multiple myeloma (SMM) to MM and occasionally extramedullary disease, is drastically affected by the tumor microenvironment (TME). Soluble factors and direct cell–cell interactions regulate MM plasma cell trafficking and homing to the BM niche. Mesenchymal stromal cells, osteoclasts, osteoblasts, myeloid and lymphoid cells present in the BM create a unique milieu that favors MM plasma cell immune evasion and promotes disease progression. Moreover, TME is implicated in malignant cell protection against anti-tumor therapy. This review describes the main cellular and non-cellular components located in the BM, which condition the immunosuppressive environment and lead the MM establishment and progression.
Collapse
|
14
|
Xu H, Niu M, Yuan X, Wu K, Liu A. CD44 as a tumor biomarker and therapeutic target. Exp Hematol Oncol 2020; 9:36. [PMID: 33303029 PMCID: PMC7727191 DOI: 10.1186/s40164-020-00192-0] [Citation(s) in RCA: 219] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 11/21/2020] [Indexed: 12/15/2022] Open
Abstract
CD44, a complex transmembrane glycoprotein, exists in multiple molecular forms, including the standard isoform CD44s and CD44 variant isoforms. CD44 participates in multiple physiological processes, and aberrant expression and dysregulation of CD44 contribute to tumor initiation and progression. CD44 represents a common biomarker of cancer stem cells, and promotes epithelial-mesenchymal transition. CD44 is involved in the regulation of diverse vital signaling pathways that modulate cancer proliferation, invasion, metastasis and therapy-resistance, and it is also modulated by a variety of molecules in cancer cells. In addition, CD44 can serve as an adverse prognostic marker among cancer population. The pleiotropic roles of CD44 in carcinoma potentially offering new molecular target for therapeutic intervention. Preclinical and clinical trials for evaluating the pharmacokinetics, efficacy and drug-related toxicity of CD44 monoclonal antibody have been carried out among tumors with CD44 expression. In this review, we focus on current data relevant to CD44, and outline CD44 structure, the regulation of CD44, functional properties of CD44 in carcinogenesis and cancer progression as well as the potential CD44-targeting therapy for cancer management.
Collapse
Affiliation(s)
- Hanxiao Xu
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Mengke Niu
- Department of Medical Oncology, The Affiliated Tumor Hospital of Zhengzhou University: Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Xun Yuan
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Kongming Wu
- Department of Medical Oncology, The Affiliated Tumor Hospital of Zhengzhou University: Henan Cancer Hospital, Zhengzhou, 450008, China. .,Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| | - Aiguo Liu
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
15
|
The role of vascular niche and endothelial cells in organogenesis and regeneration. Exp Cell Res 2020; 398:112398. [PMID: 33271129 DOI: 10.1016/j.yexcr.2020.112398] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/21/2020] [Accepted: 11/22/2020] [Indexed: 02/08/2023]
Abstract
The term vascular niche indicate the physical and biochemical microenvironment around blood vessel where endothelial cells, pericytes, and smooth muscle cells organize themselves to form blood vessels and release molecules involved in the recruitment of hematopoietic stem cells, endothelial progenitor cells and mesenchymal stem cells. The vascular niche creates a permissive environment that enables different cell types to realize their developmental or regenerative programs. In this context, the proximity between the endothelium and the new-forming cellular components of organs suggests an essential role of endothelial cells in the organs maturation. Dynamic interactions between specific organ endothelial cells and different cellular conponents are crucial for different organ morphogenesis and function. Conversely, organs provide cues shaping vascular network structure.
Collapse
|
16
|
Chen L, Fu C, Zhang Q, He C, Zhang F, Wei Q. The role of CD44 in pathological angiogenesis. FASEB J 2020; 34:13125-13139. [PMID: 32830349 DOI: 10.1096/fj.202000380rr] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 07/29/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023]
Abstract
Angiogenesis is required for normal development and occurs as a pathological step in a variety of disease settings, such as cancer, ocular diseases, and ischemia. Recent studies have revealed the role of CD44, a widely expressed cell surface adhesion molecule, in promoting pathological angiogenesis and the development of its associated diseases through its regulation of diverse function of endothelial cells, such as proliferation, migration, adhesion, invasion, and communication with the microenvironment. Conversely, the absence of CD44 expression or inhibition of its function impairs pathological angiogenesis and disease progression. Here, we summarize the current understanding of the roles of CD44 in pathological angiogenesis and the underlying cellular and molecular mechanisms.
Collapse
Affiliation(s)
- Li Chen
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P.R. China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Chenying Fu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Qing Zhang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P.R. China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Chengqi He
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P.R. China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Feng Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Quan Wei
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P.R. China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
17
|
Daniels JMA, Reuling EMBP, Dickhoff C. Endobronchial Treatment of Bronchial Carcinoid in the Elderly. CURRENT GERIATRICS REPORTS 2020. [DOI: 10.1007/s13670-020-00322-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Abstract
Purpose of the Review
Although surgical resection is considered the gold standard of curative treatment for bronchial carcinoid, endobronchial treatment (EBT) can serve as a less invasive curative or palliative treatment in a selection of patients. It is unclear whether elderly patients with bronchial carcinoid should be treated in the same way as younger patients. In order to study the characteristics and treatment of elderly patients with bronchial carcinoid, we analyzed data from a cohort of patients that have been treated for bronchial carcinoid with EBT, surgical resection, or a combination of both. We used our existing database of patients referred for EBT and defined two groups of patients: ≥ 65 and < 65 years. We compared the characteristics, treatment, and causes of death between these groups. Successful EBT was defined as definitive treatment without signs of recurrence during follow-up with CT and bronchoscopy.
Recent Findings
Thirty-five patients (19%) were ≥ 65 years. The incidence of atypical carcinoid was the same in both age groups (31%). Thirty-six of 184 patients (20%) were directly referred for surgical resection and 148 (80%) underwent EBT. There was no significant difference in success of EBT between patients <65 years (58/122, 48%) and patients ≥ 65 years (15/26, 58%) (p = 0.347). Complication rates were similar in both groups. After unsuccessful EBT, only 70% (14/20) of the elderly patients was operated, whereas 93% (85/91) of the patients < 65 years was operated (p = 0.001). Disease specific mortality was 6% (2/35) in the group ≥ 65 years and 2% (3/149) in the group < 65 years.
Summary
The incidence of atypical carcinoid is similar between the elderly and younger patients. Success rate and complication rate of EBT do not differ significantly between the age groups. After unsuccessful EBT, elderly patients were less likely to undergo surgical resection, which does not seem associated with excess disease specific mortality, although the number of events in this study is low.
Collapse
|
18
|
Abatangelo G, Vindigni V, Avruscio G, Pandis L, Brun P. Hyaluronic Acid: Redefining Its Role. Cells 2020; 9:E1743. [PMID: 32708202 PMCID: PMC7409253 DOI: 10.3390/cells9071743] [Citation(s) in RCA: 261] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/17/2020] [Accepted: 07/19/2020] [Indexed: 12/26/2022] Open
Abstract
The discovery of several unexpected complex biological roles of hyaluronic acid (HA) has promoted new research impetus for biologists and, the clinical interest in several fields of medicine, such as ophthalmology, articular pathologies, cutaneous repair, skin remodeling, vascular prosthesis, adipose tissue engineering, nerve reconstruction and cancer therapy. In addition, the great potential of HA in medicine has stimulated the interest of pharmaceutical companies which, by means of new technologies can produce HA and several new derivatives in order to increase both the residence time in a variety of human tissues and the anti-inflammatory properties. Minor chemical modifications of the molecule, such as the esterification with benzyl alcohol (Hyaff-11® biomaterials), have made possible the production of water-insoluble polymers that have been manufactured in various forms: membranes, gauzes, nonwoven meshes, gels, tubes. All these biomaterials are used as wound-covering, anti-adhesive devices and as scaffolds for tissue engineering, such as epidermis, dermis, micro-vascularized skin, cartilage and bone. In this review, the essential biological functions of HA and the applications of its derivatives for pharmaceutical and tissue regeneration purposes are reviewed.
Collapse
Affiliation(s)
- G. Abatangelo
- Faculty of Medicine, University of Padova, 35121 Padova, Italy
| | - V. Vindigni
- Clinic of Plastic and Reconstructive Surgery, University of Padova, 35128 Padova, Italy; (V.V.); (L.P.)
| | - G. Avruscio
- Department of Cardiac, Thoracic and Vascular Sciences, Angiology Unit, University of Padova, 35128 Padova, Italy;
| | - L. Pandis
- Clinic of Plastic and Reconstructive Surgery, University of Padova, 35128 Padova, Italy; (V.V.); (L.P.)
| | - P. Brun
- Department of Molecular Medicine, Histology unit, University of Padova, 35121 Padova, Italy;
| |
Collapse
|
19
|
Jiang P, Xia L, Jin Z, Ali S, Wang M, Li X, Yang R, Fang X, Zhao Z. New function of the CD44 gene: Lipid metabolism regulation in bovine mammary epithelial cells. J Dairy Sci 2020; 103:6661-6671. [DOI: 10.3168/jds.2019-17415] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 02/24/2020] [Indexed: 12/11/2022]
|
20
|
Ma J, Klemm J, Gerardo-Ramírez M, Frappart L, Castven D, Becker D, Zoch A, Parent R, Bartosch B, Minnich K, Giovannini M, Danckwardt S, Hartmann N, Morrison H, Herrlich P, Marquardt JU, Hartmann M. Cluster of differentiation 44 promotes osteosarcoma progression in mice lacking the tumor suppressor Merlin. Int J Cancer 2020; 147:2564-2577. [PMID: 32525563 DOI: 10.1002/ijc.33144] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 01/15/2023]
Abstract
Merlin is a versatile tumor suppressor protein encoded by the NF2 gene. Several lines of evidence suggest that Merlin exerts its tumor suppressor activity, at least in part, by forming an inhibitory complex with cluster of differentiation 44 (CD44). Consistently, numerous NF2 mutations in cancer patients are predicted to perturb the interaction of Merlin with CD44. We hypothesized that disruption of the Merlin-CD44 complex through loss of Merlin, unleashes putative tumor- or metastasis-promoting functions of CD44. To evaluate the relevance of the Merlin-CD44 interaction in vivo, we compared tumor growth and progression in Cd44-positive and Cd44-negative Nf2-mutant mice. Heterozygous Nf2-mutant mice were prone to developing highly metastatic osteosarcomas. Importantly, while the absence of the Cd44 gene had no effect on the frequency of primary osteosarcoma development, it strongly diminished osteosarcoma metastasis formation in the Nf2-mutant mice. In vitro assays identified transendothelial migration as the most prominent cellular phenotype dependent on CD44. Adhesion to endothelial cells was blocked by interfering with integrin α4β1 (very late antigen-4, VLA-4) on osteosarcoma cells and CD44 upregulated levels of integrin VLA-4 β1 subunit. Among other putative functions of CD44, which may contribute to the metastatic behavior, the passage through the endothelial cells also appears to be critical in vivo, as CD44 significantly promoted formation of lung metastasis upon intravenous injection of osteosarcoma cells into immunocompromised mice. Altogether, our results strongly suggest that CD44 plays a metastasis-promoting role in the absence of Merlin.
Collapse
Affiliation(s)
- Junzhi Ma
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Janina Klemm
- First Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Monserrat Gerardo-Ramírez
- First Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Lucien Frappart
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Darko Castven
- First Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Diana Becker
- First Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Ansgar Zoch
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Romain Parent
- Cancer Research Center of Lyon, INSERM U1052 and CNRS UMR5286, University of Lyon, Lyon, France
| | - Birke Bartosch
- Cancer Research Center of Lyon, INSERM U1052 and CNRS UMR5286, University of Lyon, Lyon, France
| | - Kerstin Minnich
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Marco Giovannini
- Department of Head and Neck Surgery, David Geffen School of Medicine at University of California, Los Angeles (UCLA) and Jonsson Comprehensive Cancer Center (JCCC), Los Angeles, California, USA
| | - Sven Danckwardt
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University, Mainz, Germany.,Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Nils Hartmann
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Helen Morrison
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Peter Herrlich
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Jens U Marquardt
- First Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Monika Hartmann
- First Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
21
|
Kim H, Takegahara N, Walsh MC, Choi Y. CD44 Can Compensate for IgSF11 Deficiency by Associating with the Scaffold Protein PSD-95 during Osteoclast Differentiation. Int J Mol Sci 2020; 21:ijms21072646. [PMID: 32290171 PMCID: PMC7177690 DOI: 10.3390/ijms21072646] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 01/07/2023] Open
Abstract
Differentiation of osteoclasts, which are specialized multinucleated macrophages capable of bone resorption, is driven primarily by receptor activator of NF-κB ligand (RANKL). Additional signaling from cell surface receptors, such as cell adhesion molecules (CAMs), is also required for osteoclast maturation. Previously, we have demonstrated that immunoglobulin superfamily 11 (IgSF11), a member of the immunoglobulin-CAM (IgCAM) family, plays an important role in osteoclast differentiation through association with the scaffold protein postsynaptic density protein 95 (PSD-95). Here, we demonstrate that the osteoclast-expressed CAM CD44 can compensate for IgSF11 deficiency when cell-cell interaction conditions are suboptimal by associating with PSD-95. Impaired osteoclast differentiation in IgSF11-deficient (IgSF11-/-) cultures was rescued by antibody-mediated stimulation of CD44 or by treatment with low-molecular-weight hyaluronan (LMW-HA), a CD44 ligand. Biochemical analysis revealed that PSD-95, which is required for osteoclast differentiation, associates with CD44 in osteoclasts regardless of the presence or absence of IgSF11. RNAi-mediated knockdown of PSD-95 abrogated the effects of either CD44 stimulation or LMW-HA treatment on osteoclast differentiation, suggesting that CD44, similar to IgSF11, is functionally associated with PSD-95 during osteoclast differentiation. Taken together, these results reveal that CD44 can compensate for IgSF11 deficiency in osteoclasts through association with PSD-95.
Collapse
Affiliation(s)
| | | | | | - Yongwon Choi
- Correspondence: ; Tel.: +215-746-6404; Fax: +215-573-0888
| |
Collapse
|
22
|
Oatley M, Bölükbası ÖV, Svensson V, Shvartsman M, Ganter K, Zirngibl K, Pavlovich PV, Milchevskaya V, Foteva V, Natarajan KN, Baying B, Benes V, Patil KR, Teichmann SA, Lancrin C. Single-cell transcriptomics identifies CD44 as a marker and regulator of endothelial to haematopoietic transition. Nat Commun 2020; 11:586. [PMID: 31996681 PMCID: PMC6989687 DOI: 10.1038/s41467-019-14171-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 12/18/2019] [Indexed: 12/13/2022] Open
Abstract
The endothelial to haematopoietic transition (EHT) is the process whereby haemogenic endothelium differentiates into haematopoietic stem and progenitor cells (HSPCs). The intermediary steps of this process are unclear, in particular the identity of endothelial cells that give rise to HSPCs is unknown. Using single-cell transcriptome analysis and antibody screening, we identify CD44 as a marker of EHT enabling us to isolate robustly the different stages of EHT in the aorta-gonad-mesonephros (AGM) region. This allows us to provide a detailed phenotypical and transcriptional profile of CD44-positive arterial endothelial cells from which HSPCs emerge. They are characterized with high expression of genes related to Notch signalling, TGFbeta/BMP antagonists, a downregulation of genes related to glycolysis and the TCA cycle, and a lower rate of cell cycle. Moreover, we demonstrate that by inhibiting the interaction between CD44 and its ligand hyaluronan, we can block EHT, identifying an additional regulator of HSPC development. The endothelial to haematopoietic transition (EHT) is the process where haemogenic endothelium differentiates into haematopoietic stem and progenitor cells (HSPCs). Here the authors use single cell transcriptomics and antibody screening to identify CD44 as a marker of EHT that is required for EHT and HSPC development.
Collapse
Affiliation(s)
- Morgan Oatley
- European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015, Monterotondo, Italy
| | - Özge Vargel Bölükbası
- European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015, Monterotondo, Italy.,Stem Cell and Regenerative Biology Department, Harvard University, 7 Divinity Avenue, Cambridge, MA, 02138, USA
| | - Valentine Svensson
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK.,European Molecular Biology Laboratory, EMBL-EBI, Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SD, UK.,Pachter Lab, Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA, USA
| | - Maya Shvartsman
- European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015, Monterotondo, Italy
| | - Kerstin Ganter
- European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015, Monterotondo, Italy
| | - Katharina Zirngibl
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Polina V Pavlovich
- European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015, Monterotondo, Italy.,Moscow Institute of Physics and Technology, Institutskii Per. 9, Moscow Region, Dolgoprudny, 141700, Russia.,Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, D-79108, Freiburg, Germany
| | - Vladislava Milchevskaya
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Meyerhofstrasse 1, 69117, Heidelberg, Germany.,Institut für Medizinische Statistik und Bioinformatik, Bachemer Strasse 86, 50931, Köln, Germany
| | - Vladimira Foteva
- European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015, Monterotondo, Italy
| | - Kedar N Natarajan
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK.,Department of Biochemistry and Molecular Biology, The University of Southern Denmark, Danish Institute for Advanced Study, Campusvej 55, 5230, Odense M, Denmark
| | - Bianka Baying
- European Molecular Biology Laboratory, Genomics Core Facility, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Vladimir Benes
- European Molecular Biology Laboratory, Genomics Core Facility, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Kiran R Patil
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Sarah A Teichmann
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Christophe Lancrin
- European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015, Monterotondo, Italy.
| |
Collapse
|
23
|
Cardoso BA. The Bone Marrow Niche - The Tumor Microenvironment That Ensures Leukemia Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:259-293. [PMID: 32130704 DOI: 10.1007/978-3-030-34025-4_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The human body requires a constant delivery of fresh blood cells that are needed to maintain body homeostasis. Hematopoiesis is the process that drives the formation of new blood cells from a single stem cell. This is a complex, orchestrated and tightly regulated process that occurs within the bone marrow. When such process is faulty or deregulated, leukemia arises, develops and thrives by subverting normal hematopoiesis and availing the supplies of this rich milieu.In this book chapter we will describe and characterize the bone marrow microenvironment and its key importance for leukemia expansion. The several components of the bone marrow niche, their interaction with the leukemic cells and the cellular pathways activated within the malignant cells will be emphasized. Finally, novel therapeutic strategies to target this sibling interaction will also be discussed.
Collapse
Affiliation(s)
- Bruno António Cardoso
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
24
|
Petukhov D, Richter-Dayan M, Fridlender Z, Breuer R, Wallach-Dayan SB. Increased Regeneration Following Stress-Induced Lung Injury in Bleomycin-Treated Chimeric Mice with CD44 Knockout Mesenchymal Cells. Cells 2019; 8:E1211. [PMID: 31591327 PMCID: PMC6829612 DOI: 10.3390/cells8101211] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 01/24/2023] Open
Abstract
CD44, an adhesion-molecule promoting cell-migration, is shown here to increase in stress conditions following bleomycin-induced apoptosis in alveolar epithelial cells (AECs), a main target of lung injury. In vivo, it inhibits tissue regeneration and leads to fibrosis. We show that some AECs survive by the ataxia-telangiectasia mutated kinase/ATM pathway, and undergo a CD44-mediated epithelial-mesenchymal transdifferentiation (EMT) with migratory capacities in vitro, and in vivo. We assessed apoptosis vs. proliferation of AECs following bleomycin, ATM/P53 signaling pathway in AECs, and CD44 involvement in EMT, cell motility and tissue regeneration in vitro and in vivo. Expression of survival genes, CD44, and ATM/p53 pathway was elevated in AECs surviving bleomycin injury, as were the markers of EMT (downregulation of E-cadherin, upregulation of N-cadherin and vimentin, nuclear translocation of β-catenin). Inhibition of CD44 decreased AECs transdifferentiation. Bleomycin-treated chimeric CD44KO-mice had decreased EMT markers, ATM, and mesenchymal cells (α-SMA+) accumulation in lung, increased surfactant-b, diminished lung mesenchymal cell motility, and increased lung tissue regenerative capacity following bleomycin injury, as indicated by lung collagen content and semiquantitave morphological index scoring. Thus, AECs surviving lung injury are plastic and undergo ATM-mediated, CD44-dependent transdifferentiation, preventing tissue regeneration and promoting fibrosis. Synthetic or natural compounds that downregulate CD44 may improve tissue regeneration following injury.
Collapse
Affiliation(s)
- Dmytro Petukhov
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, PO Box 12000, Qiryat Hadassah, Jerusalem 91120, Israel.
| | - Mark Richter-Dayan
- Department of Emergency Medicine, Hadassah-Hebrew University Medical Center, PO Box 12000, Qiryat Hadassah, Jerusalem 91120, Israel.
| | - Zvi Fridlender
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, PO Box 12000, Qiryat Hadassah, Jerusalem 91120, Israel.
| | - Raphael Breuer
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, PO Box 12000, Qiryat Hadassah, Jerusalem 91120, Israel.
- Department of Pathology, Boston University School of Medicine, 72 East Concord St., Boston, MA 02118, USA.
| | - Shulamit B Wallach-Dayan
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, PO Box 12000, Qiryat Hadassah, Jerusalem 91120, Israel.
| |
Collapse
|
25
|
Yang YM, Noureddin M, Liu C, Ohashi K, Kim SY, Ramnath D, Powell EE, Sweet MJ, Roh YS, Hsin IF, Deng N, Liu Z, Liang J, Mena E, Shouhed D, Schwabe RF, Jiang D, Lu SC, Noble PW, Seki E. Hyaluronan synthase 2-mediated hyaluronan production mediates Notch1 activation and liver fibrosis. Sci Transl Med 2019; 11:eaat9284. [PMID: 31189722 PMCID: PMC6589184 DOI: 10.1126/scitranslmed.aat9284] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 05/14/2019] [Indexed: 01/18/2023]
Abstract
Hyaluronan (HA), a major extracellular matrix glycosaminoglycan, is a biomarker for cirrhosis. However, little is known about the regulatory and downstream mechanisms of HA overproduction in liver fibrosis. Hepatic HA and HA synthase 2 (HAS2) expression was elevated in both human and murine liver fibrosis. HA production and liver fibrosis were reduced in mice lacking HAS2 in hepatic stellate cells (HSCs), whereas mice overexpressing HAS2 had exacerbated liver fibrosis. HAS2 was transcriptionally up-regulated by transforming growth factor-β through Wilms tumor 1 to promote fibrogenic, proliferative, and invasive properties of HSCs via CD44, Toll-like receptor 4 (TLR4), and newly identified downstream effector Notch1. Inhibition of HA synthesis by 4-methylumbelliferone reduced HSC activation and liver fibrosis in mice. Our study provides evidence that HAS2 actively synthesizes HA in HSCs and that it promotes HSC activation and liver fibrosis through Notch1. Targeted HA inhibition may have potential to be an effective therapy for liver fibrosis.
Collapse
Affiliation(s)
- Yoon Mee Yang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea
| | - Mazen Noureddin
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Cheng Liu
- Department of Infectious Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Koichiro Ohashi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - So Yeon Kim
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Divya Ramnath
- Institute for Molecular Bioscience (IMB) and IMB Centre for Inflammation and Disease Research, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Elizabeth E Powell
- Centre for Liver Disease Research, University of Queensland, Brisbane, Queensland, Australia
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, Queensland 4102, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience (IMB) and IMB Centre for Inflammation and Disease Research, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Yoon Seok Roh
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Pharmacy, Chungbuk National University College of Pharmacy, Cheongju, Chungbuk 28160, South Korea
| | - I-Fang Hsin
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Nan Deng
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Zhenqiu Liu
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jiurong Liang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Edward Mena
- California Liver Research Institute, Pasadena, CA 91105, USA
| | - Daniel Shouhed
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Robert F Schwabe
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Dianhua Jiang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Shelly C Lu
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Paul W Noble
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ekihiro Seki
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
26
|
Sieger D, Korzinskas T, Jung O, Stojanovic S, Wenisch S, Smeets R, Gosau M, Schnettler R, Najman S, Barbeck M. The Addition of High Doses of Hyaluronic Acid to a Biphasic Bone Substitute Decreases the Proinflammatory Tissue Response. Int J Mol Sci 2019; 20:E1969. [PMID: 31013636 PMCID: PMC6515558 DOI: 10.3390/ijms20081969] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/20/2019] [Accepted: 04/21/2019] [Indexed: 02/06/2023] Open
Abstract
Biphasic bone substitutes (BBS) are currently well-established biomaterials. Through their constant development, even natural components like hyaluronic acid (HY) have been added to improve both their handling and also their regenerative properties. However, little knowledge exists regarding the consequences of the addition of HY to their biocompatibility and the inflammatory tissue reactions. Thus, the present study was conducted, aiming to analyze the influence of two different amounts of high molecular weight HY (HMWHY), combined with a BBS, on in vitro biocompatibility and in vivo tissue reaction. Established in vitro procedures, using L929 cells, were used for cytocompatibility analyses under the test conditions of DIN EN:ISO 10993-5. For the in vivo part of the study, calvarial defects were created in 20 Wistar rats and subsequently filled with BBS, and BBS combined with two different HMWHY amounts, i.e., BBS + HY(L) and BBS + HY(H). As controls, empty defects were used. Established histological, immunohistochemical, and histomorphometrical methods were applied to analyze the tissue reactions to the three different materials, including the induction of pro- and anti-inflammatory macrophages and multinucleated giant cells (BMGCs). The in vitro results showed that none of the materials or compositions caused biological damage to the L929 cells and can be considered to be non-toxic. The in vivo results showed that only the addition of high doses of HY to a biphasic bone substitute significantly decreases the occurrence of pro-inflammatory macrophages (* p < 0.05), comparable to the numbers found in the control group, while no significant differences within the three study groups for M2-macrophages nor BMGCs were detected. In conclusion, the addition of different amounts of HMWHY does not seem to affect the inflammation response to BBS, while improving the material handling properties.
Collapse
Affiliation(s)
- Dominik Sieger
- Department of Oral and Maxillofacial Surgery, Division for Regenerative Orofacial Medicine, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Tadas Korzinskas
- Department of Oral and Maxillofacial Surgery, Division for Regenerative Orofacial Medicine, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Ole Jung
- Department of Oral and Maxillofacial Surgery, Division for Regenerative Orofacial Medicine, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Sanja Stojanovic
- Department for Cell and Tissue Engineering, Institute of Biology and Human Genetics, University of Niš, Faculty of Medicine, Niš 18106, Serbia.
| | - Sabine Wenisch
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, 35392 Giessen, Germany.
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, Division for Regenerative Orofacial Medicine, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
- Department of Oral Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Martin Gosau
- Department of Oral and Maxillofacial Surgery, Division for Regenerative Orofacial Medicine, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
- Department of Oral Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Reinhard Schnettler
- Department of Oral and Maxillofacial Surgery, Division for Regenerative Orofacial Medicine, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
- Department of Oral Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Stevo Najman
- Department for Cell and Tissue Engineering, Institute of Biology and Human Genetics, University of Niš, Faculty of Medicine, Niš 18106, Serbia.
| | - Mike Barbeck
- Department of Oral and Maxillofacial Surgery, Division for Regenerative Orofacial Medicine, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
- BerlinAnalytix GmbH, 12109 Berlin, Germany.
| |
Collapse
|
27
|
Hayashi Y, Sezaki M, Takizawa H. Development of the hematopoietic system: Role of inflammatory factors. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 8:e341. [PMID: 30916895 DOI: 10.1002/wdev.341] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 12/24/2022]
Abstract
Hematopoietic stem cells (HSCs) have two defining features, multipotency and self-renewal, both of which are tightly controlled by cell autonomous programs and environmental factors throughout the lifetime of an organism. During development, HSCs are born in the aorta-gonad-mesonephros region, and migrate to distinct hematopoietic organs such as the placenta, fetal liver and spleen, continuously self-renewing and expanding to reach a homeostatic number. HSCs ultimately seed the bone marrow around the time of birth and become dormant to sustain lifelong hematopoiesis. In this review, we will summarize the recent findings on the role of inflammatory factors regulating HSC development, that is, emergence, trafficking and differentiation. An understanding of HSC kinetics during developmental processes will provide useful knowledge on HSC behavior under physiological and pathophysiological conditions. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration Adult Stem Cells, Tissue Renewal, and Regeneration > Tissue Stem Cells and Niches Adult Stem Cells, Tissue Renewal, and Regeneration > Environmental Control of Stem Cells.
Collapse
Affiliation(s)
- Yoshikazu Hayashi
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Maiko Sezaki
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hitoshi Takizawa
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
28
|
CD44s Assembles Hyaluronan Coat on Filopodia and Extracellular Vesicles and Induces Tumorigenicity of MKN74 Gastric Carcinoma Cells. Cells 2019; 8:cells8030276. [PMID: 30909497 PMCID: PMC6468717 DOI: 10.3390/cells8030276] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/14/2019] [Accepted: 03/20/2019] [Indexed: 12/27/2022] Open
Abstract
CD44 is a multifunctional adhesion molecule typically upregulated in malignant, inflamed and injured tissues. Due to its ability to bind multiple ligands present in the tumor microenvironment, it promotes multiple cellular functions related to tumorigenesis. Recent data has shown that CD44 and its principal ligand hyaluronan (HA) are carried by extracellular vesicles (EV) derived from stem and tumor cells, but the role of CD44 in EV shedding has not been studied so far. To answer this question, we utilized CD44-negative human gastric carcinoma cell line MKN74 manipulated to stably express CD44 standard form (CD44s). The effect of CD44s expression on HA metabolism, EV secretion, morphology and growth of these cells was studied. Interestingly, HAS2 and HYAL2 expression levels were significantly upregulated in CD44s-expressing cells. Cell-associated HA levels were significantly increased, while HA levels in the culture medium of CD44s-positive cells was lower compared to CD44s-negative MOCK cells. CD44s expression had no significant effect on the proliferation capacity of cells, but cells showed diminished contact inhibition. Superresolution imaging revealed that CD44s and HA were accumulated on filopodia and EVs secreted from CD44s-positive cells, but no differences in total numbers of secreted EV between CD44s-negative and -positive cells was detected. In 3D cultures, CD44s-expressing cells had an enhanced invasion capacity in BME gel and increased spheroidal growth when cultured in collagen I gel. No significant differences in mitotic activity, tumor size or morphology were detected in CAM assays. However, a significant increase in HA staining coverage was detected in CD44s-positive tumors. Interestingly, CD44s-positive EVs embedded in HA-rich matrix were detected in the stromal areas of tumors. The results indicate that CD44s expression significantly increases the HA binding capacity of gastric cancer cells, while the secreted HA is downregulated. CD44s is also carried by EVs secreted by CD44s-expressing cells. These findings highlight the potential usefulness of CD44s and its ligands as multipurpose EV biomarkers, because they are upregulated in inflammatory, injured, and cancer cells and accumulate on the surface of EVs secreted in these situations.
Collapse
|
29
|
Al-Rekabi Z, Fura AM, Juhlin I, Yassin A, Popowics TE, Sniadecki NJ. Hyaluronan-CD44 interactions mediate contractility and migration in periodontal ligament cells. Cell Adh Migr 2019; 13:138-150. [PMID: 30676222 PMCID: PMC6527381 DOI: 10.1080/19336918.2019.1568140] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The role of hyaluronan (HA) in periodontal healing has been speculated via its interaction with the CD44 receptor. While HA-CD44 interactions have previously been implicated in numerous cell types; effect and mechanism of exogenous HA on periodontal ligament (PDL) cells is less clear. Herein, we examine the effect of exogenous HA on contractility and migration in human and murine PDL cells using arrays of microposts and time-lapse microscopy. Our findings observed HA-treated human PDL cells as more contractile and less migratory than untreated cells. Moreover, the effect of HA on contractility and focal adhesion area was abrogated when PDL cells were treated with Y27632, an inhibitor of rho-dependent kinase, but not when these cells were treated with ML-7, an inhibitor of myosin light chain kinase. Our results provide insight into the mechanobiology of PDL cells, which may contribute towards the development of therapeutic strategies for periodontal healing and tissue regeneration.
Collapse
Affiliation(s)
- Zeinab Al-Rekabi
- a Department of Mechanical Engineering , University of Washington , Seattle , WA , USA
| | - Adriane M Fura
- b Department of Bioengineering , University of Washington , Seattle , WA , USA
| | - Ilsa Juhlin
- a Department of Mechanical Engineering , University of Washington , Seattle , WA , USA
| | - Alaa Yassin
- c Department of Periodontics , University of Washington , Seattle , WA , USA
| | - Tracy E Popowics
- d Department of Oral Health Sciences , University of Washington , Seattle , WA , USA
| | - Nathan J Sniadecki
- a Department of Mechanical Engineering , University of Washington , Seattle , WA , USA.,b Department of Bioengineering , University of Washington , Seattle , WA , USA.,e Institute for Stem Cell and Regenerative Medicine , University of Washington , Seattle , WA , USA
| |
Collapse
|
30
|
Govindaraju P, Todd L, Shetye S, Monslow J, Puré E. CD44-dependent inflammation, fibrogenesis, and collagenolysis regulates extracellular matrix remodeling and tensile strength during cutaneous wound healing. Matrix Biol 2019; 75-76:314-330. [PMID: 29894820 PMCID: PMC6286871 DOI: 10.1016/j.matbio.2018.06.004] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/21/2018] [Accepted: 06/08/2018] [Indexed: 12/19/2022]
Abstract
Cutaneous wound healing consists of three main phases: inflammation, re-epithelialization, and tissue remodeling. During normal wound healing, these processes are tightly regulated to allow restoration of skin function and biomechanics. In many instances, healing leads to an excess accumulation of fibrillar collagen (the principal protein found in the extracellular matrix - ECM), and the formation of scar tissue, which has compromised biomechanics, tested using ramp to failure tests, compared to normal skin (Corr and Hart, 2013 [1]). Alterations in collagen accumulation and architecture have been attributed to the reduced tensile strength found in scar tissue (Brenda et al., 1999; Eleswarapu et al., 2011). Defining mechanisms that govern cellular functionality and ECM remodeling are vital to understanding normal versus pathological healing and developing approaches to prevent scarring. CD44 is a cell surface adhesion receptor expressed on nearly all cell types present in dermis. Although CD44 has been implicated in an array of inflammatory and fibrotic processes such as leukocyte recruitment, T-cell extravasation, and hyaluronic acid (the principal glycosaminoglycan found in the ECM) metabolism, the role of CD44 in cutaneous wound healing and scarring remains unknown. We demonstrate that in an excisional biopsy punch wound healing model, CD44-null mice have increased inflammatory and reduced fibrogenic responses during early phases of wound healing. At wound closure, CD44-null mice exhibit reduced collagen degradation leading to increased accumulation of fibrillar collagen, which persists after wound closure leading to reduced tensile strength resulting in a more severe scarring phenotype compared to WT mice. These data indicate that CD44 plays a previously unknown role in fibrillar collagen accumulation and wound healing during the injury response.
Collapse
Affiliation(s)
- Priya Govindaraju
- Department of Biomedical Sciences of the University of Pennsylvania, Philadelphia, PA, United States of America; Pharmacology Graduate Group of the University of Pennsylvania, Philadelphia, PA, United States of America
| | - Leslie Todd
- Department of Biomedical Sciences of the University of Pennsylvania, Philadelphia, PA, United States of America
| | - Snehal Shetye
- McKay Orthopaedic Research Laboratory of the University of Pennsylvania, Philadelphia, PA, United States of America
| | - James Monslow
- Department of Biomedical Sciences of the University of Pennsylvania, Philadelphia, PA, United States of America
| | - Ellen Puré
- Department of Biomedical Sciences of the University of Pennsylvania, Philadelphia, PA, United States of America; Pharmacology Graduate Group of the University of Pennsylvania, Philadelphia, PA, United States of America.
| |
Collapse
|
31
|
Schultz K, Grieger (Lindner) C, Li Y, Urbánek P, Ruschel A, Minnich K, Bruder D, Gereke M, Sechi A, Herrlich P. Gamma secretase dependent release of the CD44 cytoplasmic tail upregulates IFI16 in cd44-/- tumor cells, MEFs and macrophages. PLoS One 2018; 13:e0207358. [PMID: 30540779 PMCID: PMC6291121 DOI: 10.1371/journal.pone.0207358] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 10/30/2018] [Indexed: 01/01/2023] Open
Abstract
The adhesion molecule and co-receptor of receptor tyrosine kinases, CD44, is expressed in all cells of the immune system, but also in numerous non-immune cells. CD44 plays roles in the cellular response to different pathogens. The molecular actions of CD44 during these processes are by and large still unknown. The CD44 molecule undergoes a sequential proteolytic cleavage which leads to the release of a soluble intracellular domain (CD44-ICD). Previous reports had shown that the CD44-ICD is taken up into the nucleus where it enhances transcription of specific target genes. By RNA profiling we identified a CD44-dependent transcriptional increase of interferon-responsive genes, among them IFI16. IFI16 is important in the innate immune response. It senses and binds pathogenic DNA and, together with cGAS, activates the cGAS-cGAMP-STING pathway and induces the expression of genes relevant for the response, e.g. IFN-β. Our results show that the enhancement of IFI16 expression depended on CD44 cleavage. A CD44-negative tumor cell line, embryonic fibroblasts and bone marrow-derived macrophages from cd44-/- mice were reduced in their response to IFN-γ, to viral DNA fragments and to Listeria monocytogenes infection. We could rescue the deficiency of CD44 negative RPM-MC cells and cd44-/- MEFs by expressing only the soluble CD44-ICD in the absence of any other CD44 domain. Expression of the CD44-ICD carrying a mutation that prevented the uptake into the nucleus, could not rescue the absence of CD44. This molecular aspect of regulation by CD44 may explain part of the immune phenotypes of mice with cd44 gene disruption.
Collapse
Affiliation(s)
- Kristin Schultz
- Helmholtz Centre for Infection Research, Immune Regulation Group, Braunschweig, Germany
- Otto-von-Guericke-University Magdeburg, Institute of Medical Microbiology, Infection Prevention and Control, Magdeburg, Germany
| | | | - Yong Li
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Pavel Urbánek
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Anne Ruschel
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Kerstin Minnich
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Dunja Bruder
- Helmholtz Centre for Infection Research, Immune Regulation Group, Braunschweig, Germany
- Otto-von-Guericke-University Magdeburg, Institute of Medical Microbiology, Infection Prevention and Control, Magdeburg, Germany
| | - Marcus Gereke
- Helmholtz Centre for Infection Research, Immune Regulation Group, Braunschweig, Germany
- Otto-von-Guericke-University Magdeburg, Institute of Medical Microbiology, Infection Prevention and Control, Magdeburg, Germany
| | - Antonio Sechi
- Institute of Biomedical Engineering, Dept. of Cell Biology, Aachen, Germany
| | - Peter Herrlich
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
- * E-mail:
| |
Collapse
|
32
|
Leng Y, Abdullah A, Wendt MK, Calve S. Hyaluronic acid, CD44 and RHAMM regulate myoblast behavior during embryogenesis. Matrix Biol 2018; 78-79:236-254. [PMID: 30130585 DOI: 10.1016/j.matbio.2018.08.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/24/2018] [Accepted: 08/17/2018] [Indexed: 12/17/2022]
Abstract
Hyaluronic acid (HA) is an extracellular matrix (ECM) component that has been shown to play a significant role in regulating muscle cell behavior during repair and regeneration. For instance, ECM remodeling after muscle injury involves an upregulation in HA expression that is coupled with skeletal muscle precursor cell recruitment. However, little is known about the role of HA during skeletal muscle development. To gain insight into the way in which HA mediates embryonic myogenesis, we first determined the spatial distribution and gene expression of CD44, RHAMM and other HA related proteins in embryonic day (E)10.5 to E12.5 murine forelimbs. While HA and CD44 expression remained high, RHAMM decreased at both the protein (via immunohistochemistry) and RNA (via qPCR) levels. Next, we determined that 4-methylumbelliferone-mediated knockdown of HA synthesis inhibited the migration and proliferation of E11.5/E12.5 forelimb-derived cells. Then, the influence of CD44 and RHAMM on myoblast and connective tissue cell behavior was investigated using antibodies against these receptors. Anti-RHAMM, but not anti-CD44, significantly decreased the total distance myogenic progenitors migrated over 24 h, whereas both inhibited connective tissue cell migration. In contrast, anti-CD44 inhibited the proliferation of connective tissue cells and muscle progenitors, but anti-RHAMM had no effect. However, when myoblasts and connective tissue cells were depleted of CD44 and RHAMM by shRNA, motility and proliferation were significantly inhibited in both cells indicating that blocking cell surface-localized CD44 and RHAMM does not have as pronounced effect as global shRNA-mediated depletion of these receptors. These results show, for the first time, the distribution and activity of RHAMM in the context of skeletal muscle. Furthermore, our data indicate that HA, through interactions with CD44 and RHAMM, promotes myogenic progenitor migration and proliferation. Confirmation of the role of HA and its receptors in directing myogenesis will be useful for the design of regenerative therapies that aim to promote the restoration of damaged or diseased muscle.
Collapse
Affiliation(s)
- Yue Leng
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, IN 47907, United States of America
| | - Ammara Abdullah
- Medicinal Chemistry and Molecular Pharmacology, Hansen Life Sciences Research Building, Purdue University, 201 S University St, West Lafayette, IN 47907, United States of America
| | - Michael K Wendt
- Medicinal Chemistry and Molecular Pharmacology, Hansen Life Sciences Research Building, Purdue University, 201 S University St, West Lafayette, IN 47907, United States of America
| | - Sarah Calve
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, IN 47907, United States of America.
| |
Collapse
|
33
|
The survival of fetal and bone marrow monocyte-derived alveolar macrophages is promoted by CD44 and its interaction with hyaluronan. Mucosal Immunol 2018; 11:601-614. [PMID: 29067996 DOI: 10.1038/mi.2017.83] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 08/07/2017] [Accepted: 08/16/2017] [Indexed: 02/04/2023]
Abstract
Alveolar macrophages maintain lung homeostasis by performing important roles in immunosurveillance and lung surfactant catabolism. They express high levels of CD44 and are one of the few macrophage populations that constitutively bind hyaluronan, a ligand for CD44 and component of pericellular and extracellular matrices. Using adoptive transfer experiments and a mouse model of inflammation, we found that alveolar macrophages are initially depleted after an inflammatory insult then rapidly self-renew and return to original numbers after the resolution phase. Monocytes recruited to an inflamed lung differentiate and contribute to the alveolar macrophage pool, but this occurs over a much slower time frame than alveolar macrophage self-renewal. CD44 expression on both fetal and bone marrow-derived alveolar macrophages promoted their survival and provided a competitive advantage over CD44-deficient alveolar macrophages at homeostasis and after inflammation. CD44-mediated hyaluronan binding was induced by the alveolar environment, and this interaction promoted alveolar macrophage survival both ex vivo and in vivo. Without CD44, alveolar macrophages lacked a hyaluronan coat, were more susceptible to death, and were present at lower numbers in the alveolar space. This demonstrates a new role for CD44 and hyaluronan in promoting alveolar macrophage survival.
Collapse
|
34
|
Lee-Sayer SSM, Dougan MN, Cooper J, Sanderson L, Dosanjh M, Maxwell CA, Johnson P. CD44-mediated hyaluronan binding marks proliferating hematopoietic progenitor cells and promotes bone marrow engraftment. PLoS One 2018; 13:e0196011. [PMID: 29684048 PMCID: PMC5912764 DOI: 10.1371/journal.pone.0196011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/04/2018] [Indexed: 12/15/2022] Open
Abstract
CD44 is a widely expressed cell adhesion molecule that binds to the extracellular matrix component, hyaluronan. However, this interaction is not constitutive in most immune cells at steady state, as the ability of CD44 to engage hyaluronan is highly regulated. While activated T cells and macrophages gain the ability to bind hyaluronan by CD44, the status in other immune cells is less studied. Here we found a percentage of murine eosinophils, natural killer and natural killer T cells were capable of interacting with hyaluronan at steady state. To further investigate the consequences of hyaluronan binding by CD44 in the hematopoietic system, point mutations of CD44 that either cannot bind hyaluronan (LOF-CD44) or have an increased affinity for hyaluronan (GOF-CD44) were expressed in CD44-deficient bone marrow. Competitive bone marrow reconstitution of irradiated mice revealed an early preference for GOF-CD44 over WT-CD44 expressing cells, and for WT-CD44 over LOF-CD44 expressing cells, in the hematopoietic progenitor cell compartment. The advantage of the hyaluronan-binding cells was observed in the hematopoietic stem and progenitor populations, and was maintained throughout the immune system. Hematopoietic stem cells bound minimal hyaluronan at steady state, and this was increased when the cells were induced to proliferate whereas multipotent progenitors had an increased ability to bind hyaluronan at steady state. In vitro, the addition of hyaluronan promoted their proliferation. Thus, proliferating hematopoietic progenitors bind hyaluronan, and hyaluronan binding cells have a striking competitive advantage in bone marrow engraftment.
Collapse
Affiliation(s)
- Sally S. M. Lee-Sayer
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C., Canada
| | - Meghan N. Dougan
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C., Canada
- Department of Pediatrics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, B.C., Canada
| | - Jesse Cooper
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C., Canada
| | - Leslie Sanderson
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C., Canada
| | - Manisha Dosanjh
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C., Canada
| | - Christopher A. Maxwell
- Department of Pediatrics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, B.C., Canada
| | - Pauline Johnson
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C., Canada
- * E-mail:
| |
Collapse
|
35
|
Salustri A, Campagnolo L, Klinger FG, Camaioni A. Molecular organization and mechanical properties of the hyaluronan matrix surrounding the mammalian oocyte. Matrix Biol 2018; 78-79:11-23. [PMID: 29408277 DOI: 10.1016/j.matbio.2018.02.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 10/18/2022]
Abstract
Successful ovulation and oocyte fertilization are essential prerequisites for the beginning of life in sexually reproducing animals. In mammalian fertilization, the relevance of the protein coat surrounding the oocyte plasma membrane, known as zona pellucida, has been widely recognized, while, until not too long ago, the general belief was that the cumulus oophorus, consisting of follicle cells embedded in a hyaluronan rich extracellular matrix, was not essential. This opinion was based on in vitro fertilization procedures, in which a large number of sperms are normally utilized and the oocyte can be fertilized even if depleted of cumulus cells. Conversely, in vivo, only very few sperm cells reach the fertilization site, arguing against the possibility of a coincidental encounter with the oocyte. In the last two decades, proteins required for HA organization in the cumulus extracellular matrix have been identified and the study of fertility in mice deprived of the corresponding genes have provided compelling evidence that this jelly-like coat is critical for fertilization. This review focuses on the advances in understanding the molecular interactions making the cumulus environment suitable for oocyte and sperm encounter. Most of the studies on the molecular characterization of the cumulus extracellular matrix have been performed in the mouse and we will refer essentially to findings obtained in this animal model.
Collapse
Affiliation(s)
- Antonietta Salustri
- Department of Biomedicine and Prevention, Histology and Embryology Section, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| | - Luisa Campagnolo
- Department of Biomedicine and Prevention, Histology and Embryology Section, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Francesca Gioia Klinger
- Department of Biomedicine and Prevention, Histology and Embryology Section, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Antonella Camaioni
- Department of Biomedicine and Prevention, Histology and Embryology Section, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
36
|
López-Ortega O, Santos-Argumedo L. Myosin 1g Contributes to CD44 Adhesion Protein and Lipid Rafts Recycling and Controls CD44 Capping and Cell Migration in B Lymphocytes. Front Immunol 2017; 8:1731. [PMID: 29321775 PMCID: PMC5732150 DOI: 10.3389/fimmu.2017.01731] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/23/2017] [Indexed: 12/30/2022] Open
Abstract
Cell migration and adhesion are critical for immune system function and involve many proteins, which must be continuously transported and recycled in the cell. Recycling of adhesion molecules requires the participation of several proteins, including actin, tubulin, and GTPases, and of membrane components such as sphingolipids and cholesterol. However, roles of actin motor proteins in adhesion molecule recycling are poorly understood. In this study, we identified myosin 1g as one of the important motor proteins that drives recycling of the adhesion protein CD44 in B lymphocytes. We demonstrate that the lack of Myo1g decreases the cell-surface levels of CD44 and of the lipid raft surrogate GM1. In cells depleted of Myo1g, the recycling of CD44 was delayed, the delay seems to be caused at the level of formation of recycling complex and entry into recycling endosomes. Moreover, a defective lipid raft recycling in Myo1g-deficient cells had an impact both on the capping of CD44 and on cell migration. Both processes required the transportation of lipid rafts to the cell surface to deliver signaling components. Furthermore, the extramembrane was essential for cell expansion and remodeling of the plasma membrane topology. Therefore, Myo1g is important during the recycling of lipid rafts to the membrane and to the accompanied proteins that regulate plasma membrane plasticity. Thus, Myosin 1g contributes to cell adhesion and cell migration through CD44 recycling in B lymphocytes.
Collapse
Affiliation(s)
- Orestes López-Ortega
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| | - Leopoldo Santos-Argumedo
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
37
|
Joosten SPJ, Zeilstra J, van Andel H, Mijnals RC, Zaunbrecher J, Duivenvoorden AAM, van de Wetering M, Clevers H, Spaargaren M, Pals ST. MET Signaling Mediates Intestinal Crypt-Villus Development, Regeneration, and Adenoma Formation and Is Promoted by Stem Cell CD44 Isoforms. Gastroenterology 2017; 153:1040-1053.e4. [PMID: 28716720 DOI: 10.1053/j.gastro.2017.07.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 06/12/2017] [Accepted: 07/05/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Resistance of metastatic human colorectal cancer cells to drugs that block epidermal growth factor (EGF) receptor signaling could be caused by aberrant activity of other receptor tyrosine kinases, activating overlapping signaling pathways. One of these receptor tyrosine kinases could be MET, the receptor for hepatocyte growth factor (HGF). We investigated how MET signaling, and its interaction with CD44 (a putative MET coreceptor regulated by Wnt signaling and highly expressed by intestinal stem cells [ISCs] and adenomas) affects intestinal homeostasis, regeneration, and adenoma formation in mini-gut organoids and mice. METHODS We established organoid cultures from ISCs stimulated with HGF or EGF and assessed intestinal differentiation by immunohistochemistry. Mice with total epithelial disruption of MET (AhCre/Metfl/fl/LacZ) or ISC-specific disruption of MET (Lgr5Creert2/Metfl/fl/LacZ) and control mice (AhCre/Met+/+/LacZ, Lgr5Creert2/Met+/+/LacZ) were exposed to 10 Gy total body irradiation; intestinal tissues were collected, and homeostasis and regeneration were assessed by immunohistochemistry. We investigated adenoma organoid expansion stimulated by HGF or EGF using adenomas derived from Lgr5Creert2/Metfl/fl/Apcfl/fl and Lgr5Creert2/Met+/+/Apcfl/fl mice. The same mice were evaluated for adenoma prevalence and size. We also quantified adenomas in AhCre/Metfl/fl/Apcfl/+ mice compared with AhCre/Met+/+/Apcfl/+ control mice. We studied expansion of organoids generated from crypts and adenomas, stimulated by HGF or EGF, that were derived from mice expressing different CD44 splice variants (Cd44+/+, Cd44-/-, Cd44s/s, or Cd44v4-10/v4-10 mice). RESULTS Crypts incubated with EGF or HGF expanded into self-organizing mini-guts with similar levels of efficacy and contained all differentiated cell lineages. MET-deficient mice did not have defects in intestinal homeostasis. Total body irradiation reduced numbers of proliferating crypts in AhCre/Metfl/fl/LacZ mice. Lgr5Creert2/Metfl/fl/LacZ mice had impaired regeneration of MET-deficient ISCs. Adenoma organoids stimulated with EGF or HGF expanded to almost twice the size of nonstimulated organoids. MET-deficient adenoma organoids did not respond to HGF stimulation, but did respond to EGF. ISC-specific disruption of Met (Lgr5Creert2/Metfl/fl/Apcfl/fl mice) caused a twofold increase in apoptosis in microadenomas, resulting in an approximately 50% reduction of microadenoma numbers and significantly reduced average adenoma size. Total epithelial disruption of Met (AhCre/Metfl/fl/Apcfl/+ mice) resulted in an approximate 50% reduction in (micro)adenoma numbers. Intestinal crypts from Cd44-/- mice did not expand to the same extent as crypts from Cd44+/+ mice on stimulation with HGF, but had the same response to EGF. The negative effect on HGF-mediated growth was overcome by expression of CD44v4-10, but not by CD44s. Similarly, HGF-mediated expansion of adenoma organoids required CD44v4-10. CONCLUSIONS In studies of intestinal organoid cultures and mice with inducible deletion of MET, we found HGF receptor signaling to regulate intestinal homeostasis and regeneration, as well as adenoma formation. These activities of MET are promoted by the stem cell CD44 isoform CD44v4-10. Our findings provide rationale for targeting signaling via MET and CD44 during anti-EGF receptor therapy of patients with colorectal cancer or in patients resistant to EGF receptor inhibitors.
Collapse
Affiliation(s)
- Sander P J Joosten
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jurrit Zeilstra
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Harmen van Andel
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - R Clinton Mijnals
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Joost Zaunbrecher
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Annet A M Duivenvoorden
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Marc van de Wetering
- Hubrecht Institute and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hans Clevers
- Hubrecht Institute and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcel Spaargaren
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Steven T Pals
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
38
|
Hoggatt J, Kfoury Y, Scadden DT. Hematopoietic Stem Cell Niche in Health and Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 11:555-81. [PMID: 27193455 DOI: 10.1146/annurev-pathol-012615-044414] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Regulation of stem cells in adult tissues is a key determinant of how well an organism can respond to the stresses of physiological challenge and disease. This is particularly true of the hematopoietic system, where demands on host defenses can call for an acute increase in cell production. Hematopoietic stem cells receive the regulatory signals for cell production in adult mammals in the bone marrow, a tissue with higher-order architectural and functional organization than previously appreciated. Here, we review the data defining particular structural components and heterologous cells in the bone marrow that participate in hematopoietic stem cell function. Further, we explore the case for stromal-hematopoietic cell interactions contributing to neoplastic myeloid disease. As the hematopoietic regulatory networks in the bone marrow are revealed, it is anticipated that strategies will emerge for how to enhance or inhibit production of specific blood cells. In that way, the control of hematopoiesis will enter the domain of therapies to modulate broad aspects of hematopoiesis, both normal and malignant.
Collapse
Affiliation(s)
- Jonathan Hoggatt
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138.,Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114;
| | - Youmna Kfoury
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138.,Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114;
| | - David T Scadden
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138.,Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114;
| |
Collapse
|
39
|
Abstract
Several recent studies in a number of model systems including zebrafish, Arabidopsis, and mouse have revealed phenotypic differences between knockouts (i.e., mutants) and knockdowns (e.g., antisense-treated animals). These differences have been attributed to a number of reasons including off-target effects of the antisense reagents. An alternative explanation was recently proposed based on a zebrafish study reporting that genetic compensation was observed in egfl7 mutant but not knockdown animals. Dosage compensation was first reported in Drosophila in 1932, and genetic compensation in response to a gene knockout was first reported in yeast in 1969. Since then, genetic compensation has been documented many times in a number of model organisms; however, our understanding of the underlying molecular mechanisms remains limited. In this review, we revisit studies reporting genetic compensation in higher eukaryotes and outline possible molecular mechanisms, which may include both transcriptional and posttranscriptional processes.
Collapse
Affiliation(s)
- Mohamed A. El-Brolosy
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- * E-mail:
| |
Collapse
|
40
|
Johnson LA, Banerji S, Lawrance W, Gileadi U, Prota G, Holder KA, Roshorm YM, Hanke T, Cerundolo V, Gale NW, Jackson DG. Dendritic cells enter lymph vessels by hyaluronan-mediated docking to the endothelial receptor LYVE-1. Nat Immunol 2017; 18:762-770. [PMID: 28504698 DOI: 10.1038/ni.3750] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/24/2017] [Indexed: 02/07/2023]
Abstract
Trafficking of tissue dendritic cells (DCs) via lymph is critical for the generation of cellular immune responses in draining lymph nodes (LNs). In the current study we found that DCs docked to the basolateral surface of lymphatic vessels and transited to the lumen through hyaluronan-mediated interactions with the lymph-specific endothelial receptor LYVE-1, in dynamic transmigratory-cup-like structures. Furthermore, we show that targeted deletion of the gene Lyve1, antibody blockade or depletion of the DC hyaluronan coat not only delayed lymphatic trafficking of dermal DCs but also blunted their capacity to prime CD8+ T cell responses in skin-draining LNs. Our findings uncovered a previously unknown function for LYVE-1 and show that transit through the lymphatic network is initiated by the recognition of leukocyte-derived hyaluronan.
Collapse
Affiliation(s)
- Louise A Johnson
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Suneale Banerji
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - William Lawrance
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Uzi Gileadi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Gennaro Prota
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Kayla A Holder
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Yaowaluck M Roshorm
- Division of Biotechnology, School of Bioresources and Technology, King Monkut's University of Technology, Thonburi, Thailand
| | - Tomáš Hanke
- The Jenner Institute, University of Oxford, Oxford, UK
- International Research Centre for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | - David G Jackson
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| |
Collapse
|
41
|
Oliveira ML, Akkapeddi P, Alcobia I, Almeida AR, Cardoso BA, Fragoso R, Serafim TL, Barata JT. From the outside, from within: Biological and therapeutic relevance of signal transduction in T-cell acute lymphoblastic leukemia. Cell Signal 2017. [PMID: 28645565 DOI: 10.1016/j.cellsig.2017.06.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological cancer that arises from clonal expansion of transformed T-cell precursors. In this review we summarize the current knowledge on the external stimuli and cell-intrinsic lesions that drive aberrant activation of pivotal, pro-tumoral intracellular signaling pathways in T-cell precursors, driving transformation, leukemia expansion, spread or resistance to therapy. In addition to their pathophysiological relevance, receptors and kinases involved in signal transduction are often attractive candidates for targeted drug development. As such, we discuss also the potential of T-ALL signaling players as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Mariana L Oliveira
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Padma Akkapeddi
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Isabel Alcobia
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Afonso R Almeida
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Bruno A Cardoso
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Rita Fragoso
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Teresa L Serafim
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - João T Barata
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal.
| |
Collapse
|
42
|
Highly tumorigenic hepatocellular carcinoma cell line with cancer stem cell-like properties. PLoS One 2017; 12:e0171215. [PMID: 28152020 PMCID: PMC5289561 DOI: 10.1371/journal.pone.0171215] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/18/2017] [Indexed: 12/21/2022] Open
Abstract
There are limited numbers of models to study hepatocellular carcinoma (HCC) in vivo in immunocompetent hosts. In an effort to develop a cell line with improved tumorigenicity, we derived a new cell line from Hepa1-6 cells through an in vivo passage in C57BL/6 mice. The resulting Dt81Hepa1-6 cell line showed enhanced tumorigenicity compared to Hepa1-6 with more frequent (28±12 vs. 0±0 lesions at 21 days) and more rapid tumor development (21 (100%) vs. 70 days (10%)) in C57BL/6 mice. The minimal Dt81Hepa1-6 cell number required to obtain visible tumors was 100,000 cells. The Dt81Hepa1-6 cell line showed high hepatotropism with subcutaneous injection leading to liver tumors without development of tumors in lungs or spleen. In vitro, Dt81Hepa1-6 cells showed increased anchorage-independent growth (34.7±6.8 vs. 12.3±3.3 colonies; P<0.05) and increased EpCAM (8.7±1.1 folds; P<0.01) and β-catenin (5.4±1.0 folds; P<0.01) expression. A significant proportion of Dt81Hepa1-6 cells expressed EpCAM compared to Hepa1-6 (34.8±1.1% vs 0.9±0.13%; P<0.001). Enriched EpCAM+ Dt81Hepa1-6 cells led to higher tumor load than EpCAM- Dt81Hepa1-6 cells (1093±74 vs 473±100 tumors; P<0.01). The in vivo selected Dt81Hepa1-6 cell line shows high liver specificity and increased tumorigenicity compared to Hepa1-6 cells. These properties are associated with increased expression of EpCAM and β-catenin confirming that EpCAM+ HCC cells comprise a subset with characteristics of tumor-initiating cells with stem/progenitor cell features. The Dt81Hepa1-6 cell line with its cancer stem cell-like properties will be a useful tool for the study of hepatocellular carcinoma in vivo.
Collapse
|
43
|
Tamma R, Ribatti D. Bone Niches, Hematopoietic Stem Cells, and Vessel Formation. Int J Mol Sci 2017; 18:ijms18010151. [PMID: 28098778 PMCID: PMC5297784 DOI: 10.3390/ijms18010151] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/14/2016] [Accepted: 01/09/2017] [Indexed: 02/06/2023] Open
Abstract
Bone marrow (BM) is a source of hematopoietic stem cells (HSCs). HSCs are localized in both the endosteum, in the so-called endosteal niche, and close to thin-walled and fenestrated sinusoidal vessel in the center of BM, in the so-called vascular niche. HSCs give rise to all types of mature blood cells through a process finely controlled by numerous signals emerging from the bone marrow niches where HSCs reside. This review will focus on the description of the role of BM niches in the control of the fate of HSCs and will also highlight the role of the BM niches in the regulation of vasculogenesis and angiogenesis. Moreover, alterations of the signals in niche microenvironment are involved in many aspects of tumor progression and vascularization and further knowledge could provide the basis for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, 70124 Bari, Italy.
- National Cancer Institute Giovanni Paolo II, 70124 Bari, Italy.
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, 70124 Bari, Italy.
- National Cancer Institute Giovanni Paolo II, 70124 Bari, Italy.
| |
Collapse
|
44
|
Dong Y, Arif A, Olsson M, Cali V, Hardman B, Dosanjh M, Lauer M, Midura RJ, Hascall VC, Brown KL, Johnson P. Endotoxin free hyaluronan and hyaluronan fragments do not stimulate TNF-α, interleukin-12 or upregulate co-stimulatory molecules in dendritic cells or macrophages. Sci Rep 2016; 6:36928. [PMID: 27869206 PMCID: PMC5116629 DOI: 10.1038/srep36928] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 10/24/2016] [Indexed: 12/24/2022] Open
Abstract
The extracellular matrix glycosaminoglycan, hyaluronan, has been described as a regulator of tissue inflammation, with hyaluronan fragments reported to stimulate innate immune cells. High molecular mass hyaluronan is normally present in tissues, but upon inflammation lower molecular mass fragments are generated. It is unclear if these hyaluronan fragments induce an inflammatory response or are a consequence of inflammation. In this study, mouse bone marrow derived macrophages and dendritic cells (DCs) were stimulated with various sizes of hyaluronan from different sources, fragmented hyaluronan, hyaluronidases and heavy chain modified-hyaluronan (HA-HC). Key pro-inflammatory molecules, tumour necrosis factor alpha, interleukin-1 beta, interleukin-12, CCL3, and the co-stimulatory molecules, CD40 and CD86 were measured. Only human umbilical cord hyaluronan, bovine testes and Streptomyces hyaluronlyticus hyaluronidase stimulated macrophages and DCs, however, these reagents were found to be contaminated with endotoxin, which was not fully removed by polymyxin B treatment. In contrast, pharmaceutical grade hyaluronan and hyaluronan fragments failed to stimulate in vitro-derived or ex vivo macrophages and DCs, and did not induce leukocyte recruitment after intratracheal instillation into mouse lungs. Hence, endotoxin-free pharmaceutical grade hyaluronan does not stimulate macrophages and DCs in our inflammatory models. These results emphasize the importance of ensuring hyaluronan preparations are endotoxin free.
Collapse
Affiliation(s)
- Yifei Dong
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C. V6T 1Z3, Canada
| | - Arif Arif
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C. V6T 1Z3, Canada
| | - Mia Olsson
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C. V6T 1Z3, Canada.,Department of Pediatrics, British Columbia Children's Hospital Research Institute, Vancouver, B.C. Canada
| | - Valbona Cali
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio 44195, U.S.A
| | - Blair Hardman
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C. V6T 1Z3, Canada
| | - Manisha Dosanjh
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C. V6T 1Z3, Canada
| | - Mark Lauer
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio 44195, U.S.A
| | - Ronald J Midura
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio 44195, U.S.A
| | - Vincent C Hascall
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio 44195, U.S.A
| | - Kelly L Brown
- Department of Pediatrics, British Columbia Children's Hospital Research Institute, Vancouver, B.C. Canada
| | - Pauline Johnson
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C. V6T 1Z3, Canada
| |
Collapse
|
45
|
Shatirishvili M, Burk AS, Franz CM, Pace G, Kastilan T, Breuhahn K, Hinterseer E, Dierich A, Bakiri L, Wagner EF, Ponta H, Hartmann TN, Tanaka M, Orian-Rousseau V. Epidermal-specific deletion of CD44 reveals a function in keratinocytes in response to mechanical stress. Cell Death Dis 2016; 7:e2461. [PMID: 27831556 PMCID: PMC5260879 DOI: 10.1038/cddis.2016.342] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/18/2016] [Accepted: 09/23/2016] [Indexed: 11/09/2022]
Abstract
CD44, a large family of transmembrane glycoproteins, plays decisive roles in physiological and pathological conditions. CD44 isoforms are involved in several signaling pathways essential for life such as growth factor-induced signaling by EGF, HGF or VEGF. CD44 is also the main hyaluronan (HA) receptor and as such is involved in HA-dependent processes. To allow a genetic dissection of CD44 functions in homeostasis and disease, we generated a Cd44 floxed allele allowing tissue- and time-specific inactivation of all CD44 isoforms in vivo. As a proof of principle, we inactivated Cd44 in the skin epidermis using the K14Cre allele. Although the skin of such Cd44Δker mutants appeared morphologically normal, epidermal stiffness was reduced, wound healing delayed and TPA induced epidermal thickening decreased. These phenotypes might be caused by cell autonomous defects in differentiation and HA production as well as impaired adhesion and migration on HA by Cd44Δker keratinocytes. These findings support the usefulness of the conditional Cd44 allele in unraveling essential physiological and pathological functions of CD44 isoforms.
Collapse
Affiliation(s)
- M Shatirishvili
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Karlsruhe, Germany
| | - A S Burk
- University of Heidelberg, Institute of Physical Chemistry, Heidelberg, Germany
| | - C M Franz
- Karlsruhe Institute of Technology, DFG-Center for Functional Nanostructures, Karlsruhe, Germany
| | - G Pace
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Karlsruhe, Germany
| | - T Kastilan
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Karlsruhe, Germany
| | - K Breuhahn
- Heidelberg University Hospital, Institute of Pathology, Heidelberg, Germany
| | - E Hinterseer
- Laboratory for Immunological and Molecular Cancer Research, Third Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases, and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - A Dierich
- Institut Clinique de la Souris Illkirch, Illkirch, France
| | - L Bakiri
- Spanish National Cancer Centre, Genes Development and Disease Group, Cancer Cell Biology Programme, Madrid, Spain
| | - E F Wagner
- Spanish National Cancer Centre, Genes Development and Disease Group, Cancer Cell Biology Programme, Madrid, Spain
| | - H Ponta
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Karlsruhe, Germany
| | - T N Hartmann
- Laboratory for Immunological and Molecular Cancer Research, Third Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases, and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - M Tanaka
- University of Heidelberg, Institute of Physical Chemistry, Heidelberg, Germany.,Institute for Integrated Cell-Material Sciences (WPI iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - V Orian-Rousseau
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Karlsruhe, Germany
| |
Collapse
|
46
|
Hyaluronan Depolymerization by Megakaryocyte Hyaluronidase-2 Is Required for Thrombopoiesis. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2390-403. [PMID: 27398974 DOI: 10.1016/j.ajpath.2016.05.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 04/19/2016] [Accepted: 05/10/2016] [Indexed: 12/14/2022]
Abstract
Hyaluronan is the predominant glycosaminoglycan component of the extracellular matrix with an emerging role in hematopoiesis. Modulation of hyaluronan polymer size is responsible for its control over cellular functions, and the balance of hyaluronan synthesis and degradation determines its molecular size. Although two active somatic hyaluronidases are expressed in mammals, only deficiency in hyaluronidase-2 (Hyal-2) results in thrombocytopenia of unknown mechanism. Our results reveal that Hyal-2 knockout mice accumulate hyaluronan within their bone marrow and within megakaryocytes, the cells responsible for platelet generation. Proplatelet formation by Hyal-2 knockout megakaryocytes was disrupted because of abnormal formation of the demarcation membrane system, which was dilated and poorly developed. Importantly, peptide-mediated delivery of exogenous hyaluronidase rescued deficient proplatelet formation in murine and human megakaryocytes lacking Hyal-2. Together, our data uncover a previously unsuspected mechanism of how hyaluronan and Hyal-2 control platelet generation.
Collapse
|
47
|
CD44 promotes chemoresistance in T-ALL by increased drug efflux. Exp Hematol 2016; 44:166-71.e17. [DOI: 10.1016/j.exphem.2015.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 11/28/2015] [Accepted: 12/01/2015] [Indexed: 11/20/2022]
|
48
|
Passer D, van de Vrugt A, Atmanli A, Domian IJ. Atypical Protein Kinase C-Dependent Polarized Cell Division Is Required for Myocardial Trabeculation. Cell Rep 2016; 14:1662-1672. [PMID: 26876178 DOI: 10.1016/j.celrep.2016.01.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 08/06/2015] [Accepted: 01/05/2016] [Indexed: 12/17/2022] Open
Abstract
A hallmark of cardiac development is the formation of myocardial trabeculations exclusively from the luminal surface of the primitive heart tube. Although a number of genetic defects in the endocardium and cardiac jelly disrupt myocardial trabeculation, the role of cell polarization remains unclear. Here, we demonstrate that atypical protein kinase C iota (Prkci) and its interacting partners are localized primarily to the luminal side of myocardial cells of early murine embryonic hearts. A subset of these cells undergoes polarized cell division with the cell division plane perpendicular to the heart's lumen. Disruption of the cell polarity complex by targeted gene mutations results in aberrant mitotic spindle alignment, loss of polarized cardiomyocyte division, and loss of normal myocardial trabeculation. Collectively, these results suggest that, in response to inductive signals, Prkci and its downstream partners direct polarized cell division of luminal myocardial cells to drive trabeculation in the nascent heart.
Collapse
Affiliation(s)
- Derek Passer
- Cardiovascular Research Center, Massachusetts General Hospital, Charles River Plaza/CPZN 3200, 185 Cambridge Street, Boston, MA 02114-2790, USA; Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA; Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Annebel van de Vrugt
- Cardiovascular Research Center, Massachusetts General Hospital, Charles River Plaza/CPZN 3200, 185 Cambridge Street, Boston, MA 02114-2790, USA; Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Ayhan Atmanli
- Cardiovascular Research Center, Massachusetts General Hospital, Charles River Plaza/CPZN 3200, 185 Cambridge Street, Boston, MA 02114-2790, USA; Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Ibrahim J Domian
- Cardiovascular Research Center, Massachusetts General Hospital, Charles River Plaza/CPZN 3200, 185 Cambridge Street, Boston, MA 02114-2790, USA; Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA; Harvard Stem Cell Institute, 1350 Massachusetts Avenue, Cambridge, MA 02138, USA.
| |
Collapse
|
49
|
Liang J, Jiang D, Noble PW. Hyaluronan as a therapeutic target in human diseases. Adv Drug Deliv Rev 2016; 97:186-203. [PMID: 26541745 PMCID: PMC4753080 DOI: 10.1016/j.addr.2015.10.017] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 02/07/2023]
Abstract
Accumulation and turnover of extracellular matrix is a hallmark of tissue injury, repair and remodeling in human diseases. Hyaluronan is a major component of the extracellular matrix and plays an important role in regulating tissue injury and repair, and controlling disease outcomes. The function of hyaluronan depends on its size, location, and interactions with binding partners. While fragmented hyaluronan stimulates the expression of an array of genes by a variety of cell types regulating inflammatory responses and tissue repair, cell surface hyaluronan provides protection against tissue damage from the environment and promotes regeneration and repair. The interactions of hyaluronan and its binding proteins participate in the pathogenesis of many human diseases. Thus, targeting hyaluronan and its interactions with cells and proteins may provide new approaches to developing therapeutics for inflammatory and fibrosing diseases. This review focuses on the role of hyaluronan in biological and pathological processes, and as a potential therapeutic target in human diseases.
Collapse
Affiliation(s)
- Jiurong Liang
- Department of Medicine and Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dianhua Jiang
- Department of Medicine and Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Paul W Noble
- Department of Medicine and Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
50
|
Up-regulation of CD44 in the development of metastasis, recurrence and drug resistance of ovarian cancer. Oncotarget 2016; 6:9313-26. [PMID: 25823654 PMCID: PMC4496219 DOI: 10.18632/oncotarget.3220] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 01/26/2015] [Indexed: 12/17/2022] Open
Abstract
The clinical significance of Cluster of Differentiation 44 (CD44) remains controversial in human ovarian cancer. The aim of this study is to evaluate the clinical significance of CD44 expression by using a unique tissue microarray, and then to determine the biological functions of CD44 in ovarian cancer. In this study, a unique ovarian cancer tissue microarray (TMA) was constructed with paired primary, metastatic, and recurrent tumor tissues from 26 individual patients. CD44 expression in TMA was assessed by immunohistochemistry. Both the metastatic and recurrent ovarian cancer tissues expressed higher level of CD44 than the patient-matched primary tumor. A significant association has been shown between CD44 expression and both the disease free survival and overall survival. A strong increase of CD44 was found in the tumor recurrence of mouse model. Finally, when CD44 was knocked down, proliferation, migration/invasion activity, and spheroid formation were significantly suppressed, while drug sensitivity was enhanced. Thus, up-regulation of CD44 represents a crucial event in the development of metastasis, recurrence, and drug resistance to current treatments in ovarian cancer. Developing strategies to target CD44 may prevent metastasis, recurrence, and drug resistance in ovarian cancer.
Collapse
|