1
|
Irons EE, Sajina GC, Lau JT. Sialic acid in the regulation of blood cell production, differentiation and turnover. Immunology 2024; 172:517-532. [PMID: 38503445 PMCID: PMC11223974 DOI: 10.1111/imm.13780] [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: 11/27/2023] [Accepted: 03/04/2024] [Indexed: 03/21/2024] Open
Abstract
Sialic acid is a unique sugar moiety that resides in the distal and most accessible position of the glycans on mammalian cell surface and extracellular glycoproteins and glycolipids. The potential for sialic acid to obscure underlying structures has long been postulated, but the means by which such structural changes directly affect biological processes continues to be elucidated. Here, we appraise the growing body of literature detailing the importance of sialic acid for the generation, differentiation, function and death of haematopoietic cells. We conclude that sialylation is a critical post-translational modification utilized in haematopoiesis to meet the dynamic needs of the organism by enforcing rapid changes in availability of lineage-specific cell types. Though long thought to be generated only cell-autonomously within the intracellular ER-Golgi secretory apparatus, emerging data also demonstrate previously unexpected diversity in the mechanisms of sialylation. Emphasis is afforded to the mechanism of extrinsic sialylation, whereby extracellular enzymes remodel cell surface and extracellular glycans, supported by charged sugar donor molecules from activated platelets.
Collapse
Affiliation(s)
| | | | - Joseph T.Y. Lau
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203 USA
| |
Collapse
|
2
|
Das N, Panda D, Gajendra S, Gupta R, Thakral D, Kaur G, Khan A, Singh VK, Vemprala A, Bakhshi S, Seth R, Sahoo RK, Sharma A, Rai S, Prajapati VK, Singh S. Immunophenotypic characterization of leukemic stem cells in acute myeloid leukemia using single tube 10-colour panel by multiparametric flow cytometry: Deciphering the spectrum, complexity and immunophenotypic heterogeneity. Int J Lab Hematol 2024; 46:646-656. [PMID: 38456256 DOI: 10.1111/ijlh.14250] [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: 07/12/2023] [Accepted: 01/30/2024] [Indexed: 03/09/2024]
Abstract
INTRODUCTION Despite extensive research, comprehensive characterization of leukaemic stem cells (LSC) and information on their immunophenotypic differences from normal haematopoietic stem cells (HSC) is lacking. Herein, we attempted to unravel the immunophenotypic (IPT) characteristics and heterogeneity of LSC using multiparametric flow cytometry (MFC) and single-cell sequencing. MATERIALS AND METHODS Bone marrow aspirate samples from patients with acute myeloid leukaemia (AML) were evaluated using MFC at diagnostic and post induction time points using a single tube-10-colour-panel containing LSC-associated antibodies CD123, CD45RA, CD44, CD33 and COMPOSITE (CLL-1, TIM-3, CD25, CD11b, CD22, CD7, CD56) with backbone markers that is, CD45, CD34, CD38, CD117, sCD3. Single-cell sequencing of the whole transcriptome was also done in a bone marrow sample. RESULTS LSCs and HSCs were identified in 225/255 (88.2%) and 183/255 (71.6%) samples, respectively. Significantly higher expression was noted for COMPOSITE, CD45RA, CD123, CD33, and CD44 in LSCs than HSCs (p < 0.0001). On comparing the LSC specific antigen expressions between CD34+ (n = 184) and CD34- LSCs (n = 41), no difference was observed between the groups. More than one sub-population of LSC was demonstrated in 4.4% of cases, which further revealed high concordance between MFC and single cell transcriptomic analysis in one of the cases displaying three LSC subpopulations by both methods. CONCLUSION A single tube-10-colour MFC panel is proposed as an easy and reproducible tool to identify and discriminate LSCs from HSCs. LSCs display both inter- and intra-sample heterogeneity in terms of antigen expressions, which opens the facets for single cell molecular analysis to elucidate the role of subpopulations of LSCs in AML progression.
Collapse
Affiliation(s)
- Nupur Das
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Devasis Panda
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Smeeta Gajendra
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Ritu Gupta
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Deepshi Thakral
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Gurvinder Kaur
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Aafreen Khan
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Vivek Kumar Singh
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Arushi Vemprala
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Rachna Seth
- Department of Paediatrics, Dr. BRAIRCH, AIIMS, New Delhi, India
| | | | - Atul Sharma
- Department of Medical Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Sandeep Rai
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Vijay K Prajapati
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Saroj Singh
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| |
Collapse
|
3
|
Domma AJ, Henderson LA, Nurdin JA, Kamil JP. Uncloaking the viral glycocalyx: How do viruses exploit glycoimmune checkpoints? Adv Virus Res 2024; 119:63-110. [PMID: 38897709 PMCID: PMC11192240 DOI: 10.1016/bs.aivir.2024.03.001] [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] [Indexed: 06/21/2024]
Abstract
The surfaces of cells and enveloped viruses alike are coated in carbohydrates that play multifarious roles in infection and immunity. Organisms across all kingdoms of life make use of a diverse set of monosaccharide subunits, glycosidic linkages, and branching patterns to encode information within glycans. Accordingly, sugar-patterning enzymes and glycan binding proteins play integral roles in cell and organismal biology, ranging from glycoprotein quality control within the endoplasmic reticulum to lymphocyte migration, coagulation, inflammation, and tissue homeostasis. Unsurprisingly, genes involved in generating and recognizing oligosaccharide patterns are playgrounds for evolutionary conflicts that abound in cross-species interactions, exemplified by the myriad plant lectins that function as toxins. In vertebrates, glycans bearing acidic nine-carbon sugars called sialic acids are key regulators of immune responses. Various bacterial and fungal pathogens adorn their cells in sialic acids that either mimic their hosts' or are stolen from them. Yet, how viruses commandeer host sugar-patterning enzymes to thwart immune responses remains poorly studied. Here, we review examples of viruses that interact with sialic acid-binding immunoglobulin-like lectins (Siglecs), a family of immune cell receptors that regulate toll-like receptor signaling and govern glycoimmune checkpoints, while highlighting knowledge gaps that merit investigation. Efforts to illuminate how viruses leverage glycan-dependent checkpoints may translate into new clinical treatments that uncloak viral antigens and infected cell surfaces by removing or masking immunosuppressive sialoglycans, or by inhibiting viral gene products that induce their biosynthesis. Such approaches may hold the potential to unleash the immune system to clear long intractable chronic viral infections.
Collapse
Affiliation(s)
- Anthony J Domma
- LSU Health Sciences Center at Shreveport, Shreveport, LA, United States
| | | | - Jeffery A Nurdin
- LSU Health Sciences Center at Shreveport, Shreveport, LA, United States
| | - Jeremy P Kamil
- LSU Health Sciences Center at Shreveport, Shreveport, LA, United States.
| |
Collapse
|
4
|
Cerreto M, Foà R, Natoni A. The Role of the Microenvironment and Cell Adhesion Molecules in Chronic Lymphocytic Leukemia. Cancers (Basel) 2023; 15:5160. [PMID: 37958334 PMCID: PMC10647257 DOI: 10.3390/cancers15215160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a B-cell malignancy whose progression largely depends on the lymph node and bone marrow microenvironment. Indeed, CLL cells actively proliferate in specific regions of these anatomical compartments, known as proliferation centers, while being quiescent in the blood stream. Hence, CLL cell adhesion and migration into these protective niches are critical for CLL pathophysiology. CLL cells are lodged in their microenvironment through a series of molecular interactions that are mediated by cellular adhesion molecules and their counter receptors. The importance of these adhesion molecules in the clinic is demonstrated by the correlation between the expression levels of some of them, in particular CD49d, and the prognostic likelihood. Furthermore, novel therapeutic agents, such as ibrutinib, impair the functions of these adhesion molecules, leading to an egress of CLL cells from the lymph nodes and bone marrow into the circulation together with an inhibition of homing into these survival niches, thereby preventing disease progression. Several adhesion molecules have been shown to participate in CLL adhesion and migration. Their importance also stems from the observation that they are involved in promoting, directly or indirectly, survival signals that sustain CLL proliferation and limit the efficacy of standard and novel chemotherapeutic drugs, a process known as cell adhesion-mediated drug resistance. In this respect, many studies have elucidated the molecular mechanisms underlying cell adhesion-mediated drug resistance, which have highlighted different signaling pathways that may represent potential therapeutic targets. Here, we review the role of the microenvironment and the adhesion molecules that have been shown to be important in CLL and their impact on transendothelial migration and cell-mediated drug resistance. We also discuss how novel therapeutic compounds modulate the function of this important class of molecules.
Collapse
Affiliation(s)
| | | | - Alessandro Natoni
- Hematology, Department of Translational and Precision Medicine, Sapienza University, 00100 Rome, Italy; (M.C.); (R.F.)
| |
Collapse
|
5
|
Chakraborty A, Perez M, Carroll JD, Antonopoulos A, Dell A, Ortega L, Mohammed NBB, Wells M, Staudinger C, Griswold A, Chandler KB, Marrero C, Jimenez R, Tani Y, Wilmott JS, Thompson JF, Wang W, Sackstein R, Scolyer RA, Murphy GF, Haslam SM, Dimitroff CJ. Hypoxia Controls the Glycome Signature and Galectin-8-Ligand Axis to Promote Protumorigenic Properties of Metastatic Melanoma. J Invest Dermatol 2023; 143:456-469.e8. [PMID: 36174713 PMCID: PMC10123958 DOI: 10.1016/j.jid.2022.07.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 06/29/2022] [Accepted: 07/14/2022] [Indexed: 01/14/2023]
Abstract
The prognosis for patients with metastatic melanoma (MM) involving distant organs is grim, and treatment resistance is potentiated by tumor-initiating cells (TICs) that thrive under hypoxia. MM cells, including TICs, express a unique glycome featuring i-linear poly-N-acetyllactosamines through the loss of I-branching enzyme, β1,6 N-acetylglucosaminyltransferase 2. Whether hypoxia instructs MM TIC development by modulating the glycome signature remains unknown. In this study, we explored hypoxia-dependent alterations in MM glycome‒associated genes and found that β1,6 N-acetylglucosaminyltransferase 2 was downregulated and a galectin (Gal)-8-ligand axis, involving both extracellular and cell-intrinsic Gal-8, was induced. Low β1,6 N-acetylglucosaminyltransferase 2 levels correlated with poor patient outcomes, and patient serum samples were elevated for Gal-8. Depressed β1,6 N-acetylglucosaminyltransferase 2 in MM cells upregulated TIC marker, NGFR/CD271, whereas loss of MM cell‒intrinsic Gal-8 markedly lowered NGFR and reduced TIC activity in vivo. Extracellular Gal-8 bound preferentially to i-linear poly-N-acetyllactosamines on N-glycans of the TIC marker and prometastatic molecule CD44, among other receptors, and activated prosurvival factor protein kinase B. This study reveals the importance of hypoxia governing the MM glycome by enforcing i-linear poly-N-acetyllactosamine and Gal-8 expression. This mechanistic investigation also uncovers glycome-dependent regulation of pro-MM factor, NGFR, implicating i-linear poly-N-acetyllactosamine and Gal-8 as biomarkers and therapeutic targets of MM.
Collapse
Affiliation(s)
- Asmi Chakraborty
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Mariana Perez
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Jordan D Carroll
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | | | - Anne Dell
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Liettel Ortega
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Norhan B B Mohammed
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA; Department of Medical Biochemistry, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Michael Wells
- School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Caleb Staudinger
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Anthony Griswold
- John P. Hussman Institute for Human Genomics (HIHG), Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Kevin B Chandler
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Cristina Marrero
- Miami Cancer Institute, Baptist Health-South Florida, Miami, Florida, USA
| | - Ramon Jimenez
- Miami Cancer Institute, Baptist Health-South Florida, Miami, Florida, USA
| | - Yoshihiko Tani
- Japanese Red Cross Kinki Block Blood Center, Osaka, Japan
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - John F Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Wei Wang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachussetts, USA
| | - Robert Sackstein
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, Australia; Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - George F Murphy
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stuart M Haslam
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Charles J Dimitroff
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA.
| |
Collapse
|
6
|
Abstract
Both the cascade whereby a blood-borne cell enters a tissue and the anchoring of hematopoietic stem/progenitor cells (HSPCs) within bone marrow critically pivots on cell-cell interactions mediated by E-selectin binding to its canonical carbohydrate ligand, the tetrasaccharide termed "sialylated Lewis X" (sLeX). E-selectin, a member of the selectin class of adhesion molecules that is exclusively expressed by vascular endothelium, engages sLeX-bearing glycoconjugates that adorn mature leukocytes and HSPCs, as well as malignant cells, thereby permitting these cells to extravasate into various tissues. E-selectin expression is induced on microvascular endothelial cells within inflammatory loci at all tissues. However, conspicuously, E-selectin is constitutively expressed within microvessels in skin and marrow and, additionally, is inducibly expressed at these sites. Within the marrow, E-selectin receptor/ligand interactions promote lodgment of HSPCs and their malignant counterparts within hematopoietic growth-promoting microenvironments, collectively known as "vascular niches". Indeed, E-selectin receptor/ligand interactions have been reported to regulate both hematopoietic stem, and leukemic, cell proliferative dynamics. As such, signaling induced via engagement of E-selectin ligands is gaining interest as a critical mediator of homeostatic and malignant hematopoiesis, and this review will present current perspectives on the glycoconjugates mediating E-selectin receptor/ligand interactions and their currently defined role(s) in leukemogenesis.
Collapse
Affiliation(s)
- Evan Ales
- Department of Translational Medicine & The Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Robert Sackstein
- Department of Translational Medicine & The Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States.
| |
Collapse
|
7
|
Wisnovsky S, Bertozzi CR. Reading the glyco-code: New approaches to studying protein-carbohydrate interactions. Curr Opin Struct Biol 2022; 75:102395. [PMID: 35653954 PMCID: PMC9811956 DOI: 10.1016/j.sbi.2022.102395] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/06/2022] [Accepted: 04/16/2022] [Indexed: 01/07/2023]
Abstract
The surface of all living cells is decorated with carbohydrate molecules. Hundreds of functional proteins bind to these glycosylated ligands; such binding events subsequently modulate many aspects of protein and cell function. Identifying ligands for glycan-binding proteins (GBPs) is a defining challenge of glycoscience research. Here, we review recent advances that are allowing protein-carbohydrate interactions to be dissected with an unprecedented level of precision. We specifically highlight how cell-based glycan arrays and glyco-genomic profiling are being used to define the structural determinants of glycan-protein interactions in living cells. Going forward, these methods create exciting new opportunities for the study of glycans in physiology and disease.
Collapse
Affiliation(s)
- Simon Wisnovsky
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA; Howard Hughes Medical Institute, Stanford, CA, 94305, USA.
| |
Collapse
|
8
|
Liao C, Wang Q, An J, Chen J, Li X, Long Q, Xiao L, Guan X, Liu J. CD44 Glycosylation as a Therapeutic Target in Oncology. Front Oncol 2022; 12:883831. [PMID: 35936713 PMCID: PMC9351704 DOI: 10.3389/fonc.2022.883831] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/10/2022] [Indexed: 11/27/2022] Open
Abstract
The interaction of non-kinase transmembrane glycoprotein CD44 with ligands including hyaluronic acid (HA) is closely related to the occurrence and development of tumors. Changes in CD44 glycosylation can regulate its binding to HA, Siglec-15, fibronectin, TM4SF5, PRG4, FGF2, collagen and podoplanin and activate or inhibit c-Src/STAT3/Twist1/Bmi1, PI3K/AKT/mTOR, ERK/NF-κB/NANOG and other signaling pathways, thereby having a profound impact on the tumor microenvironment and tumor cell fate. However, the glycosylation of CD44 is complex and largely unknown, and the current understanding of how CD44 glycosylation affects tumors is limited. These issues must be addressed before targeted CD44 glycosylation can be applied to treat human cancers.
Collapse
Affiliation(s)
- Chengcheng Liao
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Qian Wang
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Life Sciences Institute, Zunyi Medical University, Zunyi, China
| | - Jiaxing An
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jie Chen
- Department of Urology, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaolan Li
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Life Sciences Institute, Zunyi Medical University, Zunyi, China
| | - Qian Long
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Linlin Xiao
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- *Correspondence: Linlin Xiao, ; Xiaoyan Guan, ; Jianguo Liu,
| | - Xiaoyan Guan
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- *Correspondence: Linlin Xiao, ; Xiaoyan Guan, ; Jianguo Liu,
| | - Jianguo Liu
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- *Correspondence: Linlin Xiao, ; Xiaoyan Guan, ; Jianguo Liu,
| |
Collapse
|
9
|
Kumar R, Pereira RS, Niemann J, Azimpour AI, Zanetti C, Karantanou C, Minka W, Minciacchi VR, Kowarz E, Meister M, Godavarthy PS, Maguer-Satta V, Lefort S, Wiercinska E, Bonig H, Marschalek R, Krause DS. The differential role of the lipid raft-associated protein flotillin 2 for progression of myeloid leukemia. Blood Adv 2022; 6:3611-3624. [PMID: 35298613 PMCID: PMC9631564 DOI: 10.1182/bloodadvances.2021005992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 03/08/2022] [Indexed: 11/20/2022] Open
Abstract
Lipid raft-associated proteins play a vital role in membrane-mediated processes. The lipid microdomain-associated protein flotillin 2 (FLOT2), which has a scaffolding function, is involved in polarization, as well as in actin cytoskeletal organization of primitive and mature hematopoietic cells and has been associated with different malignancies. However, its involvement in myeloid leukemias is not well studied. Using murine transplantation models, we show here that the absence of FLOT2 from leukemia-initiating cells (LICs) altered the disease course of BCR-ABL1+ chronic myeloid leukemia (CML), but not of MLL-AF9-driven acute myeloid leukemia (AML). While FLOT2 was required for expression of the adhesion molecule CD44 on both CML- and AML-LIC, a defect in the cytoskeleton, cell polarity, and impaired homing ability of LIC was only observed in FLOT2-deficient BCR-ABL1+ compared with MLL-AF9+ cells. Downstream of CD44, BCR-ABL1 kinase-independent discrepancies were observed regarding expression, localization, and activity of cell division control protein 42 homolog (CDC42) between wild-type (WT) and FLOT2-deficient human CML and AML cells. Inhibition of CDC42 by ML141 impaired the homing of CML LIC and, thereby, CML progression. This suggested that alteration of both CD44 and CDC42 may be causative of impaired CML progression in the absence of FLOT2. In summary, our data suggest a FLOT2-CD44-CDC42 axis, which differentially regulates CML vs AML progression, with deficiency of FLOT2 impairing the development of CML.
Collapse
Affiliation(s)
- Rahul Kumar
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Raquel S. Pereira
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Julian Niemann
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Alexander I. Azimpour
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Costanza Zanetti
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Christina Karantanou
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Wahyu Minka
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Valentina R. Minciacchi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Eric Kowarz
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| | - Melanie Meister
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Parimala S. Godavarthy
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | | | - Sylvain Lefort
- CRCL, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Lyon, France
| | - Eliza Wiercinska
- German Red Cross Blood Service Baden-Württemberg-Hessen, Institute Frankfurt, Frankfurt, Germany
| | - Halvard Bonig
- German Red Cross Blood Service Baden-Württemberg-Hessen, Institute Frankfurt, Frankfurt, Germany
- Goethe University, Institute for Transfusion Medicine and Immunohematology, Frankfurt, Germany
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| | - Daniela S. Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- German Red Cross Blood Service Baden-Württemberg-Hessen, Institute Frankfurt, Frankfurt, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Germany
- Frankfurt Cancer Institute, Frankfurt, Germany; and
- Institute for General Pharmacology and Toxicology, Institute for Biochemistry II, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
10
|
Refining the migration and engraftment of short-term and long-term HSCs by enhancing homing-specific adhesion mechanisms. Blood Adv 2022; 6:4373-4391. [PMID: 35764498 DOI: 10.1182/bloodadvances.2022007465] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 06/21/2022] [Indexed: 11/20/2022] Open
Abstract
In contrast to the short-term(ST)-CD34pos stem cells, studies have suggested that long-term (LT) hematopoietic stem cells (HSC) found in the CD34neg stem cell pool have trouble migrating and engrafting when introduced intravenously. We set out to fully elucidate the adhesion mechanisms used by ST/LT-HSCs to migrate to the bone marrow in order to understand these deficiencies. Focusing on murine ST-HSCs(Flk2negCD34pos) and LT-HSCs(Flk2negCD34neg), we observed a distinctive expression pattern of bone marrow homing effectors necessary for the first step, namely sialyl Lewis-X(sLex;ligand for E-selectin), and the second step, namely CXCR4 (receptor for SDF-1). sLex expression was higher on Flk2negCD34pos ST-HSCs(>60%) compared to Flk2negCD34neg LT-HSCs(<10%), which correlated to binding to E-selectin. Higher levels of CXCR4 were observed on Flk2negCD34pos ST-HSCs compared to Flk2negCD34neg LT-HSCs. Interestingly, expression of CD26, a peptidase known to deactivate chemokines (i.e.SDF-1), was higher on Flk2negCD34neg LT-HSCs. Given that migration is compromised in Flk2negCD34neg LT-HSCs, we aimed to enhance their ability to migrate using recombinant fucosyltransferase 6 (rhFTVI) and DiprotinA (CD26-inhibitor). We observed that although LT-HSCs expressed low levels of sLex, in vivo engraftment was not compromised. Moreover, although both treaments enhanced migration in vitro, only pre-treatment of LT-HSCs with DiprotinA enhanced engraftment in vivo. Remarkably, fucosylation of Flk2negCD34pos ST-HSCs consistently led to their ability to transplant secondary recipients, the gold standard for testing functionality of LT-HSCs. These data suggest that treatments to overcome the molecular disparity in adhesion mechanisms among ST-HSCs and LT-HSCs, differentially influences their abilities to migrate and engraft in vivo and boosts ST-HSCs engraftment in vivo.
Collapse
|
11
|
Lung-Derived Selectins Enhance Metastatic Behavior of Triple Negative Breast Cancer Cells. Biomedicines 2021; 9:biomedicines9111580. [PMID: 34829810 PMCID: PMC8615792 DOI: 10.3390/biomedicines9111580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/24/2021] [Accepted: 10/28/2021] [Indexed: 12/26/2022] Open
Abstract
The lung is one of the deadliest sites of breast cancer metastasis, particularly for triple negative breast cancer (TNBC). We have previously shown that the lung produces several soluble factors that may enhance the metastatic behavior of TNBC, including E-, L-, and P-selectin. In this paper, we hypothesize that lung-derived selectins promote TNBC metastatic behavior and may serve as a potential therapeutic target. Lungs were isolated from mice and used to generate lung-conditioned media (CM). Lung-derived selectins were immunodepleted and TNBC migration and proliferation were assessed in response to native or selectin-depleted lung-CM. A 3D ex vivo pulmonary metastasis assay (PuMA) was used to assess the metastatic progression of TNBC in the lungs of wild-type versus triple-selectin (ELP-/-) knockout mice. We observed that individual lung-derived selectins enhance in vitro migration (p ≤ 0.05), but not the proliferation of TNBC cells, and that ex vivo metastatic progression is reduced in the lungs of ELP-/- mice compared to wild-type mice (p ≤ 0.05). Treatment with the pan-selectin inhibitor bimosiamose reduced in vitro lung-specific TNBC migration and proliferation (p ≤ 0.05). Taken together, these results suggest that lung-derived selectins may present a potential therapeutic target against TNBC metastasis. Future studies are aimed at elucidating the pro-metastatic mechanisms of lung-derived selectins and developing a lung-directed therapeutic approach.
Collapse
|
12
|
NK Cell-Mediated Eradication of Ovarian Cancer Cells with a Novel Chimeric Antigen Receptor Directed against CD44. Biomedicines 2021; 9:biomedicines9101339. [PMID: 34680456 PMCID: PMC8533227 DOI: 10.3390/biomedicines9101339] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/09/2022] Open
Abstract
Ovarian cancer is the most common cause of gynecological cancer-related death in the developed world. Disease recurrence and chemoresistance are major causes of poor survival rates in ovarian cancer patients. Ovarian cancer stem cells (CSCs) were shown to represent a source of tumor recurrence owing to the high resistance to chemotherapy and enhanced tumorigenicity. Chimeric antigen receptor (CAR)-based adoptive immunotherapy represents a promising strategy to reduce the risk for recurrent disease. In this study, we developed a codon-optimized third-generation CAR to specifically target CD44, a marker widely expressed on ovarian cancer cells and associated with CSC-like properties and intraperitoneal tumor spread. We equipped NK-92 cells with the anti-CD44 CAR (CD44NK) and an anti-CD19 control CAR (CD19NK) using lentiviral SIN vectors. Compared to CD19NK and untransduced NK-92 cells, CD44NK showed potent and specific cytotoxic activity against CD44-positive ovarian cancer cell lines (SKOV3 and OVCAR3) and primary ovarian cancer cells harvested from ascites. In contrast, CD44NK had less cytotoxic activity against CD44-negative A2780 cells. Specific activation of engineered NK cells was also demonstrated by interferon-γ (IFNγ) secretion assays. Furthermore, CD44NK cells still demonstrated cytotoxic activity under cisplatin treatment. Most importantly, the simultaneous treatment with CD44NK and cisplatin showed higher anti-tumor activity than sequential treatment.
Collapse
|
13
|
Szydlak R. Biological, chemical and mechanical factors regulating migration and homing of mesenchymal stem cells. World J Stem Cells 2021; 13:619-631. [PMID: 34249231 PMCID: PMC8246245 DOI: 10.4252/wjsc.v13.i6.619] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/03/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a population of primary and non-specialized cells, which can be isolated from various tissues. Currently, MSCs are key players in cellular therapy and regenerative medicine. However, the possibility of using MSCs in the treatment of many diseases needs to be preceded, though, by in-depth analysis of their properties, especially by determining the mechanism of tissue homing as well as the mechanism, due to which cells contribute to tissue regeneration. This review is intended to present information on recent findings regarding the mechanism of recruitment and tissue homing by MSCs and discuss current hypotheses for how MSCs can reach target tissues.
Collapse
Affiliation(s)
- Renata Szydlak
- Department of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kraków 31-034, Poland
| |
Collapse
|
14
|
Increased B4GALT1 expression is associated with platelet surface galactosylation and thrombopoietin plasma levels in MPNs. Blood 2021; 137:2085-2089. [PMID: 33238000 DOI: 10.1182/blood.2020007265] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 11/09/2020] [Indexed: 12/31/2022] Open
Abstract
Aberrant megakaryopoiesis is a hallmark of the myeloproliferative neoplasms (MPNs), a group of clonal hematological malignancies originating from hematopoietic stem cells, leading to an increase in mature blood cells in the peripheral blood. Sialylated derivatives of the glycan structure β4-N-acetyllactosamine (Galβ1,4GlcNAc or type-2 LacNAc, hereafter referred to as LacNAc) regulate platelet life span, hepatic thrombopoietin (TPO) production, and thrombopoiesis. We found increased TPO plasma levels in MPNs with high allele burden of the mutated clones. Remarkably, platelets isolated from MPNs had a significant increase in LacNAc expression that correlated with the high allele burden regardless of the underlying identified mutation. Megakaryocytes derived in vitro from these patients showed an increased expression of the B4GALT1 gene encoding β-1,4-galactosyltransferase 1 (β4GalT1). Consistently, megakaryocytes from MPN showed increased LacNAc expression relative to healthy controls, which was counteracted by the treatment with a Janus kinase 1/2 inhibitor. Altered expression of B4GALT1 in mutant megakaryocytes can lead to the production of platelets with aberrant galactosylation, which in turn promote hepatic TPO synthesis regardless of platelet mass. Our findings provide a new paradigm for understanding aberrant megakaryopoiesis in MPNs and identify β4GalT1 as a potential actionable target for therapy.
Collapse
|
15
|
Smith BAH, Bertozzi CR. The clinical impact of glycobiology: targeting selectins, Siglecs and mammalian glycans. Nat Rev Drug Discov 2021; 20:217-243. [PMID: 33462432 PMCID: PMC7812346 DOI: 10.1038/s41573-020-00093-1] [Citation(s) in RCA: 240] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 01/31/2023]
Abstract
Carbohydrates - namely glycans - decorate every cell in the human body and most secreted proteins. Advances in genomics, glycoproteomics and tools from chemical biology have made glycobiology more tractable and understandable. Dysregulated glycosylation plays a major role in disease processes from immune evasion to cognition, sparking research that aims to target glycans for therapeutic benefit. The field is now poised for a boom in drug development. As a harbinger of this activity, glycobiology has already produced several drugs that have improved human health or are currently being translated to the clinic. Focusing on three areas - selectins, Siglecs and glycan-targeted antibodies - this Review aims to tell the stories behind therapies inspired by glycans and to outline how the lessons learned from these approaches are paving the way for future glycobiology-focused therapeutics.
Collapse
Affiliation(s)
- Benjamin A H Smith
- Department of Chemical & Systems Biology and ChEM-H, Stanford School of Medicine, Stanford, CA, USA
| | - Carolyn R Bertozzi
- Department of Chemical & Systems Biology and ChEM-H, Stanford School of Medicine, Stanford, CA, USA.
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
16
|
Kulkarni R, Kale V. Physiological Cues Involved in the Regulation of Adhesion Mechanisms in Hematopoietic Stem Cell Fate Decision. Front Cell Dev Biol 2020; 8:611. [PMID: 32754597 PMCID: PMC7366553 DOI: 10.3389/fcell.2020.00611] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/19/2020] [Indexed: 12/16/2022] Open
Abstract
Hematopoietic stem cells (HSC) could have several fates in the body; viz. self-renewal, differentiation, migration, quiescence, and apoptosis. These fate decisions play a crucial role in maintaining homeostasis and critically depend on the interaction of the HSCs with their micro-environmental constituents. However, the physiological cues promoting these interactions in vivo have not been identified to a great extent. Intense research using various in vitro and in vivo models is going on in various laboratories to understand the mechanisms involved in these interactions, as understanding of these mechanistic would greatly help in improving clinical transplantations. However, though these elegant studies have identified the molecular interactions involved in the process, harnessing these interactions to the recipients' benefit would ultimately depend on manipulation of environmental cues initiating them in vivo: hence, these need to be identified at the earliest. HSCs reside in the bone marrow, which is a very complex tissue comprising of various types of stromal cells along with their secreted cytokines, extra-cellular matrix (ECM) molecules and extra-cellular vesicles (EVs). These components control the HSC fate decision through direct cell-cell interactions - mediated via various types of adhesion molecules -, cell-ECM interactions - mediated mostly via integrins -, or through soluble mediators like cytokines and EVs. This could be a very dynamic process involving multiple transient interactions acting concurrently or sequentially, and the adhesion molecules involved in various fate determining situations could be different. If the switch mechanisms governing these dynamic states in vivo are identified, they could be harnessed for the development of novel therapeutics. Here, in addition to reviewing the adhesion molecules involved in the regulation of HSCs, we also touch upon recent advances in our understanding of the physiological cues known to initiate specific adhesive interactions of HSCs with the marrow stromal cells or ECM molecules and EVs secreted by them.
Collapse
Affiliation(s)
- Rohan Kulkarni
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell Research, Symbiosis International University, Pune, India
| |
Collapse
|
17
|
Villatoro A, Konieczny J, Cuminetti V, Arranz L. Leukemia Stem Cell Release From the Stem Cell Niche to Treat Acute Myeloid Leukemia. Front Cell Dev Biol 2020; 8:607. [PMID: 32754595 PMCID: PMC7367216 DOI: 10.3389/fcell.2020.00607] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/19/2020] [Indexed: 01/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous, complex, and deadly disease, whose treatment has hardly evolved for decades and grounds on the use of intensive chemotherapy regimens. Chemotherapy helps reduce AML bulk, but promotes relapse in the long-run by selection of chemoresistant leukemia stem cells (LSC). These may diversify and result in progression to more aggressive forms of AML. In vivo models suggest that the bone marrow stem cell niche helps LSC stay dormant and protected from chemotherapy. Here, we summarize relevant changes in stem cell niche homing and adhesion of AML LSC vs. healthy hematopoietic stem cells, and provide an overview of clinical trials aiming at targeting these processes for AML treatment and future directions within this field. Promising results with various non-mutation-targeted novel therapies directed to LSC eradication via interference with their anchoring to the stem cell niche have encouraged on-going or future advanced phase III clinical trials. In the coming years, we may see a shift in the focus of AML treatment to LSC-directed therapies if the prospect of improved cure rates holds true. In the future, AML treatment should lean toward personalized therapies using combinations of these compounds plus mutation-targeted agents and/or targeted delivery of chemotherapy, aiming at LSC eradication with reduced side effects.
Collapse
Affiliation(s)
- Alicia Villatoro
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Joanna Konieczny
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Vincent Cuminetti
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Lorena Arranz
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway.,Norwegian Center for Molecular Medicine (NCMM), University of Oslo, Oslo, Norway
| |
Collapse
|
18
|
Tvaroška I, Selvaraj C, Koča J. Selectins-The Two Dr. Jekyll and Mr. Hyde Faces of Adhesion Molecules-A Review. Molecules 2020; 25:molecules25122835. [PMID: 32575485 PMCID: PMC7355470 DOI: 10.3390/molecules25122835] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/27/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
Selectins belong to a group of adhesion molecules that fulfill an essential role in immune and inflammatory responses and tissue healing. Selectins are glycoproteins that decode the information carried by glycan structures, and non-covalent interactions of selectins with these glycan structures mediate biological processes. The sialylated and fucosylated tetrasaccharide sLex is an essential glycan recognized by selectins. Several glycosyltransferases are responsible for the biosynthesis of the sLex tetrasaccharide. Selectins are involved in a sequence of interactions of circulated leukocytes with endothelial cells in the blood called the adhesion cascade. Recently, it has become evident that cancer cells utilize a similar adhesion cascade to promote metastases. However, like Dr. Jekyll and Mr. Hyde’s two faces, selectins also contribute to tissue destruction during some infections and inflammatory diseases. The most prominent function of selectins is associated with the initial stage of the leukocyte adhesion cascade, in which selectin binding enables tethering and rolling. The first adhesive event occurs through specific non-covalent interactions between selectins and their ligands, with glycans functioning as an interface between leukocytes or cancer cells and the endothelium. Targeting these interactions remains a principal strategy aimed at developing new therapies for the treatment of immune and inflammatory disorders and cancer. In this review, we will survey the significant contributions to and the current status of the understanding of the structure of selectins and the role of selectins in various biological processes. The potential of selectins and their ligands as therapeutic targets in chronic and acute inflammatory diseases and cancer will also be discussed. We will emphasize the structural characteristic of selectins and the catalytic mechanisms of glycosyltransferases involved in the biosynthesis of glycan recognition determinants. Furthermore, recent achievements in the synthesis of selectin inhibitors will be reviewed with a focus on the various strategies used for the development of glycosyltransferase inhibitors, including substrate analog inhibitors and transition state analog inhibitors, which are based on knowledge of the catalytic mechanism.
Collapse
Affiliation(s)
- Igor Tvaroška
- Central European Institute of Technology (CEITEC), Masaryk University, 62500 Brno, Czech Republic
- Institute of Chemistry, Slovak Academy of Sciences, 84538 Bratislava, Slovak Republic
- Correspondence: (I.T.); (J.K.); Tel.: +421-948-535-601 (I.T.); +420-731-682-606 (J.K.)
| | - Chandrabose Selvaraj
- Central European Institute of Technology (CEITEC), Masaryk University, 62500 Brno, Czech Republic
| | - Jaroslav Koča
- Central European Institute of Technology (CEITEC), Masaryk University, 62500 Brno, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic
- Correspondence: (I.T.); (J.K.); Tel.: +421-948-535-601 (I.T.); +420-731-682-606 (J.K.)
| |
Collapse
|
19
|
Birmingham KG, Robinson IE, Edwards EE, Thomas SN. Photoconversion and chromatographic microfluidic system reveals differential cellular phenotypes of adhesion velocity versus persistence in shear flow. LAB ON A CHIP 2020; 20:806-822. [PMID: 31971187 PMCID: PMC7299069 DOI: 10.1039/c9lc00923j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
An integrated photoconversion and cell sorting parallel-plate chromatography channel enabling the measurement of instantaneous and average velocities of cells mediating adhesion in flow fields was engineered to study the mechanisms underlying adhesion to selectins by metastatic cancer cells. Through the facile enrichment of cells into subfractions of differing adhesive behaviors and a fluorescent velocity probe amenable to off-chip analysis, underlying, causal molecular profiles implicated in differing adhesive phenotypes of metastatic cancer cells could be interrogated. This analytical method revealed selectin-mediated rolling adhesion to be strongly associated with expression of selectin ligands, correlations that vary with ligand type and rolling velocity magnitude. Discrete selectin ligand expression profiles were also found to underlie persistent versus non-persistent adhesion on selectins, suggestive of divergent regulatory mechanisms. This integrated cell sorting and photoconversion microfluidic platform thus enables in vitro analysis and comparisons of adhesive phenotypes as they relate to mechanisms of cancer cell metastasis in the context of selectin mediated adhesion, revealing new insights into potential cancer dissemination pathways.
Collapse
Affiliation(s)
- Katherine G Birmingham
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia. and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| | - Isaac E Robinson
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia. and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| | - Erin E Edwards
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia and Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Susan N Thomas
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia. and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia and Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia and Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
20
|
β4GALT1 controls β1 integrin function to govern thrombopoiesis and hematopoietic stem cell homeostasis. Nat Commun 2020; 11:356. [PMID: 31953383 PMCID: PMC6968998 DOI: 10.1038/s41467-019-14178-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 12/13/2019] [Indexed: 12/26/2022] Open
Abstract
Glycosylation is critical to megakaryocyte (MK) and thrombopoiesis in the context of gene mutations that affect sialylation and galactosylation. Here, we identify the conserved B4galt1 gene as a critical regulator of thrombopoiesis in MKs. β4GalT1 deficiency increases the number of fully differentiated MKs. However, the resulting lack of glycosylation enhances β1 integrin signaling leading to dysplastic MKs with severely impaired demarcation system formation and thrombopoiesis. Platelets lacking β4GalT1 adhere avidly to β1 integrin ligands laminin, fibronectin, and collagen, while other platelet functions are normal. Impaired thrombopoiesis leads to increased plasma thrombopoietin (TPO) levels and perturbed hematopoietic stem cells (HSCs). Remarkably, β1 integrin deletion, specifically in MKs, restores thrombopoiesis. TPO and CXCL12 regulate β4GalT1 in the MK lineage. Thus, our findings establish a non-redundant role for β4GalT1 in the regulation of β1 integrin function and signaling during thrombopoiesis. Defective thrombopoiesis and lack of β4GalT1 further affect HSC homeostasis.
Collapse
|
21
|
Mereiter S, Martins ÁM, Gomes C, Balmaña M, Macedo JA, Polom K, Roviello F, Magalhães A, Reis CA. O‐glycan truncation enhances cancer‐related functions of
CD
44 in gastric cancer. FEBS Lett 2019; 593:1675-1689. [DOI: 10.1002/1873-3468.13432] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/04/2019] [Accepted: 05/06/2019] [Indexed: 01/16/2023]
Affiliation(s)
- Stefan Mereiter
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Álvaro M. Martins
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Catarina Gomes
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Meritxell Balmaña
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Joana A. Macedo
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Karol Polom
- Department of Surgical Oncology Medical University of Gdansk Poland
- General Surgery and Surgical Oncology Department University of Siena Italy
| | - Franco Roviello
- General Surgery and Surgical Oncology Department University of Siena Italy
| | - Ana Magalhães
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Celso A. Reis
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
- Faculty of Medicine University of Porto Portugal
- Instituto de Ciências Biomédicas Abel Salazar University of Porto Portugal
| |
Collapse
|
22
|
Weil BR, Neelamegham S. Selectins and Immune Cells in Acute Myocardial Infarction and Post-infarction Ventricular Remodeling: Pathophysiology and Novel Treatments. Front Immunol 2019; 10:300. [PMID: 30873166 PMCID: PMC6400985 DOI: 10.3389/fimmu.2019.00300] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/05/2019] [Indexed: 12/21/2022] Open
Abstract
The glycosciences aim to understand the impact of extracellular and intracellular carbohydrate structures on biological function. These glycans primarily fall into three major groups: lipid-linked carbohydrates that are referred to as glycosphingolipids or simply glycolipids; relatively short carbohydrate chains that are often O- or N-linked to proteins yielding common glycoproteins; and extended linear polymeric carbohydrate structures that are referred to as glycosaminoglycans (GAGs). Whereas, the impact of such carbohydrate structures has been extensively examined in cancer biology, their role in acute and chronic heart disease is less studied. In this context, a growing body of evidence indicates that glycans play an important role in immune mediated cell recruitment to damaged heart tissue to initiate wound healing and repair after injury. This is particularly important following ischemia and reperfusion that occurs in the heart in the setting of acute myocardial infarction. Here, immune system-mediated repair of the damaged myocardium plays a critical role in determining post-infarction ventricular remodeling, cardiac function, and patient outcome. Further, alterations in immune cell activity can promote the development of heart failure. The present review summarizes our current understanding of the phases of immune-mediated repair following myocardial infarction. It discusses what is known regarding glycans in mediating the recruitment of circulating immune cells during the early inflammatory stage of post-infarction repair, with focus on the selectin family of adhesion molecules. It offers future directions for research aimed at utilizing our knowledge of mechanisms underlying immune cell recruitment to either modulate leukocyte recruitment to the injured tissue or enhance the targeted delivery of biologic therapeutics such as stem cells in an attempt to promote repair of the damaged heart.
Collapse
Affiliation(s)
- Brian R Weil
- Department of Physiology and Biophysics, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Sriram Neelamegham
- Department of Medicine, University at Buffalo, State University of New York, Buffalo, NY, United States.,Department of Chemical & Biological Engineering, University at Buffalo, State University of New York, Buffalo, NY, United States
| |
Collapse
|
23
|
Gruszka AM, Valli D, Restelli C, Alcalay M. Adhesion Deregulation in Acute Myeloid Leukaemia. Cells 2019; 8:E66. [PMID: 30658474 PMCID: PMC6356639 DOI: 10.3390/cells8010066] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 12/11/2022] Open
Abstract
Cell adhesion is a process through which cells interact with and attach to neighboring cells or matrix using specialized surface cell adhesion molecules (AMs). Adhesion plays an important role in normal haematopoiesis and in acute myeloid leukaemia (AML). AML blasts express many of the AMs identified on normal haematopoietic precursors. Differential expression of AMs between normal haematopoietic cells and leukaemic blasts has been documented to a variable extent, likely reflecting the heterogeneity of the disease. AMs govern a variety of processes within the bone marrow (BM), such as migration, homing, and quiescence. AML blasts home to the BM, as the AM-mediated interaction with the niche protects them from chemotherapeutic agents. On the contrary, they detach from the niches and move from the BM into the peripheral blood to colonize other sites, i.e., the spleen and liver, possibly in a process that is reminiscent of epithelial-to-mesenchymal-transition in metastatic solid cancers. The expression of AMs has a prognostic impact and there are ongoing efforts to therapeutically target adhesion in the fight against leukaemia.
Collapse
Affiliation(s)
- Alicja M Gruszka
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
| | - Debora Valli
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
| | - Cecilia Restelli
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
| | - Myriam Alcalay
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20 122 Milan, Italy.
| |
Collapse
|
24
|
Sackstein R. The First Step in Adoptive Cell Immunotherapeutics: Assuring Cell Delivery via Glycoengineering. Front Immunol 2019; 9:3084. [PMID: 30687313 PMCID: PMC6336727 DOI: 10.3389/fimmu.2018.03084] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 12/13/2018] [Indexed: 12/26/2022] Open
Abstract
Despite decades of intensive attention directed to creation of genetically altered cells (e.g., as in development of chimeric antigen receptor (CAR) T-cells) and/or to achieve requisite in vitro accumulation of desired immunologic effectors (e.g., elaboration of virus-specific T cells, expansion of NK cells, differentiation of dendritic cells, isolation, and propagation of Tregs, etc.), there has been essentially no interest in the most fundamental of all hurdles: assuring tissue-specific delivery of administered therapeutic cells to sites where they are needed. With regards to use of CAR T-cells, the absence of information on the efficacy of cell delivery is striking, especially in light of the clear association between administered cell dose and adverse events, and the obvious fact that pertinent cell acquisition/expansion costs would be dramatically curtailed with more efficient delivery of the administered cell bolus. Herein, based on information garnered from studies of human leukocytes and adult stem cells, the logic underlying the use of cell surface glycoengineering to enforce E-selectin ligand expression will be conveyed in the context of how this approach offers strategies to enhance delivery of CAR T-cells to marrow and to tumor beds. This application of glycoscience principles and techniques with intention to optimize cell therapeutics is a prime example of the emerging field of “translational glycobiology.”
Collapse
Affiliation(s)
- Robert Sackstein
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| |
Collapse
|
25
|
Pang X, Li H, Guan F, Li X. Multiple Roles of Glycans in Hematological Malignancies. Front Oncol 2018; 8:364. [PMID: 30237983 PMCID: PMC6135871 DOI: 10.3389/fonc.2018.00364] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/17/2018] [Indexed: 01/05/2023] Open
Abstract
The three types of blood cells (red blood cells for carrying oxygen, white blood cells for immune protection, and platelets for wound clotting) arise from hematopoietic stem/progenitor cells in the adult bone marrow, and function in physiological regulation and communication with local microenvironments to maintain systemic homeostasis. Hematological malignancies are relatively uncommon malignant disorders derived from the two major blood cell lineages: myeloid (leukemia) and lymphoid (lymphoma). Malignant clones lose their regulatory mechanisms, resulting in production of a large number of dysfunctional cells and destruction of normal hematopoiesis. Glycans are one of the four major types of essential biological macromolecules, along with nucleic acids, proteins, and lipids. Major glycan subgroups are N-glycans, O-glycans, glycosaminoglycans, and glycosphingolipids. Aberrant expression of glycan structures, resulting from dysregulation of glycan-related genes, is associated with cancer development and progression in terms of cell signaling and communication, tumor cell dissociation and invasion, cell-matrix interactions, tumor angiogenesis, immune modulation, and metastasis formation. Aberrant glycan expression occurs in most hematological malignancies, notably acute myeloid leukemia, myeloproliferative neoplasms, and multiple myeloma, etc. Here, we review recent research advances regarding aberrant glycans, their related genes, and their roles in hematological malignancies. Our improved understanding of the mechanisms that underlie aberrant patterns of glycosylation will lead to development of novel, more effective therapeutic approaches targeted to hematological malignancies.
Collapse
Affiliation(s)
- Xingchen Pang
- School of Biotechnology, Jiangnan University, Wuxi, China
| | - Hongjiao Li
- College of Life Science, Northwest University, Xi'an, China
| | - Feng Guan
- School of Biotechnology, Jiangnan University, Wuxi, China.,College of Life Science, Northwest University, Xi'an, China
| | - Xiang Li
- College of Life Science, Northwest University, Xi'an, China.,Wuxi Medical School, Jiangnan University, Wuxi, China
| |
Collapse
|
26
|
Videira PA, Silva M, Martin KC, Sackstein R. Ligation of the CD44 Glycoform HCELL on Culture-Expanded Human Monocyte-Derived Dendritic Cells Programs Transendothelial Migration. THE JOURNAL OF IMMUNOLOGY 2018; 201:1030-1043. [PMID: 29941663 DOI: 10.4049/jimmunol.1800188] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/23/2018] [Indexed: 12/12/2022]
Abstract
The success of dendritic cell (DC)-based immunotherapeutics critically hinges on the capacity of the vascularly administered cells to enter tissues. Transendothelial migration (TEM) is dictated by an ordered cascade of receptor/ligand interactions. In this study, we examined the key molecular effectors of TEM of human monocyte-derived DCs (mo-DCs) generated by clinically relevant methods: CD14 selection (CD14-S) and plastic adherence selection (PA-S). Without chemokine input, CD14-S cells undergo greater TEM than PA-S cells over TNF-α-stimulated HUVECs. TEM of CD14-S mo-DCs is E-selectin/very late Ag-4 (VLA-4) dependent, and engagement of E-selectin ligands activates VLA-4 on CD14-S mo-DCs but not on PA-S mo-DCs. E-selectin binding glycoforms of P-selectin glycoprotein ligand-1 (PSGL-1) (i.e., cutaneous lymphocyte Ag [CLA]) and CD44 (i.e., hematopoietic cell E-selectin/L-selectin ligand [HCELL]) are both expressed on CD14-S mo-DCs, but only CLA is expressed on PA-S mo-DCs. To elucidate the effect of CD44 or PSGL-1 engagement, mo-DCs were pretreated with their ligands. Ligation of CD44 on CD14-S mo-DCs triggers VLA-4 activation and TEM, whereas PSGL-1 ligation does not. HCELL expression on CD14-S mo-DC can be enforced by cell surface exofucosylation, yielding increased TEM in vitro and enhanced extravasation into bone marrow in vivo. These findings highlight structural and functional pleiotropism of CD44 in priming TEM of mo-DCs and suggest that strategies to enforce HCELL expression may boost TEM of systemically administered CD14-S mo-DCs.
Collapse
Affiliation(s)
- Paula A Videira
- Unidade de Ciências Biomoleculares Aplicadas, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal.,Centro de Estudos de Doenças Crónicas, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-190 Lisbon, Portugal.,Congenital Disorders of Glycosylation and Allies-Professionals and Patient Associations International Network, 2829-516 Caparica, Portugal
| | - Mariana Silva
- Centro de Estudos de Doenças Crónicas, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-190 Lisbon, Portugal.,Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115.,Program of Excellence in Glycosciences, Harvard Medical School, Boston, MA 02115; and
| | - Kyle C Martin
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115.,Program of Excellence in Glycosciences, Harvard Medical School, Boston, MA 02115; and
| | - Robert Sackstein
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115; .,Program of Excellence in Glycosciences, Harvard Medical School, Boston, MA 02115; and.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
27
|
Pearce OMT. Cancer glycan epitopes: biosynthesis, structure and function. Glycobiology 2018; 28:670-696. [DOI: 10.1093/glycob/cwy023] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/09/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- Oliver M T Pearce
- Centre for Cancer & Inflammation, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
| |
Collapse
|
28
|
Nesterova ES, Kravchenko SK, Mangasarova YK, Plastinina LV, Dvirnyk VN, Kovrigina AM, Shchupletsova IA, Obukhova TN, Gemdzhian EG, Vorobyev IA, Vorobyev AI. [Leukemization of follicular lymphoma: The features of diagnostic and clinical course of a rare form of the disease]. TERAPEVT ARKH 2017; 89:45-50. [PMID: 28766540 DOI: 10.17116/terarkh201789745-50] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
AIM To characterize a group of patients with follicular lymphoma (FL) with leukemization and to evaluate the efficiency of different therapy options (R-CHOP/R-FMC/high-dose chemotherapy (HDCT)). SUBJECTS AND METHODS 18 (7.2%) out of 250 patients diagnosed with FL, who were examined and treated at the National Research Center for Hematology, Ministry of Health of the Russian Federation, were found to have leukemic FL (tumor cells in the peripheral blood smears were detected by cytology and flow cytofluorometry. Eight of the 18 patients had extranodal foci of involvement: lung, stomach, spleen, lumbar muscles, upper jaw, and vertebrae. Bone marrow was involved in 17 of the 18 patients. Tumor biopsy specimens displayed a morphological pattern of indolent FL in the majority of patients (10 of the 18 patients had cytological grade 1-2 tumors and 14 patients had a nodular or nodular-diffuse tumor growth pattern). The patients underwent R-CHOP/R-FMC) or HDCT cycles as first-line therapy, followed by autologous stem cell transplantation (auto-SCT). RESULTS The median follow-up was 66 months (range 12-217 months). The 5-year overall survival (OS) and progression-free survival (PFS) rates were 70% (10% SEM) and 35% (15% SEM), respectively. The median OS was not reached; the median PFS was 3 years. CONCLUSION Leukemic FL is characterized by low OS and PFS rates. The most effective chemotherapy regimens were R-CHOP, followed by HDCT and auto-SCT in first remission or R-FMC. These cycles can to a greater extent achieve a complete eradication of the bone marrow tumor clone. Due to the relapsing course of FL and the aggressiveness of leukemic FL, it is expedient to carry out auto-SCT in first remission.
Collapse
Affiliation(s)
- E S Nesterova
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - S K Kravchenko
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - Ya K Mangasarova
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - L V Plastinina
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - V N Dvirnyk
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - A M Kovrigina
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - I A Shchupletsova
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - T N Obukhova
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - E G Gemdzhian
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - I A Vorobyev
- M.V. Lomonosov Moscow State University, Moscow, Russia
| | - A I Vorobyev
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
29
|
Zhao Y, Zhang H. Update on the mechanisms of homing of adipose tissue-derived stem cells. Cytotherapy 2017; 18:816-27. [PMID: 27260205 DOI: 10.1016/j.jcyt.2016.04.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 03/11/2016] [Accepted: 04/25/2016] [Indexed: 02/06/2023]
Abstract
Adipose tissue-derived stem cells (ADSCs), which resemble bone marrow mesenchymal stromal cells (BMSCs), have shown great advantages and promise in the field of regenerative medicine. They can be readily harvested in large numbers with low donor-site morbidity. To date, a great number of preclinical and clinical studies have shown ADSCs' safety and efficacy in regenerative medicine. However, a better understanding of the mechanisms of homing of ADSCs is needed to advance the clinical utility of this therapy. In this review, the reports of the homing of ADSCs were searched using Pubmed and Google Scholar to update our knowledge. ADSCs were proved to interact with endothelial cells by expressing the similar integrins with BMSCs. In addition, ADSCs do not possess the dominant ligand for P-selectin, just like BMSCs. Stromal derived factor-1 (SDF-1)/CXCR4 and CXC ligand-5 (CXCL5)/CXCR2 interactions are the two main axes governing ADSCs extravasation from bone marrow vessels. Some more signaling pathways involved in migration of ADSCs have been investigated, including LPA/LPA1 signaling pathway, MAPK/Erk1/2 signaling pathway, RhoA/Rock signaling pathway and PDGF-BB/PDGFR-β signaling pathway. Status quo of a lack of intensive studies on the details of homing of ADSCs should be improved in the near future before clinical application.
Collapse
Affiliation(s)
- Yong Zhao
- Minimally Invasive Urology Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Haiyang Zhang
- Minimally Invasive Urology Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China; Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|
30
|
Silva M, Fung RKF, Donnelly CB, Videira PA, Sackstein R. Cell-Specific Variation in E-Selectin Ligand Expression among Human Peripheral Blood Mononuclear Cells: Implications for Immunosurveillance and Pathobiology. THE JOURNAL OF IMMUNOLOGY 2017; 198:3576-3587. [PMID: 28330896 DOI: 10.4049/jimmunol.1601636] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 02/22/2017] [Indexed: 12/26/2022]
Abstract
Both host defense and immunopathology are shaped by the ordered recruitment of circulating leukocytes to affected sites, a process initiated by binding of blood-borne cells to E-selectin displayed at target endothelial beds. Accordingly, knowledge of the expression and function of leukocyte E-selectin ligands is key to understanding the tempo and specificity of immunoreactivity. In this study, we performed E-selectin adherence assays under hemodynamic flow conditions coupled with flow cytometry and Western blot analysis to elucidate the function and structural biology of glycoprotein E-selectin ligands expressed on human PBMCs. Circulating monocytes uniformly express high levels of the canonical E-selectin binding determinant sialyl Lewis X (sLeX) and display markedly greater adhesive interactions with E-selectin than do circulating lymphocytes, which exhibit variable E-selectin binding among CD4+ and CD8+ T cells but no binding by B cells. Monocytes prominently present sLeX decorations on an array of protein scaffolds, including P-selectin glycoprotein ligand-1, CD43, and CD44 (rendering the E-selectin ligands cutaneous lymphocyte Ag, CD43E, and hematopoietic cell E-selectin/L-selectin ligand, respectively), and B cells altogether lack E-selectin ligands. Quantitative PCR gene expression studies of glycosyltransferases that regulate display of sLeX reveal high transcript levels among circulating monocytes and low levels among circulating B cells, and, commensurately, cell surface α(1,3)-fucosylation reveals that acceptor sialyllactosaminyl glycans convertible into sLeX are abundantly expressed on human monocytes yet are relatively deficient on B cells. Collectively, these findings unveil distinct cell-specific patterns of E-selectin ligand expression among human PBMCs, indicating that circulating monocytes are specialized to engage E-selectin and providing key insights into the molecular effectors mediating recruitment of these cells at inflammatory sites.
Collapse
Affiliation(s)
- Mariana Silva
- Centro de Estudos de Doenças Crónicas, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal.,Department of Dermatology, Brigham and Women's Hospital, Boston, MA 02115.,Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115.,Program of Excellence in Glycosciences, Harvard Medical School, Boston, MA 02115
| | - Ronald Kam Fai Fung
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA 02115.,Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115.,Medical Training and Administration Unit, Royal Prince Alfred Hospital, Sydney, New South Wales 2050, Australia; and
| | - Conor Brian Donnelly
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA 02115.,Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115.,Program of Excellence in Glycosciences, Harvard Medical School, Boston, MA 02115
| | - Paula Alexandra Videira
- Centro de Estudos de Doenças Crónicas, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal.,Unidade de Ciências Biomoleculares Aplicadas, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Robert Sackstein
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA 02115; .,Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115.,Program of Excellence in Glycosciences, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
31
|
Robinson SN, Thomas MW, Simmons PJ, Lu J, Yang H, Javni JA, Shpall EJ, Zweidler-Mckay PA. Non-fucosylated CB CD34 + cells represent a good target for enforced fucosylation to improve engraftment following cord blood transplantation. Cytotherapy 2017; 19:285-292. [DOI: 10.1016/j.jcyt.2016.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 10/04/2016] [Accepted: 11/04/2016] [Indexed: 12/25/2022]
|
32
|
Abstract
Simple and complex carbohydrates (glycans) have long been known to play major metabolic, structural and physical roles in biological systems. Targeted microbial binding to host glycans has also been studied for decades. But such biological roles can only explain some of the remarkable complexity and organismal diversity of glycans in nature. Reviewing the subject about two decades ago, one could find very few clear-cut instances of glycan-recognition-specific biological roles of glycans that were of intrinsic value to the organism expressing them. In striking contrast there is now a profusion of examples, such that this updated review cannot be comprehensive. Instead, a historical overview is presented, broad principles outlined and a few examples cited, representing diverse types of roles, mediated by various glycan classes, in different evolutionary lineages. What remains unchanged is the fact that while all theories regarding biological roles of glycans are supported by compelling evidence, exceptions to each can be found. In retrospect, this is not surprising. Complex and diverse glycans appear to be ubiquitous to all cells in nature, and essential to all life forms. Thus, >3 billion years of evolution consistently generated organisms that use these molecules for many key biological roles, even while sometimes coopting them for minor functions. In this respect, glycans are no different from other major macromolecular building blocks of life (nucleic acids, proteins and lipids), simply more rapidly evolving and complex. It is time for the diverse functional roles of glycans to be fully incorporated into the mainstream of biological sciences.
Collapse
Affiliation(s)
- Ajit Varki
- Departments of Medicine and Cellular & Molecular Medicine, Glycobiology Research and Training Center, University of California at San Diego, La Jolla, CA 92093-0687, USA
| |
Collapse
|
33
|
Chou KJ, Lee PT, Chen CL, Hsu CY, Huang WC, Huang CW, Fang HC. CD44 fucosylation on mesenchymal stem cell enhances homing and macrophage polarization in ischemic kidney injury. Exp Cell Res 2016; 350:91-102. [PMID: 27871849 DOI: 10.1016/j.yexcr.2016.11.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 11/01/2016] [Accepted: 11/15/2016] [Indexed: 01/14/2023]
Abstract
The lack of homing ability possibly reduces the healing potential of bone-marrow-derived mesenchymal stem cells (MSCs). Therefore, transforming native CD44 on MSCs into a hematopoietic cell E-/L-selectin ligand (HCELL) that possesses potent E-selectin affinity might enhance the homing and regenerative abilities of MSCs. Through fucosyltransferase VI (FTVI) transfection, MSCs were fucosylated on N-glycans of CD44 to become HCELL positive, thus interacting with E-selectin on injured endothelial cells. HCELL expression facilitated MSC homing in kidneys within 24h after injury and reduced lung stasis. An in vitro adhesion assay revealed that transfection enhanced the association between MSCs and hypoxic endothelial cells. In mice treated with HCELL-positive MSCs, the injured kidneys exhibited clusters of homing MSCs, whereas MSCs were rarely observed in mouse kidneys treated with HCELL-negative MSCs. Most MSCs were initially localized at the renal capsule, and some MSCs later migrated inward between tubules. Most homing MSCs were in close contact with inflammatory cells without tubular transdifferentiation. Furthermore, HCELL-positive MSCs substantially alleviated renal injury, partly by enhancing the polarization of infiltrating macrophages. In conclusion, engineering the glycan of CD44 on MSCs through FTVI transfection might enhance renotropism and the regenerating ability of MSCs in ischemic kidney injury.
Collapse
Affiliation(s)
- Kang-Ju Chou
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Po-Tsang Lee
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chien-Liang Chen
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Yang Hsu
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wei-Chieh Huang
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Chien-Wei Huang
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Hua-Chang Fang
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
34
|
Levesque JP, Winkler IG. Cell Adhesion Molecules in Normal and Malignant Hematopoiesis: from Bench to Bedside. CURRENT STEM CELL REPORTS 2016. [DOI: 10.1007/s40778-016-0066-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
35
|
Mohme M, Riethdorf S, Pantel K. Circulating and disseminated tumour cells - mechanisms of immune surveillance and escape. Nat Rev Clin Oncol 2016; 14:155-167. [PMID: 27644321 DOI: 10.1038/nrclinonc.2016.144] [Citation(s) in RCA: 391] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Metastatic spread of tumour cells is the main cause of cancer-related deaths. Understanding the mechanisms of tumour-cell dissemination has, therefore, become an important focus for cancer research. In patients with cancer, disseminated cancer cells are often detectable in the peripheral blood as circulating tumour cells (CTCs) and in the bone marrow or lymph nodes as disseminated tumour cells (DTCs). The identification and characterization of CTCs and DTCs has yielded important insights into the mechanisms of metastasis, resulting in a better understanding of the molecular alterations and profiles underlying drug resistance. Given the expanding role of immunotherapies in the treatment of cancer, interactions between tumour cells and immune cells are the subject of intense research. Theoretically, cancer cells that exit the primary tumour site - leaving the protection of the typically immunosuppressive tumour microenvironment - will be more vulnerable to attack by immune effector cells; thus, the survival of tumour cells after dissemination might be the 'Achilles' heel' of metastatic progression. In this Review, we discuss findings relating to the interactions of CTCs and DTCs with the immune system, in the context of cancer immuno-editing, evasion from immune surveillance, and formation of metastases.
Collapse
Affiliation(s)
- Malte Mohme
- Department of Tumour Biology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany.,Department of Neurosurgery, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Sabine Riethdorf
- Department of Tumour Biology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Klaus Pantel
- Department of Tumour Biology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| |
Collapse
|
36
|
The effect of soluble E-selectin on tumor progression and metastasis. BMC Cancer 2016; 16:331. [PMID: 27220365 PMCID: PMC4879723 DOI: 10.1186/s12885-016-2366-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 05/17/2016] [Indexed: 12/14/2022] Open
Abstract
Background Distant metastasis resulting from vascular dissemination of cancer cells is the primary cause of mortality from breast cancer. We have previously reported that E-selectin expression on the endothelial cell surface mediates shear-resistant adhesion and migration of circulating cancer cells via interaction with CD44. As a result of shedding, soluble E-selectin (sE-selectin) from the activated endothelium is present in the serum. In this study, we aimed to understand the role of sE-selectin in tumor progression and metastasis. Methods We investigated the effect of sE-selectin on shear-resistant adhesion and migration of metastatic breast cancer cells and leukocytes in vitro and in vivo. Results We found that sE-selectin promoted migration and shear-resistant adhesion of CD44+/high breast cancer cell lines (MDA-MB-231 and MDA-MB-468) to non-activated human microvessel endothelial cells (ES-HMVECs), but not of CD44-/low breast cancer cell lines (MCF-7 and T-47D). This endothelial E-selectin independent, sE-selectin-mediated shear-resistant adhesion was also observed in a leukocyte cell line (HL-60) as well as human peripheral blood mononuclear cells (PBMCs). Additionally, the incubation of MDA-MB-231 cells with sE-selectin triggered FAK phosphorylation and shear-resistant adhesion of sE-selectin-treated cells resulted in increased endothelial permeabilization. However, CD44 knockdown in MDA-MB-231 and HL-60 cells resulted in a significant reduction of sE-selectin-mediated shear-resistant adhesion to non-activated HMVECs, suggesting the involvement of CD44/FAK. Moreover, functional blockade of ICAM-1 in non-activated HMVECs resulted in a marked reduction of sE-selectin-mediated shear-resistant adhesion. Finally, the pre-incubation of CD44+ 4 T1 murine breast cancer cells with sE-selectin augmented infiltration into the lung in E-selectin K/O mice and infusion of human PBMCs pre-incubated with sE-selectin stimulated MDA-MB-231 xenografted breast tumor growth in NSG mice. Conclusions Our data suggest that circulating sE-selectin stimulates a broad range of circulating cells via CD44 and mediates pleiotropic effects that promote migration and shear-resistant adhesion in an endothelial E-selectin independent fashion, in turn accelerating tissue infiltration of leukocytes and cancer cells. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2366-2) contains supplementary material, which is available to authorized users.
Collapse
|
37
|
Sackstein R. Fulfilling Koch's postulates in glycoscience: HCELL, GPS and translational glycobiology. Glycobiology 2016; 26:560-70. [PMID: 26933169 DOI: 10.1093/glycob/cww026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 02/23/2016] [Indexed: 12/16/2022] Open
Abstract
Glycoscience-based research that is performed expressly to address medical necessity and improve patient outcomes is called "translational glycobiology". In the 19th century, Robert Koch proposed a set of postulates to rigorously establish causality in microbial pathogenesis, and these postulates can be reshaped to guide knowledge into how naturally-expressed glycoconjugates direct molecular processes critical to human well-being. Studies in the 1990s indicated that E-selectin, an endothelial lectin that binds sialofucosylated carbohydrate determinants, is constitutively expressed on marrow microvessels, and investigations in my laboratory indicated that human hematopoietic stem cells (HSCs) uniquely express high levels of a specialized glycoform of CD44 called "hematopoietic cell E-/L-selectin ligand" (HCELL) that functions as a highly potent E-selectin ligand. To assess the role of HCELL in directing HSC migration to marrow, a method called "glycosyltransferase-programmed stereosubstitution" (GPS) was developed to custom-modify CD44 glycans to enforce HCELL expression on viable cell surfaces. Human mesenchymal stem cells (MSCs) are devoid of E-selectin ligands, but GPS-based glycoengineering of CD44 on MSCs licenses homing of these cells to marrow in vivo, providing direct evidence that HCELL serves as a "bone marrow homing receptor". This review will discuss the molecular basis of cell migration in historical context, will describe the discovery of HCELL and its function as the bone marrow homing receptor, and will inform on how glycoengineering of CD44 serves as a model for adapting Koch's postulates to elucidate the key roles that glycoconjugates play in human biology and for realizing the immense impact of translational glycobiology in clinical medicine.
Collapse
Affiliation(s)
- Robert Sackstein
- Department of Dermatology and Department of Medicine, Brigham & Women's Hospital, Boston, MA, USA Harvard Skin Disease Research Center Program of Excellence in Glycosciences, Harvard Medical School, 77 Avenue Louis Pasteur, Room 671, Boston, MA 02115, USA
| |
Collapse
|
38
|
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: 10.8] [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
|
39
|
Merzaban JS, Imitola J, Starossom SC, Zhu B, Wang Y, Lee J, Ali AJ, Olah M, Abuelela AF, Khoury SJ, Sackstein R. Cell surface glycan engineering of neural stem cells augments neurotropism and improves recovery in a murine model of multiple sclerosis. Glycobiology 2015; 25:1392-409. [PMID: 26153105 DOI: 10.1093/glycob/cwv046] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 07/02/2015] [Indexed: 02/07/2023] Open
Abstract
Neural stem cell (NSC)-based therapies offer potential for neural repair in central nervous system (CNS) inflammatory and degenerative disorders. Typically, these conditions present with multifocal CNS lesions making it impractical to inject NSCs locally, thus mandating optimization of vascular delivery of the cells to involved sites. Here, we analyzed NSCs for expression of molecular effectors of cell migration and found that these cells are natively devoid of E-selectin ligands. Using glycosyltransferase-programmed stereosubstitution (GPS), we glycan engineered the cell surface of NSCs ("GPS-NSCs") with resultant enforced expression of the potent E-selectin ligand HCELL (hematopoietic cell E-/L-selectin ligand) and of an E-selectin-binding glycoform of neural cell adhesion molecule ("NCAM-E"). Following intravenous (i.v.) injection, short-term homing studies demonstrated that, compared with buffer-treated (control) NSCs, GPS-NSCs showed greater neurotropism. Administration of GPS-NSC significantly attenuated the clinical course of experimental autoimmune encephalomyelitis (EAE), with markedly decreased inflammation and improved oligodendroglial and axonal integrity, but without evidence of long-term stem cell engraftment. Notably, this effect of NSC is not a universal property of adult stem cells, as administration of GPS-engineered mouse hematopoietic stem/progenitor cells did not improve EAE clinical course. These findings highlight the utility of cell surface glycan engineering to boost stem cell delivery in neuroinflammatory conditions and indicate that, despite the use of a neural tissue-specific progenitor cell population, neural repair in EAE results from endogenous repair and not from direct, NSC-derived cell replacement.
Collapse
Affiliation(s)
- Jasmeen S Merzaban
- Department of Dermatology Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Jaime Imitola
- Department of Neurology, Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah C Starossom
- Department of Neurology, Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Bing Zhu
- Department of Neurology, Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yue Wang
- Department of Neurology, Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Amal J Ali
- Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Marta Olah
- Department of Neurology, Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ayman F Abuelela
- Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Samia J Khoury
- Department of Neurology, Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Robert Sackstein
- Department of Dermatology Department of Medicine, Harvard Skin Disease Research Center
| |
Collapse
|
40
|
Guvench O. Revealing the Mechanisms of Protein Disorder and N-Glycosylation in CD44-Hyaluronan Binding Using Molecular Simulation. Front Immunol 2015; 6:305. [PMID: 26136744 PMCID: PMC4468915 DOI: 10.3389/fimmu.2015.00305] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 05/28/2015] [Indexed: 11/13/2022] Open
Abstract
The extracellular N-terminal hyaluronan binding domain (HABD) of CD44 is a small globular domain that confers hyaluronan (HA) binding functionality to this large transmembrane glycoprotein. When recombinantly expressed by itself, HABD exists as a globular water-soluble protein that retains the capacity to bind HA. This has enabled atomic-resolution structural biology experiments that have revealed the structure of HABD and its binding mode with oligomeric HA. Such experiments have also pointed to an order-to-disorder transition in HABD that is associated with HA binding. However, it had remained unclear how this structural transition was involved in binding since it occurs in a region of HABD distant from the HA-binding site. Furthermore, HABD is known to be N-glycosylated, and such glycosylation can diminish HA binding when the associated N-glycans are capped with sialic acid residues. The intrinsic flexibility of disordered proteins and of N-glycans makes it difficult to apply experimental structural biology approaches to probe the molecular mechanisms of how the order-to-disorder transition and N-glycosylation can modulate HA binding by HABD. We review recent results from molecular dynamics simulations that provide atomic-resolution mechanistic understanding of such modulation to help bridge gaps between existing experimental binding and structural biology data. Findings from these simulations include: Tyr42 may function as a molecular switch that converts the HA-binding site from a low affinity to a high affinity state; in the partially disordered form of HABD, basic amino acids in the C-terminal region can gain sufficient mobility to form direct contacts with bound HA to further stabilize binding; and terminal sialic acids on covalently attached N-glycans can form charge-paired hydrogen bonding interactions with basic amino acids that could otherwise bind to HA, thereby blocking HA binding to glycosylated CD44 HABD.
Collapse
Affiliation(s)
- Olgun Guvench
- Department of Pharmaceutical Sciences, University of New England College of Pharmacy , Portland, ME , USA
| |
Collapse
|
41
|
Sackstein R, Fuhlbrigge R. Western blot analysis of adhesive interactions under fluid shear conditions: the blot rolling assay. Methods Mol Biol 2015; 1312:399-410. [PMID: 26044020 DOI: 10.1007/978-1-4939-2694-7_39] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Western blotting has proven to be an important technique in the analysis of receptor-ligand interactions (i.e., by ligand blotting) and for identifying molecules mediating cell attachment (i.e., by cell blotting). Conventional ligand blotting and cell blotting methods employ non-dynamic (static) incubation conditions, whereby molecules or cells of interest are placed in suspension and overlaid on membranes. However, many cell-cell and cell-matrix adhesive interactions occur under fluid shear conditions, and shear stress itself mediates and/or facilitates the engagement of these physiologically appropriate receptors and ligands. Notably, shear forces critically influence the adhesion of circulating cells and platelets to vessel walls in physiologic cell migration and hemostasis, as well as in inflammatory and thrombotic disorders, cancer metastasis, and atherosclerosis. Use of non-dynamic blotting conditions to analyze such interactions can introduce bias, overtly missing relevant effectors and/or exaggerating the relative role(s) of non-physiologic adhesion molecules. To address this shortfall, we have developed a new technique for identifying binding interactions under fluid shear conditions, the "blot rolling assay." Using this method, molecules in a complex mixture are resolved by gel electrophoresis, transferred to a membrane that is rendered semitransparent, and the membrane is then incorporated into a parallel-plate flow chamber apparatus. Under controlled flow conditions, cells or particles bearing adhesion proteins of interest are then introduced into the chamber and interactions with individual immobilized molecules (bands) can be visualized in real time. The substrate molecule(s) supporting adhesion under fluid shear can then be identified by staining with specific antibodies or by excising the relevant band(s) and performing mass spectrometry or microsequencing of the isolated material. This method thus allows for the identification, within a complex mixture and without prior isolation or purification, of both known and previously uncharacterized adhesion molecules operational under dynamic conditions.
Collapse
Affiliation(s)
- Robert Sackstein
- Program of Excellence in glycosciences, Harvard Medical School, Boston, MA, USA,
| | | |
Collapse
|
42
|
Shirure VS, Liu T, Delgadillo LF, Cuckler CM, Tees DFJ, Benencia F, Goetz DJ, Burdick MM. CD44 variant isoforms expressed by breast cancer cells are functional E-selectin ligands under flow conditions. Am J Physiol Cell Physiol 2014; 308:C68-78. [PMID: 25339657 DOI: 10.1152/ajpcell.00094.2014] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Adhesion of circulating tumor cells to vascular endothelium is mediated by specialized molecules that are functional under shear forces exerted by hematogenous flow. Endothelial E-selectin binding to glycoforms of CD44 mediates shear-resistant cell adhesion in numerous physiological and pathological conditions. However, this pathway is poorly understood in breast cancer and is the focus of the present investigation. All breast cancer cell lines used in this study strongly expressed CD44. In particular, BT-20 cells expressed CD44s and multiple CD44v isoforms, whereas MDA-MB-231 cells predominantly expressed CD44s but weakly expressed CD44v isoforms. CD44 expressed by BT-20, but not MDA-MB-231, cells possessed E-selectin ligand activity as detected by Western blotting and antigen capture assays. Importantly, CD44 expressed by intact BT-20 cells were functional E-selectin ligands, regulating cell rolling and adhesion under physiological flow conditions, as found by shRNA-targeted silencing of CD44. Antigen capture assays strongly suggest greater shear-resistant E-selectin ligand activity of BT-20 cell CD44v isoforms than CD44s. Surprisingly, CD44 was not recognized by the HECA-452 MAb, which detects sialofucosylated epitopes traditionally expressed by selectin ligands, suggesting that BT-20 cells express a novel glycoform of CD44v as an E-selectin ligand. The activity of this glycoform was predominantly attributed to N-linked glycans. Furthermore, expression of CD44v as an E-selectin ligand correlated with high levels of fucosyltransferase-3 and -6 and epithelial, rather than mesenchymal, cell phenotype. Together, these data demonstrate that expression of CD44 as a functional E-selectin ligand may be important in breast cancer metastasis.
Collapse
Affiliation(s)
- Venktesh S Shirure
- Department of Chemical and Biomolecular Engineering, Russ College of Engineering and Technology, Ohio University, Athens, Ohio
| | - Tiantian Liu
- Department of Chemical and Biomolecular Engineering, Russ College of Engineering and Technology, Ohio University, Athens, Ohio
| | - Luis F Delgadillo
- Department of Chemical and Biomolecular Engineering, Russ College of Engineering and Technology, Ohio University, Athens, Ohio
| | - Chaz M Cuckler
- Department of Chemical and Biomolecular Engineering, Russ College of Engineering and Technology, Ohio University, Athens, Ohio
| | - David F J Tees
- Biomedical Engineering Program, Russ College of Engineering and Technology, Ohio University, Athens, Ohio; Department of Physics and Astronomy, College of Arts and Sciences, Ohio University, Athens, Ohio; and
| | - Fabian Benencia
- Biomedical Engineering Program, Russ College of Engineering and Technology, Ohio University, Athens, Ohio; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | - Douglas J Goetz
- Department of Chemical and Biomolecular Engineering, Russ College of Engineering and Technology, Ohio University, Athens, Ohio; Biomedical Engineering Program, Russ College of Engineering and Technology, Ohio University, Athens, Ohio
| | - Monica M Burdick
- Department of Chemical and Biomolecular Engineering, Russ College of Engineering and Technology, Ohio University, Athens, Ohio; Biomedical Engineering Program, Russ College of Engineering and Technology, Ohio University, Athens, Ohio;
| |
Collapse
|
43
|
Kavanagh DPJ, Robinson J, Kalia N. Mesenchymal Stem Cell Priming: Fine-tuning Adhesion and Function. Stem Cell Rev Rep 2014; 10:587-99. [DOI: 10.1007/s12015-014-9510-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
44
|
Heazlewood SY, Oteiza A, Cao H, Nilsson SK. Analyzing hematopoietic stem cell homing, lodgment, and engraftment to better understand the bone marrow niche. Ann N Y Acad Sci 2014; 1310:119-28. [PMID: 24428368 DOI: 10.1111/nyas.12329] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The existence of a bone marrow (BM) niche--the location in which hematopoietic stem cells (HSCs) reside--was proposed more than 30 years ago. Recent data suggest that the interaction of HSCs with cellular and extracellular components within the BM is critical for HSC regulation. The tracking of immunofluorescently labeled, prospectively isolated HSCs to and within the BM cavity allows the assessment of the regulatory processes involved in (1) homing, which involves transendothelial migration into the BM; (2) lodgment, including transmarrow migration through the extravascular space; and (3) BM reconstitution. Together, such analyses provide a better understanding of the cellular and extracellular components involved in the regulation of HSC quiescence and differentiation. Homing and lodgment of transplanted HSCs, the first critical steps in engraftment, involve multiple interactions between HSCs and the BM microenvironment. Herein, we describe a refined method of analyzing homing efficiency and spatial distribution of HSCs harvested from endosteal and/or central BM regions; we also review alternate methods. Using these techniques, microenvironment modifications within the recipient or surface protein-expression modifications on donor HSCs in animal models provide insights into components influencing the homing, lodgment, and engraftment processes.
Collapse
Affiliation(s)
- Shen Y Heazlewood
- Materials Science and Engineering, Commonwealth Scientific and Industrial Research Organization (CSIRO), Melbourne, Australia
| | | | | | | |
Collapse
|
45
|
Selectins and their ligands are required for homing and engraftment of BCR-ABL1+ leukemic stem cells in the bone marrow niche. Blood 2014; 123:1361-71. [PMID: 24394666 DOI: 10.1182/blood-2013-11-538694] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We investigated adhesion pathways that contribute to engraftment of breakpoint cluster region-Abelson murine leukemia viral oncogene homolog 1 (BCR-ABL1)-induced chronic myelogenous leukemia (CML)-like myeloproliferative neoplasia in a mouse retroviral transduction/transplantation model. Compared with normal stem/progenitor cells, BCR-ABL1(+) progenitors had similar expression of very late antigen-4 (VLA4), VLA5, leukocyte functional antigen-1, and CXCR4 but lower expression of P-selectin glycoprotein ligand-1 (PSGL-1) and of L-selectin. Whereas vascular cell adhesion molecule-1 and P-selectin were not required, deficiency of E-selectin in the recipient bone marrow endothelium significantly reduced engraftment by BCR-ABL1-expressing stem cells following intravenous injection, with leukemogenesis restored by direct intrafemoral injection. BCR-ABL1-expressing cells deficient for PSGL-1 or the selectin ligand-synthesizing enzymes core-2 β1,6-N-acetylglucosaminyltransferase or fucosyltransferases IV/VII were impaired for engraftment, and destruction of selectin ligands on leukemic progenitors by neuraminidase reduced engraftment. BCR-ABL1-expressing L-selectin-deficient progenitors were also defective in homing and engraftment, with leukemogenesis rescued by coexpression of chimeric E/L-selectin. Antibody to L-selectin decreased the engraftment of BCR-ABL1-transduced stem cells. These results establish that BCR-ABL1(+) leukemic stem cells rely to a greater extent on selectins and their ligands for homing and engraftment than do normal stem cells. Selectin blockade is a novel strategy to exploit differences between normal and leukemic stem cells that may be beneficial in autologous transplantation for CML and perhaps other leukemias.
Collapse
|
46
|
Fucosylation with fucosyltransferase VI or fucosyltransferase VII improves cord blood engraftment. Cytotherapy 2013; 16:84-9. [PMID: 24094497 DOI: 10.1016/j.jcyt.2013.07.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 07/07/2013] [Accepted: 07/11/2013] [Indexed: 11/20/2022]
Abstract
BACKGROUND AIMS Advantages associated with the use of cord blood (CB) transplantation include the availability of cryopreserved units, ethnic diversity and lower incidence of graft-versus-host disease compared with bone marrow or mobilized peripheral blood. However, poor engraftment remains a major obstacle. We and others have found that ex vivo fucosylation can enhance engraftment in murine models, and now ex vivo treatment of CB with fucosyltransferase (FT) VI before transplantation is under clinical evaluation (NCT01471067). However, FTVII appears to be more relevant to hematopoietic cells and may alter acceptor substrate diversity. The present study compared the ability of FTVI and FTVII to improve the rapidity, magnitude, multi-lineage and multi-tissue engraftment of human CB hematopoietic stem and progenitor cells (HSPCs) in vivo. METHODS CD34-selected CB HSPCs were treated with recombinant FTVI, FTVII or mock control and then injected into immunodeficient mice and monitored for multi-lineage and multi-tissue engraftment. RESULTS Both FTVI and FTVII fucosylated CB CD34⁺ cells in vitro, and both led to enhanced rates and magnitudes of engraftment compared with untreated CB CD34⁺ cells in vivo. Engraftment after treatment with either FT was robust at multiple time points and in multiple tissues with similar multi-lineage potential. In contrast, only FTVII was able to fucosylate T and B lymphocytes. CONCLUSIONS Although FTVI and FTVII were found to be similarly able to fucosylate and enhance the engraftment of CB CD34⁺ cells, differences in their ability to fucosylate lymphocytes may modulate graft-versus-tumor or graft-versus-host effects and may allow further optimization of CB transplantation.
Collapse
|
47
|
Cao TM, Takatani T, King MR. Effect of extracellular pH on selectin adhesion: theory and experiment. Biophys J 2013; 104:292-9. [PMID: 23442851 DOI: 10.1016/j.bpj.2012.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 11/28/2012] [Accepted: 12/03/2012] [Indexed: 10/27/2022] Open
Abstract
Selectins mediate circulatory leukocyte trafficking to sites of inflammation and trauma, and the extracellular microenvironments at these sites often become acidic. In this study, we investigated the influence of slightly acidic pH on the binding dynamics of selectins (P-, L-, and E-selectin) to P-selectin glycoprotein ligand-1 (PSGL-1) via computational modeling (molecular dynamics) and experimental rolling assays under shear in vitro. The P-selectin/PSGL-1 binding is strengthened at acidic pH, as evidenced by the formation of a new hydrogen bond (seen computationally) and the observed decrease in the rolling velocities of model cells. In the case of L-selectin/PSGL-1 binding dynamics, the binding strength and frequency increase at acidic pH, as indicated by the greater cell-rolling flux of neutrophils and slower rolling velocities of L-selectin-coated microspheres, respectively. The cell flux is most likely due to an increased population of L-selectin in the high-affinity conformation as pH decreases, whereas the velocities are due to increased L-selectin/PSGL-1 contacts. In contrast to P- and L-selectin, the E-selectin/PSGL-1 binding does not exhibit significant changes at acidic pH levels, as shown both experimentally and computationally.
Collapse
Affiliation(s)
- Thong M Cao
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | | | | |
Collapse
|
48
|
Vakhrushev SY, Steentoft C, Vester-Christensen MB, Bennett EP, Clausen H, Levery SB. Enhanced mass spectrometric mapping of the human GalNAc-type O-glycoproteome with SimpleCells. Mol Cell Proteomics 2013; 12:932-44. [PMID: 23399548 DOI: 10.1074/mcp.o112.021972] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Characterizing protein GalNAc-type O-glycosylation has long been a major challenge, and as a result, our understanding of this glycoproteome is particularly poor. Recently, we presented a novel strategy for high throughput identification of O-GalNAc glycosites using zinc finger nuclease gene-engineered "SimpleCell" lines producing homogeneous truncated O-glycosylation. Total lysates of cells were trypsinized and subjected to lectin affinity chromatography enrichment, followed by identification of GalNAc O-glycopeptides by nLC-MS/MS, with electron transfer dissociation employed to specify sites of O-glycosylation. Here, we demonstrate a substantial improvement in the SimpleCell strategy by including an additional stage of lectin affinity chromatography on secreted glycoproteins from culture media (secretome) and by incorporating pre-fractionation of affinity-enriched glycopeptides via IEF before nLC-MS/MS. We applied these improvements to three human SimpleCells studied previously, and each yielded a substantial increase in the number of O-glycoproteins and O-glycosites identified. We found that analysis of the secretome was an important independent factor for increasing identifications, suggesting that further substantial improvements can also be sought through analysis of subcellular organelle fractions. In addition, we uncovered a substantial nonoverlapping set of O-glycoproteins and O-glycosites using an alternative protease digestion (chymotrypsin). In total, the improvements led to identification of 259 glycoproteins, of which 152 (59%) were novel compared with our previous strategy using the same three cell lines. With respect to individual glycosites, we identified a total of 856 sites, of which 508 (59%) were novel compared with our previous strategy; this includes four new identifications of O-GalNAc attached to tyrosine. Furthermore, we uncovered ≈ 220 O-glycosites wherein the peptides were clearly identified, but the glycosites could not be unambiguously assigned to specific positions. The improved strategy should greatly facilitate high throughput characterization of the human GalNAc-type O-glycoproteome as well as be applicable to analysis of other O-glycoproteomes.
Collapse
Affiliation(s)
- Sergey Y Vakhrushev
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark.
| | | | | | | | | | | |
Collapse
|
49
|
Shirure VS, Reynolds NM, Burdick MM. Mac-2 binding protein is a novel E-selectin ligand expressed by breast cancer cells. PLoS One 2012; 7:e44529. [PMID: 22970241 PMCID: PMC3435295 DOI: 10.1371/journal.pone.0044529] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2012] [Accepted: 08/03/2012] [Indexed: 12/01/2022] Open
Abstract
Hematogenous metastasis involves the adhesion of circulating tumor cells to vascular endothelium of the secondary site. We hypothesized that breast cancer cell adhesion is mediated by interaction of endothelial E-selectin with its glycoprotein counter-receptor(s) expressed on breast cancer cells. At a hematogenous wall shear rate, ZR-75-1 breast cancer cells specifically adhered to E-selectin expressing human umbilical vein endothelial cells when tested in parallel plate flow chamber adhesion assays. Consistent with their E-selectin ligand activity, ZR-75-1 cells expressed flow cytometrically detectable epitopes of HECA-452 mAb, which recognizes high efficiency E-selectin ligands typified by sialofucosylated moieties. Multiple E-selectin reactive proteins expressed by ZR-75-1 cells were revealed by immunoprecipitation with E-selectin chimera (E-Ig chimera) followed by Western blotting. Mass spectrometry analysis of the 72 kDa protein, which exhibited the most prominent E-selectin ligand activity, corresponded to Mac-2 binding protein (Mac-2BP), a heretofore unidentified E-selectin ligand. Immunoprecipitated Mac-2BP expressed sialofucosylated epitopes and possessed E-selectin ligand activity when tested by Western blot analysis using HECA-452 mAb and E-Ig chimera, respectively, demonstrating that Mac-2BP is a novel high efficiency E-selectin ligand. Furthermore, silencing the expression of Mac-2BP from ZR-75-1 cells by shRNA markedly reduced their adhesion to E-selectin expressing cells under physiological flow conditions, confirming the functional E-selectin ligand activity of Mac-2BP on intact cells. In addition to ZR-75-1 cells, several other E-selectin ligand positive breast cancer cell lines expressed Mac-2BP as detected by Western blot and flow cytometry, suggesting that Mac-2BP may be an E-selectin ligand in a variety of breast cancer types. Further, invasive breast carcinoma tissue showed co-localized expression of Mac-2BP and HECA-452 antigens by fluorescence microscopy, underscoring the possible role of Mac-2BP as an E-selectin ligand. In summary, breast cancer cells express Mac-2BP as a novel E-selectin ligand, potentially revealing a new prognostic and therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Venktesh S. Shirure
- Department of Chemical and Biomolecular Engineering, Russ College of Engineering and Technology, Ohio University, Athens, Ohio, United States of America
| | - Nathan M. Reynolds
- Department of Chemical and Biomolecular Engineering, Russ College of Engineering and Technology, Ohio University, Athens, Ohio, United States of America
| | - Monica M. Burdick
- Department of Chemical and Biomolecular Engineering, Russ College of Engineering and Technology, Ohio University, Athens, Ohio, United States of America
- Biomedical Engineering Program, Russ College of Engineering and Technology, Ohio University, Athens, Ohio, United States of America
- * E-mail:
| |
Collapse
|
50
|
CD44 as a novel target for treatment of staphylococcal enterotoxin B-induced acute inflammatory lung injury. Clin Immunol 2012; 144:41-52. [DOI: 10.1016/j.clim.2012.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Revised: 05/01/2012] [Accepted: 05/03/2012] [Indexed: 01/07/2023]
|