1
|
Song X, Pang Y, Wei Y, Yu D, Wang Y, Gao J, Zhang S, Wu Q, Wang J, Zhao S, Deng W. Filamin A suppresses the expression of ribosomal protein genes by controlling the activity of an EGR1-Sp1-GCN5/PCAF pathway in human cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119914. [PMID: 39938690 DOI: 10.1016/j.bbamcr.2025.119914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/21/2024] [Accepted: 01/26/2025] [Indexed: 02/14/2025]
Abstract
Human ribosome biogenesis requires four types of rRNA molecules and almost eighty cytoplasmic ribosomal proteins (CRPs) to be assembled together. . In the previous work, we showed that cytoskeletal filamin A (FLNA) can suppress rRNA expression in human cells. Thus, we hypothesized that FLNA can modulate the expression of CRPs because human cells have to coordinate cellular ribosome biogenesis. Here, we show that the absence of FLNA enhances the expression of most CRP genes assayed in the work, whereas the presence of FLNA dampens the expression of these CRP genes in several transformed cell types. The analysis of RNA-seq data revealed that FLNA silencing activated the expression of almost all CRPs and many mitochondrial RP genes in SaOS2 cells. These results indicate that FLNA acts as a negative regulator in CRP expression in human cells. Mechanistically, FLNA inhibits the expression of GCN5 and PCAF, which consequently impedes the occupancies of GCN5, PCAF, andH3K9ac at CRP gene loci. Both GCN5 and PCAF participates in the regulation of CRP expression either mediated by FLNA or independently. We show that FLNA silencing activates Sp1 expression and the activation of Sp1 stimulates the expression of Gcn5 and Pcaf genes. Further analysis revealed that EGR1 can bind the Sp1 gene promoter and activate Sp1 transcription. Collectively, this study revealed an EGR1-Sp1-GCN5/PCAF pathway by which FLNA modulates the expression of CRP genes. These findings shed light on how human cells coordinate the expression of CRP genes during ribosomal biogenesis.
Collapse
Affiliation(s)
- Xiaoye Song
- School of Life Science and Health, Wuhan University of Science and Technology, China
| | - Yaoyu Pang
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Yun Wei
- School of Life Science and Health, Wuhan University of Science and Technology, China
| | - Deen Yu
- School of Life Science and Health, Wuhan University of Science and Technology, China
| | - Ye Wang
- School of Life Science and Health, Wuhan University of Science and Technology, China
| | - Junwei Gao
- School of Life Science and Health, Wuhan University of Science and Technology, China
| | - Shuting Zhang
- School of Life Science and Health, Wuhan University of Science and Technology, China
| | - Qiang Wu
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao
| | - Juan Wang
- School of Life Science and Health, Wuhan University of Science and Technology, China.
| | - Shasha Zhao
- School of Life Science and Health, Wuhan University of Science and Technology, China.
| | - Wensheng Deng
- School of Life Science and Health, Wuhan University of Science and Technology, China.
| |
Collapse
|
2
|
Rodríguez-Frade JM, González-Granado LI, Santiago CA, Mellado M. The complex nature of CXCR4 mutations in WHIM syndrome. Front Immunol 2024; 15:1406532. [PMID: 39035006 PMCID: PMC11257845 DOI: 10.3389/fimmu.2024.1406532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/20/2024] [Indexed: 07/23/2024] Open
Abstract
Heterozygous autosomal dominant mutations in the CXCR4 gene cause WHIM syndrome, a severe combined immunodeficiency disorder. The mutations primarily affect the C-terminal region of the CXCR4 chemokine receptor, specifically several potential phosphorylation sites critical for agonist (CXCL12)-mediated receptor internalization and desensitization. Mutant receptors have a prolonged residence time on the cell surface, leading to hyperactive signaling that is responsible for some of the symptoms of WHIM syndrome. Recent studies have shown that the situation is more complex than originally thought, as mutant WHIM receptors and CXCR4 exhibit different dynamics at the cell membrane, which also influences their respective cellular functions. This review examines the functional mechanisms of CXCR4 and the impact of WHIM mutations in both physiological and pathological conditions.
Collapse
Affiliation(s)
- José Miguel Rodríguez-Frade
- Department of Immunology and Oncology, Chemokine Signaling Group, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Luis Ignacio González-Granado
- Department of Pediatrics, 12 de Octubre Health Research Institute (imas12), Madrid, Spain
- Department of Public Health School of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - César A. Santiago
- X-ray Crystallography Unit, Centro Nacional de Biotecnología/Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Mario Mellado
- Department of Immunology and Oncology, Chemokine Signaling Group, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| |
Collapse
|
3
|
Pereira RS, Kumar R, Cais A, Paulini L, Kahler A, Bravo J, Minciacchi VR, Krack T, Kowarz E, Zanetti C, Godavarthy PS, Hoeller F, Llavona P, Stark T, Tascher G, Nowak D, Meduri E, Huntly BJP, Münch C, Pampaloni F, Marschalek R, Krause DS. Distinct and targetable role of calcium-sensing receptor in leukaemia. Nat Commun 2023; 14:6242. [PMID: 37802982 PMCID: PMC10558580 DOI: 10.1038/s41467-023-41770-0] [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: 09/30/2022] [Accepted: 09/12/2023] [Indexed: 10/08/2023] Open
Abstract
Haematopoietic stem cells (HSC) reside in the bone marrow microenvironment (BMM), where they respond to extracellular calcium [eCa2+] via the G-protein coupled calcium-sensing receptor (CaSR). Here we show that a calcium gradient exists in this BMM, and that [eCa2+] and response to [eCa2+] differ between leukaemias. CaSR influences the location of MLL-AF9+ acute myeloid leukaemia (AML) cells within this niche and differentially impacts MLL-AF9+ AML versus BCR-ABL1+ leukaemias. Deficiency of CaSR reduces AML leukaemic stem cells (LSC) 6.5-fold. CaSR interacts with filamin A, a crosslinker of actin filaments, affects stemness-associated factors and modulates pERK, β-catenin and c-MYC signaling and intracellular levels of [Ca2+] in MLL-AF9+ AML cells. Combination treatment of cytarabine plus CaSR-inhibition in various models may be superior to cytarabine alone. Our studies suggest CaSR to be a differential and targetable factor in leukaemia progression influencing self-renewal of AML LSC via [eCa2+] cues from the BMM.
Collapse
Affiliation(s)
- Raquel S Pereira
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Rahul Kumar
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Alessia Cais
- Pediatric Neurooncology, Hopp Children's Cancer Center Heidelberg (KiTZ) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lara Paulini
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Alisa Kahler
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Jimena Bravo
- 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
| | - Theresa Krack
- 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
| | - Costanza Zanetti
- University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | | | - Fabian Hoeller
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Pablo Llavona
- Institute of Molecular Biology gGmbH (IMB), Mainz, Germany
| | - Tabea Stark
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Georg Tascher
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Daniel Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Eshwar Meduri
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Brian J P Huntly
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Christian Münch
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Francesco Pampaloni
- Buchmann Institute for Molecular Life Sciences (BMLS, CEF-MC), Goethe University, Frankfurt am Main, Germany
| | - 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.
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany.
- Institute of General Pharmacology and Toxicology, Goethe-University, Frankfurt am Main, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
- Frankfurt Cancer Institute, Frankfurt, Germany.
| |
Collapse
|
4
|
Zhao X, Kiyozuka K, Konishi A, Kawabata-Iwakawa R, Minamishima YA, Obinata H. Actin-binding protein Filamin B regulates the cell-surface retention of endothelial sphingosine 1-phosphate receptor 1. J Biol Chem 2023:104851. [PMID: 37220855 PMCID: PMC10300261 DOI: 10.1016/j.jbc.2023.104851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/25/2023] Open
Abstract
Sphingosine 1-phosphate receptor 1 (S1PR1) is a G protein-coupled receptor essential for vascular development and postnatal vascular homeostasis. When exposed to sphingosine 1-phosphate (S1P) in the blood of ∼1 μM, S1PR1 in endothelial cells retains cell-surface localization, while lymphocyte S1PR1 shows almost complete internalization, suggesting the cell-surface retention of S1PR1 is endothelial cell-specific. To identify regulating factors that function to retain S1PR1 on the endothelial cell surface, here we utilized an enzyme-catalyzed proximity labeling technique followed by proteomic analyses. We identified Filamin B (FLNB), an actin-binding protein involved in F-actin cross-linking, as a candidate regulating protein. We show FLNB knockdown by RNA interference induced massive internalization of S1PR1 into early endosomes, which was partially ligand-dependent and required receptor phosphorylation. Further investigation showed FLNB was also important for the recycling of internalized S1PR1 back to the cell surface. FLNB knockdown did not affect the localization of S1PR3, another S1P receptor subtype expressed in endothelial cells, nor did it affect localization of ectopically expressed β2-adrenergic receptor. Functionally, we show FLNB knockdown in endothelial cells impaired S1P-induced intracellular phosphorylation events and directed cell migration and enhancement of the vascular barrier. Taken together, our results demonstrate that FLNB is a novel regulator critical for S1PR1 cell-surface localization and thereby proper endothelial cell function.
Collapse
Affiliation(s)
- Xian Zhao
- Department of Biochemistry, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Keisuke Kiyozuka
- Department of Biochemistry, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Akimitsu Konishi
- Department of Biochemistry, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Reika Kawabata-Iwakawa
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, Gunma University, Gunma, Japan
| | | | - Hideru Obinata
- Education and Research Support Center, Gunma University Graduate School of Medicine, Gunma, Japan.
| |
Collapse
|
5
|
Altered CXCR4 dynamics at the cell membrane impairs directed cell migration in WHIM syndrome patients. Proc Natl Acad Sci U S A 2022; 119:e2119483119. [PMID: 35588454 PMCID: PMC9173760 DOI: 10.1073/pnas.2119483119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
SignificanceNew imaging-based approaches are incorporating new concepts to our knowledge of biological processes. The analysis of receptor dynamics involved in cell movement using single-particle tracking demonstrates that cells require chemokine-mediated receptor clustering to sense appropriately chemoattractant gradients. Here, we report that this process does not occur in T cells expressing CXCR4R334X, a mutant form of CXCR4 linked to WHIM syndrome (warts, hypogammaglobulinemia, infections, myelokathexis). The underlaying molecular mechanism involves inappropriate actin cytoskeleton remodeling due to the inadequate β-arrestin1 activation by CXCR4R334X, which alters its lateral mobility and spatial organization. These defects, associated to CXCR4R334X expression, contribute to the retention of hematopoietic precursors in bone marrow niches and explain the severe immunological symptoms associated with WHIM syndrome.
Collapse
|
6
|
The G Protein-Coupled Receptor Kinases (GRKs) in Chemokine Receptor-Mediated Immune Cell Migration: From Molecular Cues to Physiopathology. Cells 2021; 10:cells10010075. [PMID: 33466410 PMCID: PMC7824814 DOI: 10.3390/cells10010075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/18/2020] [Accepted: 12/30/2020] [Indexed: 02/07/2023] Open
Abstract
Although G protein-coupled receptor kinases (GRKs) have long been known to regulate G protein-coupled receptor (GPCR) desensitization, their more recently characterized functions as scaffolds and signalling adapters underscore that this small family of proteins governs a larger array of physiological functions than originally suspected. This review explores how GRKs contribute to the complex signalling networks involved in the migration of immune cells along chemokine gradients sensed by cell surface GPCRs. We outline emerging evidence indicating that the coordinated docking of several GRKs on an active chemokine receptor determines a specific receptor phosphorylation barcode that will translate into distinct signalling and migration outcomes. The guidance cues for neutrophil migration are emphasized based on several alterations affecting GRKs or GPCRs reported to be involved in pathological conditions.
Collapse
|
7
|
Aberrant CXCR4 Signaling at Crossroad of WHIM Syndrome and Waldenstrom's Macroglobulinemia. Int J Mol Sci 2020; 21:ijms21165696. [PMID: 32784523 PMCID: PMC7460815 DOI: 10.3390/ijms21165696] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Given its pleiotropic functions, including its prominent role in inflammation, immune responses and cancer, the C-X-C chemokine receptor type 4 (CXCR4) has gained significant attention in recent years and has become a relevant target in drug development. Although the signaling properties of CXCR4 have been extensively studied, several aspects deserve deeper investigations. Mutations in the C-term tail of the CXCR4 gene cause WHIM syndrome, a rare congenital immunodeficiency associated by chronic leukopenia. Similar mutations have also been recently identified in 30% of patients affected by Waldenstrom’s macroglobulinaemia, a B-cell neoplasia with bone marrow accumulation of malignant cells. An ample body of work has been generated to define the impact of WHIM mutations on CXCR4 signaling properties and evaluate their role on pathogenesis, diagnosis, and response to therapy, although the identity of disease-causing signaling pathways and their relevance for disease development in different genetic variants are still open questions. This review discusses the current knowledge on biochemical properties of CXCR4 mutations to identify their prototypic signaling profile potentially useful to highlighting novel opportunities for therapeutic intervention.
Collapse
|
8
|
Martín‐Leal A, Blanco R, Casas J, Sáez ME, Rodríguez‐Bovolenta E, de Rojas I, Drechsler C, Real LM, Fabrias G, Ruíz A, Castro M, Schamel WWA, Alarcón B, van Santen HM, Mañes S. CCR5 deficiency impairs CD4 + T-cell memory responses and antigenic sensitivity through increased ceramide synthesis. EMBO J 2020; 39:e104749. [PMID: 32525588 PMCID: PMC7396835 DOI: 10.15252/embj.2020104749] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022] Open
Abstract
CCR5 is not only a coreceptor for HIV-1 infection in CD4+ T cells, but also contributes to their functional fitness. Here, we show that by limiting transcription of specific ceramide synthases, CCR5 signaling reduces ceramide levels and thereby increases T-cell antigen receptor (TCR) nanoclustering in antigen-experienced mouse and human CD4+ T cells. This activity is CCR5-specific and independent of CCR5 co-stimulatory activity. CCR5-deficient mice showed reduced production of high-affinity class-switched antibodies, but only after antigen rechallenge, which implies an impaired memory CD4+ T-cell response. This study identifies a CCR5 function in the generation of CD4+ T-cell memory responses and establishes an antigen-independent mechanism that regulates TCR nanoclustering by altering specific lipid species.
Collapse
Affiliation(s)
- Ana Martín‐Leal
- Department of Immunology and OncologyCentro Nacional de Biotecnología (CNB/CSIC)MadridSpain
| | - Raquel Blanco
- Department of Immunology and OncologyCentro Nacional de Biotecnología (CNB/CSIC)MadridSpain
| | - Josefina Casas
- Department of Biological ChemistryInstitute of Advanced Chemistry of Catalonia (IQAC‐CSIC)BarcelonaSpain
- CIBER Liver and Digestive Diseases (CIBER‐EDH)Instituto de Salud Carlos IIIMadridSpain
| | - María E Sáez
- Centro Andaluz de Estudios Bioinformáticos (CAEBi)SevilleSpain
| | - Elena Rodríguez‐Bovolenta
- Department of Cell Biology and ImmunologyCentro de Biología Molecular Severo Ochoa (CBMSO/CSIC)MadridSpain
| | - Itziar de Rojas
- Alzheimer Research CenterMemory Clinic of the Fundació ACEInstitut Català de Neurociències AplicadesBarcelonaSpain
| | - Carina Drechsler
- Signaling Research Centers BIOSS and CIBSSUniversity of FreiburgFreiburgGermany
- Department of ImmunologyFaculty of BiologyUniversity of FreiburgFreiburgGermany
- Institute for Pharmaceutical SciencesUniversity of FreiburgFreiburgGermany
| | - Luis Miguel Real
- Unit of Infectious Diseases and MicrobiologyHospital Universitario de ValmeSevilleSpain
- Department of Biochemistry, Molecular Biology and ImmunologySchool of MedicineUniversidad de MálagaMálagaSpain
| | - Gemma Fabrias
- Department of Biological ChemistryInstitute of Advanced Chemistry of Catalonia (IQAC‐CSIC)BarcelonaSpain
- CIBER Liver and Digestive Diseases (CIBER‐EDH)Instituto de Salud Carlos IIIMadridSpain
| | - Agustín Ruíz
- Alzheimer Research CenterMemory Clinic of the Fundació ACEInstitut Català de Neurociències AplicadesBarcelonaSpain
- CIBER Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos IIIMadridSpain
| | - Mario Castro
- Interdisciplinary Group of Complex SystemsEscuela Técnica Superior de IngenieríaUniversidad Pontificia ComillasMadridSpain
| | - Wolfgang WA Schamel
- Signaling Research Centers BIOSS and CIBSSUniversity of FreiburgFreiburgGermany
- Department of ImmunologyFaculty of BiologyUniversity of FreiburgFreiburgGermany
- Centre for Chronic Immunodeficiency (CCI)University of FreiburgFreiburgGermany
| | - Balbino Alarcón
- Department of Cell Biology and ImmunologyCentro de Biología Molecular Severo Ochoa (CBMSO/CSIC)MadridSpain
| | - Hisse M van Santen
- Department of Cell Biology and ImmunologyCentro de Biología Molecular Severo Ochoa (CBMSO/CSIC)MadridSpain
| | - Santos Mañes
- Department of Immunology and OncologyCentro Nacional de Biotecnología (CNB/CSIC)MadridSpain
| |
Collapse
|
9
|
Fumagalli A, Zarca A, Neves M, Caspar B, Hill SJ, Mayor F, Smit MJ, Marin P. CXCR4/ACKR3 Phosphorylation and Recruitment of Interacting Proteins: Key Mechanisms Regulating Their Functional Status. Mol Pharmacol 2019; 96:794-808. [PMID: 30837297 DOI: 10.1124/mol.118.115360] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 02/21/2019] [Indexed: 01/14/2023] Open
Abstract
The C-X-C motif chemokine receptor type 4 (CXCR4) and the atypical chemokine receptor 3 (ACKR3/CXCR7) are class A G protein-coupled receptors (GPCRs). Accumulating evidence indicates that GPCR subcellular localization, trafficking, transduction properties, and ultimately their pathophysiological functions are regulated by both interacting proteins and post-translational modifications. This has encouraged the development of novel techniques to characterize the GPCR interactome and to identify residues subjected to post-translational modifications, with a special focus on phosphorylation. This review first describes state-of-the-art methods for the identification of GPCR-interacting proteins and GPCR phosphorylated sites. In addition, we provide an overview of the current knowledge of CXCR4 and ACKR3 post-translational modifications and an exhaustive list of previously identified CXCR4- or ACKR3-interacting proteins. We then describe studies highlighting the importance of the reciprocal influence of CXCR4/ACKR3 interactomes and phosphorylation states. We also discuss their impact on the functional status of each receptor. These studies suggest that deeper knowledge of the CXCR4/ACKR3 interactomes along with their phosphorylation and ubiquitination status would shed new light on their regulation and pathophysiological functions.
Collapse
Affiliation(s)
- Amos Fumagalli
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Aurélien Zarca
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Maria Neves
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Birgit Caspar
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Stephen J Hill
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Federico Mayor
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Martine J Smit
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Philippe Marin
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| |
Collapse
|
10
|
Levy E, Reger R, Segerberg F, Lambert M, Leijonhufvud C, Baumer Y, Carlsten M, Childs R. Enhanced Bone Marrow Homing of Natural Killer Cells Following mRNA Transfection With Gain-of-Function Variant CXCR4 R334X. Front Immunol 2019; 10:1262. [PMID: 31231387 PMCID: PMC6560173 DOI: 10.3389/fimmu.2019.01262] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/17/2019] [Indexed: 11/13/2022] Open
Abstract
Adoptive transfer of natural killer (NK) cells can induce remission in patients with relapsed/refractory leukemia and myeloma. However, to date, clinical efficacy of NK cell immunotherapy has been limited to a sub-fraction of patients. Here we show that steps incorporated in the ex vivo manipulation/production of NK cell products used for adoptive infusion, such as over-night IL-2 activation or cryopreservation followed by ex vivo expansion, drastically decreases NK cell surface expression of the bone marrow (BM) homing chemokine receptor CXCR4. Reduced CXCR4 expression was associated with dampened in vitro NK cell migration toward its cognate ligand stromal-derived factor-1α (SDF-1α). NK cells isolated from patients with WHIM syndrome carry gain-of-function (GOF) mutations in CXCR4 (CXCR4R334X). Compared to healthy donors, we observed that NK cells expanded from WHIM patients have similar surface levels of CXCR4 but have a much stronger propensity to home to BM compartments when adoptively infused into NOD-scid IL2Rgammanull (NSG) mice. Therefore, in order to augment the capacity of adoptively infused NK cells to home to the BM, we genetically engineered ex vivo expanded NK cells to express the naturally occurring GOF CXCR4R334X receptor variant. Transfection of CXCR4R334X-coding mRNA into ex vivo expanded NK cells using a clinically applicable method consistently led to an increase in cell surface CXCR4 without altering NK cell phenotype, cytotoxic function, or compromising NK cell viability. Compared to non-transfected and wild type CXCR4-coding mRNA transfected counterparts, CXCR4R334X-engineered NK cells had significantly greater chemotaxis toward SDF-1α in vitro. Importantly, expression of CXCR4R334X on expanded NK cells resulted in significantly greater BM homing following adoptive transfer into NSG mice compared to non-transfected NK cell controls. Collectively, these data suggest up-regulation of cell surface CXCR4R334X on ex vivo expanded NK cells via mRNA transfection represents a novel approach to improve homing and target NK cell-based immunotherapies to BM where hematological malignancies reside.
Collapse
Affiliation(s)
- Emily Levy
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States.,The Department of Molecular Medicine, The George Washington University, Washington, DC, United States
| | - Robert Reger
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Filip Segerberg
- Department of Medicine, Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Melanie Lambert
- Department of Medicine, Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Caroline Leijonhufvud
- Department of Medicine, Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yvonne Baumer
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mattias Carlsten
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States.,Department of Medicine, Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Richard Childs
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
11
|
Wang L, Nakamura F. Identification of Filamin A Mechanobinding Partner I: Smoothelin Specifically Interacts with the Filamin A Mechanosensitive Domain 21. Biochemistry 2019; 58:4726-4736. [PMID: 30990690 DOI: 10.1021/acs.biochem.9b00100] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Filamin A (FLNA) is a ubiquitously expressed actin cross-linking protein and a scaffold of numerous binding partners to regulate cell proliferation, migration, and survival. FLNA is a homodimer, and each subunit has an N-terminal actin-binding domain followed by 24 immunoglobulin-like repeats (R). FLNA mediates mechanotransduction by force-induced conformational changes of its cryptic integrin-binding site on R21. Here, we identified two novel FLNA-binding partners, smoothelins (SMTN A and B) and leucine zipper protein 1 (LUZP1), using stable isotope labeling by amino acids in cell culture (SILAC)-based proteomics followed by an in silico screening for proteins having a consensus FLNA-binding domain. We found that, although SMTN does not interact with full-length FLNA, it binds to FLNA variant 1 (FLNAvar-1) that exposes the cryptic CD cleft of R21. Point mutations on the C strand that disrupt the integrin binding also block the SMTN interaction. We identified FLNA-binding domains on SMTN using mutagenesis and used the mutant SMTN to investigate the role of the FLNA-SMTN interaction on the dynamics and localization of SMTN in living cells. Fluorescence recovery after photobleaching (FRAP) of GFP-labeled SMTN in living cells demonstrated that the non-FLNA-binding mutant SMTN diffuses faster than wild-type SMTN. Moreover, inhibition of Rho-kinase using Y27632 also increases the diffusion. These data demonstrated that SMTN specifically interacts with FLNAvar-1 and mechanically activated FLNA in cells. The companion report (Wang and Nakamura, 2019) describes the interactions of FLNA with the transcript of the LUZP1 gene.
Collapse
Affiliation(s)
- Lina Wang
- School of Pharmaceutical Science and Technology, Life Science Platform , Tianjin University , 92 Weijin Road , Nankai District, Tianjin 300072 , China
| | - Fumihiko Nakamura
- School of Pharmaceutical Science and Technology, Life Science Platform , Tianjin University , 92 Weijin Road , Nankai District, Tianjin 300072 , China
| |
Collapse
|
12
|
Martínez-Muñoz L, Rodríguez-Frade JM, Barroso R, Sorzano CÓS, Torreño-Pina JA, Santiago CA, Manzo C, Lucas P, García-Cuesta EM, Gutierrez E, Barrio L, Vargas J, Cascio G, Carrasco YR, Sánchez-Madrid F, García-Parajo MF, Mellado M. Separating Actin-Dependent Chemokine Receptor Nanoclustering from Dimerization Indicates a Role for Clustering in CXCR4 Signaling and Function. Mol Cell 2019; 70:106-119.e10. [PMID: 29625032 DOI: 10.1016/j.molcel.2018.02.034] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 01/08/2018] [Accepted: 02/27/2018] [Indexed: 01/03/2023]
Abstract
A current challenge in cell motility studies is to understand the molecular and physical mechanisms that govern chemokine receptor nanoscale organization at the cell membrane, and their influence on cell response. Using single-particle tracking and super-resolution microscopy, we found that the chemokine receptor CXCR4 forms basal nanoclusters in resting T cells, whose extent, dynamics, and signaling strength are modulated by the orchestrated action of the actin cytoskeleton, the co-receptor CD4, and its ligand CXCL12. We identified three CXCR4 structural residues that are crucial for nanoclustering and generated an oligomerization-defective mutant that dimerized but did not form nanoclusters in response to CXCL12, which severely impaired signaling. Overall, our data provide new insights to the field of chemokine biology by showing that receptor dimerization in the absence of nanoclustering is unable to fully support CXCL12-mediated responses, including signaling and cell function in vivo.
Collapse
Affiliation(s)
- Laura Martínez-Muñoz
- Chemokine Signaling Group, Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain; Department of Cell Signaling, Centro Andaluz de Biología Molecular y Medicina Regenerativa (CSIC), 41092 Sevilla, Spain.
| | - José Miguel Rodríguez-Frade
- Chemokine Signaling Group, Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | - Rubén Barroso
- Chemokine Signaling Group, Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | - Carlos Óscar S Sorzano
- Biocomputing Unit, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | - Juan A Torreño-Pina
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860 Barcelona, Spain
| | - César A Santiago
- X-ray Crystallography Unit, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | - Carlo Manzo
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860 Barcelona, Spain; Universitat de Vic, Universitat Central de Catalunya (UVic-UCC), 08500 Vic, Spain
| | - Pilar Lucas
- Chemokine Signaling Group, Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | - Eva M García-Cuesta
- Chemokine Signaling Group, Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | - Enric Gutierrez
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860 Barcelona, Spain
| | - Laura Barrio
- B Cell Dynamics Group, Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | - Javier Vargas
- Biocomputing Unit, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain; Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Graciela Cascio
- Chemokine Signaling Group, Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | - Yolanda R Carrasco
- B Cell Dynamics Group, Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | | | - María F García-Parajo
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860 Barcelona, Spain; ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Mario Mellado
- Chemokine Signaling Group, Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain.
| |
Collapse
|
13
|
Lopez JJ, Albarrán L, Jardín I, Sanchez-Collado J, Redondo PC, Bermejo N, Bobe R, Smani T, Rosado JA. Filamin A Modulates Store-Operated Ca2+Entry by Regulating STIM1 (Stromal Interaction Molecule 1)–Orai1 Association in Human Platelets. Arterioscler Thromb Vasc Biol 2018; 38:386-397. [DOI: 10.1161/atvbaha.117.310139] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 12/13/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Jose J. Lopez
- From the Department of Physiology, University of Extremadura, Cáceres, Spain (J.J.L., L.A., I.J., J.S.-C., P.C.R., J.A.R.); Department of Hematology, Hospital San Pedro de Alcantara, Cáceres, Spain (N.B.); INSERM Unité Mixte de Recherche-Santé 1176, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France (R.B.); and Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Spain (T.S.)
| | - Letizia Albarrán
- From the Department of Physiology, University of Extremadura, Cáceres, Spain (J.J.L., L.A., I.J., J.S.-C., P.C.R., J.A.R.); Department of Hematology, Hospital San Pedro de Alcantara, Cáceres, Spain (N.B.); INSERM Unité Mixte de Recherche-Santé 1176, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France (R.B.); and Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Spain (T.S.)
| | - Isaac Jardín
- From the Department of Physiology, University of Extremadura, Cáceres, Spain (J.J.L., L.A., I.J., J.S.-C., P.C.R., J.A.R.); Department of Hematology, Hospital San Pedro de Alcantara, Cáceres, Spain (N.B.); INSERM Unité Mixte de Recherche-Santé 1176, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France (R.B.); and Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Spain (T.S.)
| | - Jose Sanchez-Collado
- From the Department of Physiology, University of Extremadura, Cáceres, Spain (J.J.L., L.A., I.J., J.S.-C., P.C.R., J.A.R.); Department of Hematology, Hospital San Pedro de Alcantara, Cáceres, Spain (N.B.); INSERM Unité Mixte de Recherche-Santé 1176, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France (R.B.); and Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Spain (T.S.)
| | - Pedro C. Redondo
- From the Department of Physiology, University of Extremadura, Cáceres, Spain (J.J.L., L.A., I.J., J.S.-C., P.C.R., J.A.R.); Department of Hematology, Hospital San Pedro de Alcantara, Cáceres, Spain (N.B.); INSERM Unité Mixte de Recherche-Santé 1176, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France (R.B.); and Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Spain (T.S.)
| | - Nuria Bermejo
- From the Department of Physiology, University of Extremadura, Cáceres, Spain (J.J.L., L.A., I.J., J.S.-C., P.C.R., J.A.R.); Department of Hematology, Hospital San Pedro de Alcantara, Cáceres, Spain (N.B.); INSERM Unité Mixte de Recherche-Santé 1176, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France (R.B.); and Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Spain (T.S.)
| | - Regis Bobe
- From the Department of Physiology, University of Extremadura, Cáceres, Spain (J.J.L., L.A., I.J., J.S.-C., P.C.R., J.A.R.); Department of Hematology, Hospital San Pedro de Alcantara, Cáceres, Spain (N.B.); INSERM Unité Mixte de Recherche-Santé 1176, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France (R.B.); and Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Spain (T.S.)
| | - Tarik Smani
- From the Department of Physiology, University of Extremadura, Cáceres, Spain (J.J.L., L.A., I.J., J.S.-C., P.C.R., J.A.R.); Department of Hematology, Hospital San Pedro de Alcantara, Cáceres, Spain (N.B.); INSERM Unité Mixte de Recherche-Santé 1176, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France (R.B.); and Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Spain (T.S.)
| | - Juan A. Rosado
- From the Department of Physiology, University of Extremadura, Cáceres, Spain (J.J.L., L.A., I.J., J.S.-C., P.C.R., J.A.R.); Department of Hematology, Hospital San Pedro de Alcantara, Cáceres, Spain (N.B.); INSERM Unité Mixte de Recherche-Santé 1176, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France (R.B.); and Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Spain (T.S.)
| |
Collapse
|
14
|
Lemos A, Melo R, Preto AJ, Almeida JG, Moreira IS, Cordeiro MNDS. In Silico Studies Targeting G-protein Coupled Receptors for Drug Research Against Parkinson's Disease. Curr Neuropharmacol 2018; 16:786-848. [PMID: 29521236 PMCID: PMC6080095 DOI: 10.2174/1570159x16666180308161642] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 02/16/2018] [Accepted: 02/02/2018] [Indexed: 11/22/2022] Open
Abstract
Parkinson's Disease (PD) is a long-term neurodegenerative brain disorder that mainly affects the motor system. The causes are still unknown, and even though currently there is no cure, several therapeutic options are available to manage its symptoms. The development of novel antiparkinsonian agents and an understanding of their proper and optimal use are, indeed, highly demanding. For the last decades, L-3,4-DihydrOxyPhenylAlanine or levodopa (L-DOPA) has been the gold-standard therapy for the symptomatic treatment of motor dysfunctions associated to PD. However, the development of dyskinesias and motor fluctuations (wearing-off and on-off phenomena) associated with long-term L-DOPA replacement therapy have limited its antiparkinsonian efficacy. The investigation for non-dopaminergic therapies has been largely explored as an attempt to counteract the motor side effects associated with dopamine replacement therapy. Being one of the largest cell membrane protein families, G-Protein-Coupled Receptors (GPCRs) have become a relevant target for drug discovery focused on a wide range of therapeutic areas, including Central Nervous System (CNS) diseases. The modulation of specific GPCRs potentially implicated in PD, excluding dopamine receptors, may provide promising non-dopaminergic therapeutic alternatives for symptomatic treatment of PD. In this review, we focused on the impact of specific GPCR subclasses, including dopamine receptors, adenosine receptors, muscarinic acetylcholine receptors, metabotropic glutamate receptors, and 5-hydroxytryptamine receptors, on the pathophysiology of PD and the importance of structure- and ligand-based in silico approaches for the development of small molecules to target these receptors.
Collapse
Affiliation(s)
- Agostinho Lemos
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007Porto, Portugal
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liège, 4000Liège, Belgium
| | - Rita Melo
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Rua Larga, 3004-517Coimbra, Portugal
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (ao km 139,7), 2695-066 Bobadela LRS, Portugal
| | - Antonio Jose Preto
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Rua Larga, 3004-517Coimbra, Portugal
| | - Jose Guilherme Almeida
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Rua Larga, 3004-517Coimbra, Portugal
| | - Irina Sousa Moreira
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Rua Larga, 3004-517Coimbra, Portugal
- Bijvoet Center for Biomolecular Research, Faculty of Science - Chemistry, Utrecht University, Utrecht, 3584CH, The Netherlands
| | - Maria Natalia Dias Soeiro Cordeiro
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007Porto, Portugal
| |
Collapse
|
15
|
Heusinkveld LE, Yim E, Yang A, Azani AB, Liu Q, Gao JL, McDermott DH, Murphy PM. Pathogenesis, diagnosis and therapeutic strategies in WHIM syndrome immunodeficiency. Expert Opin Orphan Drugs 2017; 5:813-825. [PMID: 29057173 DOI: 10.1080/21678707.2017.1375403] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
21 INTRODUCTION WHIM syndrome is a rare combined primary immunodeficiency disorder caused by autosomal dominant gain-of-function mutations in the chemokine receptor CXCR4. It is the only Mendelian condition known to be caused by mutation of a chemokine or chemokine receptor. As such, it provides a scientific opportunity to understand chemokine-dependent immunoregulation in humans and a medical opportunity to develop mechanism-based treatment and cure strategies. 22 AREAS COVERED This review covers the clinical features, genetics, immunopathogenesis and clinical management of WHIM syndrome. Clinical trials of targeted therapeutic agents and potential cure strategies are also included. 23 EXPERT OPINION WHIM syndrome may be particularly amenable to mechanism-based therapeutics for three reasons: 1) CXCR4 has been validated as the molecular target in the disease by Mendelian genetics; 2) the biochemical abnormality is excessive CXCR4 signaling; and 3) antagonists selective for CXCR4 have been developed. Plerixafor is FDA-approved for hematopoietic stem cell (HSC) mobilization and has shown preliminary safety and efficacy in phase I clinical trials in WHIM syndrome. Gene editing may represent a viable cure strategy, since chromothriptic deletion of the disease allele in HSCs resulted in clinical cure of a patient and because CXCR4 haploinsufficiency enhances engraftment of transplanted HSCs in mice.
Collapse
Affiliation(s)
- Lauren E Heusinkveld
- Laboratory of Molecular Immunology, Bldg 10, Room 11N113, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Erin Yim
- Laboratory of Molecular Immunology, Bldg 10, Room 11N113, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Alexander Yang
- Laboratory of Molecular Immunology, Bldg 10, Room 11N113, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Ari B Azani
- Laboratory of Molecular Immunology, Bldg 10, Room 11N113, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Qian Liu
- Laboratory of Molecular Immunology, Bldg 10, Room 11N113, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Ji-Liang Gao
- Laboratory of Molecular Immunology, Bldg 10, Room 11N113, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - David H McDermott
- Laboratory of Molecular Immunology, Bldg 10, Room 11N113, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Philip M Murphy
- Laboratory of Molecular Immunology, Bldg 10, Room 11N113, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
16
|
Lacalle RA, Blanco R, Carmona-Rodríguez L, Martín-Leal A, Mira E, Mañes S. Chemokine Receptor Signaling and the Hallmarks of Cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 331:181-244. [PMID: 28325212 DOI: 10.1016/bs.ircmb.2016.09.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The chemokines are a family of chemotactic cytokines that mediate their activity by acting on seven-transmembrane-spanning G protein-coupled receptors. Both the ability of the chemokines and their receptors to form homo- and heterodimers and the promiscuity of the chemokine-chemokine receptor interaction endow this protein family with enormous signaling plasticity and complexity that are not fully understood at present. Chemokines were initially identified as essential regulators of homeostatic and inflammatory trafficking of innate and adaptive leucocytes from lymphoid organs to tissues. Chemokines also mediate the host response to cancer. Nevertheless, chemokine function in this response is not limited to regulating leucocyte infiltration into the tumor microenvironment. It is now known that chemokines and their receptors influence most-if not all-hallmark processes of cancer; they act on both neoplastic and untransformed cells in the tumor microenvironment, including fibroblasts, endothelial cells (blood and lymphatic), bone marrow-derived stem cells, and, obviously, infiltrating leucocytes. This review begins with an overview of chemokine and chemokine receptor structure, to better define how chemokines affect the proliferation, survival, stemness, and metastatic potential of neoplastic cells. We also examine the main mechanisms by which chemokines regulate tumor angiogenesis and immune cell infiltration, emphasizing the pro- and antitumorigenic activity of this protein superfamily in these interrelated processes.
Collapse
Affiliation(s)
- R A Lacalle
- Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - R Blanco
- Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | | | - A Martín-Leal
- Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - E Mira
- Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - S Mañes
- Centro Nacional de Biotecnología/CSIC, Madrid, Spain.
| |
Collapse
|
17
|
Bernardini G, Antonangeli F, Bonanni V, Santoni A. Dysregulation of Chemokine/Chemokine Receptor Axes and NK Cell Tissue Localization during Diseases. Front Immunol 2016; 7:402. [PMID: 27766097 PMCID: PMC5052267 DOI: 10.3389/fimmu.2016.00402] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 09/21/2016] [Indexed: 01/06/2023] Open
Abstract
Chemokines are small chemotactic molecules that play key roles in physiological and pathological conditions. Upon signaling via their specific receptors, chemokines regulate tissue mobilization and trafficking of a wide array of immune cells, including natural killer (NK) cells. Current research is focused on analyzing changes in chemokine/chemokine receptor expression during various diseases to interfere with pathological trafficking of cells or to recruit selected cell types to specific tissues. NK cells are a heterogeneous lymphocyte population comprising several subsets endowed with distinct functional properties and mainly representing distinct stages of a linear development process. Because of their different functional potential, the type of subset that accumulates in a tissue drives the final outcome of NK cell-regulated immune response, leading to either protection or pathology. Correspondingly, chemokine receptors, including CXCR4, CXCR3, and CX3CR1, are differentially expressed by NK cell subsets, and their expression levels can be modulated during NK cell activation. At first, this review will summarize the current knowledge on the contribution of chemokines to the localization and generation of NK cell subsets in homeostasis. How an inappropriate chemotactic response can lead to pathology and how chemokine targeting can therapeutically affect tissue recruitment/localization of distinct NK cell subsets will also be discussed.
Collapse
Affiliation(s)
- Giovanni Bernardini
- Department of Molecular Medicine, Sapienza University, Rome, Italy; IRCCS NEUROMED - Mediterranean Neurological Institute, Isernia, Italy
| | | | - Valentina Bonanni
- Department of Molecular Medicine, Sapienza University , Rome , Italy
| | - Angela Santoni
- IRCCS NEUROMED - Mediterranean Neurological Institute, Isernia, Italy; Department of Molecular Medicine, Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
18
|
CXCR4 signaling in health and disease. Immunol Lett 2016; 177:6-15. [PMID: 27363619 DOI: 10.1016/j.imlet.2016.06.006] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 06/22/2016] [Accepted: 06/27/2016] [Indexed: 12/23/2022]
Abstract
Chemokines and chemokine receptors regulate multiple processes such morphogenesis, angiogenesis and immune responses. Among the chemokine receptors, CXCR4 stands out for its pleiotropic roles as well as for its involvement in several pathological conditions, including immune diseases, viral infections and cancer. For these reasons, CXCR4 represents a crucial target in drug development. In this review, we discuss of CXCR4 receptor properties and signaling in health and diseases, focusing on the WHIM syndrome, an inherited immunodeficiency caused by mutations of the CXCR4 gene.
Collapse
|
19
|
Pons M, Izquierdo I, Andreu-Carbó M, Garrido G, Planagumà J, Muriel O, Geli MI, Aragay AM. Regulation of chemokine receptor CCR2 recycling by filamin a phosphorylation. J Cell Sci 2016; 130:490-501. [DOI: 10.1242/jcs.193821] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/18/2016] [Indexed: 12/20/2022] Open
Abstract
Proper endosomal trafficking of ligand-activated G protein-coupled receptors (GPCRs) is essential to spatiotemporally tune their physiological responses. For the monocyte chemoattractant receptor 2 (CCR2B), endocytic recycling is important to sustain monocyte migration; while filamin A (FLNa) is essential for CCL2-induced monocyte migration. Here, we analyze the role of FLNa in the trafficking of CCR2B along the endocytic pathway. In FLNa knockdown cells, activated CCR2B accumulated in enlarged EEA-1-positive endosomes, which exhibited slow movement and fast fluorescence recovery, suggesting an imbalance between receptor entry and exit rates. Utilizing super-resolution microscopy, we observed that FLNa-GFP, CCR2B and β2-adrenergic receptor (β2AR) were present in actin-enriched endosomal microdomains. Depletion of FLNa decreased CCR2B association with these microdomains and concomitantly delayed CCR2B endosomal traffic, without apparently affecting the number of microdomains. Interestingly, CCR2B and β2AR signaling induced phosphorylation of FLNa at S2152 and this phosphorylation event was contributes to sustain receptor recycling. Thus, our data strongly suggest that CCR2B and β2AR signals to FLNa to stimulate its endocytosis and recycling to the plasma membrane.
Collapse
Affiliation(s)
- Mònica Pons
- Molecular Biology Institute of Barcelona (IBMB), Spanish National Research Council (CSIC), 08028 Barcelona, Spain
| | - Ismael Izquierdo
- Molecular Biology Institute of Barcelona (IBMB), Spanish National Research Council (CSIC), 08028 Barcelona, Spain
| | - Mireia Andreu-Carbó
- Molecular Biology Institute of Barcelona (IBMB), Spanish National Research Council (CSIC), 08028 Barcelona, Spain
| | - Georgina Garrido
- Molecular Biology Institute of Barcelona (IBMB), Spanish National Research Council (CSIC), 08028 Barcelona, Spain
- Present addresse: Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain
| | - Jesús Planagumà
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Present addresse: Department of Neuroimmunology, IDIBAPS, Barcelona, Spain
| | - Olivia Muriel
- Centro Nacional de Investigaciones Cardiovasculares, 28029 Madrid, Spain
| | - M. Isabel Geli
- Molecular Biology Institute of Barcelona (IBMB), Spanish National Research Council (CSIC), 08028 Barcelona, Spain
| | - Anna M. Aragay
- Molecular Biology Institute of Barcelona (IBMB), Spanish National Research Council (CSIC), 08028 Barcelona, Spain
| |
Collapse
|
20
|
Maciejewski-Duval A, Meuris F, Bignon A, Aknin ML, Balabanian K, Faivre L, Pasquet M, Barlogis V, Fieschi C, Bellanné-Chantelot C, Donadieu J, Schlecht-Louf G, Marin-Esteban V, Bachelerie F. Altered chemotactic response to CXCL12 in patients carrying GATA2 mutations. J Leukoc Biol 2015; 99:1065-76. [PMID: 26710799 DOI: 10.1189/jlb.5ma0815-388r] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 12/01/2015] [Indexed: 12/29/2022] Open
Abstract
GATA2 deficiency-formerly described as MonoMAC syndrome; dendritic cells, monocytes, B cells, and natural killer cell deficiency; familial myelodysplastic syndrome/acute myeloid leukemia; or Emberger syndrome-encompasses a range of hematologic and nonhematologic anomalies, mainly characterized by monocytopenia, B lymphopenia, natural killer cell cytopenia, neutropenia, immunodeficiency, and a high risk of developing acute myeloid leukemia. Herein, we present 7 patients with GATA2 deficiency recruited into the French Severe Chronic Neutropenia Registry, which enrolls patients with all kinds of congenital neutropenia. We performed extended immunophenotyping of their whole blood lymphocyte populations, together with the analysis of their chemotactic responses. Lymphopenia was recorded for B and CD4(+) T cells in 6 patients. Although only 3 patients displayed natural killer cell cytopenia, the CD56(bright) natural killer subpopulation was nearly absent in all 7 patients. Natural killer cells from 6 patients showed decreased CXCL12/CXCR4-dependent chemotaxis, whereas other lymphocytes, and most significantly B lymphocytes, displayed enhanced CXCL12-induced chemotaxis compared with healthy volunteers. Surface expression of CXCR4 was significantly diminished in the patients' natural killer cells, although the total expression of the receptor was found to be equivalent to that of natural killer cells from healthy individual controls. Together, these data reveal that GATA2 deficiency is associated with impaired membrane expression and chemotactic dysfunctions of CXCR4. These dysfunctions may contribute to the physiopathology of this deficiency by affecting the normal distribution of lymphocytes and thus potentially affecting the susceptibility of patients to associated infections.
Collapse
Affiliation(s)
- Anna Maciejewski-Duval
- UMR996 - Inflammation, Chemokines and Immunopathology, Inserm, Univ Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Floriane Meuris
- UMR996 - Inflammation, Chemokines and Immunopathology, Inserm, Univ Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Alexandre Bignon
- UMR996 - Inflammation, Chemokines and Immunopathology, Inserm, Univ Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Marie-Laure Aknin
- US31-UMS3679 -Plateforme PLAIMMO, Institut Paris-Saclay d'Innovation Thérapeutique (IPSIT), INSERM, CNRS, Université Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Karl Balabanian
- UMR996 - Inflammation, Chemokines and Immunopathology, Inserm, Univ Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Laurence Faivre
- Génétique et Anomalies du Développement, EA4271, Université de Bourgogne, Dijon, France and FHU TRANSLAD, Département de Génétique, CHU Dijon, Dijon, France
| | - Marlène Pasquet
- Département d'Hématologie du Centre Hospitalier Universitaire Toulouse Purpan and INSERM, CRCT, IUCT-Oncopole, Toulouse, France
| | - Vincent Barlogis
- Service d'Hématologie Pédiatrique, Assistance Publique, Hôpitaux de Marseille, Hôpital Timone Enfants, Marseille, France
| | - Claire Fieschi
- Département d'Immunologie Clinique, Hôpital Saint Louis and Université Denis Diderot, Paris, France
| | - Christine Bellanné-Chantelot
- US31-UMS3679 -Plateforme PLAIMMO, Institut Paris-Saclay d'Innovation Thérapeutique (IPSIT), INSERM, CNRS, Université Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Jean Donadieu
- UMR996 - Inflammation, Chemokines and Immunopathology, Inserm, Univ Paris-Sud, Université Paris-Saclay, Clamart, France; UMR996 - Inflammation, Chemokines and Immunopathology, Inserm, Univ Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Géraldine Schlecht-Louf
- UMR996 - Inflammation, Chemokines and Immunopathology, Inserm, Univ Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Viviana Marin-Esteban
- UMR996 - Inflammation, Chemokines and Immunopathology, Inserm, Univ Paris-Sud, Université Paris-Saclay, Clamart, France;
| | - Françoise Bachelerie
- UMR996 - Inflammation, Chemokines and Immunopathology, Inserm, Univ Paris-Sud, Université Paris-Saclay, Clamart, France;
| |
Collapse
|
21
|
Symptomatic Improvement in Human Papillomavirus-Induced Epithelial Neoplasia by Specific Targeting of the CXCR4 Chemokine Receptor. J Invest Dermatol 2015; 136:473-480. [PMID: 26967480 DOI: 10.1016/j.jid.2015.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/12/2015] [Accepted: 10/14/2015] [Indexed: 12/23/2022]
Abstract
Human papillomavirus (HPV) infection is estimated to be the causal agent in 5% of all human cancers and is the leading cause of genital warts, which is the most common sexually transmitted viral disease. Currently, there are no medications to treat HPV infection, and therapeutic strategies primarily target HPV-related cancer rather than viral infection. HPV infection has severe effects on patients who display selective susceptibility to the virus in the context of primary immunodeficiencies, such as the warts, hypogammaglobulinemia, infections, and myelokathexis syndrome, which is caused by dysfunctions of CXCR4, the receptor for the CXCL12 chemokine. In this study we showed in a transgenic mouse model of HPV-induced epidermal neoplasia the beneficial effects of Cxcl12/Cxcr4 pathway blockade with the selective CXCR4 antagonist AMD3100. Daily treatment with AMD3100 for 28 days potently reduced the abnormal ear epidermal thickening in all mice. This effect was associated with reductions in keratinocyte hyperproliferation and immune cell infiltration, both of which are linked to neoplastic progression. Moreover, we observed the abnormal coordinate expression of Cxcl12 and p16INK4a (a surrogate marker of HPV-induced cancers) in dysplastic epidermal keratinocytes, which was inhibited by AMD3100 treatment. These results provide strong evidence for the therapeutic potential of CXCL12/CXCR4 pathway blockade in HPV-induced pathogenesis.
Collapse
|
22
|
Tirupula KC, Ithychanda SS, Mohan ML, Naga Prasad SV, Qin J, Karnik SS. G protein-coupled receptors directly bind filamin A with high affinity and promote filamin phosphorylation. Biochemistry 2015; 54:6673-83. [PMID: 26460884 PMCID: PMC4642222 DOI: 10.1021/acs.biochem.5b00975] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Although interaction of a few G protein-coupled receptors (GPCRs) with Filamin A, a key actin cross-linking and biomechanical signal transducer protein, has been observed, a comprehensive structure-function analysis of this interaction is lacking. Through a systematic sequence-based analysis, we found that a conserved filamin binding motif is present in the cytoplasmic domains of >20% of the 824 GPCRs encoded in the human genome. Direct high-affinity interaction of filamin binding motif peptides of select GPCRs with the Ig domain of Filamin A was confirmed by nuclear magnetic resonance spectroscopy and isothermal titration calorimetric experiments. Engagement of the filamin binding motif with the Filamin A Ig domain induced the phosphorylation of filamin by protein kinase A in vitro. In transfected cells, agonist activation as well as constitutive activation of representative GPCRs dramatically elicited recruitment and phosphorylation of cellular Filamin A, a phenomenon long known to be crucial for regulating the structure and dynamics of the cytoskeleton. Our data suggest a molecular mechanism for direct GPCR-cytoskeleton coupling via filamin. Until now, GPCR signaling to the cytoskeleton was predominantly thought to be indirect, through canonical G protein-mediated signaling cascades involving GTPases, adenylyl cyclases, phospholipases, ion channels, and protein kinases. We propose that the GPCR-induced filamin phosphorylation pathway is a conserved, novel biochemical signaling paradigm.
Collapse
Affiliation(s)
- Kalyan C Tirupula
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic , Cleveland, Ohio 44195, United States
| | - Sujay S Ithychanda
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic , Cleveland, Ohio 44195, United States
| | - Maradumane L Mohan
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic , Cleveland, Ohio 44195, United States
| | - Sathyamangla V Naga Prasad
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic , Cleveland, Ohio 44195, United States
| | - Jun Qin
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic , Cleveland, Ohio 44195, United States
| | - Sadashiva S Karnik
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic , Cleveland, Ohio 44195, United States
| |
Collapse
|