1
|
Chen Z, Ford KP, Islam MBAR, Wan H, Han H, Ramakrishnan A, Brown RJ, Villanueva V, Wang Y, Davis BT, Weiss C, Cui W, Gate D, Schwulst SJ. Anti-CD49d Ab treatment ameliorates age-associated inflammatory response and mitigates CD8 + T-cell cytotoxicity after traumatic brain injury. J Neuroinflammation 2024; 21:267. [PMID: 39427160 PMCID: PMC11491007 DOI: 10.1186/s12974-024-03257-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
Patients aged 65 years and older account for an increasing proportion of patients with traumatic brain injury (TBI). Older TBI patients experience increased morbidity and mortality compared to their younger counterparts. Our prior data demonstrated that by blocking α4 integrin, anti-CD49d antibody (aCD49d Ab) abrogates CD8+ T-cell infiltration into the injured brain, improves survival, and attenuates neurocognitive deficits. Here, we aimed to uncover how aCD49d Ab treatment alters local cellular responses in the aged mouse brain. Consequently, mice incur age-associated toxic cytokine and chemokine responses long-term post-TBI. aCD49d Ab attenuates this response along with a T helper (Th)1/Th17 immunological shift and remediation of overall CD8+ T cell cytotoxicity. Furthermore, aCD49d Ab reduces CD8+ T cells exhibiting higher effector status, leading to reduced clonal expansion in aged, but not young, mouse brains with chronic TBI. Together, aCD49d Ab is a promising therapeutic strategy for treating TBI in the older people.
Collapse
Affiliation(s)
- Zhangying Chen
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Kacie P Ford
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mecca B A R Islam
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hanxiao Wan
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hyebin Han
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Abhirami Ramakrishnan
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ryan J Brown
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Veronica Villanueva
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yidan Wang
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Booker T Davis
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Craig Weiss
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Weiguo Cui
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - David Gate
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Steven J Schwulst
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Sabat M, Carney DW, Hernandez-Torres G, Gibson TS, Balakrishna D, Zou H, Xu R, Chen CH, de Jong R, Dougan DR, Qin L, Bigi-Botterill SV, Chambers A, Miura J, Johnson LK, Ermolieff J, Johns D, Selimkhanov J, Kwok L, DeMent K, Proffitt C, Vu P, Lindsey EA, Ivetac T, Jennings A, Wang H, Manam P, Santos C, Fullenwider C, Manohar R, Flick AC. Design and Discovery of a Potent and Selective Inhibitor of Integrin αvβ1. J Med Chem 2024; 67:10306-10320. [PMID: 38872300 DOI: 10.1021/acs.jmedchem.4c00743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Selective inhibition of the RGD (Arg-Gly-Asp) integrin αvβ1 has been recently identified as an attractive therapeutic approach for the treatment of liver fibrosis given its function, target expression, and safety profile. Our identification of a non-RGD small molecule lead followed by focused, systematic changes to the core structure utilizing a crystal structure, in silico modeling, and a tractable synthetic approach resulted in the identification of a potent small molecule exhibiting a remarkable affinity for αvβ1 relative to several other integrin isoforms measured. Azabenzimidazolone 25 demonstrated antifibrotic efficacy in an in vivo rat liver fibrosis model and represents a tool compound capable of further exploring the biological consequences of selective αvβ1 inhibition.
Collapse
Affiliation(s)
- Mark Sabat
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Daniel W Carney
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Gloria Hernandez-Torres
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Tony S Gibson
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Deepika Balakrishna
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Hua Zou
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Rui Xu
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Chien-Hung Chen
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Ron de Jong
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Douglas R Dougan
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Ling Qin
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Simone V Bigi-Botterill
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Alison Chambers
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Joanne Miura
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Lucas K Johnson
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Jacques Ermolieff
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Deidre Johns
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Jangir Selimkhanov
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Lily Kwok
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Kevin DeMent
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Chris Proffitt
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Phong Vu
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Erick A Lindsey
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Tony Ivetac
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Andy Jennings
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Haixia Wang
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Padma Manam
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Cipriano Santos
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Cody Fullenwider
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Rohan Manohar
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Andrew C Flick
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| |
Collapse
|
3
|
Kwarteng A, Mensah C, Osei‐Poku P. Eosinophil: An innate immune cell with anti-filarial vaccine and biomarker potential. Health Sci Rep 2023; 6:e1320. [PMID: 37283884 PMCID: PMC10240928 DOI: 10.1002/hsr2.1320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/11/2023] [Accepted: 05/24/2023] [Indexed: 06/08/2023] Open
Abstract
Background Filarial infections continue to pose a great challenge in endemic countries. One of the central goals in the fight against human filarial infections is the development of strategies that will lead to the inhibition of microfilariae (mf) transmission. Keeping mf under a certain threshold within endemic populations will stop transmission and eliminate the infection. Method A narrative review was carried out to identify the possibilities and limitations of exploring the use of eosinophil responses as an anti-filarial vaccine, and biomarker for the detection of filarial infections. An extensive literature search was performed in online scientific databases including PubMed Central, PubMed, BioMed Central, with the use of predefined search terms. Results A better understanding of the parasite-host interactions will lead to the development of improved and better treatment or vaccine strategies that could eliminate filariasis as soon as possible. Highlighted in this review is the explorative use of eosinophil-producing CLC/Galectin-10 as a potential biomarker for filarial infections. Also discussed are some genes, and pathways involved in eosinophil recruitments that could be explored for anti-filarial vaccine development. Conclusion In this short communication, we discuss how eosinophil-regulated genes, pathways, and networks could be critical in providing more information on how reliably a front-line immune player could be exploited for anti-filarial vaccine development and early infection biomarker.
Collapse
Affiliation(s)
- Alexander Kwarteng
- Department of Biochemistry and BiotechnologyKwame Nkrumah University of Science and TechnologyKumasiGhana
- Kumasi Centre for Collaborative Research in Tropical MedicineKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Caleb Mensah
- Kumasi Centre for Collaborative Research in Tropical MedicineKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Priscilla Osei‐Poku
- Department of Biochemistry and BiotechnologyKwame Nkrumah University of Science and TechnologyKumasiGhana
- Kumasi Centre for Collaborative Research in Tropical MedicineKwame Nkrumah University of Science and TechnologyKumasiGhana
| |
Collapse
|
4
|
Liu Q, Perez A. Assessing a computational pipeline to identify binding motifs to the α2 β1 integrin. Front Chem 2023; 11:1107400. [PMID: 36860646 PMCID: PMC9968975 DOI: 10.3389/fchem.2023.1107400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/27/2023] [Indexed: 02/16/2023] Open
Abstract
Integrins in the cell surface interact with functional motifs found in the extracellular matrix (ECM) that queue the cell for biological actions such as migration, adhesion, or growth. Multiple fibrous proteins such as collagen or fibronectin compose the ECM. The field of biomechanical engineering often deals with the design of biomaterials compatible with the ECM that will trigger cellular response (e.g., in tissue regeneration). However, there are a relative few number of known integrin binding motifs compared to all the possible peptide epitope sequences available. Computational tools could help identify novel motifs, but have been limited by the challenges in modeling the binding to integrin domains. We revisit a series of traditional and novel computational tools to assess their performance in identifying novel binding motifs for the I-domain of the α2β1 integrin.
Collapse
Affiliation(s)
| | - Alberto Perez
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, FL, United States
| |
Collapse
|
5
|
Corrêa T, Poswar F, Santos-Rebouças CB. Convergent molecular mechanisms underlying cognitive impairment in mucopolysaccharidosis type II. Metab Brain Dis 2022; 37:2089-2102. [PMID: 34797484 DOI: 10.1007/s11011-021-00872-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/02/2021] [Indexed: 11/26/2022]
Abstract
Mucopolysaccharidosis type II (MPS II) is a lysosomal storage disorder caused by pathogenic variants in the iduronate-2-sulfatase gene (IDS), responsible for the degradation of glycosaminoglycans (GAGs) heparan and dermatan sulfate. IDS enzyme deficiency results in the accumulation of GAGs within cells and tissues, including the central nervous system (CNS). The progressive neurological outcome in a representative number of MPSII patients (neuronopathic form) involves cognitive impairment, behavioral difficulties, and regression in developmental milestones. In an attempt to dissect part of the influence of axon guidance instability over the cognitive impairment presentation in MPS II, we used brain expression data, network propagation, and clustering algorithm to prioritize in the human interactome a disease module associated with the MPS II context. We identified new candidate genes and pathways that act in focal adhesion, integrin cell surface, laminin interactions, ECM proteoglycans, cytoskeleton, and phagosome that converge into functional mechanisms involved in early neural circuit formation defects and could indicate clues about cognitive impairment in patients with MPSII. Such molecular changes during neurodevelopment may precede the morphological and clinical evidence, emphasizing the importance of an early diagnosis and directing the development of potential drug leads. Furthermore, our data also support previous hypotheses pointing to shared pathogenic mechanisms in some neurodegenerative diseases.
Collapse
Affiliation(s)
- Thiago Corrêa
- Department of Genetics, Institute of Biosciences, Federal University of Rio Grande Do Sul, Porto Alegre, Brazil.
| | - Fabiano Poswar
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Cíntia B Santos-Rebouças
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
6
|
Banerjee S, Nara R, Chakraborty S, Chowdhury D, Haldar S. Integrin Regulated Autoimmune Disorders: Understanding the Role of Mechanical Force in Autoimmunity. Front Cell Dev Biol 2022; 10:852878. [PMID: 35372360 PMCID: PMC8971850 DOI: 10.3389/fcell.2022.852878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
The pathophysiology of autoimmune disorders is multifactorial, where immune cell migration, adhesion, and lymphocyte activation play crucial roles in its progression. These immune processes are majorly regulated by adhesion molecules at cell–extracellular matrix (ECM) and cell–cell junctions. Integrin, a transmembrane focal adhesion protein, plays an indispensable role in these immune cell mechanisms. Notably, integrin is regulated by mechanical force and exhibit bidirectional force transmission from both the ECM and cytosol, regulating the immune processes. Recently, integrin mechanosensitivity has been reported in different immune cell processes; however, the underlying mechanics of these integrin-mediated mechanical processes in autoimmunity still remains elusive. In this review, we have discussed how integrin-mediated mechanotransduction could be a linchpin factor in the causation and progression of autoimmune disorders. We have provided an insight into how tissue stiffness exhibits a positive correlation with the autoimmune diseases’ prevalence. This provides a plausible connection between mechanical load and autoimmunity. Overall, gaining insight into the role of mechanical force in diverse immune cell processes and their dysregulation during autoimmune disorders will open a new horizon to understand this physiological anomaly.
Collapse
|
7
|
Assessing Blood-Based Biomarkers to Define a Therapeutic Window for Natalizumab. J Pers Med 2021; 11:jpm11121347. [PMID: 34945819 PMCID: PMC8706232 DOI: 10.3390/jpm11121347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 12/21/2022] Open
Abstract
Natalizumab is a monoclonal antibody that binds CD49d. Although it is one of the most effective treatments for Relapsing-Remitting Multiple Sclerosis (RRMS), a dosing regimen has not been optimized for safety and efficacy in individual patients. We aimed to identify biomarkers to monitor Natalizumab treatment and to establish a personalized dose utilizing an ongoing longitudinal study in 29 RRMS patients under Natalizumab with standard interval dose (SD) of 300 mg/4wks or extended interval dose (EID) of 300 mg/6wks. Blood samples were analyzed by flow cytometry to determine CD49d saturation and expression in several T and B lymphocytes subpopulations. Each patient was analyzed at two different timepoints separated by 3 Natalizumab administrations. Natalizumab and sVCAM-1 levels in serum were also analyzed using ELISA. To determine the reproducibility of various markers, two different timepoints were compared and no significant differences were observed for CD49d expression nor for saturation; SD patients had higher saturation levels (~80%) than EID patients (~60%). A positive correlation exists between CD49d saturation and Natalizumab serum levels. CD49d expression and saturation are stable parameters that could be used as biomarkers in the immunomonitoring of Natalizumab treatment. Moreover, Natalizumab and sVCAM-1 serum levels could be used to optimize an individual's dosing schedule.
Collapse
|
8
|
The Spike Glycoprotein of SARS-CoV-2 Binds to β1 Integrins Expressed on the Surface of Lung Epithelial Cells. Viruses 2021; 13:v13040645. [PMID: 33918599 PMCID: PMC8069079 DOI: 10.3390/v13040645] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 01/12/2023] Open
Abstract
The spike glycoprotein attached to the envelope of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) binds to and exploits angiotensin-converting enzyme 2 (ACE2) as an entry receptor to infect pulmonary epithelial cells. A subset of integrins that recognize the arginyl–glycyl–aspartic acid (RGD) sequence in the cognate ligands has been predicted in silico to bind the spike glycoprotein and, thereby, to be exploited for viral infection. Here, we show experimental evidence that the β1 integrins predominantly expressed on human pulmonary epithelial cell lines and primary mouse alveolar epithelial cells bind to this spike protein. The cellular β1 integrins support adhesive interactions with the spike protein independently of ACE2, suggesting the possibility that the β1 integrins may function as an alternative receptor for SARS-CoV-2, which could be targeted for the prevention of viral infections.
Collapse
|
9
|
Dhaiban S, Al-Ani M, Elemam NM, Maghazachi AA. Targeting Chemokines and Chemokine Receptors in Multiple Sclerosis and Experimental Autoimmune Encephalomyelitis. J Inflamm Res 2020; 13:619-633. [PMID: 33061527 PMCID: PMC7532903 DOI: 10.2147/jir.s270872] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated and neurodegenerative disorder that results in inflammation and demyelination of the central nervous system (CNS). MS symptoms include walking difficulties, visual weakening, as well as learning and memory impairment, thus affecting the quality of the patient's life. Chemokines and chemokine receptors are expressed on the immune cells as well as the CNS resident cells. Several sets of chemokine receptors and their ligands tend to be pathogenic players in MS, including CCL2, CCL3, CCL4, CCL5, CCL7, CCL8, CCL11, CCL17, CCL19, CCL21, CCL22, CXCL1, CXCL8, CXCL9, CXCL10, CXCL11, and CXCL16. Furthermore, current modulatory drugs that are used in the treatment of MS and its animal model, the experimental autoimmune encephalomyelitis (EAE), affect the expression of several chemokine and chemokine receptors. In this review, we highlight the pathogenic roles of chemokines and their receptors as well as utilizing them as potential therapeutic targets through selective agents, such as specific antibodies and receptor blockers, or indirectly through MS or EAE immunomodulatory drugs.
Collapse
Affiliation(s)
- Sarah Dhaiban
- College of Medicine and Immuno-Oncology Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Mena Al-Ani
- College of Medicine and Immuno-Oncology Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Noha Mousaad Elemam
- College of Medicine and Immuno-Oncology Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Azzam A Maghazachi
- College of Medicine and Immuno-Oncology Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Crohn's disease management has changed significantly with increasing use of biologics. We review the recent literature on the clinical management of Crohn's disease and new approaches in selecting and optimizing therapy. RECENT FINDINGS Recent studies have addressed the efficacy of proactive anti-TNFα trough level monitoring, the efficacy of biosimilars, and the efficacy and immunogenicity of newer biologics including anti-integrin therapy and anti-IL12/23 therapy. Optimizing anti-TNFα therapy according to trough concentrations correlates with improved remission rates. Patients can be switched from the reference drug to a biosimilar, or vice versa, without a measurable change in efficacy, safety, or immunogenicity. Immunomodulators are effective in decreasing immunogenicity and boosting anti-TNFα drug level. The anti-integrin and anti-IL12/23 therapies are effective as induction and maintenance therapy with low immunogenicity and excellent safety profiles. Patients at high risk for post-operative recurrence should be started on a biologic therapy within 4 weeks post-op. Multiple biologic therapies are currently available for treatment of Crohn's disease including anti-TNFα therapy, anti-integrin therapy, and anti-IL12/23 therapy. The choice of first-line therapy should be based on individual risk-benefit analysis, route of administration, and patient preference. Patient with inadequate response should have their trough level checked and therapy optimized. Therapeutic prophylaxis for post-operative recurrence should be based on patient's risk factors for recurrence.
Collapse
Affiliation(s)
- Thomas X Lu
- Section of Gastroenterology, Hepatology and Nutrition, Inflammatory Bowel Disease Center, University of Chicago Medicine, Chicago, IL, 60637, USA
| | - Russell D Cohen
- Section of Gastroenterology, Hepatology and Nutrition, Inflammatory Bowel Disease Center, University of Chicago Medicine, Chicago, IL, 60637, USA.
| |
Collapse
|
11
|
Chawla R, Nath M, Moksha L, Nag TC, Velpandian T. An experimental study to evaluate safety/toxicity of intravitreal natalizumab. Indian J Ophthalmol 2018; 66:1441-1445. [PMID: 30249830 PMCID: PMC6173036 DOI: 10.4103/ijo.ijo_425_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Purpose: The purpose of this prospective experimental study was to evaluate the safety/toxicity of α4β1 integrin blockade in rabbit retina using its monoclonal antibody (Natalizumab). Methods: Twelve New Zealand albino rabbits were divided into three groups (n = 4). Unilateral intravitreal injections of three different concentrations of natalizumab were performed in every rabbit of each group (Group A: 0.625 mg, Group B: 1.25 mg, and Group C: 2.5 mg). Baseline electroretinogram (ERG) and fundus photography were performed prior to injection. At days 1, 7, and 21 postinjection, ERG and fundus photography of each eye were performed. At last follow-up, Group C animals with highest drug concentration were sacrificed and the enucleated eyes were evaluated for retinal toxicity using transmission electron microscopy (TEM). Results: No difference in ERG responses was observed in eyes injected with low and intermediate concentration of natalizumab between day 0 and day 21. Furthermore, rabbits injected intravitreally with highest dose showed reduction in amplitude of “a” wave (P = 0.0017) and a reduction in amplitude of “b” wave of ERG at day 21 (P = 0.0117). TEM revealed changes in the outer plexiform layer and inner nuclear layer, suggestive of toxicity primarily to the photoreceptor synaptic terminals and bipolar cells. Conclusion: Low-dose (0.625 mg) and intermediate-dose (1.25 mg) intravitreal injection of natalizumab appears safe for rabbit retina. However, functional and anatomical changes were observed in rabbit retina following a high-dose (2.5 mg) intravitreal injection of a monoclonal antibody blocking α4β1 integrin.
Collapse
Affiliation(s)
- Rohan Chawla
- Department of Ophthalmology, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, New Delhi, India
| | - Madhu Nath
- Department of Ocular Pharmacology and Pharmacy, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, New Delhi, India
| | - Laxmi Moksha
- Department of Ocular Pharmacology and Pharmacy, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, New Delhi, India
| | - Tapas C Nag
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Thirumurthy Velpandian
- Department of Ocular Pharmacology and Pharmacy, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, New Delhi, India
| |
Collapse
|
12
|
Maginnis MS. Virus-Receptor Interactions: The Key to Cellular Invasion. J Mol Biol 2018; 430:2590-2611. [PMID: 29924965 PMCID: PMC6083867 DOI: 10.1016/j.jmb.2018.06.024] [Citation(s) in RCA: 196] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 06/04/2018] [Accepted: 06/08/2018] [Indexed: 11/05/2022]
Abstract
Virus–receptor interactions play a key regulatory role in viral host range, tissue tropism, and viral pathogenesis. Viruses utilize elegant strategies to attach to one or multiple receptors, overcome the plasma membrane barrier, enter, and access the necessary host cell machinery. The viral attachment protein can be viewed as the “key” that unlocks host cells by interacting with the “lock”—the receptor—on the cell surface, and these lock-and-key interactions are critical for viruses to successfully invade host cells. Many common themes have emerged in virus–receptor utilization within and across virus families demonstrating that viruses often target particular classes of molecules in order to mediate these events. Common viral receptors include sialylated glycans, cell adhesion molecules such as immunoglobulin superfamily members and integrins, and phosphatidylserine receptors. The redundancy in receptor usage suggests that viruses target particular receptors or “common locks” to take advantage of their cellular function and also suggests evolutionary conservation. Due to the importance of initial virus interactions with host cells in viral pathogenesis and the redundancy in viral receptor usage, exploitation of these strategies would be an attractive target for new antiviral therapeutics. Viral receptors are key regulators of host range, tissue tropism, and viral pathogenesis. Many viruses utilize common viral receptors including sialic acid, cell adhesion molecules such as immunoglobulin superfamily members and integrins, and phosphatidylserine receptors. Detailed molecular interactions between viruses and receptors have been defined through elegant biochemical analyses including glycan array screens, structural–functional analyses, and cell-based approaches providing tremendous insights into these initial events in viral infection. Commonalities in virus–receptor interactions present promising targets for the development of broad-spectrum antiviral therapies.
Collapse
Affiliation(s)
- Melissa S Maginnis
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469-5735, USA.
| |
Collapse
|
13
|
Pandolfi F, Franza L, Altamura S, Mandolini C, Cianci R, Ansari A, Kurnick JT. Integrins: Integrating the Biology and Therapy of Cell-cell Interactions. Clin Ther 2017; 39:2420-2436. [PMID: 29203050 DOI: 10.1016/j.clinthera.2017.11.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/29/2017] [Accepted: 11/07/2017] [Indexed: 12/12/2022]
Abstract
PURPOSE Although the role of integrins has been described in a variety of diseases, these roles seem to be distinct. To date, no study has attempted to provide links to the various pathways by which such integrins can be involved in these diverse disease settings. The purpose of this review was to address this gap in our knowledge with the hypothesis that there is, in fact, a common pathway by which integrins may function. METHODS This article provides an in-depth perspective on the discovery, development, and design of therapeutics that modulate cellular function by targeting integrin:ligand interactions by reviewing the literature on this subject; the review included the most recent results of clinical and subclinical studies. A MEDLINE search was conducted for articles pertaining to the various issues related to integrins, and the most relevant articles are discussed (ie, not only those published in journals with a higher impact factor). FINDINGS It seems that the ligation of the integrins with their cognate ligands plays a major role in translating membrane dialogue into biological function. In addition, they also seem to play a major regulatory role that can enhance or inhibit biological function depending on the context within which such receptor:ligand interactions occur and the organ and tissues at which interactions occurs and is manipulated. Those studies that used statistical analyses have been included where appropriate. IMPLICATIONS Our findings show that anti-integrin treatment has the potential to become a valid coadjuvant in the treatment of several diseases including cancer, inflammatory diseases, HIv infection and cardiovascular diseases.
Collapse
Affiliation(s)
- Franco Pandolfi
- Institute of Internal Medicine, Catholic University, Rome, Italy.
| | - Laura Franza
- Institute of Internal Medicine, Catholic University, Rome, Italy
| | - Simona Altamura
- Institute of Internal Medicine, Catholic University, Rome, Italy
| | | | - Rossella Cianci
- Institute of Internal Medicine, Catholic University, Rome, Italy
| | - Aftab Ansari
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - James T Kurnick
- CytoCure LLC, Beverly, Massachusetts; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
14
|
JC Polyomavirus Attachment and Entry: Potential Sites for PML Therapeutics. CURRENT CLINICAL MICROBIOLOGY REPORTS 2017; 4:132-141. [PMID: 28989857 DOI: 10.1007/s40588-017-0069-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW JC polyomavirus (JCPyV) is a significant human pathogen that causes an asymptomatic infection in the kidney in the majority of the population. In immunosuppressed individuals, the virus can become reactivated and spread to the brain, causing the fatal, demyelinating disease progressive multifocal leukoencephalopathy (PML). There are currently limited treatment options for this fatal disease. Attachment to receptors and entry into host cells are the initiating events in JCPyV infection and therefore an attractive target for therapeutics to prevent or treat PML. This review provides the current understanding of JCPyV attachment and entry events and the potential therapeutics to target these areas. RECENT FINDINGS JCPyV attachment and entry to host cells is mediated by α2,6-linked lactoseries tetrasaccharide c (LSTc) and 5-hydroxytryptamine receptors (5-HT2Rs), respectively, and subsequent trafficking to the endoplasmic reticulum is required for infection. Recently, vaccines, monoclonal antibodies, and small molecules have shown promise as anti-viral and PML therapies. SUMMARY This review summarizes our current understanding of JCPyV attachment, entry, and trafficking and the development of potential PML therapeutics that inhibit these critical steps in JCPyV infection.
Collapse
|
15
|
Darzi L, Boshtam M, Shariati L, Kouhpayeh S, Gheibi A, Mirian M, Rahimmanesh I, Khanahmad H, Tabatabaiefar MA. The silencing effect of miR-30a on ITGA4 gene expression in vitro: an approach for gene therapy. Res Pharm Sci 2017; 12:456-464. [PMID: 29204174 PMCID: PMC5691572 DOI: 10.4103/1735-5362.217426] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Integrins are adhesion molecules which play crucial roles in cell-cell and cell-extracellular matrix interactions. Very late antigen-4 or α4β1 and lymphocyte Peyer’s patch adhesion molecule-1 or α4β7, are key factors in the invasion of tumor cells and metastasis. Based on the previous reports, integrin α4 (ITGA4) is overexpressed in some immune disorders and cancers. Thus, inhibition of ITGA4 could be a therapeutic strategy. In the present study, miR-30a was selected in order to suppress ITGA4 expression. The ITGA4 3' UTR was amplified, cloned in the Z2827-M67-(ITGA4) plasmid and named as Z2827-M67/3'UTR. HeLa cells were divided into five groups; (1) untreated without any transfection, (2) mock with Z2827-M67/3'UTR transfection and X-tremeGENE reagent, (3) negative control with Z2827-M67/3'UTR transfection alone, (4) test with miR-30a mimic and Z2827-M67/3'UTR transfection and (5) scramble with miR-30a scramble and Z2827-M67/3'UTR transfection. The MTT assay was performed to evaluate cell survival and cytotoxicity in each group. Real-time RT-PCR was applied for the ITGA4 expression analysis. The findings of this study showed that miR-30a downregulated ITGA4 expression and had no effect on the cell survival. Due to the silencing effect of miR-30a on the ITGA4 gene expression, this agent could be considered as a potential tool for cancer and immune disorders therapy.
Collapse
Affiliation(s)
- Leila Darzi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Laleh Shariati
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Shirin Kouhpayeh
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Azam Gheibi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, I.R. Iran
| | - Mina Mirian
- Department of Pharmaceutical Biotechnology and Isfahan Pharmaceutical Science Research Center, School of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Ilnaz Rahimmanesh
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran.,Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Noncommunicable Disease, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Mohammad Amin Tabatabaiefar
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran.,Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Noncommunicable Disease, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
16
|
Anjos L, Morgado I, Guerreiro M, Cardoso JCR, Melo EP, Power DM. Cartilage acidic protein 1, a new member of the beta-propeller protein family with amyloid propensity. Proteins 2016; 85:242-255. [DOI: 10.1002/prot.25210] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/06/2016] [Accepted: 11/09/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Liliana Anjos
- Comparative Endocrinology and Integrative Biology Group (CEIB), Centro de Ciencias do Mar (CCMAR), University of Algarve; Campus de Gambelas Faro 8005-139 Portugal
| | - Isabel Morgado
- Comparative Endocrinology and Integrative Biology Group (CEIB), Centro de Ciencias do Mar (CCMAR), University of Algarve; Campus de Gambelas Faro 8005-139 Portugal
| | - Marta Guerreiro
- Comparative Endocrinology and Integrative Biology Group (CEIB), Centro de Ciencias do Mar (CCMAR), University of Algarve; Campus de Gambelas Faro 8005-139 Portugal
| | - João C. R. Cardoso
- Comparative Endocrinology and Integrative Biology Group (CEIB), Centro de Ciencias do Mar (CCMAR), University of Algarve; Campus de Gambelas Faro 8005-139 Portugal
| | - Eduardo P. Melo
- Campus de Gambelas, Center for Biomedical Research, University of Algarve; Faro 8005-139 Portugal
| | - Deborah M. Power
- Comparative Endocrinology and Integrative Biology Group (CEIB), Centro de Ciencias do Mar (CCMAR), University of Algarve; Campus de Gambelas Faro 8005-139 Portugal
| |
Collapse
|
17
|
Naderi Beni S, Kouhpayeh S, Hejazi Z, Heidari Hafshejani N, Khanahmad H. Construction and Characterization of Recombinant HEK Cell Over Expressing α4 Integrin. Adv Pharm Bull 2015; 5:429-34. [PMID: 26504766 DOI: 10.15171/apb.2015.058] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 05/13/2015] [Accepted: 05/14/2015] [Indexed: 11/09/2022] Open
Abstract
PURPOSE Integrins are heterodimeric membrane proteins, which are exposed to post translational modifications in eukaryotic cells in contrast to prokaryotic cells. These modifications provide advantages for production of proper nanobody, mono and polyclonal antibody against this surface protein and also in aptamer selection process. Since the majority of diagnostic and therapeutic antibodies, target the surface epitopes, eukaryotic membrane proteins provide an appropriate model for further investigation on therapeutic agents. METHODS Escherichia coli strain top 10, was used as host for ITGA-4 expression vector encoding the human integrin α4. The plasmid was extracted and consequently, ITGA-4 vector was digested to make a linear plasmid. Human Embryonic Kidney-293 (HEK-293) cell transfected with linear plasmid and subsequently screened for stable ITGA-4 expressing Cells. Three separated clones were isolated twenty one days after transfection. Chromosomal DNA was extracted from ITGA-4-transfected cells. The presence of ITGA-4 gene in HEK-293 genome was confirmed by PCR. The expression level of ITGA-4 on HEK-293 cells was also analyzed by Flow cytometry. RESULTS Flow cytometric analysis showed that HEK-293 cells have no expression of integrin α4 on their surface while 95% of transfected HEK-293 cells with ITGA4, expressed different levels of integrin α4 on their surfaces which correlates well with genomic DNA PCR amplification results. CONCLUSION The results suggest that we have successfully constructed the integrin α4 expressing HEK293 cell, which will facilitate further research into the production of antibody, nanobody and aptamer against α4 integrin.
Collapse
Affiliation(s)
- Shamsi Naderi Beni
- Isfahan University of Medical Sciences, School of Medicine, Department of Genetic and Molecular Biology, Isfahan, Iran
| | - Shirin Kouhpayeh
- Isfahan University of Medical Sciences, School of Medicine, Department of Immunology, Isfahan, Iran
| | - Zahra Hejazi
- Isfahan University of Medical Sciences, School of Medicine, Department of Genetic and Molecular Biology, Isfahan, Iran
| | - Nahid Heidari Hafshejani
- Isfahan University of Medical Sciences, School of Medicine, Department of Genetic and Molecular Biology, Isfahan, Iran
| | - Hossein Khanahmad
- Isfahan University of Medical Sciences, School of Medicine, Department of Genetic and Molecular Biology, Isfahan, Iran
| |
Collapse
|
18
|
Vergara D, D'Alessandro M, Rizzello A, De Riccardis L, Lunetti P, Del Boccio P, De Robertis F, Trianni G, Maffia M, Giudetti AM. A lipidomic approach to the study of human CD4(+) T lymphocytes in multiple sclerosis. BMC Neurosci 2015. [PMID: 26205308 PMCID: PMC4513631 DOI: 10.1186/s12868-015-0183-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Background Lipids play different important roles in central nervous system so that dysregulation of lipid pathways has been implicated in a growing
number of neurodegenerative disorders including multiple sclerosis (MS). MS is the most prevalent autoimmune disorder of the central nervous system, with neurological symptoms caused by inflammation and demyelination. In this study, a lipidomic analysis was performed for the rapid profile of CD4+ T lymphocytes from MS patient and control samples in an untargeted approach. Methods A matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF) mass spectrometry based approach was used for the analysis of lipid extracts using 9-aminoacridine as matrix. Lipids were analyzed in negative mode and selected species fragmented using MALDI tandem mass spectrometry for their structural assignments. Results The analysis reveals some modifications in the phospholipid pattern of MS CD4+ T lymphocytes with respect to healthy controls with a significant increase of cardiolipin species in MS samples. Conclusions These results demonstrate the feasibility of a MALDI-TOF approach for the analysis of CD4+ lipid extracts and suggest how alterations in the lipid metabolism characterized lymphocytes of MS patients.
Collapse
Affiliation(s)
- Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, Lecce, Italy. .,Laboratory of Clinical Proteomic, "Giovanni Paolo II" Hospital, ASL-Lecce, Piazzetta F. Muratore, Lecce, Italy.
| | - Michele D'Alessandro
- Department of Medical, Oral and Biotechnological Sciences, Research Centre on Aging (Ce.S.I), "G. d'Annunzio" University Foundation, Chieti-Pescara, Italy.
| | - Antonia Rizzello
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, Lecce, Italy. .,Laboratory of Clinical Proteomic, "Giovanni Paolo II" Hospital, ASL-Lecce, Piazzetta F. Muratore, Lecce, Italy.
| | - Lidia De Riccardis
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, Lecce, Italy.
| | - Paola Lunetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, Lecce, Italy.
| | - Piero Del Boccio
- Department of Medical, Oral and Biotechnological Sciences, Research Centre on Aging (Ce.S.I), "G. d'Annunzio" University Foundation, Chieti-Pescara, Italy.
| | - Francesca De Robertis
- Department of Neurology, "Vito Fazzi" Hospital, ASL-Lecce, Piazzetta F. Muratore, Lecce, Italy.
| | - Giorgio Trianni
- Department of Neurology, "Vito Fazzi" Hospital, ASL-Lecce, Piazzetta F. Muratore, Lecce, Italy.
| | - Michele Maffia
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, Lecce, Italy. .,Laboratory of Clinical Proteomic, "Giovanni Paolo II" Hospital, ASL-Lecce, Piazzetta F. Muratore, Lecce, Italy.
| | - Anna M Giudetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, Lecce, Italy.
| |
Collapse
|
19
|
Progressive multifocal leukoencephalopathy-associated mutations in the JC polyomavirus capsid disrupt lactoseries tetrasaccharide c binding. mBio 2013; 4:e00247-13. [PMID: 23760462 PMCID: PMC3685208 DOI: 10.1128/mbio.00247-13] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The human JC polyomavirus (JCPyV) is the causative agent of the fatal, demyelinating disease progressive multifocal leukoencephalopathy (PML). The Mad-1 prototype strain of JCPyV uses the glycan lactoseries tetrasaccharide c (LSTc) and serotonin receptor 5-HT2A to attach to and enter into host cells, respectively. Specific residues in the viral capsid protein VP1 are responsible for direct interactions with the α2,6-linked sialic acid of LSTc. Viral isolates from individuals with PML often contain mutations in the sialic acid-binding pocket of VP1 that are hypothesized to arise from positive selection. We reconstituted these mutations in the Mad-1 strain of JCPyV and found that they were not capable of growth. The mutations were then introduced into recombinant VP1 and reconstituted as pentamers in order to conduct binding studies and structural analyses. VP1 pentamers carrying PML-associated mutations were not capable of binding to permissive cells. High-resolution structure determination revealed that these pentamers are well folded but no longer bind to LSTc due to steric clashes in the sialic acid-binding site. Reconstitution of the mutations into JCPyV pseudoviruses allowed us to directly quantify the infectivity of the mutants in several cell lines. The JCPyV pseudoviruses with PML-associated mutations were not infectious, nor were they able to engage sialic acid as measured by hemagglutination of human red blood cells. These results demonstrate that viruses from PML patients with single point mutations in VP1 disrupt binding to sialic acid motifs and render these viruses noninfectious. Infection with human JC polyomavirus (JCPyV) is common and asymptomatic in healthy individuals, but during immunosuppression, JCPyV can spread from the kidney to the central nervous system (CNS) and cause a fatal, demyelinating disease, progressive multifocal leukoencephalopathy (PML). Individuals infected with HIV, those who have AIDS, or those receiving immunomodulatory therapies for autoimmune diseases are at serious risk for PML. Recent reports have demonstrated that viral isolates from PML patients often have distinct changes within the major capsid protein. Our structural-functional approach highlights that these mutations result in abolished engagement of the carbohydrate receptor motif LSTc that is necessary for infection. Viruses with PML-associated mutations are not infectious in glial cells, suggesting that they may play an alternative role in PML pathogenesis.
Collapse
|
20
|
Abstract
Respiratory syncytial virus (RSV) is the major cause of respiratory illness in infants worldwide. Neurologic alterations, such as seizures and ataxia, have been associated with RSV infection. We demonstrate the presence of RSV proteins and RNA in zones of the brain--such as the hippocampus, ventromedial hypothalamic nucleus, and brainstem--of infected mice. One month after disease resolution, rodents showed behavioral and cognitive impairment in marble burying (MB) and Morris water maze (MWM) tests. Our data indicate that the learning impairment caused by RSV is a result of a deficient induction of long-term potentiation in the hippocampus of infected animals. In addition, immunization with recombinant bacillus Calmette-Guérin (BCG) expressing RSV nucleoprotein prevented behavioral disorders, corroborating the specific effect of RSV infection over the central nervous system. Our findings provide evidence that RSV can spread from the airways to the central nervous system and cause functional alterations to the brain, both of which can be prevented by proper immunization against RSV.
Collapse
|
21
|
PI3-kinase γ promotes Rap1a-mediated activation of myeloid cell integrin α4β1, leading to tumor inflammation and growth. PLoS One 2013; 8:e60226. [PMID: 23565202 PMCID: PMC3614555 DOI: 10.1371/journal.pone.0060226] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 02/23/2013] [Indexed: 12/24/2022] Open
Abstract
Tumor inflammation, the recruitment of myeloid lineage cells into the tumor microenvironment, promotes angiogenesis, immunosuppression and metastasis. CD11b+Gr1lo monocytic lineage cells and CD11b+Gr1hi granulocytic lineage cells are recruited from the circulation by tumor-derived chemoattractants, which stimulate PI3-kinase γ (PI3Kγ)-mediated integrin α4 activation and extravasation. We show here that PI3Kγ activates PLCγ, leading to RasGrp/CalDAG-GEF-I&II mediated, Rap1a-dependent activation of integrin α4β1, extravasation of monocytes and granulocytes, and inflammation-associated tumor progression. Genetic depletion of PLCγ, CalDAG-GEFI or II, Rap1a, or the Rap1 effector RIAM was sufficient to prevent integrin α4 activation by chemoattractants or activated PI3Kγ (p110γCAAX), while activated Rap (RapV12) promoted constitutive integrin activation and cell adhesion that could only be blocked by inhibition of RIAM or integrin α4β1. Similar to blockade of PI3Kγ or integrin α4β1, blockade of Rap1a suppressed both the recruitment of monocytes and granulocytes to tumors and tumor progression. These results demonstrate critical roles for a PI3Kγ-Rap1a-dependent pathway in integrin activation during tumor inflammation and suggest novel avenues for cancer therapy.
Collapse
|