1
|
Rijal R, Gomer RH. Pharmacological inhibition of host pathways enhances macrophage killing of intracellular bacterial pathogens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.06.647500. [PMID: 40291742 PMCID: PMC12026824 DOI: 10.1101/2025.04.06.647500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
After ingestion into macrophage phagosomes, some bacterial pathogens such as Mycobacterium tuberculosis ( Mtb ) evade killing by preventing phagosome acidification and fusion of the phagosome with a lysosome. Mtb accumulates extracellular polyphosphate (polyP), and polyP inhibits macrophage phagosome acidification and bacterial killing. In Dictyostelium discoideum , polyP also inhibits bacterial killing, and we identified some proteins in D. discoideum that polyP requires to suppress the killing of ingested bacteria. Here, we find that pharmacological inhibition of human orthologues of the D. discoideum proteins, including P2Y1 receptors, mammalian Target of Rapamycin (mTOR), and inositol hexakisphosphate kinase, enhances the killing of Mtb , Legionella pneumophila , and Listeria monocytogenes by human macrophages. Mtb inhibits phagosome acidification, expression of the proinflammatory marker CD54, and autophagy, and increases expression of the anti-inflammatory marker CD206. In Mtb -infected macrophages, the polyP-degrading enzyme polyphosphatase (ScPPX) and inhibitors reversed these effects, with ScPPX increasing CD54 expression more in female macrophages compared to male macrophages. In addition, Mtb inhibits proteasome activity, and some, but not all, inhibitors reversed these effects. While the existence of a dedicated polyP signaling pathway remains uncertain, our findings suggest that pharmacological inhibition of select host proteins can restore macrophage function and enhances the killing of intracellular pathogens. Importance Human macrophages engulf bacteria into phagosomes, which then fuse with lysosomes to kill the bacteria. However, after engulfment, pathogenic bacteria such as Mycobacterium tuberculosis , Legionella pneumophila , and Listeria monocytogenes can block phagosome-lysosome fusion, allowing their survival. Here, we show that pharmacological inhibition of specific macrophage proteins reverses these effects and enhances bacterial killing. These findings suggest that targeting host factors involved in these processes may provide a therapeutic strategy to improve macrophage function against infections such as tuberculosis, Legionnaires' disease, and listeriosis.
Collapse
|
2
|
Richter R, Sari E, Stacks D, Lewis C, Smith J, Xu N, Langster T, Finley D, Margaroli C, Genschmer KR. Protocol for the isolation of human neutrophil-derived extracellular vesicles for characterization and functional experimentation. STAR Protoc 2025; 6:103739. [PMID: 40186860 PMCID: PMC12002981 DOI: 10.1016/j.xpro.2025.103739] [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: 01/14/2025] [Revised: 02/21/2025] [Accepted: 03/12/2025] [Indexed: 04/07/2025] Open
Abstract
Small extracellular vesicles (EVs) derived from neutrophils play an important role in the remodeling of extracellular matrices in human health and disease. Here, we present a protocol for isolating EVs secreted from human neutrophils either ex vivo or within circulation. We describe steps for neutrophil and plasma isolation, neutrophil activation, and EV isolation and enumeration. We then detail procedures for isolating CD66b+ EVs using magnetic bead pull-down and subsequent characterization by flow cytometry, western blot, and neutrophil elastase activity assays. For complete information on the generation and use of this protocol, please refer to Genschmer et al.1.
Collapse
Affiliation(s)
- Robert Richter
- Department of Pediatrics, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA.
| | - Ezgi Sari
- University of Alabama at Birmingham Graduate School, Birmingham, AL 35233, USA; Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Delores Stacks
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Crystal Lewis
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Julian Smith
- Department of Medicine, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Ningyong Xu
- Department of Medicine, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Tamiajoi Langster
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA; Department of Medicine, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Dakota Finley
- Department of Medicine, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Camilla Margaroli
- University of Alabama at Birmingham Graduate School, Birmingham, AL 35233, USA
| | - Kristopher R Genschmer
- Department of Medicine, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA.
| |
Collapse
|
3
|
Alesawy A, Alotaibi N, Alalshaikh M, Aljofi FE, Aldossary N, Al-Zahrani N, Omar O, Madi M. Impact of phenytoin and valproic acid on cytotoxicity and inflammatory mediators in human mononuclear cells: with and without lipopolysaccharide stimulation. PeerJ 2025; 13:e19102. [PMID: 40115275 PMCID: PMC11925041 DOI: 10.7717/peerj.19102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/12/2025] [Indexed: 03/23/2025] Open
Abstract
Background Valproic acid (VPA) is known for its broad-spectrum antiepileptic effects and is recommended for generalized epilepsy, in contrast to phenytoin, which has a more limited spectrum. This study investigated the cytotoxic and inflammatory responses to phenytoin and VPA in peripheral blood mononuclear cells (PBMCs), with and without bacterial lipopolysaccharide (LPS) stimulation. Methods PBMCs from healthy donors were divided into 12 groups: control (Ctrl), phenytoin (Phy), and four concentrations of VPA (Val-50, Val-75, Val-100, Val-200), with and without LPS. Assessments were conducted on days 1 and 3, including total, live, and dead cell counts, cell viability, and lactic acid dehydrogenase (LDH) cytotoxicity assays. Inflammatory mediators (IL-6, IL-1β) and immune markers (IL-18, IgA) were measured using enzyme-linked immunosorbent assay (ELISA) on day 3. Statistical analysis involved two-way ANOVA, Tukey's HSD tests, and paired t-tests. Results All treatment groups showed significant declines in cell counts and viability from day 1 to day 3, which were exacerbated by LPS. Val-50 + LPS maintained higher cell counts compared to Ctrl + LPS and Phy + LPS. Elevated LDH levels were primarily observed in the Val-100 and Val-200 groups, with and without LPS. In the absence of LPS, the Val-75 and Val-100 groups showed notable reductions in IL-18 and IgA levels, while all VPA treatments reduced IL-6 levels compared to controls. This effect was enhanced under LPS exposure, although IL-1β reductions in the Val-75, Val-100, and Val-200 groups were reversed in the presence of LPS. Val-75 demonstrated lower cytotoxic and inflammatory responses compared to Phy and higher VPA doses, showing moderate LDH increases and reduced IL-18, IgA, IL-1β, and IL-6 levels, particularly under LPS challenge. Conclusion Phenytoin and VPA induced significant cytotoxic and inflammatory responses, influenced by dosage and LPS exposure. Val-75 exhibited a dose-specific immunomodulatory effect, reducing both pro-inflammatory and immune markers.
Collapse
Affiliation(s)
- Aminah Alesawy
- Department of Preventive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Norah Alotaibi
- Department of Preventive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Marwa Alalshaikh
- Department of Preventive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Faisal E. Aljofi
- Department of Preventive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Nada Aldossary
- Department of Pathology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Nada Al-Zahrani
- Blood Bank, Laboratory Medicine, King Fahad University Hospital, Al Khobar, Saudi Arabia
| | - Omar Omar
- Department of Biomedical Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Marwa Madi
- Department of Preventive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
4
|
Taccardi D, Gowdy HGM, Li VW, Wing AC, Baharnoori M, Finlayson M, Ghasemlou N. Circadian rhythmicity of symptomatic phenotypes in multiple sclerosis: the CircaMS study protocol and feasibility of biomarker collection. BMJ Open 2025; 15:e095357. [PMID: 40044201 PMCID: PMC11883553 DOI: 10.1136/bmjopen-2024-095357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/12/2025] [Indexed: 03/09/2025] Open
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic autoimmune neurological disease with a variable prognosis and unpredictable course. Fatigue, pain and low mood are common symptoms that tend to fluctuate in people with MS (pwMS). Disrupted circadian rhythms may have a role in the symptoms' variability. Distinguishing interindividual differences and temporal daily fluctuations in MS symptoms may help to define specific symptomatic phenotypes. Understanding how these phenotypes are associated with quality of life and their immunological underpinnings-immune profiles-could shape new MS management strategies. Our primary aim is to document ongoing fluctuations in fatigue, pain and mood in a cohort of pwMS to determine whether symptom variability is associated with differential quality of life. Our secondary aim is to evaluate the feasibility of our study design to identify immune profiles of circadian rhythmicity in MS. METHODS AND ANALYSIS This observational cohort study examines individual temporal fluctuations in MS symptomatology via ecological momentary assessment in a cohort of pwMS. All participants complete (1) a baseline battery of questionnaires and (2) electronic symptom-tracking diaries to rate fatigue, pain intensity and mood on a 0-10 scale at three time points (08:00, 14:00 and 20:00) for 10 days. Participants will be grouped into symptomatic phenotypes based on longitudinal data from e-diaries. We will assess whether exhibiting a specific phenotype is associated with certain baseline measures. A subgroup of 20 participants-feasibility study-will also complete blood sample collection two times within 24 hours to study immune profiles and molecular markers of circadian rhythmicity in MS. Flow cytometry, whole blood RNA sequencing and plasma analyses will be applied to determine changes in immune profiles indicative of circadian rhythmicity.This work has the potential to reduce the burden of this complex disease on a global scale. Future studies will build on our work to understand individual variability in MS symptomatology, including disease severity; identification of biomarkers underlying the association between rhythmic symptomatology profiles and symptomatic phenotypes in MS; and designing personalised interventions focused on interindividual differences in symptomatology and circadian rhythmicity. ETHICS AND DISSEMINATION The CircaMS project and its associated procedures have been reviewed and approved by the Queen's University Health Sciences and Affiliated Teaching Hospitals research ethics board (File number: 6039383). Participants provide informed consent to participate, and their data will not be identifiable in any publication or report. All documents are stored securely and only accessible by study staff and authorised personnel. The results will be presented to academic and lay audiences via national/international conferences, publications in peer-reviewed journals, social media and through an official website created to engage pwMS, caregivers, clinicians and researchers.
Collapse
Affiliation(s)
- Doriana Taccardi
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Hailey G M Gowdy
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Vina Wenyu Li
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Ana Cristina Wing
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
- Neurology, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - Moogeh Baharnoori
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
- Neurology, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - Marcia Finlayson
- School of Rehabilitation Therapy, Queen's University, Kingston, Ontario, Canada
| | - Nader Ghasemlou
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, Ontario, Canada
- Department of Anesthesiology, Queen's University, Kingston, Ontario, Canada
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
5
|
Kwon J, Kawase H, Mattonet K, Guenther S, Hahnefeld L, Shamsara J, Heering J, Kurz M, Kirchhofer S, Krasel C, Ulrich M, Persechino M, Murthy S, Orlandi C, Sadik CD, Geisslinger G, Bünemann M, Kolb P, Offermanns S, Wettschureck N. Orphan G protein-coupled receptor GPRC5B controls macrophage function by facilitating prostaglandin E receptor 2 signaling. Nat Commun 2025; 16:1448. [PMID: 39920161 PMCID: PMC11805951 DOI: 10.1038/s41467-025-56713-0] [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: 04/19/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
Macrophages express numerous G protein-coupled receptors (GPCRs) that regulate adhesion, migration, and activation, but the function of orphan receptor GPRC5B in macrophages is unknown. Both resident peritoneal and bone marrow-derived macrophages from myeloid-specific GPRC5B-deficient mice show increased migration and phagocytosis, resulting in improved bacterial clearance in a peritonitis model. In other models such as myocardial infarction, increased myeloid cell recruitment has adverse effects. Mechanistically, we found that GPRC5B physically interacts with GPCRs of the prostanoid receptor family, resulting in enhanced signaling through the prostaglandin E receptor 2 (EP2). In GPRC5B-deficient macrophages, EP2-mediated anti-inflammatory effects are diminished, resulting in hyperactivity. Using in silico modelling and docking, we identify residues potentially mediating GPRC5B/EP2 dimerization and show that their mutation results in loss of GPRC5B-mediated facilitation of EP2 signaling. Finally, we demonstrate that decoy peptides mimicking the interacting sequence are able to reduce GPRC5B-mediated facilitation of EP2-induced cAMP signaling in macrophages.
Collapse
Affiliation(s)
- Jeonghyeon Kwon
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Haruya Kawase
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Kenny Mattonet
- Imaging Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Guenther
- Deep sequencing platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Lisa Hahnefeld
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, Institute of Clinical Pharmacology, Frankfurt am Main, Germany
| | - Jamal Shamsara
- Department of Pharmaceutical Chemistry, University of Marburg, Marburg, Germany
| | - Jan Heering
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Michael Kurz
- Department of Pharmacology and Clinical Pharmacy, University of Marburg, Marburg, Germany
| | - Sina Kirchhofer
- Department of Pharmacology and Clinical Pharmacy, University of Marburg, Marburg, Germany
| | - Cornelius Krasel
- Department of Pharmacology and Clinical Pharmacy, University of Marburg, Marburg, Germany
| | - Michaela Ulrich
- Department of Pharmacology and Clinical Pharmacy, University of Marburg, Marburg, Germany
| | | | - Sripriya Murthy
- Department of Dermatology, Allergy, and Venereology, University of Lübeck, Lübeck, Germany
| | - Cesare Orlandi
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Christian D Sadik
- Department of Dermatology, Allergy, and Venereology, University of Lübeck, Lübeck, Germany
| | - Gerd Geisslinger
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, Institute of Clinical Pharmacology, Frankfurt am Main, Germany
| | - Moritz Bünemann
- Department of Pharmacology and Clinical Pharmacy, University of Marburg, Marburg, Germany
| | - Peter Kolb
- Department of Pharmaceutical Chemistry, University of Marburg, Marburg, Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Centre for Molecular Medicine, Medical Faculty, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
- Centre for Molecular Medicine, Medical Faculty, Goethe-University Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
6
|
Hoshi K, Hashim Y, Togo S, Saiwaki S, Motomura H, Sumiyoshi I, Nakazawa S, Ochi Y, Miyoshi C, Heo R, Tabe Y, Abe K, Urata Y, Takahashi K. Design of buffer property for the new enrichment method of circulating tumor cell based on immunomagnetic-negative separation. Comput Struct Biotechnol J 2024; 25:281-289. [PMID: 39720309 PMCID: PMC11667567 DOI: 10.1016/j.csbj.2024.11.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 12/26/2024] Open
Abstract
Metastasis is a significant contributor to cancer-related mortality and a critical issue in cancer. Monitoring the changes in circulating tumor cells (CTCs) with metastatic potential is a valuable prognostic and predictive biomarker. CTCs are a rare population in the peripheral blood of patients with cancer. The enrichment process is extremely important for the isolation of clinically significant CTC subpopulations, which can then be used for further analysis. The present study postulates that the buffer serves as an essential field for immunomagnetic separation, thereby enhancing the efficacy of CTC enrichment in peripheral blood. This, in turn, facilitates CTC detection. Here, we describe the design of buffers for developing a novel immunomagnetic-negative separation method for CTC enrichment. During the design process, the buffer properties of the floating and cell coatings had a synergistic effect on the efficiency of cell enrichment in blood samples. The efficacy of the method was evaluated using peripheral blood samples from patients with non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). The developed method enriched clinically relevant CTC subpopulations that expressed the epithelial-mesenchymal transition (EMT)-related molecule vimentin and/or the cancer immune checkpoint marker programmed death ligand 1 (PD-L1). Furthermore, it was applicable as a part of the enrichment process in a TelomeScan® (OBP-401)-based CTC detection assay with high sensitivity and specificity. From the perspective of methodological approaches, the design of buffer properties can be useful for developing a highly versatile enrichment method for handling CTC heterogeneity.
Collapse
Affiliation(s)
- Kazuaki Hoshi
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yasinjan Hashim
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Leading Center for the Development and Research of Cancer Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Shinsaku Togo
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Shoko Saiwaki
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Hiroaki Motomura
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Issei Sumiyoshi
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Shun Nakazawa
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yusuke Ochi
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Chieko Miyoshi
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Rihyang Heo
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yoko Tabe
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Kanae Abe
- Oncolys BioPharma, Inc., 4-1-28 Toranomon, Minato-ku, Tokyo 105-0001, Japan
| | - Yasuo Urata
- Oncolys BioPharma, Inc., 4-1-28 Toranomon, Minato-ku, Tokyo 105-0001, Japan
| | - Kazuhisa Takahashi
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
7
|
Rajput S, Gautam D, Vats A, Roshan M, Goyal P, Rana C, S M P, Ludri A, De S. Aquaporin (AQP) gene family in Buffalo and Goat: Molecular characterization and their expression analysis. Int J Biol Macromol 2024; 280:136145. [PMID: 39353522 DOI: 10.1016/j.ijbiomac.2024.136145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Aquaporins (AQPs) are essential membrane proteins facilitating water and small solute transport across cell membranes. Mammals have approximately 13 paralogs of AQPs that may have evolved through gene duplication events. These genes are present in two separate clusters within the genome. In the present study, comprehensive 13 AQP genes (AQP0-12) were cloned and characterized in buffalo and goat. The protein coding region of AQPs in both species ranged from 729 to 990 bps, corresponding to 263-330 amino acid residues. Two important residues including NPA motifs and ar/R selectivity filter were found conserved in all AQPs, except for AQP7, 11 and 12. AQP0, 2, 4, 5, 7, 9, 12 showed tissue-restricted expression, whereas AQP1, 3, 8, and 11 exhibited ubiquitous expression across several tissues. AQP10 was identified as a pseudogene in all artiodactyls. Transcript variants were identified in buffalo and goat, where some variants of goat AQP5 and 6 lacked important motifs. Evolutionary analysis indicated positive selection at or near the NPA motifs and ar/R selectivity filter of AQP0, 3, 6, 7, and 10 that may alter its structure and function. This study is crucial for future investigations aiming to study the molecular mechanisms of AQPs in response to various physiological conditions.
Collapse
Affiliation(s)
- Shiveeli Rajput
- ICAR-National Dairy Research Institute (NDRI), Animal Biotechnology Division, Animal Genomics Lab, Karnal 132001, Haryana, India
| | - Devika Gautam
- ICAR-National Dairy Research Institute (NDRI), Animal Biotechnology Division, Animal Genomics Lab, Karnal 132001, Haryana, India
| | - Ashutosh Vats
- ICAR-National Dairy Research Institute (NDRI), Animal Biotechnology Division, Animal Genomics Lab, Karnal 132001, Haryana, India
| | - Mayank Roshan
- ICAR-National Dairy Research Institute (NDRI), Animal Biotechnology Division, Animal Genomics Lab, Karnal 132001, Haryana, India
| | - Priyanka Goyal
- Animal Biochemistry Division, ICAR-National Dairy Research Institute (NDRI), Karnal 132001, Haryana, India
| | - Chanchal Rana
- ICAR-National Dairy Research Institute (NDRI), Animal Biotechnology Division, Animal Genomics Lab, Karnal 132001, Haryana, India
| | - Payal S M
- Animal Biochemistry Division, ICAR-National Dairy Research Institute (NDRI), Karnal 132001, Haryana, India
| | - Ashutosh Ludri
- Department of Physiology, ICAR-National Dairy Research Institute (NDRI), Karnal 132001, Haryana, India
| | - Sachinandan De
- ICAR-National Dairy Research Institute (NDRI), Animal Biotechnology Division, Animal Genomics Lab, Karnal 132001, Haryana, India.
| |
Collapse
|
8
|
Ghalwash AA, El-Gohary RM, El Amrousy D, Morad LM, Kassem SS, Hegab II, Okasha AH. The gut microbiota metabolite trimethylamine-N-oxide in children with β-thalassemia: potential implication for iron-induced renal tubular dysfunction. Pediatr Res 2024:10.1038/s41390-024-03639-w. [PMID: 39448817 DOI: 10.1038/s41390-024-03639-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 09/22/2024] [Accepted: 09/29/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Renal tubular dysfunction is common in transfusion-dependent β thalassemia (β-TM). Iron overload, chronic anemia, and hypoxia are precipitating factors for renal insult. However, gut microbiota engagement in the renal insult has not been explored. Our work aimed to assess the potential link between iron overload, gut leakage/dysbiosis, and kidney dysfunction in these children. METHODS We enrolled 40 children with β-TM and 40 healthy controls. Gut leakage/dysbiosis biomarkers (trimethylamine-N-oxide [TMAO] and fecal short-chain fatty acids [SCFAs]), oxidative stress and inflammatory biomarkers, TMAO-regulated proteins such as serum sirtuin 1 (S.SIRT1) and serum high mobility box group-1 (S.HMGB1), and tubular dysfunction biomarkers were assessed. Correlations and regression analysis were performed to assess the relation between different parameters. RESULTS Iron overload, redox imbalance, and generalized inflammation were evident in children with β-TM. Renal tubular dysfunction biomarkers and S.TMAO were significantly elevated in the patient group. Furthermore, fecal SCFAs were significantly lower with upregulation of the investigated genes in the patient group. The correlation studies affirmed the close relationship between circulating ferritin, TMAO, and renal dysfunction and strongly implicated SIRT1/HMGB1 axis in TMAO action. CONCLUSIONS Gut dysbiosis may have a role in the pathogenesis of renal injury in children with β-TM. IMPACT Renal tubular dysfunction is a prominent health issue in β thalassemia major (β-TM). Iron overload, chronic anemia, and hypoxia are known precipitating factors. However, gut microbiota engagement in renal insult in these patients has not yet been explored. We aimed to assess potential link between iron overload, gut leakage/dysbiosis, and kidney dysfunction in β-TM children and to highlight the SIRT1/HMGB1 axis, a signal motivated by the gut microbiota-dependent metabolite trimethylamine-N-oxide (TMAO), involvement in such insults. We found that gut leakage/dysbiosis may have a role in kidney dysfunction in β-TM children by exacerbating the iron-motivated oxidative stress, inflammation, ferroptosis, and modulating SIRT1/HMGB1 axis.
Collapse
Affiliation(s)
- Asmaa A Ghalwash
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Rehab M El-Gohary
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Doaa El Amrousy
- Pediatric Department, Faculty of Medicine, Tanta University, Tanta, Egypt.
| | - Lamia M Morad
- Pediatric Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Shaima S Kassem
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | - Asmaa H Okasha
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
9
|
Ostapchuk YO, Lushova AV, Kan SA, Abdolla N, Kali A, Tleulieva R, Perfilyeva AV, Perfilyeva YV. Long-term changes in the phenotype and cytokine production of monocytes in COVID-19 recovered and vaccinated individuals. Infect Immun 2024; 92:e0021624. [PMID: 38874358 PMCID: PMC11238551 DOI: 10.1128/iai.00216-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
Monocytes play a crucial role in the immune response against pathogens. Here, we sought to determine COVID-19 and the vaccine Gam-COVID-Vac induce long-term changes in the phenotype and cytokine production of circulating monocytes. Monocytes were purified from peripheral blood mononuclear cells of healthy donors who had not had COVID-19 or vaccination, who had received two doses of Gam-COVID-Vac, and who had mild/moderate COVID-19 in the last 6 months and evaluated by flow cytometry. To investigate the effect of SARS-CoV-2 proteins, monocytes were cultured for 2 days with or without stimulation with recombinant SARS-CoV-2 S1 and N peptides. Monocytes obtained from vaccinated and recovered individuals showed increased basal expression of HLA-DR, CD63, CXCR2, and TLR7. We also observed an increased frequency of CD63+ classical monocytes in both groups, as well as an increased frequency of HLA-DR+ non-classical monocytes in the COVID-19-recovered group compared to the control group. Monocytes from vaccinated and recovered donors produced higher basal levels of IL-6, IL-1β, and TNF-α cytokines. Ex vivo stimulation with SARS-CoV-2 antigens induced increased expression of HLA-DR and TLR7 on monocytes obtained from the control group. The challenge with SARS-CoV-2 antigens had no effect on the production of IL-6, IL-1β, and TNF-α cytokines by monocytes. The acquired data offer compelling evidence of enduring alterations in both the phenotype and functional status of circulating monocytes subsequent to vaccination with Gam-COVID-Vac and mild/moderate COVID-19 infection. At least some of these changes appear to be a consequence of exposure to SARS-CoV-2 S1 and N antigens.
Collapse
Affiliation(s)
- Yekaterina O. Ostapchuk
- Laboratory of Molecular Immunology and Immunobiotechnology, M.A. Aitkhozhin’s Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
- Almaty Branch of the National Center for Biotechnology, Almaty, Kazakhstan
- ECO-Consulting LLC, Almaty, Kazakhstan
| | - Anzhelika V. Lushova
- Laboratory of Molecular Immunology and Immunobiotechnology, M.A. Aitkhozhin’s Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
- Almaty Branch of the National Center for Biotechnology, Almaty, Kazakhstan
- Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Sofia A. Kan
- Laboratory of Molecular Immunology and Immunobiotechnology, M.A. Aitkhozhin’s Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
- Almaty Branch of the National Center for Biotechnology, Almaty, Kazakhstan
- Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Nurshat Abdolla
- Laboratory of Molecular Immunology and Immunobiotechnology, M.A. Aitkhozhin’s Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
- Almaty Branch of the National Center for Biotechnology, Almaty, Kazakhstan
| | - Aikyn Kali
- Laboratory of Molecular Immunology and Immunobiotechnology, M.A. Aitkhozhin’s Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Raikhan Tleulieva
- Laboratory of Molecular Immunology and Immunobiotechnology, M.A. Aitkhozhin’s Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | | | - Yuliya V. Perfilyeva
- Laboratory of Molecular Immunology and Immunobiotechnology, M.A. Aitkhozhin’s Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
- Almaty Branch of the National Center for Biotechnology, Almaty, Kazakhstan
| |
Collapse
|
10
|
Limanaqi F, Zecchini S, Ogno P, Artusa V, Fenizia C, Saulle I, Vanetti C, Garziano M, Strizzi S, Trabattoni D, Clerici M, Biasin M. Alpha-synuclein shapes monocyte and macrophage cell biology and functions by bridging alterations of autophagy and inflammatory pathways. Front Cell Dev Biol 2024; 12:1421360. [PMID: 39035028 PMCID: PMC11257978 DOI: 10.3389/fcell.2024.1421360] [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: 04/22/2024] [Accepted: 06/17/2024] [Indexed: 07/23/2024] Open
Abstract
Introduction: Abnormal spreading of alpha-synuclein (αS), a hallmark of Parkinson's disease, is known to promote peripheral inflammation, which occurs in part via functional alterations in monocytes/macrophages. However, underlying intracellular mechanisms remain unclear. Methods: Herein we investigate the subcellular, molecular, and functional effects of excess αS in human THP-1 monocytic cell line, THP-1-derived macrophages, and at least preliminarily, in primary monocyte-derived macrophages (MDMs). In cells cultured w/wo recombinant αS (1 μM) for 4 h and 24 h, by Confocal microscopy, Western Blot, RT-qPCR, Elisa, and Flow Cytometry we assessed: i) αS internalization; ii) cytokine/chemokine expression/secretion, and C-C motif chemokine receptor 2 (CCR2) levels; iii) autophagy (LC3II/I, LAMP1/LysoTracker, p62, pS6/total S6); and iv) lipid droplets (LDs) accumulation, and cholesterol pathway gene expression. Transwell migration assay was employed to measure THP-1 cell migration/chemotaxis, while FITC-IgG-bead assay was used to analyze phagocytic capacity, and the fate of phagocytosed cargo in THP-1-derived macrophages. Results: Extracellular αS was internalized by THP-1 cells, THP-1-derived macrophages, and MDMs. In THP1 cells, αS induced a general pro-inflammatory profile and conditioned media from αS-exposed THP-1 cells potently attracted unstimulated cells. However, CCL2 secretion peaked at 4 h αS, consistent with early internalization of its receptor CCR2, while this was blunted at 24 h αS exposure, when CCR2 recycled back to the plasma membrane. Again, 4 h αS-exposed THP-1 cells showed increased spontaneous migration, while 24 h αS-exposed cells showed reduced chemotaxis. This occurred in the absence of cell toxicity and was associated with upregulation of autophagy/lysosomal markers, suggesting a pro-survival/tolerance mechanism against stress-related inflammation. Instead, in THP-1-derived macrophages, αS time-dependently potentiated the intracellular accumulation, and release of pro-inflammatory mediators. This was accompanied by mild toxicity, reduced autophagy-lysosomal markers, defective LDs formation, as well as impaired phagocytosis, and the appearance of stagnant lysosomes engulfed with phagocytosed cargo, suggesting a status of macrophage exhaustion reminiscent of hypophagia. Discussion: In summary, despite an apparently similar pro-inflammatory phenotype, monocytes and macrophages respond differently to intracellular αS accumulation in terms of cell survival, metabolism, and functions. Our results suggest that in periphery, αS exerts cell- and context-specific biological effects bridging alterations of autophagy, lipid dynamics, and inflammatory pathways.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Pasquale Ogno
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Valentina Artusa
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Claudio Fenizia
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Irma Saulle
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Claudia Vanetti
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Micaela Garziano
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Sergio Strizzi
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Daria Trabattoni
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Mario Clerici
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Mara Biasin
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
11
|
Q.B. Alenzi F. Survivin: A key apoptosis inhibitor in COVID-19 infection and its implication for treatment protocol. Saudi J Biol Sci 2024; 31:104021. [PMID: 38831893 PMCID: PMC11145386 DOI: 10.1016/j.sjbs.2024.104021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 06/05/2024] Open
Abstract
While the relationship between cellular apoptosis and proliferation rates in COVID patients remains underexplored in existing literature, various viruses are known to impact these fundamental process to modulate response to infection. This paper aims to assess apoptosis and proliferation rates in individuals recently infected with Coronavirus, both before and after vaccination, comparing them with healthy controls. Peripheral blood cells from newly diagnosed COVID-19 patients revealed a significant increase in proliferation and apoptosis levels in fresh lymphocytes and granulocytes compared to healthy donors. Notably, as none of the patients were under corticosteroid therapy or cytotoxic drugs, the study underscores the critical role of white blood (WBC) apoptosis in viral pathogenesis, potentially contributing significantly to COVID-19's pathogenicity. Elevated levels of soluble Fas ligand (FaSL) and the pro-inflatmmatory cytokine IL-38 were identified in COVID-19 patients, indicating potential immune dysregulation. Furthermore, individual who received the vaccine or recovered from COVID-19 exhibited higher survivin rates, suggesting a protective role for survivin in migitating lung damage. These findings suggest the prospect of developing a strategy to prevent WBC apoptosis, offering potential benefits in averting lymphopenia associated with severe COVID-19 ouctomes.
Collapse
Affiliation(s)
- Faris Q.B. Alenzi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| |
Collapse
|
12
|
Parada-Kusz M, Clatworthy AE, Goering ER, Blackwood SM, Shigeta JY, Mashin E, Salm EJ, Choi C, Combs S, Lee JSW, Rodriguez-Osorio C, Clish C, Tomita S, Hung DT. 3-Hydroxykynurenine targets kainate receptors to promote defense against infection. Nat Chem Biol 2024:10.1038/s41589-024-01635-z. [PMID: 38898166 DOI: 10.1038/s41589-024-01635-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 05/07/2024] [Indexed: 06/21/2024]
Abstract
Bacterial infection involves a complex interaction between the pathogen and host where the outcome of infection is not solely determined by pathogen eradication. To identify small molecules that promote host survival by altering the host-pathogen dynamic, we conducted an in vivo chemical screen using zebrafish embryos and found that treatment with 3-hydroxykynurenine (3-HK) protects from lethal bacterial infection. 3-HK, a metabolite produced through host tryptophan metabolism, has no direct antibacterial activity but enhances host survival by restricting bacterial expansion in macrophages through a systemic mechanism that targets kainate-sensitive glutamate receptors. These findings reveal a new pathway by which tryptophan metabolism and kainate-sensitive glutamate receptors function and interact to modulate immunity, with important implications for the coordination between the immune and nervous systems in pathological conditions.
Collapse
Affiliation(s)
- Margarita Parada-Kusz
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anne E Clatworthy
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA.
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Emily R Goering
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Stephanie M Blackwood
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jack Y Shigeta
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Elizabeth J Salm
- Department of Cellular and Molecular Physiology and Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Catherine Choi
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Senya Combs
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jenny S W Lee
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carlos Rodriguez-Osorio
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Susumu Tomita
- Department of Cellular and Molecular Physiology and Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Deborah T Hung
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA.
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
13
|
Zitta K, Hummitzsch L, Lichte F, Fändrich F, Steinfath M, Eimer C, Kapahnke S, Buerger M, Hess K, Rusch M, Rusch R, Berndt R, Albrecht M. Effects of temporal IFNγ exposure on macrophage phenotype and secretory profile: exploring GMP-Compliant production of a novel subtype of regulatory macrophages (Mreg IFNγ0) for potential cell therapeutic applications. J Transl Med 2024; 22:534. [PMID: 38835045 PMCID: PMC11151567 DOI: 10.1186/s12967-024-05336-y] [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: 04/17/2024] [Accepted: 05/18/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Macrophages are involved in tissue homeostasis, angiogenesis and immunomodulation. Proangiogenic and anti-inflammatory macrophages (regulatory macrophages, Mreg) can be differentiated in-vitro from CD14+ monocytes by using a defined cell culture medium and a stimulus of IFNγ. AIM OF THE STUDY To scrutinize the potential impact of temporal IFNγ exposure on macrophage differentiation as such exposure may lead to the emergence of a distinct and novel macrophage subtype. METHODS Differentiation of human CD14+ monocytes to Mreg was performed using a GMP compliant protocol and administration of IFNγ on day 6. Monocytes from the same donor were in parallel differentiated to MregIFNγ0 using the identical protocol but with administration of IFNγ on day 0. Cell characterization was performed using brightfield microscopy, automated and metabolic cell analysis, transmission electron microscopy, flow cytometry, qPCR and secretome profiling. RESULTS Mreg and MregIFNγ0 showed no differences in cell size and volume. However, phenotypically MregIFNγ0 exhibited fewer intracellular vesicles/vacuoles but larger pseudopodia-like extensions. MregIFNγ0 revealed reduced expression of IDO and PD-L1 (P < 0.01 for both). They were positive for CD80, CD14, CD16 and CD38 (P < 0.0001vs. Mreg for all), while the majority of MregIFNγ0 did not express CD206, CD56, and CD103 on their cell surface (P < 0.01 vs. Mreg for all). In terms of their secretomes, MregIFNγ0 differed significantly from Mreg. MregIFNγ0 media exhibited reduced levels of ENA-78, Osteopontin and Serpin E1, while the amounts of MIG (CXCL9) and IP10 were increased. CONCLUSION Exposing CD14+ monocytes to an alternatively timed IFNγ stimulation results in a novel macrophage subtype which possess additional M1-like features (MregIFNγ0). MregIFNγ0 may therefore have the potential to serve as cellular therapeutics for clinical applications beyond those covered by M2-like Mreg, including immunomodulation and tumor treatment.
Collapse
Affiliation(s)
- Karina Zitta
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein (UKSH), Kiel, Germany.
| | - Lars Hummitzsch
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Frank Lichte
- Department of Anatomy, University of Kiel, Kiel, Germany
| | - Fred Fändrich
- Clinic for Applied Cell Therapy, UKSH, Kiel, Germany
| | - Markus Steinfath
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Christine Eimer
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | | | - Matthias Buerger
- Clinic for Vascular and Endovascular Surgery, UKSH, Kiel, Germany
| | | | - Melanie Rusch
- Clinic for Vascular and Endovascular Surgery, UKSH, Kiel, Germany
| | - Rene Rusch
- Clinic for Vascular and Endovascular Surgery, UKSH, Kiel, Germany
| | - Rouven Berndt
- Clinic for Vascular and Endovascular Surgery, UKSH, Kiel, Germany
| | - Martin Albrecht
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| |
Collapse
|
14
|
Mosebarger A, Vidal MS, Bento GFC, Lintao RCV, Severino MEL, Kumar Kammala A, Menon R. Immune cells at the feto-maternal interface: Comprehensive characterization and insights into term labor. J Reprod Immunol 2024; 163:104239. [PMID: 38493591 DOI: 10.1016/j.jri.2024.104239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/05/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024]
Abstract
Immune cells at the feto-maternal interface play an important role in pregnancy; starting at implantation, maintenance of pregnancy, and parturition. The role of decidual immune cells in induction of labor still needs to be understood. Published reports on this topic show heterogeneity in methods of cell isolation, assay, analysis and cellular characterization making it difficult to collate available information in order to understand the contribution of immune cells at term leading to parturition. In the present study, available literature was reviewed to study the differences in immune cells between the decidua basalis and decidua parietalis, as well as between immune cells in term and preterm labor. Additionally, immune cells at the decidua parietalis were isolated from term not in labor (TNL) or term in labor (TL) samples and characterized via flow cytometry using a comprehensive, high-dimensional antibody panel. This allowed a full view of immune cell differences without combining multiple studies, which must include variation in isolation and analysis methods, for more conclusive data. The ratio of cells found in decidua parietalis in this study generally matched those reported in the literature, although we report a lower percentage of natural killer (NK) cells at term. We report that CD4 expression on CD8- NK cells decreased in term labor compared to not in labor samples, suggesting that natural killer cells may be migrating to other sites during labor. Also, we report a decrease in CD38 expression on CD8+ CD57+ T cells in labor, indicative of cytotoxic T cell senescence. Our study provides a comprehensive status of immune cells at the decidua-chorion interface at term.
Collapse
Affiliation(s)
- Angela Mosebarger
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Manuel S Vidal
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of Philippines Manila, Manila, Philippines
| | | | - Ryan C V Lintao
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of Philippines Manila, Manila, Philippines
| | - Mary Elise L Severino
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of Philippines Manila, Manila, Philippines
| | - Ananth Kumar Kammala
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Ramkumar Menon
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA.
| |
Collapse
|
15
|
Favaretto G, Rossi MN, Cuollo L, Laffranchi M, Cervelli M, Soriani A, Sozzani S, Santoni A, Antonangeli F. Neutrophil-activating secretome characterizes palbociclib-induced senescence of breast cancer cells. Cancer Immunol Immunother 2024; 73:113. [PMID: 38693312 PMCID: PMC11063017 DOI: 10.1007/s00262-024-03695-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/30/2024] [Indexed: 05/03/2024]
Abstract
Senescent cells have a profound impact on the surrounding microenvironment through the secretion of numerous bioactive molecules and inflammatory factors. The induction of therapy-induced senescence by anticancer drugs is known, but how senescent tumor cells influence the tumor immune landscape, particularly neutrophil activity, is still unclear. In this study, we investigate the induction of cellular senescence in breast cancer cells and the subsequent immunomodulatory effects on neutrophils using the CDK4/6 inhibitor palbociclib, which is approved for the treatment of breast cancer and is under intense investigation for additional malignancies. Our research demonstrates that palbociclib induces a reversible form of senescence endowed with an inflammatory secretome capable of recruiting and activating neutrophils, in part through the action of interleukin-8 and acute-phase serum amyloid A1. The activation of neutrophils is accompanied by the release of neutrophil extracellular trap and the phagocytic removal of senescent tumor cells. These findings may be relevant for the success of cancer therapy as neutrophils, and neutrophil-driven inflammation can differently affect tumor progression. Our results reveal that neutrophils, as already demonstrated for macrophages and natural killer cells, can be recruited and engaged by senescent tumor cells to participate in their clearance. Understanding the interplay between senescent cells and neutrophils may lead to innovative strategies to cope with chronic or tumor-associated inflammation.
Collapse
Affiliation(s)
- Gabriele Favaretto
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), c/o Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161, Rome, Italy
| | | | - Lorenzo Cuollo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Mattia Laffranchi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Alessandra Soriani
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Silvano Sozzani
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Fabrizio Antonangeli
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), c/o Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161, Rome, Italy.
| |
Collapse
|
16
|
Rijal R, Gomer RH. Gallein potentiates isoniazid's ability to suppress Mycobacterium tuberculosis growth. Front Microbiol 2024; 15:1369763. [PMID: 38690363 PMCID: PMC11060752 DOI: 10.3389/fmicb.2024.1369763] [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: 01/12/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Mycobacterium tuberculosis (Mtb), the bacterium that causes tuberculosis (TB), can be difficult to treat because of drug tolerance. Increased intracellular polyphosphate (polyP) in Mtb enhances tolerance to antibiotics, and capsular polyP in Neisseria gonorrhoeae potentiates resistance to antimicrobials. The mechanism by which bacteria utilize polyP to adapt to antimicrobial pressure is not known. In this study, we found that Mtb adapts to the TB frontline antibiotic isoniazid (INH) by enhancing the accumulation of cellular, extracellular, and cell surface polyP. Gallein, a broad-spectrum inhibitor of the polyphosphate kinase that synthesizes polyP, prevents this INH-induced increase in extracellular and cell surface polyP levels. Gallein and INH work synergistically to attenuate Mtb's ability to grow in in vitro culture and within human macrophages. Mtb when exposed to INH, and in the presence of INH, gallein inhibits cell envelope formation in most but not all Mtb cells. Metabolomics indicated that INH or gallein have a modest impact on levels of Mtb metabolites, but when used in combination, they significantly reduce levels of metabolites involved in cell envelope synthesis and amino acid, carbohydrate, and nucleoside metabolism, revealing a synergistic effect. These data suggest that gallein represents a promising avenue to potentiate the treatment of TB.
Collapse
Affiliation(s)
- Ramesh Rijal
- Gomer Lab, Department of Biology, Texas A&M University, College Station, TX, United States
| | - Richard H. Gomer
- Gomer Lab, Department of Biology, Texas A&M University, College Station, TX, United States
| |
Collapse
|
17
|
Mun S, Lee HJ, Kim P. Rebuilding the microenvironment of primary tumors in humans: a focus on stroma. Exp Mol Med 2024; 56:527-548. [PMID: 38443595 PMCID: PMC10984944 DOI: 10.1038/s12276-024-01191-5] [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: 08/31/2023] [Revised: 12/05/2023] [Accepted: 12/29/2023] [Indexed: 03/07/2024] Open
Abstract
Conventional tumor models have critical shortcomings in that they lack the complexity of the human stroma. The heterogeneous stroma is a central compartment of the tumor microenvironment (TME) that must be addressed in cancer research and precision medicine. To fully model the human tumor stroma, the deconstruction and reconstruction of tumor tissues have been suggested as new approaches for in vitro tumor modeling. In this review, we summarize the heterogeneity of tumor-associated stromal cells and general deconstruction approaches used to isolate patient-specific stromal cells from tumor tissue; we also address the effect of the deconstruction procedure on the characteristics of primary cells. Finally, perspectives on the future of reconstructed tumor models are discussed, with an emphasis on the essential prerequisites for developing authentic humanized tumor models.
Collapse
Affiliation(s)
- Siwon Mun
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, South Korea
| | - Hyun Jin Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, South Korea
| | - Pilnam Kim
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, South Korea.
- Institute for Health Science and Technology, KAIST, Daejeon, 34141, South Korea.
| |
Collapse
|
18
|
Zhang J, Ma J, Xu Y, Wu Y, Miao M. A fully automated Lab-on-a-Disc platform integrated a high-speed triggered siphon valve for PBMCs extraction. Talanta 2024; 268:125292. [PMID: 37857105 DOI: 10.1016/j.talanta.2023.125292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/19/2023] [Accepted: 10/06/2023] [Indexed: 10/21/2023]
Abstract
Human Peripheral Blood Mononuclear Cells (PBMCs) are isolated from peripheral blood and identified as any blood cell with a round nucleus that exhibits immune responses and undergoes immunophenotypic changes upon exposure to various pathophysiological stimuli. Obtaining high-recovery and clinical-grade PBMCs without decreasing cell viability and causing stress is crucial for disease diagnosis and successful immunotherapy. However, traditional manual PBMCs extraction methods rely on manual intervention with less recovery rate and reliability. In this study, we introduced a novel and efficient strategy for the fully automated extraction of PBMCs based on a Lab-on-a-Disk (LoaD) platform. The centrifugal chip used percoll as density gradient media (DGM) for separation and extraction on account of the density difference of cells in whole blood, without labeling and any additional extra cellular filtration or cell lysis steps. Above all, we proposed a high-speed triggered siphon valve, which was closed under the speed of cell sedimentation and subsequently opened by increasing speed to complete the extraction of PBMCs. It can avoid the problem that previous siphon valves rely on unstable hydrophilic surface treatment and prime under low/zero speed conditions. With valves and the clock channel integrated on the chip, users can achieve fully automated collection of PBMCs. Compared with the clinical laboratory results, the recovery rate of extracted PBMCs was 80 %. The experimental results prove that the high-speed triggered siphon valve improves the extraction efficiency of PBMCs. The robust chips, which are not only simple to manufacture and assemble but also stable and reliable to use, have great potential in biomedical and clinical applications.
Collapse
Affiliation(s)
- Jiahao Zhang
- State Key Laboratory of Applied Optics, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, Jilin, 130033, China; Key Laboratory of Optical System Advanced Manufacturing Technology, Chinese Academy of Sciences, Changchun Institute of Optics, Fine Mechanics and Physics, Changchun, Jilin, 130033, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junyu Ma
- State Key Laboratory of Applied Optics, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, Jilin, 130033, China; Key Laboratory of Optical System Advanced Manufacturing Technology, Chinese Academy of Sciences, Changchun Institute of Optics, Fine Mechanics and Physics, Changchun, Jilin, 130033, China
| | - Yang Xu
- State Key Laboratory of Applied Optics, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, Jilin, 130033, China; Key Laboratory of Optical System Advanced Manufacturing Technology, Chinese Academy of Sciences, Changchun Institute of Optics, Fine Mechanics and Physics, Changchun, Jilin, 130033, China; GD Changguang Zhongke Bio Co., Ltd., Foshan, Guangdong, 528200, China
| | - Yihui Wu
- State Key Laboratory of Applied Optics, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, Jilin, 130033, China; Key Laboratory of Optical System Advanced Manufacturing Technology, Chinese Academy of Sciences, Changchun Institute of Optics, Fine Mechanics and Physics, Changchun, Jilin, 130033, China; GD Changguang Zhongke Bio Co., Ltd., Foshan, Guangdong, 528200, China.
| | - Mingshu Miao
- Department of Clinical Laboratory, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| |
Collapse
|
19
|
Rijal R, Gomer RH. Gallein and isoniazid act synergistically to attenuate Mycobacterium tuberculosis growth in human macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.574965. [PMID: 38260681 PMCID: PMC10802476 DOI: 10.1101/2024.01.10.574965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Mycobacterium tuberculosis (Mtb), the bacterium that causes tuberculosis (TB), can be difficult to treat because of drug resistance. Increased intracellular polyphosphate (polyP) in Mtb enhances resistance to antibiotics, and capsular polyP in Neisseria gonorrhoeae potentiates resistance to antimicrobials. The mechanism by which bacteria utilize polyP to adapt to antimicrobial pressure is not known. In this study, we found that Mtb adapts to the TB frontline antibiotic isoniazid (INH) by enhancing the accumulation of cellular, extracellular, and cell surface polyP. Gallein, a broad-spectrum inhibitor of the polyphosphate kinase that synthesizes polyP, prevents this INH-induced increase in extracellular and cell surface polyP levels. Gallein and INH work synergistically to attenuate Mtb's ability to grow in in vitro culture and within human macrophages. Mtb when exposed to INH, and in the presence of INH, gallein inhibits cell envelope formation in most but not all Mtb cells. Metabolomics indicated that INH or gallein have a modest impact on levels of Mtb metabolites, but when used in combination, they significantly reduce levels of metabolites involved in cell envelope synthesis and amino acid, carbohydrate, and nucleoside metabolism, revealing a synergistic effect. These data suggest that gallein represents a promising avenue to potentiate the treatment of TB.
Collapse
Affiliation(s)
- Ramesh Rijal
- Department of Biology, Texas A&M University, College Station, TX 77843-3474, USA
| | - Richard H. Gomer
- Department of Biology, Texas A&M University, College Station, TX 77843-3474, USA
| |
Collapse
|
20
|
Roselletti E, Pericolini E, Nore A, Takacs P, Kozma B, Sala A, De Seta F, Comar M, Usher J, Brown GD, Wilson D. Zinc prevents vaginal candidiasis by inhibiting expression of an inflammatory fungal protein. Sci Transl Med 2023; 15:eadi3363. [PMID: 38055800 PMCID: PMC7616067 DOI: 10.1126/scitranslmed.adi3363] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 11/15/2023] [Indexed: 12/08/2023]
Abstract
Candida causes an estimated half-billion cases of vulvovaginal candidiasis (VVC) every year. VVC is most commonly caused by Candida albicans, which, in this setting, triggers nonprotective neutrophil infiltration, aggressive local inflammation, and symptomatic disease. Despite its prevalence, little is known about the molecular mechanisms underpinning the immunopathology of this fungal infection. In this study, we describe the molecular determinant of VVC immunopathology and a potentially straightforward way to prevent disease. In response to zinc limitation, C. albicans releases a trace mineral binding molecule called Pra1 (pH-regulated antigen). Here, we show that the PRA1 gene is strongly up-regulated during vaginal infections and that its expression positively correlated with proinflammatory cytokine concentrations in women. Genetic deletion of PRA1 prevented vaginal inflammation in mice, and application of a zinc solution down-regulated expression of the gene and also blocked immunopathology. We also show that treatment of women suffering from recurrent VVC with a zinc gel prevented reinfections. We have therefore identified a key mediator of symptomatic VVC, giving us an opportunity to develop a range of preventative measures for combatting this disease.
Collapse
Affiliation(s)
- Elena Roselletti
- Medical Research Council Centre for Medical Mycology at The University of Exeter, University of Exeter, Geoffrey Pope Building Stocker Road, Exeter, UK, EX4 4QD
| | - Eva Pericolini
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy, 41125
| | - Alexandre Nore
- Medical Research Council Centre for Medical Mycology at The University of Exeter, University of Exeter, Geoffrey Pope Building Stocker Road, Exeter, UK, EX4 4QD
| | - Peter Takacs
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary, 4032
- Division of Female Pelvic Medicine and Reconstructive Surgery, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA, 23507
| | - Bence Kozma
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary, 4032
| | - Arianna Sala
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy, 41125
| | - Francesco De Seta
- Department of Medical Sciences, University of Trieste, Institute for Maternal and Child Health-IRCCS, Burlo Garofolo, Trieste, Italy, 34137
| | - Manola Comar
- Unit of Advanced Microbiology Diagnosis and Translational Research, Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, University of Trieste, Trieste, Italy, 34137
| | - Jane Usher
- Medical Research Council Centre for Medical Mycology at The University of Exeter, University of Exeter, Geoffrey Pope Building Stocker Road, Exeter, UK, EX4 4QD
| | - Gordon D Brown
- Medical Research Council Centre for Medical Mycology at The University of Exeter, University of Exeter, Geoffrey Pope Building Stocker Road, Exeter, UK, EX4 4QD
| | - Duncan Wilson
- Medical Research Council Centre for Medical Mycology at The University of Exeter, University of Exeter, Geoffrey Pope Building Stocker Road, Exeter, UK, EX4 4QD
| |
Collapse
|
21
|
Fahmy HA, Mohamed MA, Mekkawy MH, Taha EFS. Role of TLR4 signaling pathway in the mitigation of damaged lung by low-dose gamma irradiation. Cell Biochem Funct 2023; 41:1188-1199. [PMID: 37732723 DOI: 10.1002/cbf.3851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/22/2023] [Accepted: 08/28/2023] [Indexed: 09/22/2023]
Abstract
Organisms frequently suffer negative effects from large doses of ionizing radiation. However, radiation is not as hazardous at lower doses as was once believed. The current study aims to evaluate the possible radio-adaptive effect induced by low-dose radiation (LDR) in modulating high-dose radiation (HDR) and N-nitrosodiethylamine (NDEA)-induced lung injury in male albino rats. Sixty-four male rats were randomly divided into four groups: Group 1 (control): normal rats; Group 2 (D): rats given NDEA in drinking water; Group 3 (DR): rats administered with NDEA then exposed to fractionated HDR; and Group 4 (DRL): rats administered with NDEA then exposed to LDR + HDR. In the next stage, malondialdehyde (MDA), glutathione reduced (GSH), catalase (CAT), and superoxide dismutase (SOD) levels in the lung tissues were measured. Furthermore, the enzyme-linked immunoassay analysis technique was performed to assess the Toll-like receptor 4 (TLR4), interleukin-1 receptor-associated kinase 4 (IRAK4), and mitogen-activated protein kinases (MAPK) expression levels. Histopathological and DNA fragmentation analyses in lung tissue, in addition to hematological and apoptosis analyses of the blood samples, were also conducted. Results demonstrated a significant increase in antioxidant defense and a reduction in MDA levels were observed in LDR-treated animals compared to the D and DR groups. Additionally, exposure to LDR decreased TLR4, IRAK4, and MAPK levels, decreased apoptosis, and restored all the alterations in the histopathological, hematological parameters, and DNA fragmentation, indicating its protective effects on the lung when compared with untreated rats. Taken together, LDR shows protective action against the negative effects of subsequent HDR and NDEA. This impact may be attributable to the adaptive response induced by LDR, which decreases DNA damage in lung tissue and activates the antioxidative, antiapoptotic, and anti-inflammatory systems in the affected animals, enabling them to withstand the following HDR exposure.
Collapse
Affiliation(s)
- Hanan A Fahmy
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (AEA), Cairo, Egypt
| | - Marwa A Mohamed
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (AEA), Cairo, Egypt
| | - Mai H Mekkawy
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (AEA), Cairo, Egypt
| | - Eman F S Taha
- Health Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
22
|
Phatale V, Famta P, Srinivasarao DA, Vambhurkar G, Jain N, Pandey G, Kolipaka T, Khairnar P, Shah S, Singh SB, Raghuvanshi RS, Srivastava S. Neutrophil membrane-based nanotherapeutics: Propitious paradigm shift in the management of cancer. Life Sci 2023; 331:122021. [PMID: 37582468 DOI: 10.1016/j.lfs.2023.122021] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/03/2023] [Accepted: 08/10/2023] [Indexed: 08/17/2023]
Abstract
Cancer is the leading cause of death across the globe, with 19.3 million new cancer cases and 10 million deaths in the year 2020. Conventional treatment modalities have numerous pitfalls, such as off-site cytotoxicity and poor bioavailability. Nanocarriers (NCs) have been explored to deliver various therapeutic moieties such as chemotherapeutic agents and photothermal agents, etc. However, several limitations, such as rapid clearance by the reticuloendothelial system, poor extravasation into the tumor microenvironment, and low systemic half-life are roadblocks to successful clinical translation. To circumvent the pitfalls of currently available treatment modalities, neutrophil membrane (NM)-based nanotherapeutics have emerged as a promising platform for cancer management. Their versatile features such as natural tumor tropism, tumor-specific accumulation, and prevention from rapid clearance owing to their autologous nature make them an effective anticancer NCs. In this manuscript, we have discussed various methods for isolation, coating and characterization of NM. We have discussed the role of NM-coated nanotherapeutics as neoadjuvant and adjuvant in different treatment modalities, such as chemotherapy, photothermal and photodynamic therapies with rationales behind their inclusion. Clinical hurdles faced during the bench-to-bedside translation with possible solutions have been discussed. We believe that in the upcoming years, NM-coated nanotherapeutics will open a new horizon in cancer management.
Collapse
Affiliation(s)
- Vivek Phatale
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Paras Famta
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Dadi A Srinivasarao
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Ganesh Vambhurkar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Naitik Jain
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Giriraj Pandey
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Tejaswini Kolipaka
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pooja Khairnar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Shah
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Rajeev Singh Raghuvanshi
- Central Drugs Standard Control Organization (CDSCO), Directorate General of Health Services, Ministry of Health & Family Welfare, Government of India, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
23
|
Hamdan F, Feodoroff M, Russo S, Fusciello M, Feola S, Chiaro J, Antignani G, Greco F, Leusen J, Ylösmäki E, Grönholm M, Cerullo V. Controlled release of enhanced cross-hybrid IgGA Fc PD-L1 inhibitors using oncolytic adenoviruses. Mol Ther Oncolytics 2023; 28:264-276. [PMID: 36911070 PMCID: PMC9995465 DOI: 10.1016/j.omto.2023.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 01/31/2023] [Indexed: 02/07/2023] Open
Abstract
Immune checkpoint inhibitors have clinical success in prolonging the life of many cancer patients. However, only a minority of patients benefit from such therapy, calling for further improvements. Currently, most PD-L1 checkpoint inhibitors in the clinic do not elicit Fc effector mechanisms that would substantially increase their efficacy. To gain potency and circumvent off-target effects, we previously designed an oncolytic adenovirus (Ad-Cab) expressing an Fc fusion peptide against PD-L1 on a cross-hybrid immunoglobulin GA (IgGA) Fc. Ad-Cab elicited antibody effector mechanisms of IgG1 and IgA, which led to higher tumor killing compared with each isotype alone and with clinically approved PD-L1 checkpoint inhibitors. In this study, we further improved the therapy to increase the IgG1 Fc effector mechanisms of the IgGA Fc fusion peptide (Ad-Cab FT) by adding four somatic mutations that increase natural killer (NK) cell activation. Ad-Cab FT was shown to work better at lower concentrations compared with Ad-Cab in vitro and in vivo and to have better tumor- and myeloid-derived suppressor cell killing, likely because of higher NK cell activation. Additionally, the biodistribution of the Fc fusion peptide demonstrated targeted release in the tumor microenvironment with minimal or no leakage to the peripheral blood and organs in mice. These data demonstrate effective and safe use of Ad-Cab FT, bidding for further clinical investigation.
Collapse
Affiliation(s)
- Firas Hamdan
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Michaela Feodoroff
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.,iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Salvatore Russo
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Manlio Fusciello
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Sara Feola
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jacopo Chiaro
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Gabriella Antignani
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Francesca Greco
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jeanette Leusen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Erkko Ylösmäki
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Mikaela Grönholm
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Vincenzo Cerullo
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland.,Department of Molecular Medicine and Medical Biotechnology and CEINGE, Naples University Federico II, Naples, Italy
| |
Collapse
|
24
|
Riça IG, Joughin BA, Teke ME, Emmons TR, Griffith A, Cahill LA, Banner-Goodspeed V, Robson SC, Hernandez JM, Segal BH, Otterbein LE, Hauser CJ, Lederer JA, Yaffe MB. Neutrophil heterogeneity and emergence of a distinct population of CD11b/CD18-activated low-density neutrophils after trauma. J Trauma Acute Care Surg 2023; 94:187-196. [PMID: 36694330 PMCID: PMC9881754 DOI: 10.1097/ta.0000000000003823] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Multiple large clinical trauma trials have documented an increased susceptibility to infection after injury. Although neutrophils (polymorphonuclear leukocytes [PMNs]) were historically considered a homogeneous cell type, we hypothesized that injury could alter neutrophil heterogeneity and predispose to dysfunction. To explore whether trauma modifies PMN heterogeneity, we performed an observational mass-spectrometry-based cytometry study on total leukocytes and low-density PMNs found in the peripheral blood mononuclear cell fraction of leukocytes from healthy controls and trauma patients. METHODS A total of 74 samples from 12 trauma patients, each sampled at 1 or more time points, and matched controls were fractionated and profiled by mass-spectrometry-based cytometry using a panel of 44 distinct markers. After deconvolution and conservative gating on neutrophils, data were analyzed using Seurat, followed by clustering of principal components. RESULTS Eleven distinct neutrophil populations were resolved in control and trauma neutrophils based on differential protein surface marker expression. Trauma markedly altered the basal heterogeneity of neutrophil subgroups seen in the control samples, with loss of a dominant population of resting neutrophils marked by high expression of C3AR and low levels of CD63, CD64, and CD177 (cluster 1), and expansion of two alternative neutrophil populations, one of which is marked by high expression of CD177 with suppression of CD10, CD16, C3AR, CD63, and CD64 (cluster 6). Remarkably, following trauma, a substantially larger percentage of neutrophils sediment in the monocyte fraction. These low-density neutrophils bear markers of functional exhaustion and form a unique trauma-induced population (cluster 9) with markedly upregulated expression of active surface adhesion molecules (activated CD11b/CD18), with suppression of nearly all other surface markers, including receptors for formyl peptides, leukotrienes, chemokines, and complement. CONCLUSION Circulating neutrophils demonstrate considerable evidence of functional heterogeneity that is markedly altered by trauma. Trauma induces evolution of a novel, exhausted, low-density neutrophil population with immunosuppressive features.
Collapse
Affiliation(s)
- Ingred Goretti Riça
- Departments of Biological Engineering and Biology, David H. Koch Institute for Integrative Cancer Research, and Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Brian A. Joughin
- Departments of Biological Engineering and Biology, David H. Koch Institute for Integrative Cancer Research, and Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Martha E. Teke
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Tiffany R. Emmons
- Departments of Biological Engineering and Biology, David H. Koch Institute for Integrative Cancer Research, and Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alec Griffith
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Laura A. Cahill
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Valerie Banner-Goodspeed
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115 USA
| | - Simon C. Robson
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115 USA
| | - Jonathan M. Hernandez
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Brahm H. Segal
- Department of Medicine, Roswell Park Comprehensive Cancer Center, University of Buffalo School of Medicine, Buffalo, NY14263 USA
| | - Leo E. Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115 USA
| | - Carl J. Hauser
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115 USA
| | - James A. Lederer
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Michael B. Yaffe
- Departments of Biological Engineering and Biology, David H. Koch Institute for Integrative Cancer Research, and Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115 USA
| |
Collapse
|
25
|
Johnson RK, Overlee BL, Sagen JA, Howe CL. Peripheral blood mononuclear cell phenotype and function are maintained after overnight shipping of whole blood. Sci Rep 2022; 12:19920. [PMID: 36402888 PMCID: PMC9675784 DOI: 10.1038/s41598-022-24550-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022] Open
Abstract
Same day processing of biospecimens such as blood is not always feasible, which presents a challenge for research programs seeking to study a broad population or to characterize patients with rare diseases. Recruiting sites may not be equipped to process blood samples and variability in timing and technique employed to isolate peripheral blood mononuclear cells (PBMCs) at local sites may compromise reproducibility across patients. One solution is to send whole blood collected by routine phlebotomy via overnight courier to the testing site under ambient conditions. Determining the impact of shipping on subsequent leukocyte responses is a necessary prerequisite to any experimental analysis derived from transported samples. To this end, whole blood was collected from healthy control subjects and processed fresh or at 6, 24 and 48 h after collection and handling under modeled shipping conditions. At endpoint, whole blood was assessed via a complete blood count with differential and immunophenotyped using a standardized panel of antibodies [HLADR, CD66b, CD3, CD14, CD16]. PBMCs and neutrophils were isolated from whole blood and subjected to ex vivo stimulation with lipopolysaccharide and heat-killed Staphylococcus aureus. Stimulated release of cytokines and chemokines was assessed by cytometric bead array. RNA was also isolated from PBMCs to analyze transcriptional changes induced by shipping. The complete blood count with differential revealed that most parameters were maintained in shipped blood held for 24 h at ambient temperature. Immunophenotyping indicated preservation of cellular profiles at 24 h, although with broadening of some populations and a decrease in CD16 intensity on classical monocytes. At the transcriptional level, RNAseq analysis identified upregulation of a transcription factor module associated with inflammation in unstimulated PBMCs derived from whole blood shipped overnight. However, these changes were limited in both scale and number of impacted genes. Ex vivo stimulation of PBMCs further revealed preservation of functional responses in cells isolated from shipped blood held for 24 h at ambient temperature. However, neutrophil responses were largely abrogated by this time. By 48 h neither cell population responded within normal parameters. These findings indicate that robust immunophenotyping and PBMC stimulated response profiles are maintained in whole blood shipped overnight and processed within 24 h of collection, yielding results that are representative of those obtained from the sample immediately following venipuncture. This methodology is feasible for many patient recruitment sites to implement and allows for sophisticated immunological analysis of patient populations derived from large geographic areas. With regard to rare disease research, this meets a universal need to enroll patients in sufficient numbers for immunoprofiling and discovery of underlying pathogenic mechanisms.
Collapse
Affiliation(s)
- Renee K Johnson
- Translational Neuroimmunology Lab, Mayo Clinic, Guggenheim 1542C, 200 First St SW, Rochester, MN, 55905, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Brittany L Overlee
- Translational Neuroimmunology Lab, Mayo Clinic, Guggenheim 1542C, 200 First St SW, Rochester, MN, 55905, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jessica A Sagen
- Translational Neuroimmunology Lab, Mayo Clinic, Guggenheim 1542C, 200 First St SW, Rochester, MN, 55905, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
- Center for MS and Autoimmune Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Charles L Howe
- Translational Neuroimmunology Lab, Mayo Clinic, Guggenheim 1542C, 200 First St SW, Rochester, MN, 55905, USA.
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA.
- Center for MS and Autoimmune Neurology, Mayo Clinic, Rochester, MN, 55905, USA.
- Division of Experimental Neurology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|