1
|
Duhaini M, Fares P, Hafezi L, El-Zein H, Kondapalli KC. Sodium proton exchanger NHE9 pHine-tunes exosome production by impairing Rab7 activity. J Biol Chem 2025; 301:108264. [PMID: 39909375 PMCID: PMC11929068 DOI: 10.1016/j.jbc.2025.108264] [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: 11/12/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
Cell-to-cell communication is mediated by vesicles ranging from 30 to 150 nm, known as exosomes. These exosomes shuttle bioactive molecules such as proteins, lipids, and nucleic acids, thus playing crucial roles in both health and disease mechanisms. Exosomes form within the endocytic pathway through the process of inward budding of the endosomal membrane, facilitated by the progressive acidification of the endosomal lumen. Although endosomal pH is known to be critical for exosome production, the precise molecular mechanisms involved remain poorly defined. Maintaining optimal endosomal pH involves meticulous coordination between proton pumping and leakage mechanisms. The sodium-proton exchanger NHE9, located on the endosomal membrane, modulates endosomal pH by transporting protons out of the endosomes in exchange for sodium or potassium ions. Here, we use genetic engineering, biochemistry, and advanced microscopy to demonstrate that the sodium-proton exchanger NHE9 significantly affects exosome production by regulating endosomal pH. NHE9-mediated endosomal alkalization impairs Rab7 activation, thereby disrupting the delivery of multivesicular endosomes to lysosomes. Moreover, luminal alkalization promotes the recruitment of Rab27b. This enhances the targeting of multivesicular endosomes to the cell periphery, their fusion with the plasma membrane, and subsequent exosome secretion. Our findings reveal the detailed molecular mechanisms through which endosomal pH regulates exosome production. Additionally, we identify NHE9 as a potential target for therapeutic strategies aimed at controlling exosome dynamics.
Collapse
Affiliation(s)
- Mariam Duhaini
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Perla Fares
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Lili Hafezi
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Hadi El-Zein
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Kalyan C Kondapalli
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, USA.
| |
Collapse
|
2
|
Hook JL, Kuebler WM. CFTR as a therapeutic target for severe lung infection. Am J Physiol Lung Cell Mol Physiol 2025; 328:L229-L238. [PMID: 39772994 DOI: 10.1152/ajplung.00289.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Lung infection is one of the leading causes of morbidity and mortality worldwide. Even with appropriate antibiotic and antiviral treatment, mortality in hospitalized patients often exceeds 10%, highlighting the need for the development of new therapeutic strategies. Of late, cystic fibrosis transmembrane conductance regulator (CFTR) is-in addition to its well-established roles in the lung airway and extrapulmonary organs-increasingly recognized as a key regulator of alveolar homeostasis and defense. In the alveolar epithelium, CFTR mediates alveolar fluid secretion and liquid homeostasis; in the microvascular endothelium, CFTR maintains vascular barrier function. CFTR also contributes to alveolar immunity. Yet, in lung infection, diverse molecular mechanisms reduce CFTR abundance and otherwise impair its function, promoting alveolar inflammation, edema, and cell death. Preservation or restoration of CFTR function by CFTR modulator drugs thus presents a promising avenue to combat lung infection in a pathogen-independent manner.
Collapse
Affiliation(s)
- Jaime L Hook
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Bercusson A, Williams TJ, Simmonds NJ, Alton EWFW, Griesenbach U, Shah A, Warris A, Armstrong-James D. Increased NFAT and NFκB signalling contribute to the hyperinflammatory phenotype in response to Aspergillus fumigatus in a mouse model of cystic fibrosis. PLoS Pathog 2025; 21:e1012784. [PMID: 39903773 PMCID: PMC11957335 DOI: 10.1371/journal.ppat.1012784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/31/2025] [Accepted: 01/16/2025] [Indexed: 02/06/2025] Open
Abstract
Aspergillus fumigatus (Af) is a major mould pathogen found ubiquitously in the air. It commonly infects the airways of people with cystic fibrosis (CF) leading to Aspergillus bronchitis or allergic bronchopulmonary aspergillosis. Resident alveolar macrophages and recruited neutrophils are important first lines of defence for clearance of Af in the lung. However, their contribution to the inflammatory phenotype in CF during Af infection is not well understood. Here, utilising CFTR deficient mice we describe a hyperinflammatory phenotype in both acute and allergic murine models of pulmonary aspergillosis. We show that during aspergillosis, CFTR deficiency leads to increased alveolar macrophage death and persistent inflammation of the airways in CF, accompanied by impaired fungal control. Utilising CFTR deficient murine cells and primary human CF cells we show that at a cellular level there is increased activation of NFκB and NFAT in response to Af which, as in in vivo models, is associated with increased cell death and reduced fungal control. Taken together, these studies indicate that CFTR deficiency promotes increased activation of inflammatory pathways, the induction of macrophage cell death and reduced fungal control contributing to the hyper-inflammatory of pulmonary aspergillosis phenotypes in CF.
Collapse
Affiliation(s)
- Amelia Bercusson
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London, United Kingdom
- Cystic Fibrosis Unit, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Thomas J. Williams
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London, United Kingdom
- Department of Cardiothoracic Transplantation and Mechanical Support, Harefield Hospital, Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Nicholas J. Simmonds
- Adult Cystic Fibrosis Centre, Royal Brompton Hospital, London, United Kingdom
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Eric WFW Alton
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Uta Griesenbach
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Anand Shah
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London, United Kingdom
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Darius Armstrong-James
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London, United Kingdom
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
4
|
Kovacs T, Cs. Szabo B, Kothalawala RC, Szekelyhidi V, Nagy P, Varga Z, Panyi G, Zakany F. Inhibition of the H V1 voltage-gated proton channel compromises the viability of human polarized macrophages in a polarization- and ceramide-dependent manner. Front Immunol 2024; 15:1487578. [PMID: 39742270 PMCID: PMC11685079 DOI: 10.3389/fimmu.2024.1487578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
The human voltage-gated proton channel (HV1) provides an efficient proton extrusion pathway from the cytoplasm contributing to the intracellular pH regulation and the oxidative burst. Although its pharmacological inhibition was previously shown to induce cell death in various cell types, no such effects have been examined in polarized macrophages albeit HV1 was suggested to play important roles in these cells. This study highlights that 5-chloro-2-guanidinobenzimidazole (ClGBI), the most widely applied HV1 inhibitor, reduces the viability of human THP-1-derived polarized macrophages at biologically relevant doses with M1 macrophages being the most, and M2 cells the least sensitive to this compound. ClGBI may exert this effect principally by blocking HV1 since the sensitivity of polarized macrophages correlates well with their HV1 expression levels; inhibitors of other macrophage ion channels that may be susceptible for off-target ClGBI effects cause no viability reductions; and Zn2+, another non-specific HV1 blocker, exerts similar effects. As a potential mechanism behind the ClGBI-induced cell death, we identify a complex pH dysregulation involving acidification of the cytoplasm and alkalinization of the lysosomes, which eventually result in membrane ceramide accumulation. Furthermore, ClGBI effects are alleviated by ARC39, a selective acid sphingomyelinase inhibitor supporting the unequivocal significance of ceramide accumulation in the process. Altogether, our results suggest that HV1 inhibition leads to cellular toxicity in polarized macrophages in a polarization-dependent manner, which occurs due to a pH dysregulation and concomitant ceramide overproduction mainly depending on the activity of acid sphingomyelinase. The reduced macrophage viability and plausible concomitant changes in homeostatic M1-M2 balance could contribute to both the therapeutic and potential side effects of HV1 inhibitors that show great promise in the treatment of neuroinflammation and malignant diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Florina Zakany
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
5
|
Galué-Parra A, de Moraes LS, Hage AAP, Castro de Sena CB, Nascimento JLMD, da Silva EO. In vitro immunomodulatory effects of Caryocar villosum oil on murine macrophages. Biomed Pharmacother 2024; 179:117360. [PMID: 39232387 DOI: 10.1016/j.biopha.2024.117360] [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: 07/04/2024] [Revised: 08/16/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024] Open
Abstract
Macrophages undergo activation in response to multiple stimuli, including pathogens, growth factors and natural products. The inflammatory response and oxidative stress play critical roles in such macrophage activation. Some natural products reportedly promote immunoregulatory effects and the control of macrophage activation. Caryocar villosum (Cv), a native amazon plant, contains compounds that are an important source of molecules capable of macrophage activation. Herein, we demonstrate the immunomodulatory effects of oil obtained from Caryocar villosum (CvO) on macrophages. Macrophages were treated with varying concentrations of CvO, and resulting cellular morphological and functional changes were evaluated, including the production of nitric oxide (NO), reactive oxygen species (ROS), cytokines and phagocytic activity. Treatment of cells with 50 and 100 μg/mL CvO induced morphological and physiological alterations in the macrophages, such as increased cell surface and phagocytic activity. Additionally, treatment increased the productions of inflammatory cytokines (INF-γ, TNF-α, IL-6) and anti-inflammatory cytokines (IL-17 and IL-10) by macrophages, and significantly decreased ROS levels. In conclusion, these data suggest that, due to molecular diversity, CvO promoted an immunomodulatory effect on macrophages, mediated by an increased production of cytokines, and inhibition of ROS generation and phagocytic activity. Thus, CvO presents potential as a therapeutic agent for the treatment of inflammatory and non-inflammatory diseases.
Collapse
Affiliation(s)
- Adan Galué-Parra
- Laboratory of Structural Biology, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil; Postgraduate Program in Biology of Infectious and Parasitic Agents, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil; National Institute of Science and Technology in Structural Biology and Bioimaging, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lienne Silveira de Moraes
- Laboratory of Structural Biology, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil
| | - Amanda Anastácia Pinto Hage
- Laboratory of Structural Biology, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil
| | - Chubert Bernardo Castro de Sena
- Laboratory of Structural Biology, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil; National Institute of Science and Technology in Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jose Luiz Martins do Nascimento
- Laboratory of Molecular and Cellular Neurochemistry, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil; National Institute of Science and Technology in Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Edilene Oliveira da Silva
- Laboratory of Structural Biology, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil; Postgraduate Program in Biology of Infectious and Parasitic Agents, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil; National Institute of Science and Technology in Structural Biology and Bioimaging, Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
6
|
Schnell A, Tamm S, Hedtfeld S, Rodriguez Gonzalez C, Hoerning A, Lachmann N, Stanke F, Dittrich AM, Munder A. Analysis of CFTR mRNA and Protein in Peripheral Blood Mononuclear Cells via Quantitative Real-Time PCR and Western Blot. Int J Mol Sci 2024; 25:6367. [PMID: 38928073 PMCID: PMC11203434 DOI: 10.3390/ijms25126367] [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: 12/21/2023] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
The Cystic Fibrosis Conductance Transmembrane Regulator gene encodes for the CFTR ion channel, which is responsible for the transport of chloride and bicarbonate across the plasma membrane. Mutations in the gene result in impaired ion transport, subsequently leading to perturbed secretion in all exocrine glands and, therefore, the multi-organ disease cystic fibrosis (CF). In recent years, several studies have reported on CFTR expression in immune cells as demonstrated by immunofluorescence, flow cytometry, and immunoblotting. However, these data are mainly restricted to single-cell populations and show significant variation depending on the methodology used. Here, we investigated CFTR transcription and protein expression using standardized protocols in a comprehensive panel of immune cells. Methods: We applied a high-resolution Western blot protocol using a combination of highly specific monoclonal CFTR antibodies that have been optimized for the detection of CFTR in epithelial cells and healthy primary immune cell subpopulations sorted by flow cytometry and used immortalized cell lines as controls. The specificity of CFTR protein detection was controlled by peptide competition and enzymatic Peptide-N-Glycosidase-F (PNGase) digest. CFTR transcripts were analyzed using quantitative real-time PCR and normalized to the level of epithelial T84 cells as a reference. Results: CFTR mRNA expression could be shown for primary CD4+ T cells, NK cells, as well as differentiated THP-1 and Jurkat T cells. In contrast, we failed to detect CFTR transcripts for CD14+ monocytes and undifferentiated THP-1 cells, as well as for B cells and CD8+ T cells. Prominent immunoreactive bands were detectable by immunoblotting with the combination of four CFTR antibodies targeting different epitopes of the CFTR protein. However, in biosamples of non-epithelial origin, these CFTR-like protein bands could be unmasked as false positives through peptide competition or PNGase digest, meaning that the observed mRNA transcripts were not necessarily translated into CFTR proteins, which could be detected via immunoblotting. Our results confirm that mRNA expression in immune cells is many times lower than in that cells of epithelial origin. The immunoreactive signals in immune cells turned out to be false positives, and may be provoked by the presence of a high-affinity protein with a similar epitope. Non-specific binding (e.g., Fab-interaction with glycosyl branches) might also contribute to false positive signals. Our findings highlight the necessity of accurate controls, such as CFTR-negative cells, as well as peptide competition and glycolytic digest in order to identify genuine CFTR protein by immunoblotting. Our data suggest, furthermore, that CFTR protein expression data from techniques such as histology, for which the absence of a molecular weight or other independent control prevents the unmasking of false positive immunoreactive signals, must be interpreted carefully as well.
Collapse
Affiliation(s)
- Alexander Schnell
- Department of Pediatric and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany;
| | - Stephanie Tamm
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, 30625 Hannover, Germany
| | - Silke Hedtfeld
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, 30625 Hannover, Germany
| | - Claudio Rodriguez Gonzalez
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
| | - Andre Hoerning
- Department of Pediatric and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany;
| | - Nico Lachmann
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, 30625 Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany
| | - Frauke Stanke
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, 30625 Hannover, Germany
| | - Anna-Maria Dittrich
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, 30625 Hannover, Germany
| | - Antje Munder
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
7
|
Yu Y, Zhang Z, Yu Y. Timing of Phagosome Maturation Depends on Their Transport Switching from Actin to Microtubule Tracks. J Phys Chem B 2023; 127:9312-9322. [PMID: 37871280 PMCID: PMC10759163 DOI: 10.1021/acs.jpcb.3c05647] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Phagosomes, specialized membrane compartments responsible for digesting internalized pathogens, undergo sequential dynamic and biochemical changes as they mature from nascent phagosomes to degradative phagolysosomes. Maturation of phagosomes depends on their transport along actin filaments and microtubules. However, the specific quantitative relationship between the biochemical transformation and transport dynamics remains poorly characterized. The autonomous nature of phagosomes, moving and maturing at different rates, makes understanding this relationship challenging. Addressing this challenge, in this study we engineered particle sensors to image and quantify single phagosomes' maturation. We found that as phagosomes move from the actin cortex to microtubule tracks, the timing of their actin-to-microtubule transition governs the duration of the early phagosome stage before acquiring degradative capacities. Prolonged entrapment of phagosomes in the actin cortex extends the early phagosome stage by delaying the dissociation of early endosome markers and phagosome acidification. Conversely, a shortened transition from actin- to microtubule-based movements causes the opposite effect on phagosome maturation. These results suggest that the actin- and microtubule-based transport of phagosomes functions like a "clock" to coordinate the timing of biochemical events during phagosome maturation, which is crucial for effective pathogen degradation.
Collapse
Affiliation(s)
- Yanqi Yu
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405-7102, United States
| | - Zihan Zhang
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405-7102, United States
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405-7102, United States
| |
Collapse
|
8
|
Leon-Icaza SA, Bagayoko S, Vergé R, Iakobachvili N, Ferrand C, Aydogan T, Bernard C, Sanchez Dafun A, Murris-Espin M, Mazières J, Bordignon PJ, Mazères S, Bernes-Lasserre P, Ramé V, Lagarde JM, Marcoux J, Bousquet MP, Chalut C, Guilhot C, Clevers H, Peters PJ, Molle V, Lugo-Villarino G, Cam K, Berry L, Meunier E, Cougoule C. Druggable redox pathways against Mycobacterium abscessus in cystic fibrosis patient-derived airway organoids. PLoS Pathog 2023; 19:e1011559. [PMID: 37619220 PMCID: PMC10449475 DOI: 10.1371/journal.ppat.1011559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 07/13/2023] [Indexed: 08/26/2023] Open
Abstract
Mycobacterium abscessus (Mabs) drives life-shortening mortality in cystic fibrosis (CF) patients, primarily because of its resistance to chemotherapeutic agents. To date, our knowledge on the host and bacterial determinants driving Mabs pathology in CF patient lung remains rudimentary. Here, we used human airway organoids (AOs) microinjected with smooth (S) or rough (R-)Mabs to evaluate bacteria fitness, host responses to infection, and new treatment efficacy. We show that S Mabs formed biofilm, and R Mabs formed cord serpentines and displayed a higher virulence. While Mabs infection triggers enhanced oxidative stress, pharmacological activation of antioxidant pathways resulted in better control of Mabs growth and reduced virulence. Genetic and pharmacological inhibition of the CFTR is associated with better growth and higher virulence of S and R Mabs. Finally, pharmacological activation of antioxidant pathways inhibited Mabs growth, at least in part through the quinone oxidoreductase NQO1, and improved efficacy in combination with cefoxitin, a first line antibiotic. In conclusion, we have established AOs as a suitable human system to decipher mechanisms of CF-driven respiratory infection by Mabs and propose boosting of the NRF2-NQO1 axis as a potential host-directed strategy to improve Mabs infection control.
Collapse
Affiliation(s)
- Stephen Adonai Leon-Icaza
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Salimata Bagayoko
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Romain Vergé
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Nino Iakobachvili
- M4i Nanoscopy Division, Maastricht University, Maastricht, Netherlands
| | - Chloé Ferrand
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Talip Aydogan
- Laboratory of Pathogen Host Interactions (LPHI), Université Montpellier, CNRS, Montpellier, France
| | - Célia Bernard
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Angelique Sanchez Dafun
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Marlène Murris-Espin
- Service de Pneumologie, Hôpital Larrey, CHU de Toulouse, Toulouse, France
- Centre de ressource et de compétence pour la mucoviscidose de l’adulte (CRCM adulte), CHU de Toulouse, Toulouse, France
| | - Julien Mazières
- Service de Pneumologie, Hôpital Larrey, CHU de Toulouse, Toulouse, France
| | - Pierre Jean Bordignon
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Serge Mazères
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | | | - Victoria Ramé
- Imactiv-3D SAS, 1 Place Pierre POTIER, Toulouse, France
| | | | - Julien Marcoux
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Marie-Pierre Bousquet
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Christian Chalut
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Christophe Guilhot
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Utrecht, Netherlands
| | - Peter J. Peters
- M4i Nanoscopy Division, Maastricht University, Maastricht, Netherlands
| | - Virginie Molle
- Laboratory of Pathogen Host Interactions (LPHI), Université Montpellier, CNRS, Montpellier, France
| | - Geanncarlo Lugo-Villarino
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Kaymeuang Cam
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Laurence Berry
- Laboratory of Pathogen Host Interactions (LPHI), Université Montpellier, CNRS, Montpellier, France
| | - Etienne Meunier
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Céline Cougoule
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| |
Collapse
|
9
|
Iazzi M, Sadeghi S, Gupta GD. A Proteomic Survey of the Cystic Fibrosis Transmembrane Conductance Regulator Surfaceome. Int J Mol Sci 2023; 24:11457. [PMID: 37511222 PMCID: PMC10380767 DOI: 10.3390/ijms241411457] [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: 06/07/2023] [Revised: 07/08/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
The aim of this review article is to collate recent contributions of proteomic studies to cystic fibrosis transmembrane conductance regulator (CFTR) biology. We summarize advances from these studies and create an accessible resource for future CFTR proteomic efforts. We focus our attention on the CFTR interaction network at the cell surface, thus generating a CFTR 'surfaceome'. We review the main findings about CFTR interactions and highlight several functional categories amongst these that could lead to the discovery of potential biomarkers and drug targets for CF.
Collapse
Affiliation(s)
| | | | - Gagan D. Gupta
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada
| |
Collapse
|
10
|
Pawelec KM, Tu E, Chakravarty S, Hix JML, Buchanan L, Kenney L, Buchanan F, Chatterjee N, Das S, Alessio A, Shapiro EM. Incorporating Tantalum Oxide Nanoparticles into Implantable Polymeric Biomedical Devices for Radiological Monitoring. Adv Healthc Mater 2023; 12:e2203167. [PMID: 36848875 PMCID: PMC10460461 DOI: 10.1002/adhm.202203167] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/19/2023] [Indexed: 03/01/2023]
Abstract
Longitudinal radiological monitoring of biomedical devices is increasingly important, driven by the risk of device failure following implantation. Polymeric devices are poorly visualized with clinical imaging, hampering efforts to use diagnostic imaging to predict failure and enable intervention. Introducing nanoparticle contrast agents into polymers is a potential method for creating radiopaque materials that can be monitored via computed tomography. However, the properties of composites may be altered with nanoparticle addition, jeopardizing device functionality. Thus, the material and biomechanical responses of model nanoparticle-doped biomedical devices (phantoms), created from 0-40 wt% tantalum oxide (TaOx ) nanoparticles in polycaprolactone and poly(lactide-co-glycolide) 85:15 and 50:50, representing non, slow, and fast degrading systems, respectively, are investigated. Phantoms degrade over 20 weeks in vitro in simulated physiological environments: healthy tissue (pH 7.4), inflammation (pH 6.5), and lysosomal conditions (pH 5.5), while radiopacity, structural stability, mechanical strength, and mass loss are monitored. The polymer matrix determines overall degradation kinetics, which increases with lower pH and higher TaOx content. Importantly, all radiopaque phantoms could be monitored for a full 20 weeks. Phantoms implanted in vivo and serially imaged demonstrate similar results. An optimal range of 5-20 wt% TaOx nanoparticles balances radiopacity requirements with implant properties, facilitating next-generation biomedical devices.
Collapse
Affiliation(s)
- Kendell M. Pawelec
- Department of RadiologyMichigan State University846 Service RdEast LansingMI48824USA
| | - Ethan Tu
- Department of Biomedical EngineeringMichigan State University775 Woodlot DrEast LansingMI48824USA
| | - Shatadru Chakravarty
- Department of RadiologyMichigan State University846 Service RdEast LansingMI48824USA
- Present address:
TechInsightsSuite 500, 1891 Robertson RdNepeanONK2H 5B7Canada
| | - Jeremy M. L. Hix
- Department of RadiologyMichigan State University846 Service RdEast LansingMI48824USA
- Institute for Quantitative Health Science and Engineering (IQ)Michigan State University775 Woodlot DrEast LansingMI48824USA
| | - Lane Buchanan
- Department of RadiologyMichigan State University846 Service RdEast LansingMI48824USA
| | - Legend Kenney
- Department of Biomedical EngineeringMichigan State University775 Woodlot DrEast LansingMI48824USA
| | - Foster Buchanan
- Department of RadiologyMichigan State University846 Service RdEast LansingMI48824USA
- Present address:
Lake Erie College of Osteopathic Medicine1858 W Grandview BlvdEriePA16509USA
| | - Nandini Chatterjee
- Department of RadiologyMichigan State University846 Service RdEast LansingMI48824USA
| | - Subhashri Das
- Department of RadiologyMichigan State University846 Service RdEast LansingMI48824USA
| | - Adam Alessio
- Department of RadiologyMichigan State University846 Service RdEast LansingMI48824USA
- Department of Biomedical EngineeringMichigan State University775 Woodlot DrEast LansingMI48824USA
- Department of Computational Mathematics Science EngineeringMichigan State University428 S. Shaw LnEast LansingMI48824USA
| | - Erik M. Shapiro
- Department of RadiologyMichigan State University846 Service RdEast LansingMI48824USA
| |
Collapse
|
11
|
Abstract
Cystic fibrosis (CF) is a monogenic recessive genetic disorder caused by mutations in the CF Transmembrane-conductance Regulator gene (CFTR). Remarkable progress in basic research has led to the discovery of highly effective CFTR modulators. Now ~90% of CF patients are treatable. However, these modulator therapies are not curative and do not cover the full spectrum of CFTR mutations. Thus, there is a continued need to develop a complete and durable therapy that can treat all CF patients once and for all. As CF is a genetic disease, the ultimate therapy would be in-situ repair of the genetic lesions in the genome. Within the past few years, new technologies, such as CRISPR/Cas gene editing, have emerged as an appealing platform to revise the genome, ushering in a new era of genetic therapy. This review provided an update on this rapidly evolving field and the status of adapting the technology for CF therapy.
Collapse
Affiliation(s)
- Guoshun Wang
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, CSRB 607, 533 Bolivar Street, New Orleans, LA 70112, USA
| |
Collapse
|
12
|
Zhang Q, Li Y, Jian Y, Li M, Wang X. Lysosomal chloride transporter CLH-6 protects lysosome membrane integrity via cathepsin activation. J Cell Biol 2023; 222:e202210063. [PMID: 37058288 PMCID: PMC10114921 DOI: 10.1083/jcb.202210063] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 02/11/2023] [Accepted: 03/10/2023] [Indexed: 04/15/2023] Open
Abstract
Lysosomal integrity is vital for cell homeostasis, but the underlying mechanisms are poorly understood. Here, we identify CLH-6, the C. elegans ortholog of the lysosomal Cl-/H+ antiporter ClC-7, as an important factor for protecting lysosomal integrity. Loss of CLH-6 affects lysosomal degradation, causing cargo accumulation and membrane rupture. Reducing cargo delivery or increasing CPL-1/cathepsin L or CPR-2/cathepsin B expression suppresses these lysosomal defects. Inactivation of CPL-1 or CPR-2, like CLH-6 inactivation, affects cargo digestion and causes lysosomal membrane rupture. Thus, loss of CLH-6 impairs cargo degradation, leading to membrane damage of lysosomes. In clh-6(lf) mutants, lysosomes are acidified as in wild type but contain lower chloride levels, and cathepsin B and L activities are significantly reduced. Cl- binds to CPL-1 and CPR-2 in vitro, and Cl- supplementation increases lysosomal cathepsin B and L activities. Altogether, these findings suggest that CLH-6 maintains the luminal chloride levels required for cathepsin activity, thus facilitating substrate digestion to protect lysosomal membrane integrity.
Collapse
Affiliation(s)
- Qianqian Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Youli Jian
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Meijiao Li
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan, and Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Xiaochen Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
13
|
Dolman NJ, Kilgore JA. A Review of Reagents for Fluorescence Microscopy of Cellular Compartments and Structures, Part I: BacMam Labeling and Reagents for Vesicular Structures. Curr Protoc 2023; 3:e751. [PMID: 37311031 DOI: 10.1002/cpz1.751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Fluorescent labeling of vesicular structures in cultured cells, particularly for live cells, can be challenging for a number of reasons. The first challenge is to identify a reagent that will be specific enough where some structures have a number of potential reagents and others very few options. The emergence of BacMam constructs has provided more easy-to-use choices. Presented here is a discussion of BacMam constructs as well as a review of commercially available reagents for labeling vesicular structures in cells, including endosomes, peroxisomes, lysosomes, and autophagosomes, complete with a featured reagent, recommended protocol, troubleshooting guide, and example image for each structure. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Delivering targeted fluorescent proteins using pre-made, high-titer BacMam constructs Alternate Protocol 1: Non-pseudo-typed BacMam viruses in standard cell types and pseudo-typed BacMam viruses in hard-to-transduce cell types Basic Protocol 2: Labeling endosomes: pHrodo™-10k-dextran Basic Protocol 3: Labeling peroxisomes: BacMam 2.0 CellLight™ Peroxisome-GFP Alternate Protocol 2: Labeling peroxisomes using antibodies Basic Protocol 4: Labeling autophagosomes: Transduction of cells with Premo™ Autophagy Sensor GFP-LC3B Alternate Protocol 3: Labeling autophagosomes using antibodies Basic Protocol 5: Labeling lysosomes: LysoTracker Red DND-99.
Collapse
|
14
|
Lotti V, Lagni A, Diani E, Sorio C, Gibellini D. Crosslink between SARS-CoV-2 replication and cystic fibrosis hallmarks. Front Microbiol 2023; 14:1162470. [PMID: 37250046 PMCID: PMC10213757 DOI: 10.3389/fmicb.2023.1162470] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/21/2023] [Indexed: 05/31/2023] Open
Abstract
SARS-CoV-2, the etiological cause of the COVID-19 pandemic, can cause severe illness in certain at-risk populations, including people with cystic fibrosis (pwCF). Nevertheless, several studies indicated that pwCF do not have higher risks of SARS-CoV-2 infection nor do they demonstrate worse clinical outcomes than those of the general population. Recent in vitro studies indicate cellular and molecular processes to be significant drivers in pwCF lower infection rates and milder symptoms than expected in cases of SARS-CoV-2 infection. These range from cytokine releases to biochemical alterations leading to morphological rearrangements inside the cells associated with CFTR impairment. Based on available data, the reported low incidence of SARS-CoV-2 infection among pwCF is likely a result of several variables linked to CFTR dysfunction, such as thick mucus, IL-6 reduction, altered ACE2 and TMPRSS2 processing and/or functioning, defective anions exchange, and autophagosome formation. An extensive analysis of the relation between SARS-CoV-2 infection and pwCF is essential to elucidate the mechanisms involved in this lower-than-expected infection impact and to possibly suggest potential new antiviral strategies.
Collapse
Affiliation(s)
- Virginia Lotti
- Microbiology Section, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Anna Lagni
- Microbiology Section, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Erica Diani
- Microbiology Section, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Claudio Sorio
- General Pathology Section, Department of Medicine, University of Verona, Verona, Italy
| | - Davide Gibellini
- Microbiology Section, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| |
Collapse
|
15
|
Pandey S, Anang V, Singh S, Seth S, Bhatt AN, Kalra N, Manda K, Soni R, Roy BG, Natarajan K, Dwarakanath BS. Dietary administration of the glycolytic inhibitor 2-deoxy-D-glucose reduces endotoxemia-induced inflammation and oxidative stress: Implications in PAMP-associated acute and chronic pathology. Front Pharmacol 2023; 14:940129. [PMID: 37234710 PMCID: PMC10206263 DOI: 10.3389/fphar.2023.940129] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Pathogen-associated molecular patterns (PAMPs) like bacterial cell wall components and viral nucleic acids are known ligands of innate inflammatory receptors that trigger multiple inflammatory pathways that may result in acute inflammation and oxidative stress-driven tissue and organ toxicity. When dysregulated, this inflammation may lead to acute toxicity and multiorgan failure. Inflammatory events are often driven by high energy demands and macromolecular biosynthesis. Therefore, we proposed that targeting the metabolism of lipopolysaccharide (LPS)-driven inflammatory events, using an energy restriction approach, can be an effective strategy to prevent the acute or chronic detrimental effects of accidental or seasonal bacterial and other pathogenic exposures. In the present study, we investigated the potential of energy restriction mimetic agent (ERMA) 2-deoxy-D-glucose (2-DG) in targeting the metabolism of inflammatory events during LPS-elicited acute inflammatory response. Mice fed with 2-DG as a dietary component in drinking water showed reduced LPS-driven inflammatory processes. Dietary 2-DG reduced LPS-induced lung endothelial damage and oxidative stress by strengthening the antioxidant defense system and limiting the activation and expression of inflammatory proteins, viz., P-Stat-3, NfκΒ, and MAP kinases. This was accompanied by decreased TNF, IL-1β, and IL-6 levels in peripheral blood and bronchoalveolar lavage fluid (BALF). 2-DG also reduced the infiltration of PMNCs (polymorphonuclear cells) in inflamed tissues. Altered glycolysis and improved mitochondrial activity in 2-DG-treated RAW 264.7 macrophage cells suggested possible impairment of macrophage metabolism and, therefore, activation in macrophages. Taken together, the present study suggests that inclusion of glycolytic inhibitor 2-DG as a part of the diet can be helpful in preventing the severity and poor prognosis associated with inflammatory events during bacterial and other pathogenic exposures.
Collapse
Affiliation(s)
- Sanjay Pandey
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Vandana Anang
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Saurabh Singh
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Saurabh Seth
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Anant Narayan Bhatt
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Namita Kalra
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Kailash Manda
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Ravi Soni
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Bal Gangadhar Roy
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - K. Natarajan
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Bilikere S. Dwarakanath
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| |
Collapse
|
16
|
Pawelec KM, Tu E, Chakravarty S, Hix JM, Buchanan L, Kenney L, Buchanan F, Chatterjee N, Das S, Alessio A, Shapiro EM. Incorporating Radiopacity into Implantable Polymeric Biomedical Devices for Clinical Radiological Monitoring. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.06.523025. [PMID: 36711467 PMCID: PMC9881976 DOI: 10.1101/2023.01.06.523025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Longitudinal radiological monitoring of biomedical devices is increasingly important, driven by risk of device failure following implantation. Polymeric devices are poorly visualized with clinical imaging, hampering efforts to use diagnostic imaging to predict failure and enable intervention. Introducing nanoparticle contrast agents into polymers is a potential method for creating radiopaque materials that can be monitored via computed tomography. However, properties of composites may be altered with nanoparticle addition, jeopardizing device functionality. This, we investigated material and biomechanical response of model nanoparticle-doped biomedical devices (phantoms), created from 0-40wt% TaO x nanoparticles in polycaprolactone, poly(lactide-co-glycolide) 85:15 and 50:50, representing non-, slow and fast degrading systems, respectively. Phantoms degraded over 20 weeks in vitro, in simulated physiological environments: healthy tissue (pH 7.4), inflammation (pH 6.5), and lysosomal conditions (pH 5.5), while radiopacity, structural stability, mechanical strength and mass loss were monitored. The polymer matrix determined overall degradation kinetics, which increased with lower pH and higher TaO x content. Importantly, all radiopaque phantoms could be monitored for a full 20-weeks. Phantoms implanted in vivo and serially imaged, demonstrated similar results. An optimal range of 5-20wt% TaO x nanoparticles balanced radiopacity requirements with implant properties, facilitating next-generation biomedical devices.
Collapse
Affiliation(s)
| | - Ethan Tu
- Michigan State University, Dept Biomedical Engineering, East Lansing, MI 48823
| | | | - Jeremy Ml Hix
- Michigan State University, Dept Radiology, East Lansing, MI 48823
- Michigan State University, Institute for Quantitative Health Science and Engineering (IQ), East Lansing, MI 48823
| | - Lane Buchanan
- Michigan State University, Dept Radiology, East Lansing, MI 48823
| | - Legend Kenney
- Michigan State University, Dept Biomedical Engineering, East Lansing, MI 48823
| | - Foster Buchanan
- Michigan State University, Dept Radiology, East Lansing, MI 48823
| | | | - Subhashri Das
- Michigan State University, Dept Radiology, East Lansing, MI 48823
| | - Adam Alessio
- Michigan State University, Dept Radiology, East Lansing, MI 48823
- Michigan State University, Dept Biomedical Engineering, East Lansing, MI 48823
- Michigan State University, Dept of Computational Mathematics Science Engineering, East Lansing, MI 48823
| | - Erik M Shapiro
- Michigan State University, Dept Radiology, East Lansing, MI 48823
| |
Collapse
|
17
|
Carey RM, Palmer JN, Adappa ND, Lee RJ. Loss of CFTR function is associated with reduced bitter taste receptor-stimulated nitric oxide innate immune responses in nasal epithelial cells and macrophages. Front Immunol 2023; 14:1096242. [PMID: 36742335 PMCID: PMC9890060 DOI: 10.3389/fimmu.2023.1096242] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023] Open
Abstract
Introduction Bitter taste receptors (T2Rs) are G protein-coupled receptors identified on the tongue but expressed all over the body, including in airway cilia and macrophages, where T2Rs serve an immune role. T2R isoforms detect bitter metabolites (quinolones and acyl-homoserine lactones) secreted by gram negative bacteria, including Pseudomonas aeruginosa, a major pathogen in cystic fibrosis (CF). T2R activation by bitter bacterial products triggers calcium-dependent nitric oxide (NO) production. In airway cells, the NO increases mucociliary clearance and has direct antibacterial properties. In macrophages, the same pathway enhances phagocytosis. Because prior studies linked CF with reduced NO, we hypothesized that CF cells may have reduced T2R/NO responses, possibly contributing to reduced innate immunity in CF. Methods Immunofluorescence, qPCR, and live cell imaging were used to measure T2R localization, calcium and NO signaling, ciliary beating, and antimicrobial responses in air-liquid interface cultures of primary human nasal epithelial cells and immortalized bronchial cell lines. Immunofluorescence and live cell imaging was used to measure T2R signaling and phagocytosis in primary human monocyte-derived macrophages. Results Primary nasal epithelial cells from both CF and non-CF patients exhibited similar T2R expression, localization, and calcium signals. However, CF cells exhibited reduced NO production also observed in immortalized CFBE41o- CF cells and non-CF 16HBE cells CRISPR modified with CF-causing mutations in the CF transmembrane conductance regulator (CFTR). NO was restored by VX-770/VX-809 corrector/potentiator pre-treatment, suggesting reduced NO in CF cells is due to loss of CFTR function. In nasal cells, reduced NO correlated with reduced ciliary and antibacterial responses. In primary human macrophages, inhibition of CFTR reduced NO production and phagocytosis during T2R stimulation. Conclusions Together, these data suggest an intrinsic deficiency in T2R/NO signaling caused by loss of CFTR function that may contribute to intrinsic susceptibilities of CF patients to P. aeruginosa and other gram-negative bacteria that activate T2Rs.
Collapse
Affiliation(s)
- Ryan M Carey
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - James N Palmer
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Nithin D Adappa
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Robert J Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
18
|
Li X, Kolling FW, Aridgides D, Mellinger D, Ashare A, Jakubzick CV. ScRNA-seq expression of IFI27 and APOC2 identifies four alveolar macrophage superclusters in healthy BALF. Life Sci Alliance 2022; 5:e202201458. [PMID: 35820705 PMCID: PMC9275597 DOI: 10.26508/lsa.202201458] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 12/15/2022] Open
Abstract
Alveolar macrophages (AMs) reside on the luminal surface of the airways and alveoli, ensuring proper gas exchange by ingesting cellular debris and pathogens, and regulating inflammatory responses. Therefore, understanding the heterogeneity and diverse roles played by AMs, interstitial macrophages, and recruited monocytes is critical for treating airway diseases. We performed single-cell RNA sequencing on 113,213 bronchoalveolar lavage cells from four healthy and three uninflamed cystic fibrosis subjects and identified two MARCKS+LGMN+IMs, FOLR2+SELENOP+ and SPP1+PLA2G7+ IMs, monocyte subtypes, DC1, DC2, migDCs, plasmacytoid DCs, lymphocytes, epithelial cells, and four AM superclusters (families) based on the gene expression of IFI27 and APOC2 These four AM families have at least eight distinct functional members (subclusters) named after their differentially expressed gene(s): IGF1, CCL18, CXCL5, cholesterol, chemokine, metallothionein, interferon, and small-cluster AMs. Interestingly, the chemokine cluster further divides with each subcluster selectively expressing a unique combination of chemokines. One of the most striking observations, besides the heterogeneity, is the conservation of AM family members in relatively equal ratio across all AM superclusters and individuals. Transcriptional data and TotalSeq technology were used to investigate cell surface markers that distinguish resident AMs from recruited monocytes. Last, other AM datasets were projected onto our dataset. Similar AM superclusters and functional subclusters were observed, along with a significant increase in chemokine and IFN AM subclusters in individuals infected with COVID-19. Overall, functional specializations of the AM subclusters suggest that there are highly regulated AM niches with defined programming states, highlighting a clear division of labor.
Collapse
Affiliation(s)
- Xin Li
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH, USA
| | - Fred W Kolling
- Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Hanover, NH, USA
| | - Daniel Aridgides
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Diane Mellinger
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Alix Ashare
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH, USA
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Claudia V Jakubzick
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH, USA
| |
Collapse
|
19
|
Meoli A, Eickmeier O, Pisi G, Fainardi V, Zielen S, Esposito S. Impact of CFTR Modulators on the Impaired Function of Phagocytes in Cystic Fibrosis Lung Disease. Int J Mol Sci 2022; 23:12421. [PMID: 36293274 PMCID: PMC9604330 DOI: 10.3390/ijms232012421] [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: 08/31/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
Cystic fibrosis (CF), the most common genetically inherited disease in Caucasian populations, is a multi-systemic life-threatening autosomal recessive disorder caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. In 2012, the arrival of CFTR modulators (potentiators, correctors, amplifiers, stabilizers, and read-through agents) revolutionized the therapeutic approach to CF. In this review, we examined the physiopathological mechanism of chronic dysregulated innate immune response in the lungs of CF patients with pulmonary involvement with particular reference to phagocytes, critically analyzing the role of CFTR modulators in influencing and eventually restoring their function. Our literature review highlighted that the role of CFTR in the lungs is crucial not only for the epithelial function but also for host defense, with particular reference to phagocytes. In macrophages and neutrophils, the CFTR dysfunction compromises both the intricate process of phagocytosis and the mechanisms of initiation and control of inflammation which then reverberates on the epithelial environment already burdened by the chronic colonization of pathogens leading to irreversible tissue damage. In this context, investigating the impact of CFTR modulators on phagocytic functions is therefore crucial not only for explaining the underlying mechanisms of pleiotropic effects of these molecules but also to better understand the physiopathological basis of this disease, still partly unexplored, and to develop new complementary or alternative therapeutic approaches.
Collapse
Affiliation(s)
- Aniello Meoli
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Division of Allergy, Pulmonology and Cystic Fibrosis, Department for Children and Adolescents, University Hospital, 60431 Frankfurt, Germany
| | - Olaf Eickmeier
- Division of Allergy, Pulmonology and Cystic Fibrosis, Department for Children and Adolescents, University Hospital, 60431 Frankfurt, Germany
| | - Giovanna Pisi
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Valentina Fainardi
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Stefan Zielen
- Division of Allergy, Pulmonology and Cystic Fibrosis, Department for Children and Adolescents, University Hospital, 60431 Frankfurt, Germany
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| |
Collapse
|
20
|
Yu Y, Zhang Z, Walpole GFW, Yu Y. Kinetics of phagosome maturation is coupled to their intracellular motility. Commun Biol 2022; 5:1014. [PMID: 36163370 PMCID: PMC9512794 DOI: 10.1038/s42003-022-03988-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 09/12/2022] [Indexed: 11/09/2022] Open
Abstract
Immune cells degrade internalized pathogens in phagosomes through sequential biochemical changes. The degradation must be fast enough for effective infection control. The presumption is that each phagosome degrades cargos autonomously with a distinct but stochastic kinetic rate. However, here we show that the degradation kinetics of individual phagosomes is not stochastic but coupled to their intracellular motility. By engineering RotSensors that are optically anisotropic, magnetic responsive, and fluorogenic in response to degradation activities in phagosomes, we monitored cargo degradation kinetics in single phagosomes simultaneously with their translational and rotational dynamics. We show that phagosomes that move faster centripetally are more likely to encounter and fuse with lysosomes, thereby acidifying faster and degrading cargos more efficiently. The degradation rates increase nearly linearly with the translational and rotational velocities of phagosomes. Our results indicate that the centripetal motion of phagosomes functions as a clock for controlling the progression of cargo degradation.
Collapse
Affiliation(s)
- Yanqi Yu
- Department of Chemistry, Indiana University, Bloomington, IN, 47405-7102, USA
| | - Zihan Zhang
- Department of Chemistry, Indiana University, Bloomington, IN, 47405-7102, USA
| | - Glenn F W Walpole
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, IN, 47405-7102, USA.
| |
Collapse
|
21
|
Jaganathan D, Bruscia EM, Kopp BT. Emerging Concepts in Defective Macrophage Phagocytosis in Cystic Fibrosis. Int J Mol Sci 2022; 23:7750. [PMID: 35887098 PMCID: PMC9319215 DOI: 10.3390/ijms23147750] [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] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 01/18/2023] Open
Abstract
Cystic fibrosis (CF) is caused by mutations of the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Chronic inflammation and decline in lung function are major reasons for morbidity in CF. Mutant CFTR expressed in phagocytic cells such as macrophages contributes to persistent infection, inflammation, and lung disease in CF. Macrophages play a central role in innate immunity by eliminating pathogenic microbes by a process called phagocytosis. Phagocytosis is required for tissue homeostasis, balancing inflammation, and crosstalk with the adaptive immune system for antigen presentation. This review focused on (1) current understandings of the signaling underlying phagocytic mechanisms; (2) existing evidence for phagocytic dysregulation in CF; and (3) the emerging role of CFTR modulators in influencing CF phagocytic function. Alterations in CF macrophages from receptor initiation to phagosome formation are linked to disease progression in CF. A deeper understanding of macrophages in the context of CFTR and phagocytosis proteins at each step of phagosome formation might contribute to the new therapeutic development of dysregulated innate immunity in CF. Therefore, the review also indicates future areas of research in the context of CFTR and macrophages.
Collapse
Affiliation(s)
- Devi Jaganathan
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
| | - Emanuela M. Bruscia
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06510, USA;
| | - Benjamin T. Kopp
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
- Division of Pulmonary Medicine, Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
| |
Collapse
|
22
|
Margaroli C, Horati H, Garratt LW, Giacalone VD, Schofield C, Dittrich AS, Rosenow T, Dobosh BS, Lim HS, Frey DL, Veltman M, Silva GL, Brown MR, Schultz C, Tiddens HAWM, Ranganathan S, Chandler JD, Qiu P, Peng L, Scholte BJ, Mall MA, Kicic A, Guglani L, Stick SM, Janssens HM, Tirouvanziam R. Macrophage PD-1 associates with neutrophilia and reduced bacterial killing in early cystic fibrosis airway disease. J Cyst Fibros 2022; 21:967-976. [PMID: 35732550 DOI: 10.1016/j.jcf.2022.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/17/2022] [Accepted: 06/02/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND Macrophages are the major resident immune cells in human airways coordinating responses to infection and injury. In cystic fibrosis (CF), neutrophils are recruited to the airways shortly after birth, and actively exocytose damaging enzymes prior to chronic infection, suggesting a potential defect in macrophage immunomodulatory function. Signaling through the exhaustion marker programmed death protein 1 (PD-1) controls macrophage function in cancer, sepsis, and airway infection. Therefore, we sought to identify potential associations between macrophage PD-1 and markers of airway disease in children with CF. METHODS Blood and bronchoalveolar lavage fluid (BALF) were collected from 45 children with CF aged 3 to 62 months and structural lung damage was quantified by computed tomography. The phenotype of airway leukocytes was assessed by flow cytometry, while the release of enzymes and immunomodulatory mediators by molecular assays. RESULTS Airway macrophage PD-1 expression correlated positively with structural lung damage, neutrophilic inflammation, and infection. Interestingly, even in the absence of detectable infection, macrophage PD-1 expression was elevated and correlated with neutrophilic inflammation. In an in vitro model mimicking leukocyte recruitment into CF airways, soluble mediators derived from recruited neutrophils directly induced PD-1 expression on recruited monocytes/macrophages, suggesting a causal link between neutrophilic inflammation and macrophage PD-1 expression in CF. Finally, blockade of PD-1 in a short-term culture of CF BALF leukocytes resulted in improved pathogen clearance. CONCLUSION Taken together, these findings suggest that in early CF lung disease, PD-1 upregulation associates with airway macrophage exhaustion, neutrophil takeover, infection, and structural damage.
Collapse
Affiliation(s)
- Camilla Margaroli
- Department of Pediatrics, Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, IMPEDE-CF Program, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Hamed Horati
- Department of Pediatrics, Div. of Respiratory Medicine and Allergology, I-BALL program, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, Rotterdam, The Netherlands
| | - Luke W Garratt
- AREST-CF Program, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Vincent D Giacalone
- Department of Pediatrics, Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, IMPEDE-CF Program, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Craig Schofield
- AREST-CF Program, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - A Susanne Dittrich
- Department of Translational Pulmonology, Translational Lung Research Center (TLRC), German Center for Lung Research (DZL) and Department of Pulmonology, and Critical Care Medicine, Thoraxklinik at the University of Heidelberg, Heidelberg, Germany
| | - Tim Rosenow
- AREST-CF Program, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Brian S Dobosh
- Department of Pediatrics, Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, IMPEDE-CF Program, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Hong S Lim
- Department of Biomedical engineering, The Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America
| | - Dario L Frey
- Department of Translational Pulmonology, Translational Lung Research Center (TLRC), German Center for Lung Research (DZL) and Department of Pulmonology, and Critical Care Medicine, Thoraxklinik at the University of Heidelberg, Heidelberg, Germany
| | - Mieke Veltman
- Department of Pediatrics, Div. of Respiratory Medicine and Allergology, I-BALL program, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, Rotterdam, The Netherlands
| | - George L Silva
- Department of Pediatrics, Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, IMPEDE-CF Program, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Milton R Brown
- Department of Pediatrics, Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, IMPEDE-CF Program, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Carsten Schultz
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR, United States of America
| | - Harm A W M Tiddens
- Department of Pediatrics, Div. of Respiratory Medicine and Allergology, I-BALL program, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, Rotterdam, The Netherlands
| | - Sarath Ranganathan
- Department of Pediatrics, University of Melbourne, Melbourne, Australia; Murdoch Children's Research Institute, and Department of Respiratory and Sleep Medicine, Royal Children's Hospital, Parkville, Australia
| | - Joshua D Chandler
- Department of Pediatrics, Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, IMPEDE-CF Program, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Peng Qiu
- Department of Biomedical engineering, The Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America
| | - Limin Peng
- Department of Pediatrics, Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, IMPEDE-CF Program, Emory University School of Medicine, Atlanta, GA, United States of America; Department of Biostatistics, Emory University School of Public Health, Atlanta, GA, United States of America
| | - Bob J Scholte
- Department of Pediatrics, Div. of Respiratory Medicine and Allergology, I-BALL program, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, Rotterdam, The Netherlands
| | - Marcus A Mall
- Department of Translational Pulmonology, Translational Lung Research Center (TLRC), German Center for Lung Research (DZL) and Department of Pulmonology, and Critical Care Medicine, Thoraxklinik at the University of Heidelberg, Heidelberg, Germany; Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Anthony Kicic
- AREST-CF Program, Telethon Kids Institute, University of Western Australia, Perth, Australia; Department of Respiratory and Sleep Medicine, Perth Children's Hospital and Faculty of Medicine, University of Western Australia, Perth, Western Australia, Australia; School of Public Heath, Curtin University, Perth, Western Australia, Australia
| | - Lokesh Guglani
- Department of Pediatrics, Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, IMPEDE-CF Program, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Stephen M Stick
- AREST-CF Program, Telethon Kids Institute, University of Western Australia, Perth, Australia; Department of Respiratory and Sleep Medicine, Perth Children's Hospital and Faculty of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Hettie M Janssens
- Department of Pediatrics, Div. of Respiratory Medicine and Allergology, I-BALL program, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, Rotterdam, The Netherlands
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, IMPEDE-CF Program, Emory University School of Medicine, Atlanta, GA, United States of America.
| |
Collapse
|
23
|
Malla JA, Upadhyay A, Ghosh P, Mondal D, Mondal A, Sharma S, Talukdar P. Chloride Transport across Liposomes and Cells by Nontoxic 3-(1 H-1,2,3-Triazol-1-yl)benzamides. Org Lett 2022; 24:4124-4128. [PMID: 35657329 DOI: 10.1021/acs.orglett.2c01219] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Synthetic anion transmembrane transporters are adding new aspirations for treating channelopathies by replacing defective ion channels. The availability of such suitable candidates is still infrequent due to the associated toxicity. Here, we report 3-(1H-1,2,3-triazol-1-yl)benzamides as transmembrane anion carriers, nontoxic to cells. The selective and electrogenic chloride transport activity was established by fluorescence and ion selective electrode-based assays. MQAE assay confirmed the chloride uptake into the cells by the nontoxic compounds.
Collapse
Affiliation(s)
- Javid Ahmad Malla
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, 411008 Pune, Maharashtra, India
| | - Avisikta Upadhyay
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, 411008 Pune, Maharashtra, India
| | - Pulak Ghosh
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, 411008 Pune, Maharashtra, India
| | - Debashis Mondal
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, 411008 Pune, Maharashtra, India
| | - Abhishek Mondal
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, 411008 Pune, Maharashtra, India
| | - Shilpy Sharma
- Department of Biotechnology, Savitribai Phule Pune University, 411007 Pune, Maharashtra, India
| | - Pinaki Talukdar
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, 411008 Pune, Maharashtra, India
| |
Collapse
|
24
|
Rezende GL, Nakanishi M, Couto SCP, Martins CLFS, Sampaio ALL, Albuquerque LFF, Kückelhaus SAS, Muniz-Junqueira MI. Alterations in innate immune responses of patients with chronic rhinosinusitis related to cystic fibrosis. PLoS One 2022; 17:e0267986. [PMID: 35522627 PMCID: PMC9075614 DOI: 10.1371/journal.pone.0267986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/19/2022] [Indexed: 11/19/2022] Open
Abstract
The role of phagocytes of children with cystic fibrosis (CF) associated with different phenotypes of chronic rhinosinusitis (CRS) is unclear. The aim of this study was to evaluate the phagocytic capacity of blood neutrophils and monocytes and production of superoxide anion by phagocytes in patients with CF with or without chronic rhinosinusitis and with or without nasal polyps (NP). This cross-sectional study was established in 2015-2017 in a tertiary reference center to the CF treatment, Brasilia, Brazil. Sample included 30 children volunteers with CRS related to CF (n = 16) and control subjects (n = 14). Epidemiological and clinical data were compared. Collection of 15 mL of peripheral blood and nasal endoscopy to identify the presence or absence of nasal polyps (NP) were performed. Phagocytosis of Saccharomyces cerevisiae by pathogen-associated molecular pattern receptors and opsonin receptors was assessed. Superoxide anion production was evaluated. The control group showed a higher phagocytic index to monocytes and neutrophils than to the CF or CF+CRS with NP groups [Kruskal-Wallis p = 0.0025] when phagocytosis were evaluated by pathogen-associated molecular pattern receptors (5 yeasts/cell). The phagocytic index of the CF+CRS without NP group was higher than in the CF+CRS with NP group (Kruskal-Wallis p = 0.0168). In the control group, the percentage of phagocytes involved in phagocytosis and superoxide anion production (74.0 ± 9.6%) were higher in all CF groups (p < 0,0001). The innate immune response, represented by phagocytic activity and superoxide anion production by monocytes and neutrophils was more impaired in patients with CF related or not related to CRS than in the control group. However, the phagocytic function of patients without NP showed less impairment.
Collapse
Affiliation(s)
- Gustavo L. Rezende
- Hospital de Base, Brasília, Federal District, Brazil
- Laboratory of Cellular Immunology, Pathology, Faculty of Medicine, University of Brasilia, Brasília, Federal District, Brazil
- Nucleus of Research in Applied Morphology and Immunology, Morphology, Faculty of Medicine, University de Brasília, Brasília, Federal District, Brazil
| | - Marcio Nakanishi
- Laboratory of Cellular Immunology, Pathology, Faculty of Medicine, University of Brasilia, Brasília, Federal District, Brazil
- Department of Otolaryngology, Faculty of Medicine, University of Brasilia, Brasília, Federal District, Brazil
- D’Or Institute for Research and Education (IDOR), Brasília, Federal District, Brazil
| | - Shirley C. P. Couto
- Laboratory of Cellular Immunology, Pathology, Faculty of Medicine, University of Brasilia, Brasília, Federal District, Brazil
| | - Carmen L. F. S. Martins
- Laboratory of Cellular Immunology, Pathology, Faculty of Medicine, University of Brasilia, Brasília, Federal District, Brazil
- Department of Pediatric, Faculty of Medicine, University of Brasilia, Brasilia, Distrito Federal, Brazil
| | - André L. L. Sampaio
- Department of Otolaryngology, Faculty of Medicine, University of Brasilia, Brasília, Federal District, Brazil
| | - Lucas F. F. Albuquerque
- Nucleus of Research in Applied Morphology and Immunology, Morphology, Faculty of Medicine, University de Brasília, Brasília, Federal District, Brazil
| | - Selma A. S. Kückelhaus
- Laboratory of Cellular Immunology, Pathology, Faculty of Medicine, University of Brasilia, Brasília, Federal District, Brazil
- Nucleus of Research in Applied Morphology and Immunology, Morphology, Faculty of Medicine, University de Brasília, Brasília, Federal District, Brazil
| | - Maria I. Muniz-Junqueira
- Laboratory of Cellular Immunology, Pathology, Faculty of Medicine, University of Brasilia, Brasília, Federal District, Brazil
- Nucleus of Research in Applied Morphology and Immunology, Morphology, Faculty of Medicine, University de Brasília, Brasília, Federal District, Brazil
| |
Collapse
|
25
|
Badr A, Eltobgy M, Krause K, Hamilton K, Estfanous S, Daily KP, Abu Khweek A, Hegazi A, Anne MNK, Carafice C, Robledo-Avila F, Saqr Y, Zhang X, Bonfield TL, Gavrilin MA, Partida-Sanchez S, Seveau S, Cormet-Boyaka E, Amer AO. CFTR Modulators Restore Acidification of Autophago-Lysosomes and Bacterial Clearance in Cystic Fibrosis Macrophages. Front Cell Infect Microbiol 2022; 12:819554. [PMID: 35252032 PMCID: PMC8890004 DOI: 10.3389/fcimb.2022.819554] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 01/19/2022] [Indexed: 12/17/2022] Open
Abstract
Cystic fibrosis (CF) human and mouse macrophages are defective in their ability to clear bacteria such as Burkholderia cenocepacia. The autophagy process in CF (F508del) macrophages is halted, and the underlying mechanism remains unclear. Furthermore, the role of CFTR in maintaining the acidification of endosomal and lysosomal compartments in CF cells has been a subject of debate. Using 3D reconstruction of z-stack confocal images, we show that CFTR is recruited to LC3-labeled autophagosomes harboring B. cenocepacia. Using several complementary approaches, we report that CF macrophages display defective lysosomal acidification and degradative function for cargos destined to autophagosomes, whereas non-autophagosomal cargos are effectively degraded within acidic compartments. Notably, treatment of CF macrophages with CFTR modulators (tezacaftor/ivacaftor) improved the autophagy flux, lysosomal acidification and function, and bacterial clearance. In addition, CFTR modulators improved CFTR function as demonstrated by patch-clamp. In conclusion, CFTR regulates the acidification of a specific subset of lysosomes that specifically fuse with autophagosomes. Therefore, our study describes a new biological location and function for CFTR in autophago-lysosomes and clarifies the long-standing discrepancies in the field.
Collapse
Affiliation(s)
- Asmaa Badr
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
- Clinical Pathology Department, College of Medicine, Mansoura University, Mansoura, Egypt
| | - Mostafa Eltobgy
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Kathrin Krause
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
- Max Planck Unit for the Science of Pathogens, Berlin, Germany
| | - Kaitlin Hamilton
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Shady Estfanous
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Kylene P. Daily
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Arwa Abu Khweek
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
- Department of Biology and Biochemistry, Birzeit University, West Bank, Palestine
| | - Ahmad Hegazi
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Midhun N. K. Anne
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Cierra Carafice
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Frank Robledo-Avila
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Youssra Saqr
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Xiaoli Zhang
- Center for Biostatistics, Ohio State University, Columbus, OH, United States
| | - Tracey L. Bonfield
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Mikhail A. Gavrilin
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Columbus, OH, United States
| | | | - Stephanie Seveau
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Estelle Cormet-Boyaka
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Amal O. Amer
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
26
|
Riazanski V, Mauleon G, Zimnicka AM, Chen S, Nelson DJ. Phagosomal chloride dynamics in the alveolar macrophage. iScience 2022; 25:103636. [PMID: 35024579 PMCID: PMC8733233 DOI: 10.1016/j.isci.2021.103636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 10/11/2021] [Accepted: 12/14/2021] [Indexed: 12/04/2022] Open
Abstract
Acidification in intracellular organelles is tightly linked to the influx of Cl- counteracting proton translocation by the electrogenic V-ATPase. We quantified the dynamics of Cl- transfer accompanying cargo incorporation into single phagosomes in alveolar macrophages (AMs). Phagosomal Cl- concentration and acidification magnitude were followed in real time with maximal acidification achieved at levels of approximately 200 mM. Live cell confocal microscopy verified that phagosomal Cl- influx utilized predominantly the Cl- channel CFTR. Relative levels of elemental chlorine (Cl) in hard X-ray fluorescence microprobe (XFM) analysis within single phagosomes validated the increase in Cl- content. XFM revealed the complex interplay between elemental K content inside the phagosome and changes in Cl- during phagosomal particle uptake. Cl- -dependent changes in phagosomal membrane potential were obtained using second harmonic generation (SHG) microscopy. These studies provide a mechanistic insight for screening studies in drug development targeting pulmonary inflammatory disease.
Collapse
Affiliation(s)
- Vladimir Riazanski
- The University of Chicago, Department of Pharmacological and Physiological Sciences, 947 E. 58th Street, MC 0926, Chicago, IL 60637, USA
| | - Gerardo Mauleon
- The University of Chicago, Department of Pharmacological and Physiological Sciences, 947 E. 58th Street, MC 0926, Chicago, IL 60637, USA
| | - Adriana M. Zimnicka
- The University of Chicago, Department of Pharmacological and Physiological Sciences, 947 E. 58th Street, MC 0926, Chicago, IL 60637, USA
| | - Si Chen
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Deborah J. Nelson
- The University of Chicago, Department of Pharmacological and Physiological Sciences, 947 E. 58th Street, MC 0926, Chicago, IL 60637, USA
| |
Collapse
|
27
|
Riazanski V, Mauleon G, Lucas K, Walker S, Zimnicka AM, McGrath JL, Nelson DJ. Real time imaging of single extracellular vesicle pH regulation in a microfluidic cross-flow filtration platform. Commun Biol 2022; 5:13. [PMID: 35013561 PMCID: PMC8748679 DOI: 10.1038/s42003-021-02965-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 12/13/2021] [Indexed: 11/22/2022] Open
Abstract
Extracellular vesicles (EVs) are cell-derived membranous structures carrying transmembrane proteins and luminal cargo. Their complex cargo requires pH stability in EVs while traversing diverse body fluids. We used a filtration-based platform to capture and stabilize EVs based on their size and studied their pH regulation at the single EV level. Dead-end filtration facilitated EV capture in the pores of an ultrathin (100 nm thick) and nanoporous silicon nitride (NPN) membrane within a custom microfluidic device. Immobilized EVs were rapidly exposed to test solution changes driven across the backside of the membrane using tangential flow without exposing the EVs to fluid shear forces. The epithelial sodium-hydrogen exchanger, NHE1, is a ubiquitous plasma membrane protein tasked with the maintenance of cytoplasmic pH at neutrality. We show that NHE1 identified on the membrane of EVs is functional in the maintenance of pH neutrality within single vesicles. This is the first mechanistic description of EV function on the single vesicle level. Riazanski et al describe a platform to capture extracellular vesicles (EVs) using a nanoporous silicon nitride membrane, investigate the expression of NHE1 protein on the surface of EVs and monitor the transport of Na+ and H+ at the single EV level. The authors report a mechanistic function of the proteins found in EVs and specifically identify NHE1 on a single EV, where it maintains pH neutrality within single vesicles.
Collapse
Affiliation(s)
- Vladimir Riazanski
- Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - Gerardo Mauleon
- Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - Kilean Lucas
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Samuel Walker
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Adriana M Zimnicka
- Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Deborah J Nelson
- Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
28
|
Meijer L, Hery-Arnaud G, Leven C, Nowak E, Hillion S, Renaudineau Y, Durieu I, Chiron R, Prevotat A, Fajac I, Hubert D, Murris-Espin M, Huge S, Danner-Boucher I, Ravoninjatovo B, Leroy S, Macey J, Urban T, Rault G, Mottier D, Berre RL. Safety and pharmacokinetics of Roscovitine (Seliciclib) in cystic fibrosis patients chronically infected with Pseudomonas aeruginosa, a randomized, placebo-controlled study. J Cyst Fibros 2021; 21:529-536. [PMID: 34961705 DOI: 10.1016/j.jcf.2021.10.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/17/2021] [Accepted: 10/19/2021] [Indexed: 01/20/2023]
Abstract
BACKGROUND The orally available kinase inhibitor R-roscovitine has undergone clinical trials against various cancers and is currently under clinical evaluation against Cushing disease and rheumatoid arthritis. Roscovitine displays biological properties suggesting potential benefits in CF: it partially corrects F508del-CFTR trafficking, stimulates the bactericidal properties of CF alveolar macrophages, and displays anti-inflammatory properties and analgesic effects. METHODS A phase 2 trial study (ROSCO-CF) was launched to evaluate the safety and effects of roscovitine in Pseudomonas aeruginosa infected adult CF patients carrying two CF causing mutations (at least one F508del-CFTR mutation) and harboring a FEV1 ≥40%. ROSCO-CF was a multicenter, double-blind, placebo-controlled, dose-ranging study (200, 400, 800 mg roscovitine, orally administered daily for 4 days/week/4 weeks). RESULTS Among the 34 volunteers enrolled, randomization assigned 11/8/8/7 to receive the 0 (placebo)/ 200/400/800 mg roscovitine doses, respectively. In these subjects with polypharmacy, roscovitine was relatively safe and well-tolerated, with no significant adverse effects (AEs) other than five serious AEs (SAEs) possibly related to roscovitine. Pharmacokinetics of roscovitine were rather variable among subjects. No significant efficacy, at the levels of inflammation, infection, spirometry, sweat chloride, pain and quality of life, was detected in roscovitine-treated groups compared to the placebo-treated group. CONCLUSION Roscovitine was relatively safe and well-tolerated in CF patients especially at the 200 and 400 mg doses. However, there were 5 subject withdrawals due to SAEs in the roscovitine group and none in the placebo group. The lack of evidence for efficacy of roscovitine (despite encouraging cellular and animal results) may be due to high pharmacokinetics variability, short duration of treatment, and/or inappropriate dosing protocol.
Collapse
Affiliation(s)
- Laurent Meijer
- ManRos Therapeutics, Presqu'île de Perharidy, Roscoff 29680, France.
| | - Geneviève Hery-Arnaud
- Unité de Bactériologie, Hôpital de la Cavale Blanche, CHRU Brest, Brest cedex 29609, France; Univ Brest, Inserm, EFS, UMR 1078, GGB, Brest 29200, France
| | - Cyril Leven
- Département de Biochimie et Pharmaco-Toxicologie, CHRU Brest, Brest cedex 29609, France; Univ Brest, EA 3878, GETBO, Brest 29200, France
| | - Emmanuel Nowak
- INSERM CIC 1412, Brest University Hospital, Brest cedex 29609, France
| | - Sophie Hillion
- Laboratoire d'Immunologie et Immunothérapie, CHRU de Brest, INSERM U1227, 2 avenue Foch, BP824, 29609 Brest cedex, France
| | - Yves Renaudineau
- Laboratoire d'Immunologie et Immunothérapie, CHRU de Brest, INSERM U1227, 2 avenue Foch, BP824, 29609 Brest cedex, France
| | - Isabelle Durieu
- Research on Healthcare Performance (RESHAPE), INSERM U1290, Claude Bernard Lyon 1 University, 8 Avenue Rockefeller, 69003 Lyon, France; Department of Internal Medicine, Cystic Fibrosis Center, Hospices Civils de Lyon, Pierre-Bénite 69495, France
| | - Raphaël Chiron
- CHU Montpellier, Maladies Respiratoires, Hôpital Arnaud de Villeneuve, 371, avenue du Doyen Gaston Giraud, Montpellier 34295, France
| | - Anne Prevotat
- Service de pneumologie, CHR - Hôpital Calmette, Boulevard du Pr. Leclercq, Lille 59037, France
| | - Isabelle Fajac
- APHP.Centre - Université de Paris, 27 rue du Faubourg Saint-Jacques, Paris 75014, France
| | - Dominique Hubert
- APHP.Centre - Université de Paris, 27 rue du Faubourg Saint-Jacques, Paris 75014, France
| | - Marlène Murris-Espin
- CHU Toulouse, CRCM adulte, Service de Pneumologie, Clinique des Voies Respiratoires. Hôpital Larrey, 24 chemin de Pouvourville, Toulouse 31059, France
| | - Sandrine Huge
- Centre Hospitalier Bretagne Atlantique, CRCM Mixte 56, 20 Boulevard du général Maurice Guillaudot, Vannes cedex 56017, France
| | - Isabelle Danner-Boucher
- CHU de Nantes, Service de Pneumologie, Hôpital Nord Laennec, Boulevard Jacques-Monod, Nantes 44093, Saint-Herblain, France
| | - Bruno Ravoninjatovo
- Centre de Ressources et de Compétences de la Mucoviscidose, Maladies Respiratoires et Allergiques, Hôpital Maison Blanche - CHU Reims, 45 rue Cognacq-Jay, 51100 Reims, France
| | - Sylvie Leroy
- CHU de Nice, Hôpital Pasteur, Service de Pneumologie, Oncologie Thoracique et Soins Intensifs Respiratoires, 30 Voie Romaine, CS 51069, Nice cedex 1 06001, France
| | - Julie Macey
- CHU Bordeaux, Hôpital Haut-Lévêque, Service de Pneumologie, Avenue de Magellan, Pessac cedex 33604, France
| | - Thierry Urban
- Département de Pneumologie, CHU Angers, Site de Larrey, 4 rue de Larrey, Angers cedex 49933, France
| | - Gilles Rault
- Fondation Ildys, Centre de Perharidy, Roscoff cedex 29684, France
| | - Dominique Mottier
- Département de Biochimie et Pharmaco-Toxicologie, CHRU Brest, Brest cedex 29609, France
| | - Rozenn Le Berre
- Univ Brest, Inserm, EFS, UMR 1078, GGB, Brest 29200, France; Département de Médecine Interne et Pneumologie, CHRU Brest, Brest cedex 29609, France
| |
Collapse
|
29
|
Bojanowski CM, Lu S, Kolls JK. Mucosal Immunity in Cystic Fibrosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:2901-2912. [PMID: 35802761 PMCID: PMC9270582 DOI: 10.4049/jimmunol.2100424] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 10/21/2021] [Indexed: 05/27/2023]
Abstract
The highly complex and variable genotype-phenotype relationships observed in cystic fibrosis (CF) have been an area of growing interest since the discovery of the CF transmembrane conductance regulator (CFTR) gene >30 y ago. The consistently observed excessive, yet ineffective, activation of both the innate and adaptive host immune systems and the establishment of chronic infections within the lung, leading to destruction and functional decline, remain the primary causes of morbidity and mortality in CF. The fact that both inflammation and pathogenic bacteria persist despite the introduction of modulator therapies targeting the defective protein, CFTR, highlights that we still have much to discover regarding mucosal immunity determinants in CF. Gene modifier studies have overwhelmingly implicated immune genes in the pulmonary phenotype of the disease. In this context, we aim to review recent advances in our understanding of the innate and adaptive immune systems in CF lung disease.
Collapse
Affiliation(s)
- Christine M Bojanowski
- Section of Pulmonary Diseases, Critical Care, and Environmental Medicine, Department of Medicine, Tulane University School of Medicine, New Orleans, LA;
| | - Shiping Lu
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA; and
| | - Jay K Kolls
- Center for Translational Research in Infection and Inflammation, Department of Medicine, Tulane University School of Medicine, New Orleans, LA
| |
Collapse
|
30
|
Macrophages from gut-corrected CF mice express human CFTR and lack a pro-inflammatory phenotype. J Cyst Fibros 2021; 21:370-374. [PMID: 34799297 PMCID: PMC9097694 DOI: 10.1016/j.jcf.2021.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 11/05/2021] [Accepted: 11/06/2021] [Indexed: 11/24/2022]
Abstract
The cftrtm1unc Tg(FABP-hCFTR) mouse is a commonly-used animal model of CF. This mouse expresses human CFTR in the gut to prevent fatal intestinal obstruction. Macrophages from this mouse fail to replicate immune dysfunction seen in patient cells. We show ectopic expression of human CFTR transgene in macrophages from this CF mouse. This may help to explain anomalies in the field related to use of this model.
Macrophages represent prominent immune orchestrators of cystic fibrosis (CF) inflammation and, as such, are an ever-increasing focus of CF research with several reports of intrinsic immune dysfunction related to loss of CFTR activity in macrophages themselves. Animal models of CF have contributed, in no small part, to a deepening of our understanding of the pathophysiology of the disease and towards therapeutic development. A commonly-used animal model in CF research is the Cftrtm1Unc Tg(FABP-hCFTR) mouse, which displays gut-specific expression of a human CFTR transgene in order to rescue the high rate of early mortality in Cftr-null mice associated with severe intestinal obstruction. We find significant variation in the response to inflammatory challenge of patient macrophages and cells derived from the Cftrtm1Unc Tg(FABP-hCFTR) mouse and show that macrophages derived from this mouse exhibit aberrant expression of human CFTR. This may contribute to the absence of inflammatory changes in this model.
Collapse
|
31
|
Lukasiak A, Zajac M. The Distribution and Role of the CFTR Protein in the Intracellular Compartments. MEMBRANES 2021; 11:membranes11110804. [PMID: 34832033 PMCID: PMC8618639 DOI: 10.3390/membranes11110804] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 12/11/2022]
Abstract
Cystic fibrosis is a hereditary disease that mainly affects secretory organs in humans. It is caused by mutations in the gene encoding CFTR with the most common phenylalanine deletion at position 508. CFTR is an anion channel mainly conducting Cl− across the apical membranes of many different epithelial cells, the impairment of which causes dysregulation of epithelial fluid secretion and thickening of the mucus. This, in turn, leads to the dysfunction of organs such as the lungs, pancreas, kidney and liver. The CFTR protein is mainly localized in the plasma membrane; however, there is a growing body of evidence that it is also present in the intracellular organelles such as the endosomes, lysosomes, phagosomes and mitochondria. Dysfunction of the CFTR protein affects not only the ion transport across the epithelial tissues, but also has an impact on the proper functioning of the intracellular compartments. The review aims to provide a summary of the present state of knowledge regarding CFTR localization and function in intracellular compartments, the physiological role of this localization and the consequences of protein dysfunction at cellular, epithelial and organ levels. An in-depth understanding of intracellular processes involved in CFTR impairment may reveal novel opportunities in pharmacological agents of cystic fibrosis.
Collapse
|
32
|
Jackson L, Waters V. Factors influencing the acquisition and eradication of early Pseudomonas aeruginosa infection in cystic fibrosis. J Cyst Fibros 2020; 20:8-16. [PMID: 33172756 DOI: 10.1016/j.jcf.2020.10.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/02/2020] [Accepted: 10/27/2020] [Indexed: 12/21/2022]
Abstract
In recent years considerable improvements have been made in increasing the life expectancy of patients with cystic fibrosis. New highly effective modulator therapies targeting the underlying defect in the cystic fibrosis transmembrane conductance regulator protein are expected to enhance lifespan even further. However, chronic Pseudomonas aeruginosa pulmonary infections continue to threaten CF patient lung health and mortality rates. Early and aggressive antibiotic eradication therapies targeting P. aeruginosa are standard practice, but these eradication therapies fail in 10-40% of patients. The reasons for P. aeruginosa eradication failure remain unclear. Thus, this review summarizes the evidence to date for pseudomonal acquisition and eradication failure in the cystic fibrosis lung. A complex combination of host and bacterial factors are responsible for initial establishment of P. aeruginosa pulmonary infections. Moreover, host and pseudomonal factors, polymicrobial interactions, and antimicrobial limitations in relation to P. aeruginosa eradication therapy failure are summarized.
Collapse
Affiliation(s)
- Lindsay Jackson
- Translational Medicine, Hospital for Sick Children, Toronto, Canada.
| | - Valerie Waters
- Translational Medicine, Hospital for Sick Children, Toronto, Canada; Infectious Diseases, Department of Pediatrics, Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
33
|
Riazanski V, Sui Z, Nelson DJ. Kinetic Separation of Oxidative and Non-oxidative Metabolism in Single Phagosomes from Alveolar Macrophages: Impact on Bacterial Killing. iScience 2020; 23:101759. [PMID: 33251491 PMCID: PMC7677711 DOI: 10.1016/j.isci.2020.101759] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/07/2020] [Accepted: 10/29/2020] [Indexed: 01/10/2023] Open
Abstract
The relative contribution of the two phagosomal catabolic processes, oxidative and metabolic, was assessed in the killing of Pseudomonas aeruginosa in phagosomes of alveolar macrophages (AMs) from wild-type (p47-phox+/+) or NOX-defective (p47-phox−/−) mice. Free radical release and degradative acidification within AM phagosomes is sequential and separable. The initial NOX activity, identifiable as a transient alkalinization, leads to fast bacterial wall permeabilization by ROS. This is followed by V-ATPase-induced acidification and enzymatic bacterial degradation contributed through phagosomal-lysosomal fusion. The alkalinization/acidification ratio was variable among phagosomes within single cells of a given genotype and not as a function of macrophage M1 or M2 classification, possibly owing to uneven distribution of phagosomal transporter proteins. Irregular, excessive NOX activity prevents phago-lysosomal fusion, and the lack of V-ATPase-induced acidification leads to bacterial stasis in the phagosome. Thus, efficient phagosomal bacterial killing is a result of tightly balanced activity between two processes. Phagosomal NOX and V-ATPase activation is sequential and separable in macrophages Superoxide (O2-) inhibits lysosomal fusion thereby inhibiting phagosomal acidification Phagosomes in single cells are heterogeneous in NOX activity and thereby acidification NOX activity is the dominant factor in bactericidal efficacy in macrophage phagosomes
Collapse
Affiliation(s)
- Vladimir Riazanski
- The University of Chicago, Department of Pharmacological and Physiological Sciences, 947 E. 58th Street, MC 0926, Chicago, IL 60637, USA
- Corresponding author
| | - Zihao Sui
- The University of Chicago, Department of Pharmacological and Physiological Sciences, 947 E. 58th Street, MC 0926, Chicago, IL 60637, USA
| | - Deborah J. Nelson
- The University of Chicago, Department of Pharmacological and Physiological Sciences, 947 E. 58th Street, MC 0926, Chicago, IL 60637, USA
- Corresponding author
| |
Collapse
|
34
|
Briottet M, Shum M, Urbach V. The Role of Specialized Pro-Resolving Mediators in Cystic Fibrosis Airways Disease. Front Pharmacol 2020; 11:1290. [PMID: 32982730 PMCID: PMC7493015 DOI: 10.3389/fphar.2020.01290] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/04/2020] [Indexed: 12/26/2022] Open
Abstract
Cystic Fibrosis (CF) is a recessive genetic disease due to mutations of the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) gene encoding the CFTR chloride channel. The ion transport abnormalities related to CFTR mutation generate a dehydrated airway surface liquid (ASL) layer, which is responsible for an altered mucociliary clearance, favors infections and persistent inflammation that lead to progressive lung destruction and respiratory failure. The inflammatory response is normally followed by an active resolution phase to return to tissue homeostasis, which involves specialized pro-resolving mediators (SPMs). SPMs promote resolution of inflammation, clearance of microbes, tissue regeneration and reduce pain, but do not evoke unwanted immunosuppression. The airways of CF patients showed a decreased production of SPMs even in the absence of pathogens. SPMs levels in the airway correlated with CF patients' lung function. The prognosis for CF has greatly improved but there remains a critical need for more effective treatments that prevent excessive inflammation, lung damage, and declining pulmonary function for all CF patients. This review aims to highlight the recent understanding of CF airway inflammation and the possible impact of SPMs on functions that are altered in CF airways.
Collapse
Affiliation(s)
| | | | - Valerie Urbach
- Institut national de la santé et de la recherche médicale (Inserm) U955, Institut Mondor de Recherche Biomédicale (IMRB), Créteil, France
| |
Collapse
|
35
|
Currie AJ, Main ET, Wilson HM, Armstrong-James D, Warris A. CFTR Modulators Dampen Aspergillus-Induced Reactive Oxygen Species Production by Cystic Fibrosis Phagocytes. Front Cell Infect Microbiol 2020; 10:372. [PMID: 32793514 PMCID: PMC7393064 DOI: 10.3389/fcimb.2020.00372] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/17/2020] [Indexed: 12/17/2022] Open
Abstract
Excessive inflammation by phagocytes during Aspergillus fumigatus infection is thought to promote lung function decline in CF patients. CFTR modulators have been shown to reduce A. fumigatus colonization in vivo, however, their antifungal and anti-inflammatory mechanisms are unclear. Other treatments including azithromycin and acebilustat may dampen Aspergillus-induced inflammation due to their immunomodulatory properties. Therefore, we set out in this study to determine the effects of current CF therapies on ROS production and fungal killing, either direct or indirect by enhancing antifungal immune mechanisms in peripheral blood immune cells from CF patients upon A. fumigatus infection. Isolated peripheral blood mononuclear cells (PBMCs) and polymorphonuclear cells (PMNs) from CF patients and healthy volunteers were challenged with A. fumigatus following pre-treatment with CFTR modulators, azithromycin or acebilustat. Ivacaftor/lumacaftor treated CF and control subject PMNs resulted in a significant reduction (p < 0.05) in Aspergillus-induced ROS. For CF PBMC, Aspergillus-induced ROS was significantly reduced when pre-treated with ivacaftor alone (p < 0.01) or in combination with lumacaftor (p < 0.01), with a comparable significant reduction in control subject PBMC (p < 0.05). Azithromycin and acebilustat had no effect on ROS production by CF or control subject phagocytes. None of the treatments showed an indirect or direct antifungal activity. In summary, CFTR modulators have potential for additional immunomodulatory benefits to prevent or treat Aspergillus-induced inflammation in CF. The comparable effects of CFTR modulators observed in phagocytes from control subjects questions their exact mechanism of action.
Collapse
Affiliation(s)
- Alexander J Currie
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom.,Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Ellen T Main
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Heather M Wilson
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | | | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
36
|
Turton KB, Ingram RJ, Valvano MA. Macrophage dysfunction in cystic fibrosis: Nature or nurture? J Leukoc Biol 2020; 109:573-582. [PMID: 32678926 DOI: 10.1002/jlb.4ru0620-245r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) affect the homeostasis of chloride flux by epithelial cells. This has deleterious consequences, especially in respiratory epithelia, where the defect results in mucus accumulation distinctive of cystic fibrosis. CFTR is, however, also expressed in phagocytic cells, like macrophages. Immune cells are highly sensitive to conditioning by their environment; thus, CFTR dysfunction in epithelia influences macrophages by affecting the lung milieu, but the mutations also appear to be directly consequential for intrinsic macrophage functions. Particular mutations can alter CFTR's folding, traffic of the protein to the membrane and function. As such, understanding the intrinsic effects of CFTR mutation requires distinguishing the secondary effects of misfolded CFTR on cell stress pathways from the primary defect of CFTR dysfunction/absence. Investigations into CFTR's role in macrophages have exploited various models, each with their own advantages and limitations. This review summarizes these methodologic approaches, discussing their physiological correspondence and highlighting key findings. The controversy surrounding CFTR-dependent acidification is used as a case study to highlight difficulties in commensurability across model systems. Recent work in macrophage biology, including polarization and host-pathogen interaction studies, brought into the context of CFTR research, offers potential explanations for observed discrepancies between studies. Moreover, the rapid advancement of novel gene editing technologies and new macrophage model systems makes this assessment of the field's models and methodologies timely.
Collapse
Affiliation(s)
- Keren B Turton
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Rebecca J Ingram
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Miguel A Valvano
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
37
|
Zhang X, Mao F, Wong NK, Bao Y, Lin Y, Liu K, Li J, Xiang Z, Ma H, Xiao S, Zhang Y, Yu Z. CLIC2α Chloride Channel Orchestrates Immunomodulation of Hemocyte Phagocytosis and Bactericidal Activity in Crassostrea gigas. iScience 2020; 23:101328. [PMID: 32674055 PMCID: PMC7363696 DOI: 10.1016/j.isci.2020.101328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 06/02/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023] Open
Abstract
Chloride ion plays critical roles in modulating immunological interactions. Herein, we demonstrated that the anion channel CLIC2α mediates Cl− flux to regulate hemocytes functions in the Pacific oyster (Crassostrea gigas). Specifically, during infection by Vibrio parahemolyticus, chloride influx was activated following onset of phagocytosis. Phosphorylation of Akt was stimulated by Cl− ions entering host cells, further contributing to signal transduction regulating internalization of bacteria through the PI3K/Akt signaling pathway. Concomitantly, Cl− entered phagosomes, promoted the acidification and maturation of phagosomes, and contributed to production of HOCl to eradicate engulfed bacteria. Finally, genomic screening reveals CLIC2α as a major Cl− channel gene responsible for regulating Cl− influx in oysters. Knockdown of CLIC2α predictably impeded phagosome acidification and restricted bacterial killing in oysters. In conclusion, our work has established CLIC2α as a prominent regulator of Cl− influx and thus Cl− function in C. gigas in bacterial infection contexts. Influx of chloride ions is switched on during phagocytosis in oyster hemocytes PI3K/Akt signaling pathway mediates chloride-dependent activation of phagocytosis Cl− promotes phagosomal acidification and HOCl production CLIC2α is the principal chloride channel encoding gene within oyster genome
Collapse
Affiliation(s)
- Xiangyu Zhang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, Innovation Academy of South China Sea Ecology and Environmental Engineering (ISEE), South China Sea Institute of Oceanology, Chinese Academy of Science, Guangzhou 510301, P. R. China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 510301, P. R. China; University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Fan Mao
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, Innovation Academy of South China Sea Ecology and Environmental Engineering (ISEE), South China Sea Institute of Oceanology, Chinese Academy of Science, Guangzhou 510301, P. R. China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 510301, P. R. China
| | - Nai-Kei Wong
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, Innovation Academy of South China Sea Ecology and Environmental Engineering (ISEE), South China Sea Institute of Oceanology, Chinese Academy of Science, Guangzhou 510301, P. R. China; National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, The Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen 518112, P. R. China
| | - Yongbo Bao
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, P. R. China
| | - Yue Lin
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, Innovation Academy of South China Sea Ecology and Environmental Engineering (ISEE), South China Sea Institute of Oceanology, Chinese Academy of Science, Guangzhou 510301, P. R. China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 510301, P. R. China; University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Kunna Liu
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, Innovation Academy of South China Sea Ecology and Environmental Engineering (ISEE), South China Sea Institute of Oceanology, Chinese Academy of Science, Guangzhou 510301, P. R. China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 510301, P. R. China; University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jun Li
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, Innovation Academy of South China Sea Ecology and Environmental Engineering (ISEE), South China Sea Institute of Oceanology, Chinese Academy of Science, Guangzhou 510301, P. R. China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 510301, P. R. China
| | - Zhiming Xiang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, Innovation Academy of South China Sea Ecology and Environmental Engineering (ISEE), South China Sea Institute of Oceanology, Chinese Academy of Science, Guangzhou 510301, P. R. China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 510301, P. R. China
| | - Haitao Ma
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, Innovation Academy of South China Sea Ecology and Environmental Engineering (ISEE), South China Sea Institute of Oceanology, Chinese Academy of Science, Guangzhou 510301, P. R. China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 510301, P. R. China
| | - Shu Xiao
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, Innovation Academy of South China Sea Ecology and Environmental Engineering (ISEE), South China Sea Institute of Oceanology, Chinese Academy of Science, Guangzhou 510301, P. R. China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 510301, P. R. China
| | - Yang Zhang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, Innovation Academy of South China Sea Ecology and Environmental Engineering (ISEE), South China Sea Institute of Oceanology, Chinese Academy of Science, Guangzhou 510301, P. R. China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 510301, P. R. China.
| | - Ziniu Yu
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, Innovation Academy of South China Sea Ecology and Environmental Engineering (ISEE), South China Sea Institute of Oceanology, Chinese Academy of Science, Guangzhou 510301, P. R. China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 510301, P. R. China.
| |
Collapse
|
38
|
DNA Microsystems for Biodiagnosis. MICROMACHINES 2020; 11:mi11040445. [PMID: 32340280 PMCID: PMC7231314 DOI: 10.3390/mi11040445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 12/16/2022]
Abstract
Researchers are continuously making progress towards diagnosis and treatment of numerous diseases. However, there are still major issues that are presenting many challenges for current medical diagnosis. On the other hand, DNA nanotechnology has evolved significantly over the last three decades and is highly interdisciplinary. With many potential technologies derived from the field, it is natural to begin exploring and incorporating its knowledge to develop DNA microsystems for biodiagnosis in order to help address current obstacles, such as disease detection and drug resistance. Here, current challenges in disease detection are presented along with standard methods for diagnosis. Then, a brief overview of DNA nanotechnology is introduced along with its main attractive features for constructing biodiagnostic microsystems. Lastly, suggested DNA-based microsystems are discussed through proof-of-concept demonstrations with improvement strategies for standard diagnostic approaches.
Collapse
|
39
|
Mészáros LS, Ceccaldi P, Lorenzi M, Redman HJ, Pfitzner E, Heberle J, Senger M, Stripp ST, Berggren G. Spectroscopic investigations under whole-cell conditions provide new insight into the metal hydride chemistry of [FeFe]-hydrogenase. Chem Sci 2020; 11:4608-4617. [PMID: 34122916 PMCID: PMC8159234 DOI: 10.1039/d0sc00512f] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hydrogenases are among the fastest H2 evolving catalysts known to date and have been extensively studied under in vitro conditions. Here, we report the first mechanistic investigation of an [FeFe]-hydrogenase under whole-cell conditions. Functional [FeFe]-hydrogenase from the green alga Chlamydomonas reinhardtii is generated in genetically modified Escherichia coli cells by addition of a synthetic cofactor to the growth medium. The assembly and reactivity of the resulting semi-synthetic enzyme was monitored using whole-cell electron paramagnetic resonance and Fourier-transform Infrared difference spectroscopy as well as scattering scanning near-field optical microscopy. Through a combination of gas treatments, pH titrations, and isotope editing we were able to corroborate the formation of a number of proposed catalytic intermediates in living cells, supporting their physiological relevance. Moreover, a previously incompletely characterized catalytic intermediate is reported herein, attributed to the formation of a protonated metal hydride species.
Collapse
Affiliation(s)
- Lívia S Mészáros
- Molecular Biomimetics, Dept. of Chemistry - Ångström Laboratory, Uppsala University Lägerhyddsvägen 1 SE-75120 Uppsala Sweden
| | - Pierre Ceccaldi
- Molecular Biomimetics, Dept. of Chemistry - Ångström Laboratory, Uppsala University Lägerhyddsvägen 1 SE-75120 Uppsala Sweden
| | - Marco Lorenzi
- Molecular Biomimetics, Dept. of Chemistry - Ångström Laboratory, Uppsala University Lägerhyddsvägen 1 SE-75120 Uppsala Sweden
| | - Holly J Redman
- Molecular Biomimetics, Dept. of Chemistry - Ångström Laboratory, Uppsala University Lägerhyddsvägen 1 SE-75120 Uppsala Sweden
| | - Emanuel Pfitzner
- Institute of Experimental Physics, Experimental Molecular Biophysics, Freie Universität Berlin Arnimallee 14 Berlin DE-14195 Germany
| | - Joachim Heberle
- Institute of Experimental Physics, Experimental Molecular Biophysics, Freie Universität Berlin Arnimallee 14 Berlin DE-14195 Germany
| | - Moritz Senger
- Institute of Experimental Physics, Experimental Molecular Biophysics, Freie Universität Berlin Arnimallee 14 Berlin DE-14195 Germany
| | - Sven T Stripp
- Institute of Experimental Physics, Experimental Molecular Biophysics, Freie Universität Berlin Arnimallee 14 Berlin DE-14195 Germany
| | - Gustav Berggren
- Molecular Biomimetics, Dept. of Chemistry - Ångström Laboratory, Uppsala University Lägerhyddsvägen 1 SE-75120 Uppsala Sweden
| |
Collapse
|
40
|
Law SM, Stanfield SJ, Hardisty GR, Dransfield I, Campbell CJ, Gray RD. Human cystic fibrosis monocyte derived macrophages display no defect in acidification of phagolysosomes when measured by optical nanosensors. J Cyst Fibros 2020; 19:203-210. [PMID: 31501051 DOI: 10.1016/j.jcf.2019.09.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 09/03/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Defective macrophage phagolysosomal acidification is implicated in numerous lung diseases including Cystic Fibrosis (CF) and may contribute to defective pathogen killing. Conflicting reports relating to phagolysosomal pH in CF macrophages have been published, in part related to the use of pH-sensitive fluorescent probes where potential inadequacies in experimental design can be a contributing factor (e.g. employing probes with incorrect pKa for the cellular compartment of interest). We developed a reliable method to quantify macrophage phagolysosomal pH using surface-enhanced Raman spectroscopy-based nanosensors. METHODS Monocyte-derived macrophages from CF and healthy control participants were incubated with nanosensors. Live cell imaging identified phagocytosed nanosensors, and surface-enhanced Raman spectroscopy was performed using para-mercaptobenzoic acid functionalised gold nanoparticles which produce Raman spectra that change predictably with their environmental pH. Conventional fluorescence spectroscopy was carried out in comparison. Nanosensor localisation to phagolysosomes was confirmed by transmission electron microscopy. RESULTS Nanosensors were actively phagocytosed by macrophages into phagolysosomes and acidification occurred rapidly and remained stable for at least 60 min. There was no difference in phagolysosomal pH between healthy control and CF macrophages (5.41 ± 0.11 vs. 5.41 ± 0.20, p > .9999), further confirmed by inhibiting Cystic Fibrosis Transmembrane Conductance Regulator in healthy control monocyte-derived macrophages. CONCLUSIONS Optical nanosensors accurately measure macrophage phagolysosomal pH and demonstrate no phagolysosomal acidification defect in human CF monocyte-derived macrophages. Further studies using alveolar macrophages could extend the impact of our findings. Nanosensors represent a novel and precise means to measure organelle functions with widespread potential for the study and monitoring of several lung diseases.
Collapse
Affiliation(s)
- Sheonagh M Law
- Centre for Inflammation Research, The Queen's Medical Research Institute, 47 Little France Crescent, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Samuel J Stanfield
- Joseph Black Building, The University of Edinburgh, David Brewster Rd, Edinburgh EH9 3FJ, UK
| | - Gareth R Hardisty
- Centre for Inflammation Research, The Queen's Medical Research Institute, 47 Little France Crescent, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Ian Dransfield
- Centre for Inflammation Research, The Queen's Medical Research Institute, 47 Little France Crescent, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Colin J Campbell
- Joseph Black Building, The University of Edinburgh, David Brewster Rd, Edinburgh EH9 3FJ, UK
| | - Robert D Gray
- Centre for Inflammation Research, The Queen's Medical Research Institute, 47 Little France Crescent, The University of Edinburgh, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
41
|
Zajac M, Chakraborty K, Saha S, Mahadevan V, Infield DT, Accardi A, Qiu Z, Krishnan Y. What biologists want from their chloride reporters – a conversation between chemists and biologists. J Cell Sci 2020; 133:133/2/jcs240390. [DOI: 10.1242/jcs.240390] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
ABSTRACT
Impaired chloride transport affects diverse processes ranging from neuron excitability to water secretion, which underlie epilepsy and cystic fibrosis, respectively. The ability to image chloride fluxes with fluorescent probes has been essential for the investigation of the roles of chloride channels and transporters in health and disease. Therefore, developing effective fluorescent chloride reporters is critical to characterizing chloride transporters and discovering new ones. However, each chloride channel or transporter has a unique functional context that demands a suite of chloride probes with appropriate sensing characteristics. This Review seeks to juxtapose the biology of chloride transport with the chemistries underlying chloride sensors by exploring the various biological roles of chloride and highlighting the insights delivered by studies using chloride reporters. We then delineate the evolution of small-molecule sensors and genetically encoded chloride reporters. Finally, we analyze discussions with chloride biologists to identify the advantages and limitations of sensors in each biological context, as well as to recognize the key design challenges that must be overcome for developing the next generation of chloride sensors.
Collapse
Affiliation(s)
- Matthew Zajac
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL 60637, USA
| | - Kasturi Chakraborty
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL 60637, USA
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Sonali Saha
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Vivek Mahadevan
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Daniel T. Infield
- Department of Molecular Physiology and Biophysics, The University of Iowa, Iowa City, IA 52242, USA
| | - Alessio Accardi
- Department of Anesthesiology, Weill Cornell Medical School, New York, NY 10065, USA
- Department of Physiology and Biophysics, Weill Cornell Medical School, New York, NY 10065, USA
- Department of Biochemistry, Weill Cornell Medical School, New York, NY 10065, USA
| | - Zhaozhu Qiu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Yamuna Krishnan
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
42
|
Xue YW, Hayata A, Itoh H, Inoue M. Biological Effects of a Simplified Synthetic Analogue of Ion-Channel-Forming Polytheonamide B on Plasma Membrane and Lysosomes. Chemistry 2019; 25:15198-15204. [PMID: 31549755 DOI: 10.1002/chem.201903974] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/24/2019] [Indexed: 11/09/2022]
Abstract
Polytheonamide B (1) is a linear 48-mer natural peptide with alternating d- and l-amino acid residues. Compound 1 forms conducting channels for monovalent ions and exhibits potent cytotoxicity against MCF-7 cells. Previously, we reported that nanomolar concentrations of 1 induce plasma membrane depolarization and lysosomal pH disruption, which triggers apoptosis. Here, we report the cellular localization and biological action of a simplified synthetic analogue of 1, polytheonamide mimic 3. Compared with 1, the toxicity of 3 against MCF-7 cells is 16 times weaker. Although its plasma membrane depolarization effect is only 3.6 times lower, more 3 (20-fold) is required to neutralize lysosomal pH. Thus, the effective concentrations for lysosomal neutralization and cytotoxicity by 3 are comparable. These results strongly suggest that the activity of 3 against the lysosomal membrane is more important for apoptotic cell death than its effects on the plasma membrane, and provide valuable information regarding the unique behavior of polytheonamide-based molecules.
Collapse
Affiliation(s)
- Yun-Wei Xue
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Atsushi Hayata
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hiroaki Itoh
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Masayuki Inoue
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
43
|
Recchiuti A, Mattoscio D, Isopi E. Roles, Actions, and Therapeutic Potential of Specialized Pro-resolving Lipid Mediators for the Treatment of Inflammation in Cystic Fibrosis. Front Pharmacol 2019; 10:252. [PMID: 31001110 PMCID: PMC6454233 DOI: 10.3389/fphar.2019.00252] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/28/2019] [Indexed: 01/07/2023] Open
Abstract
Non-resolving inflammation is the main mechanism of morbidity and mortality among patients suffering from cystic fibrosis (CF), the most common life-threatening human genetic disease. Resolution of inflammation is an active process timely controlled by endogenous specialized pro-resolving lipid mediators (SPMs) produced locally in inflammatory loci to restrain this innate response, prevent further damages to the host, and permit return to homeostasis. Lipoxins, resolvins, protectins, and maresins are SPM derived from polyunsaturated fatty acids that limit excessive leukocyte infiltration and pro-inflammatory signals, stimulate innate microbial killing, and enhance resolution. Their unique chemical structures, receptors, and bioactions are being elucidated. Accruing data indicate that SPMs carry protective functions against unrelenting inflammation and infections in preclinical models and human CF systems. Here, we reviewed their roles and actions in controlling resolution of inflammation, evidence for their impairment in CF, and proofs of principle for their exploitation as innovative, non-immunosuppressive drugs to address inflammation and infections in CF.
Collapse
Affiliation(s)
- Antonio Recchiuti
- Department of Medical, Oral and Biotechnological Science, Università “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Centro di Scienze dell’Invecchiamento e Medicina Traslazionale (CeSI-MeT), Università “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Domenico Mattoscio
- Department of Medical, Oral and Biotechnological Science, Università “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Centro di Scienze dell’Invecchiamento e Medicina Traslazionale (CeSI-MeT), Università “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Elisa Isopi
- Department of Medical, Oral and Biotechnological Science, Università “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Centro di Scienze dell’Invecchiamento e Medicina Traslazionale (CeSI-MeT), Università “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| |
Collapse
|
44
|
Warris A, Bercusson A, Armstrong-James D. Aspergillus colonization and antifungal immunity in cystic fibrosis patients. Med Mycol 2019; 57:S118-S126. [PMID: 30816976 DOI: 10.1093/mmy/myy074] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/20/2018] [Accepted: 07/26/2018] [Indexed: 12/11/2022] Open
Abstract
Cystic fibrosis (CF), caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, is the most common inherited life-limiting disease in North European people affecting 90,000 people worldwide. Progressive lung damage caused by recurrent infection and chronic airway inflammation is the major determinant of survival with a median age at death of 29 years. Approximately 60% of CF patients are infected with Aspergillus fumigatus, a ubiquitous environmental fungus, and its presence has been associated with accelerated lung function decline. Half of the patients infected with Aspergillus are <18 years of age. Yet time of acquisition of this fungus and determinants of CF-related Aspergillus disease severity and progression are not known. CFTR expression has been demonstrated in cells of the innate and adaptive immune system and has shown to be critical for normal function. Research delineating the role of CFTR-deficient phagocytes in Aspergillus persistence and infection in the CF lung, has only recently received attention. In this concise review we aim to present the current understanding with respect to when people with CF acquire infection with A. fumigatus and antifungal immune responses by CF immune cells.
Collapse
Affiliation(s)
- Adilia Warris
- MRC Centre for Medical Mycology, Aberdeen Fungal Group, University of Aberdeen, United Kingdom
| | - Amelia Bercusson
- National Heart and Lung Institute, Imperial College London, United Kingdom
| | | |
Collapse
|
45
|
Olden BR, Cheng E, Cheng Y, Pun SH. Identifying key barriers in cationic polymer gene delivery to human T cells. Biomater Sci 2019; 7:789-797. [PMID: 30633266 PMCID: PMC6391219 DOI: 10.1039/c8bm01262h] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
T cells have emerged as a therapeutically-relevant target for ex vivo gene delivery and editing. However, most commercially available reagents cannot transfect T cells and designing cationic polymers for non-viral gene delivery to T cells has resulted in moderate success. Here, we assess various barriers to successful gene transfer in the Jurkat human T cell line and primary human T cells. Using two polymers previously developed by our group, we show that uptake is one barrier to gene delivery in primary human T cells but is not predictive of successful gene delivery. We then probe intracellular pathways for barriers to gene transfer including endosomal acidification, autophagy, and immune sensing pathways. We find that endosomal acidification is slower and not as robust in human T cells compared to the model HeLa human cell line commonly used to evaluate cationic polymers for gene delivery. These studies inform the future design of cationic polymers for non-viral gene delivery to T cells, specifically, to rely on alternative endosomal release mechanisms rather than on pH-triggered release.
Collapse
Affiliation(s)
- Brynn R Olden
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA.
| | | | | | | |
Collapse
|
46
|
Farkaš R, Beňová-Liszeková D, Mentelová L, Beňo M, Babišová K, Trusinová-Pečeňová L, Raška O, Chase BA, Raška I. Endosomal vacuoles of the prepupal salivary glands of Drosophila play an essential role in the metabolic reallocation of iron. Dev Growth Differ 2018; 60:411-430. [PMID: 30123964 DOI: 10.1111/dgd.12562] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 06/30/2018] [Accepted: 07/02/2018] [Indexed: 12/17/2022]
Abstract
In the recent past, we demonstrated that a great deal is going on in the salivary glands of Drosophila in the interval after they release their glycoprotein-rich secretory glue during pupariation. The early-to-mid prepupal salivary glands undergo extensive endocytosis with widespread vacuolation of the cytoplasm followed by massive apocrine secretion. Here, we describe additional novel properties of these endosomes. The use of vital pH-sensitive probes provided confirmatory evidence that these endosomes have acidic contents and that there are two types of endocytosis seen in the prepupal glands. The salivary glands simultaneously generate mildly acidic, small, basally-derived endosomes and strongly acidic, large and apical endosomes. Staining of the large vacuoles with vital acidic probes is possible only after there is ambipolar fusion of both basal and apical endosomes, since only basally-derived endosomes can bring fluorescent probes into the vesicular system. We obtained multiple lines of evidence that the small basally-derived endosomes are chiefly involved in the uptake of dietary Fe3+ iron. The fusion of basal endosomes with the larger and strongly acidic apical endosomes appears to facilitate optimal conditions for ferrireductase activity inside the vacuoles to release metabolic Fe2+ iron. While iron was not detectable directly due to limited staining sensitivity, we found increasing fluorescence of the glutathione-sensitive probe CellTracker Blue CMAC in large vacuoles, which appeared to depend on the amount of iron released by ferrireductase. Moreover, heterologous fluorescently-labeled mammalian iron-bound transferrin is actively taken up, providing direct evidence for active iron uptake by basal endocytosis. In addition, we serendipitously found that small (basal) endosomes were uniquely recognized by PNA lectin, whereas large (apical) vacuoles bound DBA lectin.
Collapse
Affiliation(s)
- Robert Farkaš
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Denisa Beňová-Liszeková
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lucia Mentelová
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia.,Department of Genetics, Comenius University, Bratislava, Slovakia
| | - Milan Beňo
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Klaudia Babišová
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia.,Department of Genetics, Comenius University, Bratislava, Slovakia
| | - Ludmila Trusinová-Pečeňová
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Otakar Raška
- Institute of Biology and Medical Genetics, 1st Faculty of Medicine, Charles University, Prague, Czech Republic.,Department of Normal, Pathological and Clinical Physiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Bruce A Chase
- Department of Biology, University of Nebraska, Omaha, Nebraska
| | - Ivan Raška
- Institute of Biology and Medical Genetics, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
47
|
Hayata A, Itoh H, Inoue M. Solid-Phase Total Synthesis and Dual Mechanism of Action of the Channel-Forming 48-mer Peptide Polytheonamide B. J Am Chem Soc 2018; 140:10602-10611. [PMID: 30040396 DOI: 10.1021/jacs.8b06755] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Polytheonamide B (1) is a unique peptide natural product because of its extremely complex structure, a channel-forming ability in vitro, and the extremely potent cytotoxicity. The 48-mer sequence of 1 comprises alternating d,l-amino acids and possesses an array of sterically bulky β-tetrasubstituted and hydrogen bond forming residues. These unusual structural features are believed to drive 1 to fold into a 4.5 nm long tube, form a transmembrane ion channel at the plasma membrane, and exert cytotoxicity. Despite its potential biological application, however, multiple substitutions by these unusual residues significantly heightened the synthetic challenges, impeding the solid-phase peptide synthesis (SPPS) of 1. In this study, we first addressed the synthesis problem by extensive optimization of various factors of the SPPS. Adaptation of a new protective group strategy allowed for elongation of a 37-mer peptide on resin, to which an N-terminal 11-mer fragment was condensed. Removal of the 18 protective groups and resin gave rise to 1 in excellent overall yield (4.5%, 76 steps from 17). The SPPS protocol is operationally simple and was proven easily amenable to total synthesis of the fluorescent 48-mer probe 2. Synthetic 1 and 2 were utilized for analysis of their cellular behavior. Reflecting its ion-channel function, the addition of 1 to MCF-7 cells rapidly diminished a potential across the plasma membrane. Furthermore, fluorescence imaging study revealed that 1 and 2 were also internalized into the cells, accumulating in acidic lysosomes and neutralizing the lysosomal pH gradient. These new findings indicated that 1 is capable of exerting two functions upon causing apoptotic cell death of mammalian cells: It induces free cation transport across the plasma as well as lysosomal membranes. The present chemical and biological studies provide valuable information for the design and synthesis of polytheonamide-based molecules with more potent and selective biological activities.
Collapse
Affiliation(s)
- Atsushi Hayata
- Graduate School of Pharmaceutical Sciences , The University of Tokyo , 7-3-1 Hongo, Bunkyo-ku , Tokyo 113-0033 , Japan
| | - Hiroaki Itoh
- Graduate School of Pharmaceutical Sciences , The University of Tokyo , 7-3-1 Hongo, Bunkyo-ku , Tokyo 113-0033 , Japan
| | - Masayuki Inoue
- Graduate School of Pharmaceutical Sciences , The University of Tokyo , 7-3-1 Hongo, Bunkyo-ku , Tokyo 113-0033 , Japan
| |
Collapse
|
48
|
Abstract
Dextrans are a versatile class of polysaccharides with applications that span medicine, cell biology, food science, and consumer goods. Here, we report on a new type of large monofunctionalized dextran that exhibits unusual properties: efficient cytosolic and nuclear uptake. This dextran permeates various human cell types without the use of transfection agents, electroporation, or membrane perturbation. Cellular uptake occurs primarily through active transport via receptor-mediated processes. These monofunctionalized dextrans could serve as intracellular delivery platforms for drugs or other cargos.
Collapse
Affiliation(s)
- Wen Chyan
- Department of Chemistry, University of Wisconsin−Madison, Madison, Wisconsin 53706, United States
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Henry R. Kilgore
- Graduate Program in Biophysics, University of Wisconsin−Madison, Madison, Wisconsin 53706, United States
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Ronald T. Raines
- Department of Chemistry, University of Wisconsin−Madison, Madison, Wisconsin 53706, United States
- Department of Biochemistry, University of Wisconsin−Madison, Madison, Wisconsin 53706, United States
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
49
|
Sun Q, Zhang Z, Zhang R, Gao R, McClements DJ. Development of Functional or Medical Foods for Oral Administration of Insulin for Diabetes Treatment: Gastroprotective Edible Microgels. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:4820-4826. [PMID: 29701967 DOI: 10.1021/acs.jafc.8b00233] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Insulin and an antacid [Mg(OH)2] were co-encapsulated inside calcium alginate microgels (diameter = 280 μm) using a vibrating nozzle injector. Confocal microscopy indicated that insulin was successfully encapsulated inside the microgels and remained inside them after they were exposed to simulated gastric conditions. Localized fluorescence intensity measurements indicated that the internal pH of the antacid-loaded microgels was around pH 7.4 after incubation in acidic gastric fluids but below the limit of detection (pH < 4) in the antacid-free microgels. After incubation in small intestine conditions, around 30% of the insulin was released from the antacid-loaded microgels over a 2 h period. Encapsulation of insulin within the antacid-loaded microgels increased its biological activity after exposure to simulated gastric conditions. In particular, the encapsulated insulin significantly increased Akt phosphorylation at both Thr308 and Ser473 in L6 myotubes when compared to free insulin.
Collapse
Affiliation(s)
- Quancai Sun
- School of Food and Biological Engineering , Jiangsu University , Zhenjiang , Jiangsu 212001 , People's Republic of China
| | - Zipei Zhang
- Department of Food Science , University of Massachusetts Amherst , Amherst , Massachusetts 01003 , United States
| | - Ruojie Zhang
- Department of Food Science , University of Massachusetts Amherst , Amherst , Massachusetts 01003 , United States
| | - Ruichang Gao
- School of Food and Biological Engineering , Jiangsu University , Zhenjiang , Jiangsu 212001 , People's Republic of China
| | - David Julian McClements
- Department of Food Science , University of Massachusetts Amherst , Amherst , Massachusetts 01003 , United States
| |
Collapse
|
50
|
Phagocytosis depends on TRPV2-mediated calcium influx and requires TRPV2 in lipids rafts: alteration in macrophages from patients with cystic fibrosis. Sci Rep 2018. [PMID: 29523858 PMCID: PMC5844937 DOI: 10.1038/s41598-018-22558-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Whereas many phagocytosis steps involve ionic fluxes, the underlying ion channels remain poorly defined. As reported in mice, the calcium conducting TRPV2 channel impacts the phagocytic process. Macrophage phagocytosis is critical for defense against pathogens. In cystic fibrosis (CF), macrophages have lost their capacity to act as suppressor cells and thus play a significant role in the initiating stages leading to chronic inflammation/infection. In a previous study, we demonstrated that impaired function of CF macrophages is due to a deficient phagocytosis. The aim of the present study was to investigate TRPV2 role in the phagocytosis capacity of healthy primary human macrophage by studying its activity, its membrane localization and its recruitment in lipid rafts. In primary human macrophages, we showed that P. aeruginosa recruits TRPV2 channels at the cell surface and induced a calcium influx required for bacterial phagocytosis. We presently demonstrate that to be functional and play a role in phagocytosis, TRPV2 might require a preferential localization in lipid rafts. Furthermore, CF macrophage displays a perturbed calcium homeostasis due to a defect in TRPV2. In this context, deregulated TRPV2-signaling in CF macrophages could explain their defective phagocytosis capacity that contribute to the maintenance of chronic infection.
Collapse
|