1
|
Chen X, Yu H, Bai X, Wu C, Yang M. Proteomics and phosphoproteomics differences between human and cow milk fat globule membrane: An innovative guide for infant formula. Food Chem 2025; 472:142890. [PMID: 39824080 DOI: 10.1016/j.foodchem.2025.142890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/19/2024] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
This study employed proteomics and phosphoproteomics to compare protein expression and phosphorylation modifications in the milk fat globule membrane (MFGM) of human and bovine mature milk. A total of 410 proteins and 73 phosphorylation sites on 49 proteins were identified in the MFGM of bovine and human mature milk, respectively. Differential analysis revealed 40 differentially expressed proteins and 8 differentially phosphorylated sites, followed by bioinformatics analysis. Seven overlapping proteins were identified, with GLYCAM1 and Alpha-S2-casein showing significant differences in both protein expression and phosphorylation modifications, particularly in relation to immune functions. The results suggest that bovine MFGM may provide unique advantages in supporting infant immune systems and promoting gut health, offering valuable insights for future innovation in infant formula development.
Collapse
Affiliation(s)
- Xinping Chen
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China
| | - Hong Yu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China
| | - Xue Bai
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China
| | - Chunshuang Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China
| | - Mei Yang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China.
| |
Collapse
|
2
|
Ghosh A, Gorain B. Mechanistic insight of neurodegeneration due to micro/nano-plastic-induced gut dysbiosis. Arch Toxicol 2025; 99:83-101. [PMID: 39370473 DOI: 10.1007/s00204-024-03875-3] [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: 08/10/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024]
Abstract
Despite offering significant conveniences, plastic materials contribute substantially in developing environmental hazards and pollutants. Plastic trash that has not been adequately managed may eventually break down into fragments caused by human or ecological factors. Arguably, the crucial element for determining the biological toxicities of plastics are micro/nano-forms of plastics (MPs/NPs), which infiltrate the mammalian tissue through different media and routes. Infiltration of MPs/NPs across the intestinal barrier leads to microbial architectural dysfunction, which further modulates the population of gastrointestinal microbes. Thereby, it triggers inflammatory mediators (e.g., IL-1α/β, TNF-α, and IFN-γ) by activating specific receptors located in the gut barrier. Mounting evidence indicates that MPs/NPs disrupt host pathophysiological function through modification of junctional proteins and effector cells. Moreover, the alteration of microbial diversity by MPs/NPs causes the breakdown of the blood-brain barrier and translocation of metabolites (e.g., SCFAs, LPS) through the vagus nerve. Potent penetration affects the neuronal networks, neuronal protein accumulation, acceleration of oxidative stress, and alteration of neurofibrillary tangles, and hinders distinctive communicating pathways. Conclusively, alterations of these neurotoxic factors are possibly responsible for the associated neurodegenerative disorders due to the exposure of MPs/NPs. In this review, the hypothesis on MPs/NPs associated with gut microbial dysbiosis has been interlinked to the distinct neurological impairment through the gut-brain axis.
Collapse
Affiliation(s)
- Arya Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India.
| |
Collapse
|
3
|
Manrique-Caballero CL, Barasch J, Zaidi SK, Bates CM, Ray EC, Kleyman TR, Al-Bataineh MM. Expression and distribution of MUC1 in the developing and adult kidney. Am J Physiol Renal Physiol 2025; 328:F107-F120. [PMID: 39588770 DOI: 10.1152/ajprenal.00206.2024] [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: 07/17/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 12/28/2024] Open
Abstract
Mucin 1 (or MUC1) is a heterodimeric transmembrane glycoprotein expressed on the apical surface of polarized epithelial cells in several tissues including the kidney. Recent studies have revealed several novel roles of MUC1 in the kidney, potentially including bacterial infection, mineral balance, and genetic interstitial kidney disease, even though MUC1 levels are reduced not only in the kidney but also in all tissues due to MUC1 mutations. A careful localization of MUC1 in discrete segments of the nephron is the first step in understanding the multiple functional roles of MUC1 in the kidney. Most published reports of MUC1 expression to date have been largely confined to cultured cells, tumor tissues, and selective nephron segments of experimental rodents, and very few studies have been performed using human kidney tissues. Given the rising attention to the role of MUC1 in differing components of renal physiology, we carefully examined the kidney distribution of MUC1 in both human and mouse kidney sections using well-defined markers for different nephron segments or cell types. We further extended our investigation to include sections of early stages of mouse kidney development and upon injury in humans. We included staining for MUC1 in urothelial cells, the highly specialized epithelial cells lining the renal pelvis and bladder. These data implicate a role for MUC1 in antimicrobial defense. Our study provides the groundwork to test the physiological relevance of MUC1 in the urinary tract.NEW & NOTEWORTHY MUC1 is a transmembrane glycoprotein expressed on the apical surface of polarized epithelial tissues and most carcinomas. MUC1 may play novel roles in the kidney including defense against infections. Here, we examine the expression of MUC1 in mouse and human kidneys. We show that the distal nephron and the urinary system are the predominant sites of expression of both message and protein, implicating segment-specific roles including distal nephron defense against ascending bacteria.
Collapse
Affiliation(s)
- Carlos L Manrique-Caballero
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Jonathan Barasch
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, United States
| | - Syed K Zaidi
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Carlton M Bates
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Evan C Ray
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Mohammad M Al-Bataineh
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
4
|
Leow SS, Khoo JS, Lee WK, Hoh CC, Fairus S, Sambanthamurthi R, Hayes KC. RNA-Seq transcriptome profiling of Nile rat livers reveals novel insights on the anti-diabetic mechanisms of Water-Soluble Palm Fruit Extract. J Appl Genet 2024; 65:867-895. [PMID: 38890243 DOI: 10.1007/s13353-024-00880-1] [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: 09/11/2023] [Revised: 04/08/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024]
Abstract
Water-Soluble Palm Fruit Extract (WSPFE) has been shown to confer anti-diabetic effects in the Nile rat (NR) (Arvicanthis niloticus). Liquid and powder WSPFE both deterred diabetes onset in NRs fed a high-carbohydrate (hiCHO) diet, but the liquid form provided better protection. In this study, NRs were fed either a hiCHO diet or the same diet added with liquid or powder WSPFE. Following feeding of the diets for 8 weeks, random blood glucose levels were measured to categorize NRs as either diabetes-resistant or diabetes-susceptible, based on a cut-off value of 75 mg/dL. Livers were then obtained for Illumina HiSeq 4000 paired end RNA-sequencing (RNA-Seq) and the data were mapped to the reference genome. Consistent with physiological and biochemical parameters, the gene expression data obtained indicated that WSPFE was associated with protection against diabetes. Among hepatic genes upregulated by WSPFE versus controls, were genes related to insulin-like growth factor binding protein, leptin receptor, and processes of hepatic metabolism maintenance, while those downregulated were related to antigen binding, immunoglobulin receptor, inflammation- and cancer-related processes. WSPFE supplementation thus helped inhibit diabetes progression in NRs by increasing insulin sensitivity and reducing both the inflammatory effects of a hiCHO diet and the related DNA-damage compensatory mechanisms contributing to liver disease progression. In addition, the genetic permissiveness of susceptible NRs to develop diabetes was potentially associated with dysregulated compensatory mechanisms involving insulin signaling and oxidative stress over time. Further studies on other NR organs associated with diabetes and its complications are warranted.
Collapse
Affiliation(s)
- Soon-Sen Leow
- Malaysian Palm Oil Board, No. 6, Persiaran Institusi, Bandar Baru Bangi, 43000, Kajang, Selangor, Malaysia.
| | - Jia-Shiun Khoo
- Codon Genomics Sdn Bhd, No. 26, Jalan Dutamas 7, Taman Dutamas Balakong, 43200, Seri Kembangan, Selangor, Malaysia
| | - Wei-Kang Lee
- Codon Genomics Sdn Bhd, No. 26, Jalan Dutamas 7, Taman Dutamas Balakong, 43200, Seri Kembangan, Selangor, Malaysia
| | - Chee-Choong Hoh
- Codon Genomics Sdn Bhd, No. 26, Jalan Dutamas 7, Taman Dutamas Balakong, 43200, Seri Kembangan, Selangor, Malaysia
| | - Syed Fairus
- Malaysian Palm Oil Board, No. 6, Persiaran Institusi, Bandar Baru Bangi, 43000, Kajang, Selangor, Malaysia
| | - Ravigadevi Sambanthamurthi
- Malaysian Palm Oil Board, No. 6, Persiaran Institusi, Bandar Baru Bangi, 43000, Kajang, Selangor, Malaysia
- Academy of Sciences Malaysia, Level 20, West Wing, MATRADE Tower, Jalan Sultan Haji Ahmad Shah, Off Jalan Tuanku Abdul Halim, 50480, Kuala Lumpur, Malaysia
| | - K C Hayes
- Brandeis University, 415 South Street, Waltham, MA, 02454, USA
| |
Collapse
|
5
|
Achille A, Guarnieri G, Vianello A. Krebs von den Lungen-6 (KL-6) as a diagnostic and prognostic biomarker for non-neoplastic lung diseases. Clin Chem Lab Med 2024:cclm-2024-1089. [PMID: 39589105 DOI: 10.1515/cclm-2024-1089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 11/11/2024] [Indexed: 11/27/2024]
Abstract
Important advancements have been made in understanding the pathogenetic mechanisms underlying acute and chronic lung disorders. But although a wide variety of innovative biomarkers have and are being investigated, they are not largely employed to evaluate non-neoplastic lung diseases. The current work aims to examine the use of Krebs von den Lungen-6 (KL-6), a mucin-like glycoprotein predominantly expressed on the surface of type II alveolar epithelial cells (AEC2s), to evaluate the stage, response to treatment, and prognosis in patients with non-neoplastic lung disorders. Data analysis suggests that KL-6 can be utilized as an effective diagnostic and prognostic biomarker in individuals with interstitial lung disease and as a predictor of clinical outcomes in subjects with SARS-CoV-2-related pneumonia. Moreover, KL-6 can be reliably used in routine clinical settings to diagnose and predict the outcome of patients with chronic obstructive pulmonary disease (COPD) exacerbation. The optimal cut-off points within the European population should be defined to improve KL-6's diagnostic efficacy.
Collapse
Affiliation(s)
- Alessia Achille
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Gabriella Guarnieri
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Andrea Vianello
- UOC Fisiopatologia Respiratoria Ospedale-Università di Padova Via Giustiniani, Padova, Italy
| |
Collapse
|
6
|
Święch E, Barszcz M, Taciak M, Gawin K, Tuśnio A, Konopka A, Skomiał J. Mucin gene expression in the large intestine of young pigs: The effect of dietary level of two types of chicory inulin. Nutrition 2024; 131:112635. [PMID: 39689616 DOI: 10.1016/j.nut.2024.112635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 10/09/2024] [Accepted: 11/06/2024] [Indexed: 12/19/2024]
Abstract
OBJECTIVES Inulin is a plant polysaccharide that may affect the gut barrier depending on its degree of polymerization (DP). Therefore, this study aimed to determine the effect of dietary level of two inulin types differing in DP on mucin content and MUC gene expression in the colon mucosa of pigs. METHODS The 40-d experiment involved 56 piglets divided into seven groups fed a diet with 0%, 1%, 2%, or 3% inulin with DP of 10 or 23. Mucin content was measured fluorometrically, gene expression was determined by quantitative polymerase chain reaction, and data were analyzed by one-way analysis of variance and three-way analysis of variance. RESULTS In the proximal colon, mucin content was decreased by the diet with 1% inulin with DP of 23 (IN23), whereas in the middle colon mucin content was decreased by the other IN23 diets. The diet with 3% inulin with DP of 10 upregulated MUC2 and MUC12 in the proximal colon and MUC2 and MUC13 in the middle colon. In the proximal colon, all IN23 diets increased MUC2 expression, whereas the 2% and 3% IN23 diets upregulated MUC1 and MUC12. In the middle colon, MUC2 was increased by the 1% and 2% IN23 diets, MUC4 by the 1% diet, and MUC13 by the 3% diet. In the distal colon, the 3% IN23 diet upregulated MUC1, whereas all IN23 diets downregulated MUC2, MUC4, and MUC12. With the exception of MU13 and MUC20, the expression of MUC genes was the highest in the proximal colon. CONCLUSIONS Inulin, particularly with higher DP, may exert a beneficial effect on the colon mucosa by modulation of MUC gene expression.
Collapse
Affiliation(s)
- Ewa Święch
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland.
| | - Marcin Barszcz
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| | - Marcin Taciak
- Division of Animal Nutrition, Institute of Animal Sciences, Warsaw University of Life Sciences, Warsaw, Poland
| | - Kamil Gawin
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| | - Anna Tuśnio
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| | - Adrianna Konopka
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| | - Jacek Skomiał
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| |
Collapse
|
7
|
Surve D, Fish A, Debnath M, Pinjari A, Lorenzana A, Piya S, Peyton S, Kulkarni A. Sprayable inflammasome-inhibiting lipid nanorods in a polymeric scaffold for psoriasis therapy. Nat Commun 2024; 15:9035. [PMID: 39426974 PMCID: PMC11490495 DOI: 10.1038/s41467-024-53396-x] [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: 03/03/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024] Open
Abstract
Localized delivery of inflammasome inhibitors in phagocytic macrophages could be promising for psoriasis treatment. The present work demonstrates the development of non-spherical lipid nanoparticles, mimicking pathogen-like shapes, consisting of an anti-inflammatory inflammasome inhibiting lipid (pyridoxine dipalmitate) as a trojan horse. The nanorods inhibit inflammasome by 3.8- and 4.5-fold compared with nanoellipses and nanospheres, respectively. Nanorods reduce apoptosis-associated speck-like protein and lysosomal rupture, restrain calcium influx, and mitochondrial reactive oxygen species. Dual inflammasome inhibitor (NLRP3/AIM-2-IN-3) loaded nanorods cause synergistic inhibition by 21.5- and 59-folds compared with nanorods and free drug, respectively alongside caspase-1 inhibition. The NLRP3/AIM-2-IN-3 nanorod when transformed into a polymeric scaffold, simultaneously and effectively inhibits RNA levels of NLRP3, AIM2, caspase-1, chemokine ligand-2, gasdermin-D, interleukin-1β, toll-like receptor 7/ 8, and IL-17A by 6.4-, 1.6-, 2.0-, 13.0-, 4.2-, 24.4-, 4.3-, and 1.82-fold, respectively in psoriatic skin in comparison to Imiquimod positive control group in an in-vivo psoriasis-like mice model.
Collapse
Affiliation(s)
- Dhanashree Surve
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Adam Fish
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Maharshi Debnath
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Aniruddha Pinjari
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Adrian Lorenzana
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Sumi Piya
- Pathology Department, University of Massachusetts-Chan Medical School, Baystate Medical Center, Springfield, MA, 01199, USA
| | - Shelly Peyton
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA.
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
8
|
Pisanu L, Mucaj K, Conio V, Bertuccio F, Giana I, Arlando L, Russo M, Montini S, Bortolotto C, Corsico AG, Stella GM. Lung bronchiectasisas a paradigm of the interplay between infection and colonization on plastic modulation of the pre-metastatic niche. Front Oncol 2024; 14:1480777. [PMID: 39469649 PMCID: PMC11513253 DOI: 10.3389/fonc.2024.1480777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/24/2024] [Indexed: 10/30/2024] Open
Abstract
The lungs are most often a preferential target organ for malignant spreading and growth. It is well known that chronic parenchymal inflammation and prolonged injuries represents an independent risk factor for cancer onset. Growing evidence supports the implication of lung microbiota in the pathogenesis of lung cancer. However, the full interplay between chronic inflammation, bacterial colonization, pathologic condition as bronchiectasis and malignant growth deserves better clarification. We here aim at presenting and analyzing original data and discussing the state-of-the-art on the knowledge regarding how this complex milieu acts on the plasticity of the lung pre-metastatic niche to point out the rationale for early diagnosis and therapeutic targeting.
Collapse
Affiliation(s)
- Lucrezia Pisanu
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Klodjana Mucaj
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Valentina Conio
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Francesco Bertuccio
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Ilaria Giana
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Lorenzo Arlando
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Marianna Russo
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Simone Montini
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Chandra Bortolotto
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia Medical School, Pavia, Italy
- Radiology Institute, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Angelo Guido Corsico
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Giulia Maria Stella
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
9
|
Xiao L, Pi X, Goss AC, El-Baba T, Ehrmann JF, Grinkevich E, Bazua-Valenti S, Padovano V, Alper SL, Carey D, Udeshi ND, Carr SA, Pablo JL, Robinson CV, Greka A, Wu H. Molecular basis of TMED9 oligomerization and entrapment of misfolded protein cargo in the early secretory pathway. SCIENCE ADVANCES 2024; 10:eadp2221. [PMID: 39303030 PMCID: PMC11414720 DOI: 10.1126/sciadv.adp2221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/14/2024] [Indexed: 09/22/2024]
Abstract
Intracellular accumulation of misfolded proteins causes serious human proteinopathies. The transmembrane emp24 domain 9 (TMED9) cargo receptor promotes a general mechanism of cytotoxicity by entrapping misfolded protein cargos in the early secretory pathway. However, the molecular basis for this TMED9-mediated cargo retention remains elusive. Here, we report cryo-electron microscopy structures of TMED9, which reveal its unexpected self-oligomerization into octamers, dodecamers, and, by extension, even higher-order oligomers. The TMED9 oligomerization is driven by an intrinsic symmetry mismatch between the trimeric coiled coil domain and the tetrameric transmembrane domain. Using frameshifted Mucin 1 as an example of aggregated disease-related protein cargo, we implicate a mode of direct interaction with the TMED9 luminal Golgi-dynamics domain. The structures suggest and we confirm that TMED9 oligomerization favors the recruitment of coat protein I (COPI), but not COPII coatomers, facilitating retrograde transport and explaining the observed cargo entrapment. Our work thus reveals a molecular basis for TMED9-mediated misfolded protein retention in the early secretory pathway.
Collapse
Affiliation(s)
- Le Xiao
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Xiong Pi
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Alissa C. Goss
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Tarick El-Baba
- Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford OX1 3QZ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Julian F. Ehrmann
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Elizabeth Grinkevich
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Silvana Bazua-Valenti
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Seth L. Alper
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Division of Nephrology, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Dominique Carey
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Steven A. Carr
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Juan Lorenzo Pablo
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Carol V. Robinson
- Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford OX1 3QZ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Anna Greka
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
10
|
Barlas FB, Olceroglu B, Ag Seleci D, Gumus ZP, Siyah P, Dabbek M, Garnweitne G, Stahl F, Scheper T, Timur S. Enhancing chemotherapeutic efficacy: Niosome-encapsulated Dox-Cis with MUC-1 aptamer. Cancer Med 2024; 13:e70079. [PMID: 39118454 PMCID: PMC11310550 DOI: 10.1002/cam4.70079] [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: 06/10/2024] [Revised: 07/10/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Cancer remains a formidable global health challenge, currently affecting nearly 20 million individuals worldwide. Due to the absence of universally effective treatments, ongoing research explores diverse strategies to combat this disease. Recent efforts have concentrated on developing combined drug regimens and targeted therapeutic approaches. OBJECTIVE This study aimed to investigate the anticancer efficacy of a conjugated drug system, consisting of doxorubicin and cisplatin (Dox-Cis), encapsulated within niosomes and modified with MUC-1 aptamers to enhance biocompatibility and target specific cancer cells. METHODS The chemical structure of the Dox-Cis conjugate was characterized using Fourier Transform Infrared Spectroscopy (FTIR) and Liquid Chromatography Quadrupole Time-of-Flight Mass Spectrometry (LC-Q-TOF/MS). The zeta potential and morphological parameters of the niosomal vesicles were determined through Dynamic Light Scattering (DLS) and Transmission Electron Microscopy (TEM). In vitro assessments of cell viability and apoptosis were conducted on MUC-1 positive HeLa cells and MUC-1 negative U87 cells. RESULTS The findings confirmed the successful conjugation of Dox and Cis within the niosomes. The Nio/Dox-Cis/MUC-1 formulation demonstrated enhanced efficacy compared to the individual drugs and their unencapsulated combination in both cell lines. Notably, the Nio/Dox-Cis/MUC-1 formulation exhibited greater effectiveness on HeLa cells (38.503 ± 1.407) than on U87 cells (46.653 ± 1.297). CONCLUSION The study underscores the potential of the Dox-Cis conjugate as a promising strategy for cancer treatment, particularly through platforms that facilitate targeted drug delivery to cancer cells. This targeted approach could lead to more effective and personalized cancer therapies.
Collapse
Affiliation(s)
- Firat Baris Barlas
- Institute for Technical ChemistryLeibniz University HannoverHannoverGermany
- Institue of Nanotechnology and Biotechnologyİstanbul University‐CerrahpaşaİstanbulTurkey
| | - Bilge Olceroglu
- Institue of Nanotechnology and Biotechnologyİstanbul University‐CerrahpaşaİstanbulTurkey
| | - Didem Ag Seleci
- Institute for Particle Technology (iPAT)Technische Universität BraunschweigBraunschweigGermany
| | - Zinar Pinar Gumus
- Central Research Test and Analysis Laboratory Application and Research CenterEge UniversityIzmirTurkey
| | - Pinar Siyah
- Department of Biochemistry, School of PharmacyBahçeşehir UniversityIstanbulTurkey
| | - Meriam Dabbek
- Institute for Technical ChemistryLeibniz University HannoverHannoverGermany
| | - Georg Garnweitne
- Institute for Particle Technology (iPAT)Technische Universität BraunschweigBraunschweigGermany
| | - Frank Stahl
- Institute for Technical ChemistryLeibniz University HannoverHannoverGermany
| | - Thomas Scheper
- Institute for Technical ChemistryLeibniz University HannoverHannoverGermany
| | - Suna Timur
- Central Research Test and Analysis Laboratory Application and Research CenterEge UniversityIzmirTurkey
- Department of Biochemistry, Faculty of ScienceEge UniversityIzmirTurkey
| |
Collapse
|
11
|
Weaver JJ, Smith AM. Quantitatively Mapping Immune Control during Influenza. CURRENT OPINION IN SYSTEMS BIOLOGY 2024; 38:100516. [PMID: 39430368 PMCID: PMC11488648 DOI: 10.1016/j.coisb.2024.100516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Host immune responses play a pivotal role in defending against influenza viruses. The activation of various immune components, such as interferon, macrophages, and CD8+ T cells, works to limit viral spread while maintaining lung integrity. Recent mathematical modeling studies have investigated these responses, describing their regulation, efficacy, and movement within the lung. Here, we discuss these studies and their emphasis on identifying nonlinearities and multifaceted roles of different cell phenotypes that could be responsible for spatially heterogeneous infection patterns.
Collapse
Affiliation(s)
- Jordan J.A. Weaver
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163 USA
| | - Amber M. Smith
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163 USA
| |
Collapse
|
12
|
Zhao A, Pan Y, Gao Y, Zhi Z, Lu H, Dong B, Zhang X, Wu M, Zhu F, Zhou S, Ma S. MUC1 promotes cervical squamous cell carcinoma through ERK phosphorylation-mediated regulation of ITGA2/ITGA3. BMC Cancer 2024; 24:559. [PMID: 38702644 PMCID: PMC11069143 DOI: 10.1186/s12885-024-12314-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 04/26/2024] [Indexed: 05/06/2024] Open
Abstract
In contrast to the decreasing trends in developed countries, the incidence and mortality rates of cervical squamous cell carcinoma in China have increased significantly. The screening and identification of reliable biomarkers and candidate drug targets for cervical squamous cell carcinoma are urgently needed to improve the survival rate and quality of life of patients. In this study, we demonstrated that the expression of MUC1 was greater in neoplastic tissues than in non-neoplastic tissues of the cervix, and cervical squamous cell carcinoma patients with high MUC1 expression had significantly worse overall survival than did those with low MUC1 expression, indicating its potential for early diagnosis of cervical squamous cell carcinoma. Next, we explored the regulatory mechanism of MUC1 in cervical squamous cell carcinoma. MUC1 could upregulate ITGA2 and ITGA3 expression via ERK phosphorylation, promoting the proliferation and metastasis of cervical cancer cells. Further knockdown of ITGA2 and ITGA3 significantly inhibited the tumorigenesis of cervical cancer cells. Moreover, we designed a combination drug regimen comprising MUC1-siRNA and a novel ERK inhibitor in vivo and found that the combination of these drugs achieved better results in animals with xenografts than did MUC1 alone. Overall, we discovered a novel regulatory pathway, MUC1/ERK/ITGA2/3, in cervical squamous cell carcinoma that may serve as a potential biomarker and therapeutic target in the future.
Collapse
Affiliation(s)
- Aiqin Zhao
- Department of Obstetrics and Gynecology, The People's Hospital of Suzhou New District, Suzhou, 215129, China
| | - Yunzhi Pan
- Department of Pharmacy, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou, 215131, China
| | - Yingyin Gao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
- Key Laboratory of New Drug Delivery Systems of Chinese Materia Medica, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Zheng Zhi
- Department of Pathology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China
| | - Haiying Lu
- Department of Obstetrics and Gynecology, The People's Hospital of Suzhou New District, Suzhou, 215129, China
| | - Bei Dong
- Department of Obstetrics and Gynecology, The People's Hospital of Suzhou New District, Suzhou, 215129, China
| | - Xuan Zhang
- Department of Obstetrics and Gynecology, The People's Hospital of Suzhou New District, Suzhou, 215129, China
| | - Meiying Wu
- Department of Tuberculosis, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou, 215131, China
| | - Fenxia Zhu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
- Key Laboratory of New Drug Delivery Systems of Chinese Materia Medica, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Sufang Zhou
- Department of Obstetrics and Gynecology, The People's Hospital of Suzhou New District, Suzhou, 215129, China.
| | - Sai Ma
- Department of Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China.
- Gusu School, Nanjing Medical University, Suzhou, 215008, China.
| |
Collapse
|
13
|
D'Agostino GD, Chaudhari SN, Devlin AS. Host-microbiome orchestration of the sulfated metabolome. Nat Chem Biol 2024; 20:410-421. [PMID: 38347214 DOI: 10.1038/s41589-023-01526-9] [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: 12/23/2022] [Accepted: 12/08/2023] [Indexed: 04/01/2024]
Abstract
Recent studies have demonstrated that metabolites produced by commensal bacteria causally influence health and disease. The sulfated metabolome is one class of molecules that has recently come to the forefront due to efforts to understand the role of these metabolites in host-microbiome interactions. Sulfated compounds have canonically been classified as waste products; however, studies have revealed a variety of physiological roles for these metabolites, including effects on host metabolism, immune response and neurological function. Moreover, recent research has revealed that commensal bacteria either chemically modify or synthesize a variety of sulfated compounds. In this Review, we explore how host-microbiome collaborative metabolism transforms the sulfated metabolome. We describe bacterial and mammalian enzymes that sulfonate and desulfate biologically relevant carbohydrates, amino acid derivatives and cholesterol-derived metabolites. We then discuss outstanding questions and future directions in the field, including potential roles of sulfated metabolites in disease detection, prevention and treatment. We hope that this Review inspires future research into sulfated compounds and their effects on physiology.
Collapse
Affiliation(s)
- Gabriel D D'Agostino
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Snehal N Chaudhari
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
- Department of Biochemistry, University of Wisconsin, Madison, WI, USA.
| | - A Sloan Devlin
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Choi M, Tichenor AA. Regional Conjunctival Differences in Glycocalyx Mucin Expression in Dry Eye and Normal Subjects. Invest Ophthalmol Vis Sci 2024; 65:20. [PMID: 38334701 PMCID: PMC10860684 DOI: 10.1167/iovs.65.2.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 01/10/2024] [Indexed: 02/10/2024] Open
Abstract
Purpose To compare regional conjunctival expression of membrane-associated mucins (MAMs) MUC1, MUC4, and MUC16 in normal and dry eye (DE) subjects. Methods Adults with and without signs and symptoms of DE were recruited. Impression cytology was performed to collect MAMs from four bulbar and upper eyelid palpebral conjunctival regions of both eyes. After protein extraction, samples from both eyes of a single subject were pooled by region, and expression was analyzed using a capillary electrophoresis nano-immunoassay system. The chemiluminescence intensity of each antigen binding signal was calculated after normalization to the total protein amount. Statistical analyses were conducted using GraphPad Prime 9. Results Samples from thirteen to sixteen DE and seven to eleven normal subjects were analyzed. In normal samples, MUC1 expression from the nasal bulbar conjunctiva was significantly greater than superior (P = 0.004) and inferior (P = 0.005). In DE samples, MUC1 expression was highest superiorly. Significant differences in MUC4 and MUC16 expression were not seen in normal samples. MUC4 and MUC16 expression was upregulated superiorly (P < 0.0001) and inferiorly (P < 0.0001) in DE compared with those regions in normal samples. Conclusions Although MAMs form a hydrophilic barrier called the glycocalyx, each mucin may have unique functions that are currently unexplored. All MAMs were expressed in the upper palpebral conjunctiva. Increased MUC1 expression nasally in healthy subjects suggests a functional need for increased protection. When comparing DE with normal eyes, upregulation of MUC1 superiorly, and in both MUC4 and MUC16 both superiorly and inferiorly, may indicate a need to decrease eyelid friction during blinking, especially in DE.
Collapse
Affiliation(s)
- Moonjung Choi
- New England College of Optometry, Boston, Massachusetts, United States
| | | |
Collapse
|
15
|
Tong X, Dong C, Liang S. Mucin1 as a potential molecule for cancer immunotherapy and targeted therapy. J Cancer 2024; 15:54-67. [PMID: 38164273 PMCID: PMC10751670 DOI: 10.7150/jca.88261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/23/2023] [Indexed: 01/03/2024] Open
Abstract
Mucin1 is a highly glycosylated type 1 transmembrane mucin that ranks second among 75 tumor-related antigens published by the National Cancer Institute, and has been identified as a possible therapeutic target over the past 30 years. MUC1 plays an important role in malignant transformation and disease evolution, including cell proliferation, survival, self-renewal, and metastatic invasion. MUC1 has been shown to interact with diverse effectors such as β-catenin, receptor tyrosine kinases, and cellular-abelsongene, which are of importance in the pathogenesis of various malignant tumors. Targeting MUC1 has been shown to be an effective way to induce tumor cell death in vivo and in vitro models. In recent years, a number of therapeutic strategies targeting MUC1 have been developed and their value for tumor therapy have been demonstrated experimentally. This review summarizes recent findings on the structure of MUC1, its expression in different tumors and its involved mechanism pathways, with emphasis on new progress in cancer therapy which related MUC1 in the past decade and evaluates their therapeutic effect.
Collapse
Affiliation(s)
| | - Chunyan Dong
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Shujing Liang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
16
|
Mun D, Kang M, Shin M, Choi HJ, Kang AN, Ryu S, Unno T, Maburutse BE, Oh S, Kim Y. Alleviation of DSS-induced colitis via bovine colostrum-derived extracellular vesicles with microRNA let-7a-5p is mediated by regulating Akkermansia and β-hydroxybutyrate in gut environments. Microbiol Spectr 2023; 11:e0012123. [PMID: 37966243 PMCID: PMC10714758 DOI: 10.1128/spectrum.00121-23] [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: 01/09/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023] Open
Abstract
IMPORTANCE Even though studying on the possible involvement of extracellular vesicles (EVs) in host-microbe interactions, how these relationships mediate host physiology has not clarified yet. Our current findings provide insights into the encouraging benefits of dietary source-derived EVs and microRNAs (miRNAs) on organic acid production and ultimately stimulating gut microbiome for human health, suggesting that supplementation of dietary colostrum EVs and miRNAs is a novel preventive strategy for the treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Daye Mun
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| | - Minkyoung Kang
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, South Korea
| | - Minhye Shin
- Department of Microbiology, College of Medicine, Inha University, Incheon, South Korea
| | - Hye Jin Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| | - An Na Kang
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| | - Sangdon Ryu
- Division of Evironmental Meterials, Honam National Institute of Biological Resources, Mokpo, South Korea
| | - Tatsuya Unno
- Department of Microbiology, Chungbuk National University, Cheongju, South Korea
| | - Brighton E. Maburutse
- Department of Animal Production Sciences, Marondera University of Agricultural Sciences & Technology, Marondera, Zimbabwe
| | - Sangnam Oh
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, South Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| |
Collapse
|
17
|
Jin Y, Zhang D, Deng K, Wu P, Yang D, Xie Z, Qiu W, Yu G. Role of the cAMP-PKA-NF-κB pathway in Mucin1 over-expression in A549 cells during Respiratory syncytial virus infection. BMC Infect Dis 2023; 23:845. [PMID: 38036963 PMCID: PMC10687811 DOI: 10.1186/s12879-023-08837-1] [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: 05/22/2023] [Accepted: 11/22/2023] [Indexed: 12/02/2023] Open
Abstract
Respiratory syncytial virus (RSV) is the most common pathogen associated with acute lower respiratory tract infections in infants and young children worldwide. RSV commonly presents as bronchiolitis in young children; however, it can sometimes progress to pneumonia, respiratory failure, apnoea and even death. Although mucin1 (MUC1), a type of transmembrane glycoprotein present on airway epithelial surfaces, plays a crucial anti-inflammatory role in airway infections; however, its roles in RSV-associated acute lower respiratory tract infections have rarely been explored. In this study, we first revealed very high MUC1 protein levels in the exacerbation phase in sputum samples from children with RSV bronchiolitis. Because MUC1 is the downstream target of tumour necrosis factor-alpha (TNF-α) in RSV-infected A549 cells, we observed the inhibition of NF-κB activity, main downstream signalling of TNF-α and remarkably reduced levels of MUC1 in RSV-infected and TNF-α treated A549 cells. Furthermore, the cyclic adenosine monophosphate (cAMP) analogue (dbcAMP) downregulated the protein levels of p-IκBα and MUC1 in TNF-α-treated A549 cells. By contrast, a protein kinase A inhibitor (KT5720) up-regulated the levels of those proteins. dbcAMP and KT5720 had the same effects on MUC1 protein levels in RSV-infected A549 cells. In conclusion, we found that the cAMP-PKA-NF-κB pathway may play a role in the regulation of MUC-1 over-expression during RSV infection.
Collapse
Affiliation(s)
- Yingkang Jin
- Department of Respiratory Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Dongwei Zhang
- Department of Respiratory Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Kuimiao Deng
- Department of Respiratory Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Peiqiong Wu
- Department of Respiratory Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Diyuan Yang
- Department of Respiratory Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Zhiwei Xie
- Department of Respiratory Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Wenjun Qiu
- Department of Respiratory Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Guangyuan Yu
- Department of Respiratory Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China.
| |
Collapse
|
18
|
Tóth Š, Fagová Z, Holodová M, Zeidan D, Hartel P, Čurgali K, Mechírová E, Maretta M, Nemcová R, Gancarčíková S, Danková M. Influence of Escherichia coli infection on intestinal mucosal barrier integrity of germ-free piglets. Life Sci 2023; 331:122036. [PMID: 37633417 DOI: 10.1016/j.lfs.2023.122036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/11/2023] [Accepted: 08/21/2023] [Indexed: 08/28/2023]
Abstract
AIMS We focused on investigating the influence of Escherichia coli (E. coli) on the intestinal barrier. MATERIAL AND METHODS We studied changes in the distribution and secretory activities of goblet cells and enteroendocrine cells (EECs), as well as changes in the population of mast cells (MCs) in the jejunal and colonic mucosa of germ-free (GF) piglets as a healthy control group and GF piglets whose intestines were colonised with E. coli bacteria on day 5. KEY FINDINGS The results suggest that the colon of GF piglets is more resistant and less prone to coliform bacterial infection compared to the jejunum. This can be confirmed by a lower degree of histopathological injury index as well as an improvement of the morphometric parameters of the colonic mucosa, together with a significantly increased (p < 0.05) expression of MUC1/EMA, and ZO-3. We also observed a significant decrease in the population of activated MCs (p < 0.001) and EECs (p < 0.001). These findings may indicate a rapid response and better preparation of the intestinal barrier for possible pathological attacks and the subsequent development of mucosal lesions during the development and progression of the intestinal diseases. SIGNIFICANCE To date, gut-targeted therapeutic approaches that can modulate bacterial translocation and chronic inflammation are still in their infancy but represent one of the most promising areas of research for the development of new effective treatments or clinical strategies in the future. Therefore, a better understanding of these processes can significantly contribute to the development of these targeted strategies for disease prevention and treatment.
Collapse
Affiliation(s)
- Štefan Tóth
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Histology and Embryology, Šrobárova 2, 040 01 Košice, Slovak Republic
| | - Zuzana Fagová
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Histology and Embryology, Šrobárova 2, 040 01 Košice, Slovak Republic
| | - Monika Holodová
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Histology and Embryology, Šrobárova 2, 040 01 Košice, Slovak Republic
| | - Dema Zeidan
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Histology and Embryology, Šrobárova 2, 040 01 Košice, Slovak Republic
| | - Patrick Hartel
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Histology and Embryology, Šrobárova 2, 040 01 Košice, Slovak Republic
| | - Kristína Čurgali
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Histology and Embryology, Šrobárova 2, 040 01 Košice, Slovak Republic
| | - Eva Mechírová
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Histology and Embryology, Šrobárova 2, 040 01 Košice, Slovak Republic
| | - Milan Maretta
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Neurology and L. Pasteur University Hospital, Trieda SNP 1, 040 01 Košice, Slovak Republic
| | - Radomíra Nemcová
- University of Veterinary Medicine and Pharmacy in Košice, Department of Microbiology and Immunology, Komenského 73, 041 70 Košice, Slovak Republic
| | - Soňa Gancarčíková
- University of Veterinary Medicine and Pharmacy in Košice, Department of Microbiology and Immunology, Komenského 73, 041 70 Košice, Slovak Republic
| | - Marianna Danková
- Comenius University in Bratislava, Faculty of Medicine, Institute of Histology and Embryology, Sasinkova 4, 811 04 Bratislava, Slovak Republic.
| |
Collapse
|
19
|
Atwell B, Chalasani P, Schroeder J. Nuclear epidermal growth factor receptor as a therapeutic target. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:616-629. [PMID: 37720348 PMCID: PMC10501894 DOI: 10.37349/etat.2023.00156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/09/2023] [Indexed: 09/19/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) is one of the most well-studied oncogenes with roles in proliferation, growth, metastasis, and therapeutic resistance. This intense study has led to the development of a range of targeted therapeutics including small-molecule tyrosine kinase inhibitors (TKIs), monoclonal antibodies, and nanobodies. These drugs are excellent at blocking the activation and kinase function of wild-type EGFR (wtEGFR) and several common EGFR mutants. These drugs have significantly improved outcomes for patients with cancers including head and neck, glioblastoma, colorectal, and non-small cell lung cancer (NSCLC). However, therapeutic resistance is often seen, resulting from acquired mutations or activation of compensatory signaling pathways. Additionally, these therapies are ineffective in tumors where EGFR is found predominantly in the nucleus, as can be found in triple negative breast cancer (TNBC). In TNBC, EGFR is subjected to alternative trafficking which drives the nuclear localization of the receptor. In the nucleus, EGFR interacts with several proteins to activate transcription, DNA repair, migration, and chemoresistance. Nuclear EGFR (nEGFR) correlates with metastatic disease and worse patient prognosis yet targeting its nuclear localization has proved difficult. This review provides an overview of current EGFR-targeted therapies and novel peptide-based therapies that block nEGFR, as well as their clinical applications and potential for use in oncology.
Collapse
Affiliation(s)
- Benjamin Atwell
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Pavani Chalasani
- Department of Medicine, University of Arizona, Tucson, AZ 85721, USA
- University of Arizona Cancer Center, Tucson, AZ 85721, USA
| | - Joyce Schroeder
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
- University of Arizona Cancer Center, Tucson, AZ 85721, USA
- Bio5 Institute, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
20
|
Fang J, Lai S, Yu H, Ma L. Suppression of MUC1-Overexpressing Tumors by a Novel MUC1/CD3 Bispecific Antibody. Antibodies (Basel) 2023; 12:47. [PMID: 37489369 PMCID: PMC10366937 DOI: 10.3390/antib12030047] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 07/26/2023] Open
Abstract
Mucin1 (MUC1) is abnormally glycosylated and overexpressed in a variety of epithelial cancers and plays a critical role in tumor progression. MUC1 has received remark attention as an oncogenic molecule and is considered a valuable tumor target for immunotherapy, while many monoclonal antibodies (mAbs) targeting MUC1-positive cancers in clinical studies lack satisfactory results. It would be highly desirable to develop an effective therapy against MUC1-expressing cancers. In this study, we constructed a novel T cell-engaging bispecific antibody (BsAb) targeting MUC1 and CD3 with the Fab-ScFv-IgG format. A high quality of MUC1-CD3 BsAb can be acquired through a standard method. Our study suggested that this BsAb could specifically bind to MUC1- and CD3-positive cells and efficiently enhance T cell activation, cytokine release, and cytotoxicity. Furthermore, our study demonstrated that this BsAb could potently redirect T cells to eliminate MUC1-expressing tumor cells in vitro and significantly suppress MUC1-positive tumor growth in a xenograft mouse model. Thus, T cell-engaging MUC1/CD3 BsAb could be an effective therapeutic approach to combat MUC1-positive tumors and our MUC1/CD3 BsAb could be a promising candidate in clinical applications for the treatment of MUC1-positive cancer patients.
Collapse
Affiliation(s)
- Jun Fang
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, No. 10, Lishan Road, Nanshan District, Shenzhen 518055, China
| | - Shifa Lai
- BenHealth Biopharmaceutical (Shenzhen) Co., Ltd., No. 10, Gaoxinzhong First Avenue, Nanshan District, Shenzhen 518055, China
| | - Haoyang Yu
- BenHealth Biopharmaceutical (Shenzhen) Co., Ltd., No. 10, Gaoxinzhong First Avenue, Nanshan District, Shenzhen 518055, China
| | - Lan Ma
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, No. 10, Lishan Road, Nanshan District, Shenzhen 518055, China
| |
Collapse
|
21
|
Martínez-Ortega JI, Gómez-Torres RE. Oral Focal Mucinosis of the Tongue: A Rare Localization for a Rare Entity. Am J Dermatopathol 2023; 45:00000372-990000000-00218. [PMID: 37462208 DOI: 10.1097/dad.0000000000002490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
ABSTRACT Oral focal mucinosis (OFM) is a rare connective tissue disorder that is characterized by the excessive production of hyaluronic acid due to myxoid degeneration of submucosal connective tissue. The disorder typically presents as an asymptomatic nodule or mass in the gingiva or hard palate, and OFM of the tongue is even more unusual. In this report, we present a case of OFM on the tongue in a 72-year-old female patient who presented with a symptomatic lump that had been growing for 6 months on the dorsum of her tongue. The patient reported discomfort and pain while speaking and swallowing, and the lump was visually apparent on examination. OFM is a benign condition that does not have any specific clinical or radiographical features that distinguish it from other more common oral lesions, such as lipoma or fibroma. Therefore, histopathological examination is essential for a definitive diagnosis. The management of OFM typically involves surgical excision of the lesion. In this case, complete surgical removal of the lesion under general anesthesia was performed, and the patient was followed up for 10 months postoperatively. During the follow-up period, there was no evidence of recurrence, and the patient reported significant improvement in her symptoms. In conclusion, OFM is a rare connective tissue disorder that can occur in the oral cavity. Although OFM of the tongue is even rarer, it should be considered in the differential diagnosis of oral lesions. Histopathological examination is essential for definitive diagnosis, and surgical excision is typically the preferred treatment modality.
Collapse
Affiliation(s)
| | - Rosa E Gómez-Torres
- Research Institute in Dentistry, Center of Health Sciences, University of Guadalajara, Guadalajara, México
| |
Collapse
|
22
|
Cheng M, Zhang Y, Yan J, Huang Y, Wang M, Zhai Z, Liu G, Liu C, Li J, Zhang Y, Xiao Y, Wang C, Ban C, Ren Z, Song L. Inhibiting virus replication and excessive inflammatory response: Mechanism of combined prescription of Ma-Xing-Shi-Gan decoction and Xiao-Chai-Hu decoction against influenza virus. JOURNAL OF ETHNOPHARMACOLOGY 2023; 313:116481. [PMID: 37072090 DOI: 10.1016/j.jep.2023.116481] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/24/2023] [Accepted: 04/08/2023] [Indexed: 05/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The combined prescription of two classical decoctions (Ma-Xing-Shi-Gan decoction with Xiao-Chai-Hu decoction), named as San-Yang-He-Zhi (SYHZ) decoction, has been widely used for the treatment of influenza virus (IFV) infections for decades. AIM OF THE STUDY This study aimed to evaluate the anti-influenza effect of SYHZ decoction and explore the underlying mechanism. MATERIALS AND METHODS The ingredients of SYHZ decoction were analyzed by mass spectrometry. An animal model of IFV infection was established by challenging C57BL/6J mice with PR8 virus. Three groups of mice were infected with lethal or non-lethal doses of IFV, then followed by oral administration of phosphate-buffered saline (PBS), or SYHZ, or oseltamir; blank control mice (without IFV infection) were treated with PBS. Survival rate, Lung index, colon length, body weight loss and IFV viral load were measured 7 days post infection; histology and electron-microscopy examinations of lung tissue were performed; cytokine and chemokine levels in lung and serum were measured; and the intestinal metagenome, the cecum metabolome, and the lung transcriptome were analyzed. RESULTS SYHZ treatment significantly improved survival rate compared with PBS (40% vs 0%); improved lung index, colon length, and body weight loss; and alleviated lung histological damage and viral load. SYHZ-treated mice had significantly lower levels of IL-1β, TNF-α, IL-6, CCL2, CXCL10 in lung and serum, and increased levels of multiple bioactive components in cecum. Pro-inflammatory cytokines, Toll- and NOD-like receptors, pro-apoptosis molecules, and lung-injury-related proteins were downregulated in SYHZ mice, whereas surfactant protein and mucin were upregulated. The NOD-like receptor pathway, Toll-like receptor pathway, and NF-κB pathway were downregulated by SYHZ treatment. CONCLUSIONS SYHZ decoction alleviated IFV infection in a mouse model. Multiple bioactive ingredients of SYHZ may inhibit replication of IFV and suppress excessive immune response.
Collapse
Affiliation(s)
- Miao Cheng
- Respiratory Department, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yanan Zhang
- Respiratory Department, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Jun Yan
- Respiratory Department, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yuanming Huang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, 102206, China
| | - Mingzhe Wang
- Respiratory Department, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Zhiguang Zhai
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medicine Science, Beijing, 100700, China
| | - Guoxing Liu
- Traditional Chinese Medicine Department, Linwei Liu Zunji Clinic of Traditional Chinese Medicine, Weinan, 714000, China
| | - Chang Liu
- Gulou Hospital of Traditional Chinese Medicine of Beijing, 100009, China
| | - Jintong Li
- Respiratory Department, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yue Zhang
- Respiratory Department, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, 100029, China
| | - Yuchun Xiao
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, 102206, China
| | - Chengxiang Wang
- Respiratory Department, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, 100029, China
| | - Chengjun Ban
- Respiratory Department, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Zhihong Ren
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, 102206, China.
| | - Liqiong Song
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, 102206, China.
| |
Collapse
|
23
|
Sattari Fard F, Jalilzadeh N, Mehdizadeh A, Sajjadian F, Velaei K. Understanding and targeting anoikis in metastasis for cancer therapies. Cell Biol Int 2023; 47:683-698. [PMID: 36453448 DOI: 10.1002/cbin.11970] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022]
Abstract
The development of effective treatments for cancers requires investigations for a more detailed and comprehensive understanding of the basic cellular mechanisms involved in carcinogenesis, cancer progression, and metastasis. One of those driving mechanisms is anoikis, a special type of apoptosis, which is induced by losing anchorage from the extracellular matrix (ECM). In other words, resisting death in detached cells (cells without ECM) forms an anoikis-resistant phenotype. Since the anoikis-resistance state compensates for the initial steps of cancer metastasis, this review aimed to discuss mechanisms of gaining anoikis/anoikis resistance phenotype in tumor cells. Finally, we highlighted the significance of anoikis in malignancies so as to provide clear insight into cancer diagnosis and therapy development.
Collapse
Affiliation(s)
- Farzad Sattari Fard
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, School of Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Jalilzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fakhrosadat Sajjadian
- Department of Radiology, Faculty of Para-Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kobra Velaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, School of Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
24
|
Hamza FN, Daher S, Fakhoury HMA, Grant WB, Kvietys PR, Al-Kattan K. Immunomodulatory Properties of Vitamin D in the Intestinal and Respiratory Systems. Nutrients 2023; 15:nu15071696. [PMID: 37049536 PMCID: PMC10097244 DOI: 10.3390/nu15071696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Vitamin D plays a crucial role in modulating the innate immune response by interacting with its intracellular receptor, VDR. In this review, we address vitamin D/VDR signaling and how it contributes to the regulation of intestinal and respiratory microbiota. We additionally review some components of the innate immune system, such as the barrier function of the pulmonary and intestinal epithelial membranes and secretion of mucus, with their respective modulation by vitamin D. We also explore the mechanisms by which this vitamin D/VDR signaling mounts an antimicrobial response through the transduction of microbial signals and the production of antimicrobial peptides that constitute one of the body’s first lines of defense against pathogens. Additionally, we highlight the role of vitamin D in clinical diseases, namely inflammatory bowel disease and acute respiratory distress syndrome, where excessive inflammatory responses and dysbiosis are hallmarks. Increasing evidence suggests that vitamin D supplementation may have potentially beneficial effects on those diseases.
Collapse
Affiliation(s)
- Fatheia N. Hamza
- College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| | - Sarah Daher
- College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| | - Hana M. A. Fakhoury
- College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
- Correspondence:
| | - William B. Grant
- Sunlight, Nutrition, and Health Research Center, P.O. Box 641603, San Francisco, CA 94164-1603, USA
| | - Peter R. Kvietys
- College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| | - Khaled Al-Kattan
- College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| |
Collapse
|
25
|
Kyoung H, Kim E, Cho JH, Lee H, Kim Y, Park KI, Kim HB, Song M. Dietary yeast cell wall enhanced intestinal health of broiler chickens by modulating intestinal integrity, immune responses, and microbiota. Poult Sci 2023; 102:102660. [PMID: 37043955 PMCID: PMC10140172 DOI: 10.1016/j.psj.2023.102660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/05/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
This study was conducted to determine the effects of dietary yeast cell wall (YCW) on growth performance, intestinal health, and immune responses of broiler chickens. In a randomized completely block design (block: initial body weight), a total of 800 broilers (Ross 308; 45.18 ± 3.13 g of initial body weight) were assigned to 2 dietary treatments (40 birds/pen; 10 replicates/treatment) and fed for 5 wk: 1) a basal broiler diet based on corn-soybean meal (CON) and 2) CON + 0.05% dietary YCW. Growth performance was measured at intervals in 3 phase feed program. On the final day of the study, one bird per pen was randomly selected and euthanized for sample collection. Broilers fed YCW had decreased (P < 0.05) feed conversion ratio during the grower phase compared with those fed CON. The YCW increased (P < 0.05) villus height to crypt depth ratio in the duodenum, jejunum, and ileum compared with the CON. In addition, the YCW tended to higher (P < 0.10) number of goblet cells in the duodenum than in the CON. Broilers fed YCW had increased (P < 0.05) serum TGF- β1, ileal gene expression of the claudin family, and relative abundance of Lactobacillus, Prevotella, and Enterococcus compared with the CON, but decreased serum TNF-α (P < 0.05), IL-1β (P < 0.05), and IL-6 (P < 0.10), ileal gene expression of IL-6 (P < 0.05), and relative abundance of Clostridium (P < 0.05). The present study demonstrated that the addition of dietary YCW in broiler diets enhanced the intestinal health of broiler chickens and may be associated with modulated intestinal morphology and integrity by upregulating tight junction-related protein gene expression and modifying the ileal microbiota. In addition, dietary YCW modulated immune responses and inflammatory cytokine gene expression in the ileum.
Collapse
Affiliation(s)
- Hyunjin Kyoung
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Eunjoo Kim
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, Republic of Korea; School of Environmental and Rural Science, University of New England, Armidale, NSW 2351, Australia
| | - Jin Ho Cho
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Hanbae Lee
- Pathway Intermediates, Seoul 06253, Republic of Korea
| | - Yonghee Kim
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Kyeong Il Park
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Hyeun Bum Kim
- Department of Animal Resources Science, Dankook University, Cheonan 31116, Republic of Korea
| | - Minho Song
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, Republic of Korea.
| |
Collapse
|
26
|
Supimon K, Sangsuwannukul T, Sujjitjoon J, Chieochansin T, Junking M, Yenchitsomanus PT. Cytotoxic activity of anti-mucin 1 chimeric antigen receptor T cells expressing PD-1-CD28 switch receptor against cholangiocarcinoma cells. Cytotherapy 2023; 25:148-161. [PMID: 36396553 DOI: 10.1016/j.jcyt.2022.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 09/20/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND AIMS Cholangiocarcinoma (CCA) is a lethal bile-duct cancer that is difficult to treat by current standard procedures. This drawback has prompted us to develop adoptive T-cell therapy for CCA, which requires an appropriate target antigen for binding of chimeric antigen receptor (CAR) T cells. Mucin 1 (MUC1), an overexpressed protein in CCA cells, is a potential target antigen for the CAR T-cell development. However, MUC1 overexpression also is associated with the upregulation of programmed death-ligand 1 (PD-L1), an immune checkpoint protein that prohibits anti-tumor functions of T cells, probably causing poor overall survival of patients with CCA. METHODS To overcome this problem, we developed anti-MUC1-CAR T cells containing PD-1-CD28 switch receptor (SR), namely αM.CAR/SR T cells, to target MUC1 and switch on the inhibitory signal of PD-1/PD-L1 interaction to activate CD28 signaling. Our lentiviral construct contains the sequences that encode anti-MUC1-single chain variable fragment, CD137 and CD3ζ, linked with P2A, PD-1 and CD28. RESULTS Initially, the upregulations of MUC1 and PD-L1 proteins were confirmed in CCA cell lines. αM.CAR and SR were co-expressed in 53.53 ± 13.89% of transduced T cells, mainly CD8+ T cells (85.7 ± 0.75%, P<0.0001) with the effector memory phenotype (59.22 ± 16.31%, P < 0.01). αM.CAR/SR T cells produced high levels of intracellular tumor necrosis factor-α and interferon-γ in response to the activation by CCA cells expressing MUC1, including KKU-055 (27.18 ± 4.38% and 27.33 ± 5.55%, respectively, P < 0.05) and KKU-213A (47.37 ± 12.67% and 54.55 ± 8.66%, respectively, P < 0.01). Remarkably, the cytotoxic function of αM.CAR/SR T cells against KKU-213A cells expressing PD-L1 was significantly enhanced compared with the αM.CAR T cells (70.69 ± 14.38% versus 47.15 ± 8.413%, respectively; P = 0.0301), correlated with increased granzyme B production (60.6 ± 9.89% versus 43.2 ± 8.95%, respectively; P = 0.0402). Moreover, the significantly enhanced disruption of KKU-213A spheroids by αM.CAR/SR T cells (P = 0.0027), compared with αM.CAR T cells, was also observed. CONCLUSION Taken together, the cytotoxic function of αM.CAR/SR T cells was enhanced over the αM.CAR T cells, which are potential to be further tested for CCA treatment.
Collapse
Affiliation(s)
- Kamonlapat Supimon
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Thanich Sangsuwannukul
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jatuporn Sujjitjoon
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Thaweesak Chieochansin
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Mutita Junking
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Pa-Thai Yenchitsomanus
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
27
|
Däullary T, Imdahl F, Dietrich O, Hepp L, Krammer T, Fey C, Neuhaus W, Metzger M, Vogel J, Westermann AJ, Saliba AE, Zdzieblo D. A primary cell-based in vitro model of the human small intestine reveals host olfactomedin 4 induction in response to Salmonella Typhimurium infection. Gut Microbes 2023; 15:2186109. [PMID: 36939013 PMCID: PMC10038062 DOI: 10.1080/19490976.2023.2186109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
Infection research largely relies on classical cell culture or mouse models. Despite having delivered invaluable insights into host-pathogen interactions, both have limitations in translating mechanistic principles to human pathologies. Alternatives can be derived from modern Tissue Engineering approaches, allowing the reconstruction of functional tissue models in vitro. Here, we combined a biological extracellular matrix with primary tissue-derived enteroids to establish an in vitro model of the human small intestinal epithelium exhibiting in vivo-like characteristics. Using the foodborne pathogen Salmonella enterica serovar Typhimurium, we demonstrated the applicability of our model to enteric infection research in the human context. Infection assays coupled to spatio-temporal readouts recapitulated the established key steps of epithelial infection by this pathogen in our model. Besides, we detected the upregulation of olfactomedin 4 in infected cells, a hitherto unrecognized aspect of the host response to Salmonella infection. Together, this primary human small intestinal tissue model fills the gap between simplistic cell culture and animal models of infection, and shall prove valuable in uncovering human-specific features of host-pathogen interplay.
Collapse
Affiliation(s)
- Thomas Däullary
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg (UKW), Würzburg, Germany
- Faculty of Biology, Biocenter, Chair of Microbiology, Julius-Maximilians-Universität Würzburg (JMU), Würzburg, Germany
| | - Fabian Imdahl
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Oliver Dietrich
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Laura Hepp
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg (UKW), Würzburg, Germany
| | - Tobias Krammer
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Christina Fey
- Fraunhofer Institute for Silicate Research (ISC),Translational Center Regenerative Therapies (TLC-RT), Würzburg, Germany
| | - Winfried Neuhaus
- Austrian Institute of Technology (AIT), Vienna, Austria
- Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University (DPU), Krems, Austria
| | - Marco Metzger
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg (UKW), Würzburg, Germany
- Fraunhofer Institute for Silicate Research (ISC),Translational Center Regenerative Therapies (TLC-RT), Würzburg, Germany
- Fraunhofer Institute for Silicate Research, Project Center for Stem Cell Process Engineering, Würzburg, Germany
| | - Jörg Vogel
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
- Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Alexander J Westermann
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
- Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Daniela Zdzieblo
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg (UKW), Würzburg, Germany
- Fraunhofer Institute for Silicate Research (ISC),Translational Center Regenerative Therapies (TLC-RT), Würzburg, Germany
- Fraunhofer Institute for Silicate Research, Project Center for Stem Cell Process Engineering, Würzburg, Germany
| |
Collapse
|
28
|
Barmania F, Mellet J, Holborn MA, Pepper MS. Genetic Associations with Coronavirus Susceptibility and Disease Severity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1412:119-140. [PMID: 37378764 DOI: 10.1007/978-3-031-28012-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is responsible for the coronavirus disease 2019 (COVID-19) global public health emergency, and the disease it causes is highly variable in its clinical presentation. Host genetic factors are increasingly recognised as a determinant of infection susceptibility and disease severity. Several initiatives and groups have been established to analyse and review host genetic epidemiology associated with COVID-19 outcomes. Here, we review the genetic loci associated with COVID-19 susceptibility and severity focusing on the common variants identified in genome-wide association studies.
Collapse
Affiliation(s)
- Fatima Barmania
- Institute for Cellular and Molecular Medicine, Department of Immunology, SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Juanita Mellet
- Institute for Cellular and Molecular Medicine, Department of Immunology, SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Megan A Holborn
- Institute for Cellular and Molecular Medicine, Department of Immunology, SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Michael S Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology, SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
29
|
Production of Intestinal Mucins, sIgA, and Metallothionein after Administration of Zinc and Infection of Ascaridia galli in Chickens: Preliminary Data. Life (Basel) 2022; 13:life13010067. [PMID: 36676016 PMCID: PMC9862470 DOI: 10.3390/life13010067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/30/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
The effect of inorganic zinc and Ascaridia galli infection was studied on MUC1, MUC2 (mucin), sIgA (secretory immunoglobulin A), and metallothionein in the intestines of broilers. Thirty-five-day-old chickens (n = 24), COBB 500 breed, were included in a 14-day experiment. Chickens were divided into 4 groups of 6 chickens each: control ©, Ascaridia galli (AG), Zinc group (Zn), and combined group (AG + Zn). Samples from the intestine for determination of MUC1, MUC2, sIgA, and metallothionein were taken at 7 and 14 days during necropsy. Samples from the jejunum for determination of MUC1, MUC2, sIgA, and metallothionein were taken at 7 and 14 days during necropsy. The results demonstrated that 12 days’ administration of inorganic zinc increased production of MUC1 (p < 0.0001) and MUC2 (p < 0.001) in the Ascaridia galli-infected group (Ag + Zn) in comparison to control (C). The beneficial effect of zinc was also revealed in the production of sIgA (p < 0.0001) in the combined group (AG + Zn) at 7 days. The concentration of metallothionein increased mainly in the zinc group (p < 0.01) of first sampling and was upregulated in Zn and AG + Zn groups. The obtained data indicate the use of inorganic zinc as a suitable immunomodulator of intestinal immunity in Ascaridia galli-infected chickens.
Collapse
|
30
|
Kohout VR, Wardzala CL, Kramer JR. Synthesis and biomedical applications of mucin mimic materials. Adv Drug Deliv Rev 2022; 191:114540. [PMID: 36228896 PMCID: PMC10066857 DOI: 10.1016/j.addr.2022.114540] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 08/17/2022] [Accepted: 09/13/2022] [Indexed: 02/09/2023]
Abstract
Mucin glycoproteins are the major component of mucus and coat epithelial cell surfaces forming the glycocalyx. The glycocalyx and mucus are involved in the transport of nutrients, drugs, gases, and pathogens toward the cell surface. Mucins are also involved in diverse diseases such as cystic fibrosis and cancer. Due to inherent heterogeneity in native mucin structure, many synthetic materials have been designed to probe mucin chemistry, biology, and physics. Such materials include various glycopolymers, low molecular weight glycopeptides, glycopolypeptides, polysaccharides, and polysaccharide-protein conjugates. This review highlights advances in the area of design and synthesis of mucin mimic materials, and their biomedical applications in glycan binding, epithelial models of infection, therapeutic delivery, vaccine formulation, and beyond.
Collapse
Affiliation(s)
- Victoria R Kohout
- Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT 84112, USA
| | - Casia L Wardzala
- Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT 84112, USA
| | - Jessica R Kramer
- Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT 84112, USA.
| |
Collapse
|
31
|
An In Vitro Comparison of Costimulatory Domains in Chimeric Antigen Receptor T Cell for Breast Cancer Treatment. J Immunol Res 2022; 2022:2449373. [DOI: 10.1155/2022/2449373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/27/2022] [Accepted: 11/04/2022] [Indexed: 11/23/2022] Open
Abstract
Adoptive cellular therapy with chimeric antigen receptor (CAR) T cells has emerged as a potential novel treatment for various cancers. In this study, we have generated CAR T cells targeting mucin-1 (MUC1), which is an aberrantly glycosylated antigen overexpressed on breast cancer cells. Two different signaling domains, including CD28 and 41BB, were incorporated and directly compared the superiority of different costimulatory signals. Two different CAR MUC1 constructs were transduced into primary T cells and evaluated their characteristics and antitumor activities against MUC1+ cancer cells. CAR MUC1 T cells showed high transduction efficiency and antigen specificity toward MUC1+ cancer cell lines and primary breast cancer cells. When coculturing with target cells, the transduced cells exhibited potent antitumor activity in vitro and secrete proinflammatory cytokines. Upon antigen stimulation, incorporation of the 41BB signaling domain was able to improve T cell proliferation and reduce surface PD1 expression and the upregulation of suppressive cytokines, when compared with CAR MUC1 containing the CD28 domain. Our findings show that CAR T cell targeting MUC1 can be effective against MUC1+ breast cancer cell and support the further development of CAR MUC1 T cells containing 41BB signaling in preclinical and clinical studies of breast cancer treatment.
Collapse
|
32
|
Taibe NS, Kord MA, Badawy MA, Shytaj IL, Elhefnawi MM. Progress, pitfalls, and path forward of drug repurposing for COVID-19 treatment. Ther Adv Respir Dis 2022; 16:17534666221132736. [PMID: 36282077 PMCID: PMC9597285 DOI: 10.1177/17534666221132736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
On 30 January 2020, the World Health Organization (WHO) declared the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) epidemic a public health emergency of international concern. The viral outbreak led in turn to an exponential growth of coronavirus disease 2019 (COVID-19) cases, that is, a multiorgan disease that has led to more than 6.3 million deaths worldwide, as of June 2022. There are currently few effective drugs approved for treatment of SARS-CoV-2/COVID-19 patients. Many of the compounds tested so far have been selected through a drug repurposing approach, that is, by identifying novel indications for drugs already approved for other conditions. We here present an up-to-date review of the main Food and Drug Administration (FDA)-approved drugs repurposed against SARS-CoV-2 infection, discussing their mechanism of action and their most important preclinical and clinical results. Reviewed compounds were chosen to privilege those that have been approved for use in SARS-CoV-2 patients or that have completed phase III clinical trials. Moreover, we also summarize the evidence on some novel and promising repurposed drugs in the pipeline. Finally, we discuss the current stage and possible steps toward the development of broadly effective drug combinations to suppress the onset or progression of COVID-19.
Collapse
Affiliation(s)
- Noha Samir Taibe
- Biotechnology-Biomolecular Chemistry Program, Chemistry Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Maimona A. Kord
- Department of Botany, Faculty of Science, Cairo University, Giza, Egypt
| | | | | | | |
Collapse
|
33
|
The ulcerative colitis-associated gene FUT8 regulates the quantity and quality of secreted mucins. Proc Natl Acad Sci U S A 2022; 119:e2205277119. [PMID: 36252012 PMCID: PMC9618082 DOI: 10.1073/pnas.2205277119] [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: 11/18/2022] Open
Abstract
Mucins are the main macrocomponents of the mucus layer that protects the digestive tract from pathogens. Fucosylation of mucins increases mucus viscoelasticity and its resistance to shear stress. These properties are altered in patients with ulcerative colitis (UC), which is marked by a chronic inflammation of the distal part of the colon. Here, we show that levels of Fucosyltransferase 8 (FUT8) and specific mucins are increased in the distal inflamed colon of UC patients. Recapitulating this FUT8 overexpression in mucin-producing HT29-18N2 colonic cell line increases delivery of MUC1 to the plasma membrane and extracellular release of MUC2 and MUC5AC. Mucins secreted by FUT8 overexpressing cells are more resistant to removal from the cell surface than mucins secreted by FUT8-depleted cells (FUT8 KD). FUT8 KD causes intracellular accumulation of MUC1 and alters the ratio of secreted MUC2 to MUC5AC. These data fit well with the Fut8-/- mice phenotype, which are protected from UC. Fut8-/- mice exhibit a thinner proximal colon mucus layer with an altered ratio of neutral to acidic mucins. Together, our data reveal that FUT8 modifies the biophysical properties of mucus by controlling levels of cell surface MUC1 and quantity and quality of secreted MUC2 and MUC5AC. We suggest that these changes in mucus viscoelasticity likely facilitate bacterial-epithelial interactions leading to inflammation and UC progression.
Collapse
|
34
|
Qing L, Li Q, Dong Z. MUC1: An emerging target in cancer treatment and diagnosis. Bull Cancer 2022; 109:1202-1216. [DOI: 10.1016/j.bulcan.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/26/2022] [Accepted: 08/01/2022] [Indexed: 10/14/2022]
|
35
|
Rebendenne A, Roy P, Bonaventure B, Chaves Valadão AL, Desmarets L, Arnaud-Arnould M, Rouillé Y, Tauziet M, Giovannini D, Touhami J, Lee Y, DeWeirdt P, Hegde M, Urbach S, Koulali KE, de Gracia FG, McKellar J, Dubuisson J, Wencker M, Belouzard S, Moncorgé O, Doench JG, Goujon C. Bidirectional genome-wide CRISPR screens reveal host factors regulating SARS-CoV-2, MERS-CoV and seasonal HCoVs. Nat Genet 2022; 54:1090-1102. [PMID: 35879413 PMCID: PMC11627114 DOI: 10.1038/s41588-022-01110-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 05/26/2022] [Indexed: 12/23/2022]
Abstract
CRISPR knockout (KO) screens have identified host factors regulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication. Here, we conducted a meta-analysis of these screens, which showed a high level of cell-type specificity of the identified hits, highlighting the necessity of additional models to uncover the full landscape of host factors. Thus, we performed genome-wide KO and activation screens in Calu-3 lung cells and KO screens in Caco-2 colorectal cells, followed by secondary screens in four human cell lines. This revealed host-dependency factors, including AP1G1 adaptin and ATP8B1 flippase, as well as inhibitors, including mucins. Interestingly, some of the identified genes also modulate Middle East respiratory syndrome coronavirus (MERS-CoV) and seasonal human coronavirus (HCoV) (HCoV-NL63 and HCoV-229E) replication. Moreover, most genes had an impact on viral entry, with AP1G1 likely regulating TMPRSS2 activity at the plasma membrane. These results demonstrate the value of multiple cell models and perturbational modalities for understanding SARS-CoV-2 replication and provide a list of potential targets for therapeutic interventions.
Collapse
Affiliation(s)
| | - Priyanka Roy
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Lowiese Desmarets
- Lille University, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, Lille, France
| | | | - Yves Rouillé
- Lille University, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, Lille, France
| | | | - Donatella Giovannini
- IGMM, CNRS, Montpellier University, Montpellier, France
- Metafora Biosystems, Paris, France
| | - Jawida Touhami
- IGMM, CNRS, Montpellier University, Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Yenarae Lee
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Peter DeWeirdt
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mudra Hegde
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Serge Urbach
- IGF, Montpellier University, CNRS, INSERM, Montpellier, France
| | | | | | - Joe McKellar
- IRIM, CNRS, Montpellier University, Montpellier, France
| | - Jean Dubuisson
- Lille University, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, Lille, France
| | | | - Sandrine Belouzard
- Lille University, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, Lille, France
| | | | - John G Doench
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | | |
Collapse
|
36
|
Human Milk Extracellular Vesicles: A Biological System with Clinical Implications. Cells 2022; 11:cells11152345. [PMID: 35954189 PMCID: PMC9367292 DOI: 10.3390/cells11152345] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 12/10/2022] Open
Abstract
The consumption of human milk by a breastfeeding infant is associated with positive health outcomes, including lower risk of diarrheal disease, respiratory disease, otitis media, and in later life, less risk of chronic disease. These benefits may be mediated by antibodies, glycoproteins, glycolipids, oligosaccharides, and leukocytes. More recently, human milk extracellular vesicles (hMEVs) have been identified. HMEVs contain functional cargos, i.e., miRNAs and proteins, that may transmit information from the mother to promote infant growth and development. Maternal health conditions can influence hMEV composition. This review summarizes hMEV biogenesis and functional contents, reviews the functional evidence of hMEVs in the maternal–infant health relationship, and discusses challenges and opportunities in hMEV research.
Collapse
|
37
|
Membrane-Tethered Mucin 1 Is Stimulated by Interferon and Virus Infection in Multiple Cell Types and Inhibits Influenza A Virus Infection in Human Airway Epithelium. mBio 2022; 13:e0105522. [PMID: 35699372 PMCID: PMC9426523 DOI: 10.1128/mbio.01055-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Influenza A virus (IAV) causes significant morbidity and mortality in the human population. Tethered mucin 1 (MUC1) is highly expressed in airway epithelium, the primary site of IAV replication, and also by other cell types that influence IAV infection, including macrophages. MUC1 has the potential to influence infection dynamics through physical interactions and/or signaling activity, yet MUC1 modulation and its impact during viral pathogenesis remain unclear. Thus, we investigated MUC1-IAV interactions in an in vitro model of human airway epithelium (HAE). Our data indicate that a recombinant IAV hemagglutinin (H3) and H3N2 virus can bind endogenous HAE MUC1. Notably, infection of HAE with H1N1 or H3N2 IAV strains does not trigger MUC1 shedding but instead stimulates an increase in cell-associated MUC1 protein. We observed a similar increase after type I or III interferon (IFN) stimulation; however, inhibition of IFN signaling during H1N1 infection only partially abrogated this increase, indicating that multiple soluble factors contribute to MUC1 upregulation during the antiviral response. In addition to HAE, primary human monocyte-derived macrophages also upregulated MUC1 protein in response to IFN treatment and conditioned media from IAV-infected HAE. Then, to determine the impact of MUC1 on IAV pathogenesis, we developed HAE genetically depleted of MUC1 and found that MUC1 knockout cultures exhibited enhanced viral growth compared to control cultures for several IAV strains. Together, our data support a model whereby MUC1 inhibits productive uptake of IAV in HAE. Infection then stimulates MUC1 expression on multiple cell types through IFN-dependent and -independent mechanisms that further impact infection dynamics.
Collapse
|
38
|
Lu CH, Pedram K, Tsai CT, Jones T, Li X, Nakamoto ML, Bertozzi CR, Cui B. Membrane curvature regulates the spatial distribution of bulky glycoproteins. Nat Commun 2022; 13:3093. [PMID: 35654773 PMCID: PMC9163104 DOI: 10.1038/s41467-022-30610-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 05/10/2022] [Indexed: 01/11/2023] Open
Abstract
The glycocalyx is a shell of heavily glycosylated proteins and lipids distributed on the cell surface of nearly all cell types. Recently, it has been found that bulky transmembrane glycoproteins such as MUC1 can modulate membrane shape by inducing membrane protrusions. In this work, we examine the reciprocal relationship of how membrane shape affects MUC1's spatial distribution on the cell membrane and its biological significance. By employing nanopatterned surfaces and membrane-sculpting proteins to manipulate membrane curvature, we show that MUC1 avoids positively-curved membranes (membrane invaginations) and accumulates on negatively-curved membranes (membrane protrusions). MUC1's curvature sensitivity is dependent on the length and the extent of glycosylation of its ectodomain, with large and highly glycosylated forms preferentially staying out of positive curvature. Interestingly, MUC1's avoidance of positive membrane curvature enables it to escape from endocytosis and being removed from the cell membrane. These findings also suggest that the truncation of MUC1's ectodomain, often observed in breast and ovarian cancers, may enhance its endocytosis and potentiate its intracellular accumulation and signaling.
Collapse
Affiliation(s)
- Chih-Hao Lu
- grid.168010.e0000000419368956Department of Chemistry, Stanford University, Stanford, CA 94305 USA
| | - Kayvon Pedram
- grid.168010.e0000000419368956Department of Chemistry, Stanford University, Stanford, CA 94305 USA ,grid.443970.dPresent Address: Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147 USA
| | - Ching-Ting Tsai
- grid.168010.e0000000419368956Department of Chemistry, Stanford University, Stanford, CA 94305 USA
| | - Taylor Jones
- grid.168010.e0000000419368956Department of Chemistry, Stanford University, Stanford, CA 94305 USA
| | - Xiao Li
- grid.168010.e0000000419368956Department of Chemistry, Stanford University, Stanford, CA 94305 USA ,grid.43169.390000 0001 0599 1243Present Address: School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
| | - Melissa L. Nakamoto
- grid.168010.e0000000419368956Department of Chemistry, Stanford University, Stanford, CA 94305 USA
| | - Carolyn R. Bertozzi
- grid.168010.e0000000419368956Department of Chemistry, Stanford University, Stanford, CA 94305 USA ,grid.168010.e0000000419368956Stanford ChEM-H, Stanford University, Stanford, CA 94305 USA ,grid.168010.e0000000419368956Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305 USA
| | - Bianxiao Cui
- grid.168010.e0000000419368956Department of Chemistry, Stanford University, Stanford, CA 94305 USA
| |
Collapse
|
39
|
Singh N, Diebold Y, Sahu SK, Leonardi A. Epithelial barrier dysfunction in ocular allergy. Allergy 2022; 77:1360-1372. [PMID: 34757631 PMCID: PMC9300009 DOI: 10.1111/all.15174] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 12/14/2022]
Abstract
The epithelial barrier is the first line of defense that forms a protective barrier against pathogens, pollutants, and allergens. Epithelial barrier dysfunction has been recently implicated in the development of allergic diseases such as asthma, atopic dermatitis, food allergy, and rhinitis. However, there is limited knowledge on epithelial barrier dysfunction in ocular allergy (OA). Since the ocular surface is directly exposed to the environment, it is important to understand the role of ocular epithelia and their dysfunction in OA. Impaired epithelial barrier enhances allergen uptake, which lead to activation of immune responses and development of chronic inflammation as seen in allergies. Abnormal expression of tight junction proteins that helps to maintain epithelial integrity has been reported in OA but sufficient data not available in chronic atopic (AKC) and vernal keratoconjunctivitis (VKC), the pathophysiology of which is not just complex, but also the current treatments are not completely effective. This review provides an overview of studies, which indicates the role of barrier dysfunction in OA, and highlights how ocular barrier dysfunction possibly contributes to the disease pathogenesis. The review also explores the potential of ocular epithelial barrier repair strategies as preventive and therapeutic approach.
Collapse
Affiliation(s)
- Neera Singh
- ProCyto Labs Pvt. Ltd. KIIT‐TBI KIIT University Patia, Bhubaneswar India
| | - Yolanda Diebold
- Ocular Surface Group Instituto Universitario de Oftalmobiología Aplicada (IOBA) Universidad de Valladolid Valladolid Spain
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER‐BBN) Valladolid Spain
| | - Srikant K. Sahu
- LV Prasad Eye Institute, Cornea and Anterior Segment, MTC Campus Patia, Bhubaneswar India
| | - Andrea Leonardi
- Ophthalmology Unit Department of Neuroscience University of Padova Padova Italy
| |
Collapse
|
40
|
MUC1 (CA27.29) before and after Chemotherapy and Prognosis in High-Risk Early Breast Cancer Patients. Cancers (Basel) 2022; 14:cancers14071721. [PMID: 35406491 PMCID: PMC8997086 DOI: 10.3390/cancers14071721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 03/05/2022] [Accepted: 03/09/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary CA27.29 (MUC1) is a well described biomarker for prediction of prognosis and treatment efficacy. CA27.29 is mainly evaluated in the preoperative setting. However, testing of postoperative levels and additional assessment after chemotherapy might be more informative for analyzing the usefulness of CA27.29 in relation to the efficacy of chemotherapy. Thus, both pre- and post-chemotherapy values were assessed from patients enrolled in the breast cancer SUCCESS-A trial. Pre-chemotherapy assessment was associated with disease-free survival. It had no prognostic value in node-negative patients, but there was a clear association in node-positive patients. Furthermore, it was shown that post-chemotherapy CA27.29 assessment did not add any prognostic value, either on its own or in addition to pre-chemotherapy assessment. In conclusion, this indicates that pre- and post-chemotherapy values do not provide additional information. However, pre-chemotherapy CA27.29 could be a suitable tool to identify a group with unfavorable prognosis among node-positive patients. Abstract Soluble MUC1 has been discussed as a biomarker for predicting prognosis, treatment efficacy, and monitoring disease activity in breast cancer (BC) patients. Most studies in adjuvant settings have used preoperative assessment. This study, part of the SUCCESS-A trial (NCT02181101), assessed the prognostic value of soluble MUC1 before and after standard adjuvant chemotherapy. Patients with high-risk BC were treated within the SUCCESS-A trial with either three cycles of 5-fluorouracil, epirubicin, and cyclophosphamide followed by three cycles of docetaxel or three cycles of FEC followed by three cycles of docetaxel and gemcitabine. Cox regression analyses were performed to investigate the prognostic value of CA27.29 before and after chemotherapy relative to disease-free survival (DFS), along with established BC prognostic factors such as age, body mass index, tumor size, nodal status, estrogen receptor, progesterone receptor, HER2 status, and grading. Pre-chemotherapy and post-chemotherapy CA27.29 assessments were available for 2687 patients of 3754 randomized patients. Pre-chemotherapy CA27.29 assessment was associated with DFS in addition to established prognostic factors. It had no prognostic value in node-negative patients, but there was a clear association in node-positive patients. Post-chemotherapy CA27.29 assessment did not add any prognostic value, either on its own or in addition to pre-chemotherapy CA27.29 assessment.
Collapse
|
41
|
Identified human breastmilk compositions effectively inhibit SARS-CoV-2 and variants infection and replication. iScience 2022; 25:104136. [PMID: 35342878 PMCID: PMC8937612 DOI: 10.1016/j.isci.2022.104136] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/09/2022] [Accepted: 03/17/2022] [Indexed: 01/08/2023] Open
Abstract
The global pandemic of COVID-19 caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection confers great threat to the public health. Human breastmilk is a complex with nutritional composition to nourish infants and protect them from different kinds of infectious diseases including COVID-19. Here, we identified lactoferrin (LF), mucin1 (MUC1) and α-lactalbumin (α-LA) from human breastmilk inhibit SARS-CoV-2 infection using a SARS-CoV-2 pseudovirus system and transcription and replication-competent SARS-CoV-2 virus-like-particles (trVLP). Additionally, LF and MUC1 inhibited multiple steps including viral attachment, entry and post-entry replication, while α-LA inhibited viral attachment and entry. Importantly, LF, MUC1 and α-LA possessed potent antiviral activities towards variants such as B.1.1.7 (alpha), B.1.351 (beta), P.1 (gamma) and B.1.617.1 (kappa). Taken together, our study provides evidence that human breastmilk components (LF, MUC1 and α-LA) are promising antiviral and potential therapeutic candidates warranting further development or treating COVID-19.
Collapse
|
42
|
Lithopoulos MA, Strueby L, O'Reilly M, Zhong S, Möbius MA, Eaton F, Fung M, Hurskainen M, Cyr-Depauw C, Suen C, Xu L, Collins JJP, Vadivel A, Stewart DJ, Burger D, Thébaud B. Pulmonary and Neurologic Effects of Mesenchymal Stromal Cell Extracellular Vesicles in a Multifactorial Lung Injury Model. Am J Respir Crit Care Med 2022; 205:1186-1201. [PMID: 35286238 DOI: 10.1164/rccm.202012-4520oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Bronchopulmonary dysplasia, a chronic respiratory condition originating from preterm birth, is associated with abnormal neurodevelopment. Currently, there is an absence of effective therapies for bronchopulmonary dysplasia and its associated brain injury. In preclinical trials mesenchymal stromal cell therapies demonstrate promise as a therapeutic for bronchopulmonary dysplasia. OBJECTIVES To investigate whether a multifactorial neonatal mouse model of lung injury perturbs neural progenitor cell function and to assess the ability of human umbilical cord-derived mesenchymal stromal cell extracellular vesicles to mitigate pulmonary and neurologic injury. METHODS Mice at postnatal day 7/8 were injected intraperitoneally with lipopolysaccharide and ventilated with 40% oxygen at postnatal day 9/10 for 8 hours. Treated animals received umbilical cord-mesenchymal stromal cell-derived extracellular vesicles intratracheally preceding ventilation. Lung morphology, vascularity, and inflammation were quantified. Neural progenitor cells were isolated from the subventricular zone/hippocampus and assessed for self-renewal, in vitro differentiation ability, and transcriptional profiles. MEASUREMENTS AND MAIN RESULTS The multifactorial lung injury model produced alveolar and vascular rarefaction mimicking bronchopulmonary dysplasia. Neural progenitor cells from lung injury mice showed reduced neurosphere and oligodendrocyte formation, as well as inflammatory transcriptional signatures. Mice treated with mesenchymal stromal cell extracellular vesicles showed significant improvement in lung architecture, vessel formation, and inflammatory modulation. Additionally, we observed significantly increased in vitro neurosphere formation and altered neural progenitor cell transcriptional signatures. CONCLUSIONS Our multifactorial lung injury model impairs neural progenitor cell function. Observed pulmonary and neurologic alterations are mitigated by intratracheal treatment with mesenchymal stromal cell-derived extracellular vesicles.
Collapse
Affiliation(s)
- Marissa A Lithopoulos
- Ottawa Hospital Research Institute, 10055, Regenerative Medicine Program, Ottawa, Ontario, Canada.,University of Ottawa, 6363, Department of Cellular and Molecular Medicine, Ottawa, Ontario, Canada
| | - Lannae Strueby
- University of Saskatchewan, 7235, Department of Pediatrics, Saskatoon, Saskatchewan, Canada
| | - Megan O'Reilly
- University of Alberta, 3158, Department of Pediatrics, Edmonton, Alberta, Canada
| | - Shumei Zhong
- Ottawa Hospital Research Institute, 10055, Regenerative Medicine Program, Ottawa, Ontario, Canada
| | - Marius A Möbius
- Universitätsklinikum Carl Gustav Carus, 39063, Department of Neonatalogy and Pediatric Critical Care Medicine, Dresden, Germany
| | - Farah Eaton
- University of Alberta, 3158, Faculty of Pharmacy and Pharmaceutical Sciences, Edmonton, Alberta, Canada
| | - Moses Fung
- University of Alberta, 3158, Department of Pediatrics, Edmonton, Alberta, Canada
| | - Maria Hurskainen
- Helsinki University Central Hospital, 159841, Department of Pediatric Cardiology, Helsinki, Finland.,University of Helsinki, 3835, Pediatric Research Center, Helsinki, Finland
| | - Chanèle Cyr-Depauw
- Ottawa Hospital Research Institute, 10055, Regenerative Medicine Program, Ottawa, Ontario, Canada.,University of Ottawa, 6363, Department of Cellular and Molecular Medicine, Ottawa, Ontario, Canada
| | - Colin Suen
- Ottawa Hospital Research Institute, 10055, Regenerative Medicine Program, Ottawa, Canada.,University of Ottawa, 6363, Department of Cellular and Molecular Medicine, Ottawa, Ontario, Canada
| | - Liqun Xu
- Ottawa Hospital Research Institute, 10055, Regenerative Medicine Program, Ottawa, Ontario, Canada
| | - Jennifer J P Collins
- Ottawa Hospital Research Institute, 10055, Regenerative Medicine Program, Ottawa, Ontario, Canada.,University of Ottawa, 6363, Department of Cellular and Molecular Medicine, Ottawa, Ontario, Canada
| | - Arul Vadivel
- Ottawa Hospital Research Institute, 10055, Regenerative Medicine Program, Ottawa, Ontario, Canada
| | - Duncan J Stewart
- Ottawa Hospital Research Institute, 10055, Regenerative Medicine Program, Ottawa, Ontario, Canada.,University of Ottawa, 6363, Department of Cellular and Molecular Medicine, Ottawa, Ontario, Canada
| | - Dylan Burger
- University of Ottawa, 6363, Department of Cellular and Molecular Medicine, Ottawa, Ontario, Canada.,Ottawa Hospital Research Institute, 10055, Kidney Research Centre, Chronic Disease Program, Ottawa, Ontario, Canada
| | - Bernard Thébaud
- Ottawa Hospital Research Institute, 10055, Regenerative Medicine Program, Ottawa, Ontario, Canada.,University of Ottawa, 6363, Department of Cellular and Molecular Medicine, Ottawa, Ontario, Canada.,Children's Hospital of Eastern Ontario Research Institute, 274065, Ottawa, Ontario, Canada;
| |
Collapse
|
43
|
Renga G, Nunzi E, Pariano M, Puccetti M, Bellet MM, Pieraccini G, D'Onofrio F, Santarelli I, Stincardini C, Aversa F, Riuzzi F, Antognelli C, Gargaro M, Bereshchenko O, Ricci M, Giovagnoli S, Romani L, Costantini C. Optimizing therapeutic outcomes of immune checkpoint blockade by a microbial tryptophan metabolite. J Immunother Cancer 2022; 10:jitc-2021-003725. [PMID: 35236743 PMCID: PMC8896050 DOI: 10.1136/jitc-2021-003725] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/01/2022] [Indexed: 12/11/2022] Open
Abstract
Background Despite the great success, the therapeutic benefits of immune checkpoint inhibitors (ICIs) in cancer immunotherapy are limited by either various resistance mechanisms or ICI-associated toxic effects including gastrointestinal toxicity. Thus, novel therapeutic strategies that provide manageable side effects to existing ICIs would enhance and expand their therapeutic efficacy and application. Due to its proven role in cancer development and immune regulation, gut microbiome has gained increasing expectation as a potential armamentarium to optimize immunotherapy with ICI. However, much has to be learned to fully harness gut microbiome for clinical applicability. Here we have assessed whether microbial metabolites working at the interface between microbes and the host immune system may optimize ICI therapy. Methods To this purpose, we have tested indole-3-carboxaldehyde (3-IAld), a microbial tryptophan catabolite known to contribute to epithelial barrier function and immune homeostasis in the gut via the aryl hydrocarbon receptor (AhR), in different murine models of ICI-induced colitis. Epithelial barrier integrity, inflammation and changes in gut microbiome composition and function were analyzed. AhR, indoleamine 2,3-dioxygenase 1, interleukin (IL)-10 and IL-22 knockout mice were used to investigate the mechanism of 3-IAld activity. The function of the microbiome changes induced by 3-IAld was evaluated on fecal microbiome transplantation (FMT). Finally, murine tumor models were used to assess the effect of 3-IAld treatment on the antitumor activity of ICI. Results On administration to mice with ICI-induced colitis, 3-IAld protected mice from intestinal damage via a dual action on both the host and the microbes. Indeed, paralleling the activation of the host AhR/IL-22-dependent pathway, 3-IAld also affected the composition and function of the microbiota such that FMT from 3-IAld-treated mice protected against ICI-induced colitis with the contribution of butyrate-producing bacteria. Importantly, while preventing intestinal damage, 3-IAld did not impair the antitumor activity of ICI. Conclusions This study provides a proof-of-concept demonstration that moving past bacterial phylogeny and focusing on bacterial metabolome may lead to a new class of discrete molecules, and that working at the interface between microbes and the host immune system may optimize ICI therapy.
Collapse
Affiliation(s)
- Giorgia Renga
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Emilia Nunzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Matteo Puccetti
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | | | | | - Fiorella D'Onofrio
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Ilaria Santarelli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Franco Aversa
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Francesca Riuzzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cinzia Antognelli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marco Gargaro
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Oxana Bereshchenko
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Maurizio Ricci
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
44
|
Labetoulle M, Benitez-del-Castillo JM, Barabino S, Herrero Vanrell R, Daull P, Garrigue JS, Rolando M. Artificial Tears: Biological Role of Their Ingredients in the Management of Dry Eye Disease. Int J Mol Sci 2022; 23:ijms23052434. [PMID: 35269576 PMCID: PMC8910031 DOI: 10.3390/ijms23052434] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/17/2021] [Accepted: 12/18/2021] [Indexed: 12/21/2022] Open
Abstract
Dry eye disease (DED) is the most common ocular surface disease, characterized by insufficient production and/or instability of the tear film. Tear substitutes are usually the first line of treatment for patients with DED. Despite the large variety of tear substitutes available on the market, few studies have been performed to compare their performance. There is a need to better understand the specific mechanical and pharmacological roles of each ingredient composing the different formulations. In this review, we describe the main categories of ingredients composing tear substitutes (e.g., viscosity-enhancing agents, electrolytes, osmo-protectants, antioxidants, lipids, surfactants and preservatives) as well as their effects on the ocular surface, and we provide insight into how certain components of tear substitutes may promote corneal wound healing, and/or counteract inflammation. Based on these considerations, we propose an approach to select the most appropriate tear substitute formulations according to the predominant etiological causes of DED.
Collapse
Affiliation(s)
- Marc Labetoulle
- Service d’Ophtalmologie, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, 94270 Le Kremlin Bicêtre, France;
| | | | - Stefano Barabino
- Centro Superficie Oculare e Occhio Secco, ASST Fatebenefratelli-Sacco, Ospedale L. Sacco, Università di Milano, 20157 Milan, Italy;
| | - Rocio Herrero Vanrell
- Research Group (UCM 920415), Innovation, Therapy and Pharmaceutical Development in Ophthalmology (InnOftal), Faculty of Pharmacy, Complutense University, 28040 Madrid, Spain;
| | - Philippe Daull
- Ophthalmic Innovation Center, Santen SAS, 91058 Evry, France;
| | | | - Maurizio Rolando
- Ocular Surface Centre, ISPRE (Instituto di Medicina Oftalmica) Ophthalmic, 16129 Genoa, Italy;
| |
Collapse
|
45
|
Filatova L, Emelianov G, Balabushevich N, Klyachko N. Supramolecular assemblies of mucin and lysozyme: Formation and physicochemical characterization. Process Biochem 2022. [DOI: 10.1016/j.procbio.2021.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
46
|
Ropartz D, Fanuel M, Ollivier S, Lissarrague A, Benkoulouche M, Mulard LA, André I, Guieysse D, Rogniaux H. Combination of High-Resolution Multistage Ion Mobility and Tandem MS with High Energy of Activation to Resolve the Structure of Complex Chemoenzymatically Synthesized Glycans. Anal Chem 2022; 94:2279-2287. [PMID: 35049286 DOI: 10.1021/acs.analchem.1c04982] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Carbohydrates, in particular microbial glycans, are highly structurally diverse biomolecules, the recognition of which governs numerous biological processes. Of special interest, glycans of known monosaccharide composition feature multiple possible isomers, differentiated by the anomerism and position of their glycosidic linkages. Robust analytical tools able to circumvent this extreme structural complexity are increasing in demand to ensure not only the correct determination of naturally occurring glycans but also to support the rapid development of enzymatic and chemoenzymatic glycan synthesis. In support to the later, we report the use of complementary strategies based on mass spectrometry (MS) to evaluate the ability of 14 engineered mutants of sucrose-utilizing α-transglucosylases to produce type/group-specific Shigella flexneri pentasaccharide bricks from a single lightly protected non-natural tetrasaccharide acceptor substrate. A first analysis of the reaction media by UHPLC coupled to high-accuracy MS led to detect six reaction products of enzymatic glucosylation out of the eight possible ones. A seventh structure was evidenced by an additional step of ion mobility at a resolving power (Rp) of approximately 100. Finally, a Rp of about 250 in ion mobility made it possible to detect the eighth and last of the expected structures. Complementary to these measurements, tandem MS with high activation energy charge transfer dissociation (CTD) allowed us to unambiguously characterize seven regioisomers out of the eight possible products of enzymatic glucosylation. This work illustrates the potential of the recently described powerful IMS and CTD-MS methods for the precise structural characterization of complex glycans.
Collapse
Affiliation(s)
- David Ropartz
- INRAE, UR BIA, F-44316 Nantes, France.,INRAE, BIBS Facility, F-44316 Nantes, France
| | - Mathieu Fanuel
- INRAE, UR BIA, F-44316 Nantes, France.,INRAE, BIBS Facility, F-44316 Nantes, France
| | - Simon Ollivier
- INRAE, UR BIA, F-44316 Nantes, France.,INRAE, BIBS Facility, F-44316 Nantes, France
| | - Adrien Lissarrague
- INRAE, UR BIA, F-44316 Nantes, France.,INRAE, BIBS Facility, F-44316 Nantes, France
| | - Mounir Benkoulouche
- Toulouse Biotechnology Institute, TBI, Université de Toulouse, CNRS, INRAE, INSA, 135 Avenue de Rangueil, F-31077 Toulouse Cedex 04, France
| | - Laurence A Mulard
- Institut Pasteur, Université de Paris, CNRS UMR3523, Unité de Chimie des Biomolécules, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Isabelle André
- Toulouse Biotechnology Institute, TBI, Université de Toulouse, CNRS, INRAE, INSA, 135 Avenue de Rangueil, F-31077 Toulouse Cedex 04, France
| | - David Guieysse
- Toulouse Biotechnology Institute, TBI, Université de Toulouse, CNRS, INRAE, INSA, 135 Avenue de Rangueil, F-31077 Toulouse Cedex 04, France
| | - Hélène Rogniaux
- INRAE, UR BIA, F-44316 Nantes, France.,INRAE, BIBS Facility, F-44316 Nantes, France
| |
Collapse
|
47
|
Bai X, Zhu M, He Y, Wang T, Tian D, Shu J. The impacts of probiotics in eradication therapy of Helicobacter pylori. Arch Microbiol 2022; 204:692. [PMID: 36344628 PMCID: PMC9640438 DOI: 10.1007/s00203-022-03314-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/25/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022]
Abstract
Helicobacter pylori (H. pylori) is a well-known pathogen that infects approximately half of the world's population. It is a pathogenic agent with potential health hazards related to diverse diseases, especially digestive diseases, such as chronic gastritis, peptic ulcer, and gastric carcinoma. In clinical, antibiotics are commonly applied in eradication therapy of H. pylori. However, the increase in antibiotic resistance and side effects has induced the failure of eradication therapy. Recent studies have shown that probiotic supplementation has promising application prospects. It can restore the gastrointestinal microbiota balance and prevent dysbacteriosis caused by antibiotics. Furthermore, it has been reported to have direct or indirect inhibitory effects on H. pylori. Probiotics may have a beneficial effect on H. pylori eradication. However, the strain, dosages, duration times, and safety of probiotic supplementation need further study before clinical applications.
Collapse
Affiliation(s)
- Xiaofen Bai
- Department of Gastroenterology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
- Department of Gastroenterology, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Minjie Zhu
- Department of Gastroenterology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Yajun He
- Department of Clinic Laboratory, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Tengyan Wang
- Department of Gastroenterology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Da Tian
- Department of Gastroenterology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Jianchang Shu
- Department of Gastroenterology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China.
| |
Collapse
|
48
|
A Novel Pseudoalteromonas xiamenensis Marine Isolate as a Potential Probiotic: Anti-Inflammatory and Innate Immune Modulatory Effects against Thermal and Pathogenic Stresses. Mar Drugs 2021; 19:md19120707. [PMID: 34940706 PMCID: PMC8707914 DOI: 10.3390/md19120707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/11/2021] [Accepted: 12/12/2021] [Indexed: 11/16/2022] Open
Abstract
A marine bacterial strain was isolated from seawater and characterized for it beneficial probiotic effects using zebrafish as a model system. The strain was identified by morphological, physiological, biochemical, and phylogenetic analyses. The strain was most closely related to Pseudoalteromonas xiamenensis Y2, with 99.66% similarity; thus, we named it Pseudoalteromonas xiamenensis S1131. Improvement of host disease tolerance for the P. xiamenensis isolate was adapted in a zebrafish model using Edwardsiella piscicida challenge. The larvae were pre-exposed to P. xiamenensis prior to E. piscicida challenge, resulting in a 73.3% survival rate compared to a 46.6% survival for the control. The treated larvae tolerated elevated temperatures at 38 °C, with 85% survival, compared to 60% survival for the control. Assessment of immunomodulatory responses at the mRNA level demonstrated the suppression of pro-inflammatory markers tnfα and il6, and upregulation of heat shock protein hsp90 and mucin genes. The same effect was corroborated by immunoblot analysis, revealing significant inhibition of Tnfα and an enhanced expression of the Hsp90 protein. The antibacterial activity of P. xiamenensis may be related to mucin overexpression, which can suppress bacterial biofilm formation and enhance macrophage uptake. This phenomenon was evaluated using nonstimulated macrophage RAW264.7 cells. Further studies may be warranted to elucidate a complete profile of the probiotic effects, to expand the potential applications of the present P. xiamenensis isolate.
Collapse
|
49
|
Aminu AJ, Petkova M, Atkinson AJ, Yanni J, Morris AD, Simms RT, Chen W, Yin Z, Kuniewicz M, Holda MK, Kuzmin VS, Perde F, Molenaar P, Dobrzynski H. Further insights into the molecular complexity of the human sinus node - The role of 'novel' transcription factors and microRNAs. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:86-104. [PMID: 34004232 DOI: 10.1016/j.pbiomolbio.2021.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023]
Abstract
RESEARCH PURPOSE The sinus node (SN) is the heart's primary pacemaker. Key ion channels (mainly the funny channel, HCN4) and Ca2+-handling proteins in the SN are responsible for its function. Transcription factors (TFs) regulate gene expression through inhibition or activation and microRNAs (miRs) do this through inhibition. There is high expression of macrophages and mast cells within the SN connective tissue. 'Novel'/unexplored TFs and miRs in the regulation of ion channels and immune cells in the SN are not well understood. Using RNAseq and bioinformatics, the expression profile and predicted interaction of key TFs and cell markers with key miRs in the adult human SN vs. right atrial tissue (RA) were determined. PRINCIPAL RESULTS 68 and 60 TFs significantly more or less expressed in the SN vs. RA respectively. Among those more expressed were ISL1 and TBX3 (involved in embryonic development of the SN) and 'novel' RUNX1-2, CEBPA, GLI1-2 and SOX2. These TFs were predicted to regulate HCN4 expression in the SN. Markers for different cells: fibroblasts (COL1A1), fat (FABP4), macrophages (CSF1R and CD209), natural killer (GZMA) and mast (TPSAB1) were significantly more expressed in the SN vs. RA. Interestingly, RUNX1-3, CEBPA and GLI1 also regulate expression of these cells. MiR-486-3p inhibits HCN4 and markers involved in immune response. MAJOR CONCLUSIONS In conclusion, RUNX1-2, CSF1R, TPSAB1, COL1A1 and HCN4 are highly expressed in the SN but not miR-486-3p. Their complex interactions can be used to treat SN dysfunction such as bradycardia. Interestingly, another research group recently reported miR-486-3p is upregulated in blood samples from severe COVID-19 patients who suffer from bradycardia.
Collapse
Affiliation(s)
- Abimbola J Aminu
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Maria Petkova
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Andrew J Atkinson
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Joseph Yanni
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Alex D Morris
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Robert T Simms
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Weixuan Chen
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Zeyuan Yin
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Marcin Kuniewicz
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom; Department of Anatomy, Jagiellonian University Medical College, Krakow, Poland
| | - Mateusz K Holda
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom; Department of Anatomy, Jagiellonian University Medical College, Krakow, Poland
| | - Vladislav S Kuzmin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Moscow, Russia
| | - Filip Perde
- National Institute of Legal Medicine, Bucharest, Romania
| | - Peter Molenaar
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia; Cardiovascular Molecular & Therapeutics Translational Research Group, University of Queensland, The Prince Charles Hospital, Brisbane, Australia
| | - Halina Dobrzynski
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom; Department of Anatomy, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
50
|
Moriichi K, Fujiya M, Okumura T. The endoscopic diagnosis of mucosal healing and deep remission in inflammatory bowel disease. Dig Endosc 2021; 33:1008-1023. [PMID: 33020947 DOI: 10.1111/den.13863] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022]
Abstract
The therapeutic goal in inflammatory bowel disease (IBD) patients has shifted from controlling the clinical activity alone to managing other associated problems. The concept of mucosal healing (MH) and deep remission (DR) are advocated and regarded as new therapeutic goals in IBD. However, the definition of MH still remains controversial. It is unclear whether or not the histological structures or functional factors should be included in the definition of DR in addition to clinical remission and MH. The classifications of white-light imaging (e.g. Mayo endoscopic subscore, UCEIS, CD Endoscopic Index of Severity, simple Endoscopic Score-CD) have been proposed and are now widely used to assess the severity as well as the MH of inflammation in IBD. In ulcerative colitis, magnifying chromoendoscopy has been shown to be useful to assess the MH of inflammation while other types of image-enhanced endoscopy, such as narrow-band imaging, have not. Endocytoscopy and confocal laser endomicroscopy (CLE) are also applied to assess the activity in IBD. These endoscopic procedures can estimate MH with more precision through observing the details of superficial structures, such as crypt openings. In addition, CLE can partially assess the mucosal function by detecting fluorescence leakage. Molecular imaging can possibly detect the molecules associated with inflammation, intestinal regeneration and differentiation, and various functions including the intestinal barrier and mucus secretion. These novel procedures may improve the diagnosis strategy of DR through the assessment of DR-associated factors such as the histological structures and functional factors in the near future.
Collapse
Affiliation(s)
- Kentaro Moriichi
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Hokkaido, Japan
| | - Mikihiro Fujiya
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Hokkaido, Japan
| | - Toshikatsu Okumura
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Hokkaido, Japan
| |
Collapse
|