1
|
Wang X, Yang H, Sun T, Zhang J, Wang L, Zhang Y, Zhou N. A fluorescence and SERS dual-mode biosensor for quantification and imaging of Mucin1 in living cells. Biosens Bioelectron 2025; 270:116964. [PMID: 39579679 DOI: 10.1016/j.bios.2024.116964] [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: 09/26/2024] [Revised: 11/06/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
Mucin1 (MUC1) is a cell surface transmembrane protein overexpressed in multiple types of tumor cells, which is generally considered as a tumor-associated biomarker. Thus, quantifying and imaging of MUC1 in tumor cells is of great significance for the diagnosis and biological therapy of tumors. Herein, a fluorescence (FL) and surface-enhanced Raman scattering (SERS) dual-mode biosensor was developed for sensitive detection and imaging of MUC1 in living cells. The FL-SERS biosensor was based on self-assembled satellite structures mediated by the competition of MUC1 and complementary DNA modified on Au nanostars (AuNS-cDNA-PEG) with aptamer modified on quantum dots (QD-PDDA-Apt). This biosensor achieved dual-mode quantification and imaging of MUC1 by quenching the FL signal of QD and significantly enhancing the SERS signal of PDDA through the metal FL quenching effect and hotspot effect of AuNS, respectively. The dual-mode biosensor exhibited high sensitivity to MUC1, with a detection limit of 1.19 fg/mL under FL mode and 1.16 fg/mL under SERS mode. Moreover, this biosensor displayed good selectivity, nice biological stability and low cytotoxicity. Importantly, this biosensor possessed an excellent MUC1 dual-mode imaging capability with high specificity in different tumor cells, providing a new idea for clinical diagnosis of tumors.
Collapse
Affiliation(s)
- Xiaoli Wang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi, 214122, China
| | - Huiru Yang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi, 214122, China
| | - Tao Sun
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi, 214122, China
| | - Jiale Zhang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi, 214122, China
| | - Lixuan Wang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi, 214122, China
| | - Yuting Zhang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi, 214122, China.
| | - Nandi Zhou
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
2
|
Kobayashi Y, Hanh CH, Yagi N, Le NKT, Yun Y, Shimamura A, Fukui K, Mitani A, Suzuki K, Kanda A, Iwai H. CCL4 Affects Eosinophil Survival via the Shedding of the MUC1 N-Terminal Domain in Airway Inflammation. Cells 2024; 14:33. [PMID: 39791734 PMCID: PMC11719767 DOI: 10.3390/cells14010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/24/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025] Open
Abstract
Eosinophilic chronic rhinosinusitis (ECRS), a CRS with nasal polyps (CRSwNP), is characterized by eosinophilic infiltration with type 2 inflammation and is highly associated with bronchial asthma. Intractable ECRS with poorly controlled asthma is recognized as a difficult-to-treat eosinophilic airway inflammation. Although eosinophils are activated and coincubation with airway epithelial cells prolongs their survival, the interaction mechanism between eosinophils and epithelial cells is unclear. This study examined the effect of eosinophils on mucin glycoprotein 1 (MUC1), a member of membrane-bound mucin, in the airway epithelial cells, to elucidate the mechanisms of the eosinophil-airway epithelial cell interaction. Nasal polyp samples from patients with CRSwNP and BEAS-2B airway epithelial cells, coincubated with purified eosinophils, were stained with two MUC1 antibodies. To confirm the involvement of CCL4, an anti-CCL4 neutralizing antibody or recombinant CCL4 was used as needed. The immunofluorescence results revealed a negative correlation between the expression of full-length MUC1 and eosinophil count in nasal polyps. In BEAS-2B coincubated with eosinophils, full-length MUC1, but not the C-terminal domain, was reduced, and eosinophil survival was prolonged, which was concomitant with CCL4 increase, whereas the anti-CCL4 neutralizing antibody decreased these reactions. The survival of eosinophils that contacted recombinant MUC1 without the N-terminal domain was prolonged, and recombinant CCL4 increased the expression of metalloproteases. Increased CCL4 induces the contact between eosinophils and airway epithelial cells by shedding the MUC1 N-terminal domain and enhances eosinophil survival in eosinophilic airway inflammation. This novel mechanism may be a therapeutic target for difficult-to-treat eosinophilic airway inflammation.
Collapse
Affiliation(s)
- Yoshiki Kobayashi
- Airway Disease Section, Department of Otorhinolaryngology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan; (C.H.H.); (N.K.T.L.); (Y.Y.); (A.S.); (K.F.); (A.M.); (K.S.); (A.K.); (H.I.)
- Allergy Center, Kansai Medical University Hospital, Osaka 573-1010, Japan
| | - Chu Hong Hanh
- Airway Disease Section, Department of Otorhinolaryngology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan; (C.H.H.); (N.K.T.L.); (Y.Y.); (A.S.); (K.F.); (A.M.); (K.S.); (A.K.); (H.I.)
| | - Naoto Yagi
- Third Department of Internal Medicine, Kansai Medical University, Osaka 573-1010, Japan;
| | - Nhi Kieu Thi Le
- Airway Disease Section, Department of Otorhinolaryngology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan; (C.H.H.); (N.K.T.L.); (Y.Y.); (A.S.); (K.F.); (A.M.); (K.S.); (A.K.); (H.I.)
| | - Yasutaka Yun
- Airway Disease Section, Department of Otorhinolaryngology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan; (C.H.H.); (N.K.T.L.); (Y.Y.); (A.S.); (K.F.); (A.M.); (K.S.); (A.K.); (H.I.)
- Allergy Center, Kansai Medical University Hospital, Osaka 573-1010, Japan
| | - Akihiro Shimamura
- Airway Disease Section, Department of Otorhinolaryngology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan; (C.H.H.); (N.K.T.L.); (Y.Y.); (A.S.); (K.F.); (A.M.); (K.S.); (A.K.); (H.I.)
| | - Kenta Fukui
- Airway Disease Section, Department of Otorhinolaryngology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan; (C.H.H.); (N.K.T.L.); (Y.Y.); (A.S.); (K.F.); (A.M.); (K.S.); (A.K.); (H.I.)
| | - Akitoshi Mitani
- Airway Disease Section, Department of Otorhinolaryngology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan; (C.H.H.); (N.K.T.L.); (Y.Y.); (A.S.); (K.F.); (A.M.); (K.S.); (A.K.); (H.I.)
| | - Kensuke Suzuki
- Airway Disease Section, Department of Otorhinolaryngology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan; (C.H.H.); (N.K.T.L.); (Y.Y.); (A.S.); (K.F.); (A.M.); (K.S.); (A.K.); (H.I.)
| | - Akira Kanda
- Airway Disease Section, Department of Otorhinolaryngology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan; (C.H.H.); (N.K.T.L.); (Y.Y.); (A.S.); (K.F.); (A.M.); (K.S.); (A.K.); (H.I.)
- Allergy Center, Kansai Medical University Hospital, Osaka 573-1010, Japan
| | - Hiroshi Iwai
- Airway Disease Section, Department of Otorhinolaryngology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan; (C.H.H.); (N.K.T.L.); (Y.Y.); (A.S.); (K.F.); (A.M.); (K.S.); (A.K.); (H.I.)
| |
Collapse
|
3
|
Kidd KO, Williams AH, Taylor A, Martin L, Robins V, Sayer JA, Olinger E, Mabillard HR, Papagregoriou G, Deltas C, Stavrou C, Conlon PJ, Hogan RE, Elhassan EAE, Springer D, Zima T, Izzi C, Vrbacká A, Piherová L, Pohludka M, Radina M, Vylet'al P, Hodanova K, Zivna M, Kmoch S, Bleyer AJ. Eight-fold increased COVID-19 mortality in autosomal dominant tubulointerstitial kidney disease due to MUC1 mutations: an observational study. BMC Nephrol 2024; 25:449. [PMID: 39696072 DOI: 10.1186/s12882-024-03896-1] [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: 07/10/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND MUC1 and UMOD pathogenic variants cause autosomal dominant tubulointerstitial kidney disease (ADTKD). MUC1 is expressed in kidney, nasal mucosa and respiratory tract, while UMOD is expressed only in kidney. Due to haplo-insufficiency ADTKD-MUC1 patients produce approximately 50% of normal mucin-1. METHODS To determine whether decreased mucin-1 production was associated with an increased COVID-19 risk, we sent a survey to members of an ADTKD registry in September 2021, after the initial, severe wave of COVID-19. We linked results to previously obtained ADTKD genotype and plasma CA15-3 (mucin-1) levels and created a longitudinal registry of COVID-19 related deaths. RESULTS Surveys were emailed to 637 individuals, with responses from 89 ADTKD-MUC1 and 132 ADTKD-UMOD individuals. 19/83 (23%) ADTKD-MUC1 survey respondents reported a prior COVID-19 infection vs. 14/125 (11%) ADTKD-UMOD respondents (odds ratio (OR) 2.35 (95%CI 1.60-3.11, P = 0.0260). Including additional familial cases reported from survey respondents, 10/41 (24%) ADTKD-MUC1 individuals died of COVID-19 vs. 1/30 (3%) with ADTKD-UMOD, with OR 9.21 (95%CI 1.22-69.32), P = 0.03. The mean plasma mucin-1 level prior to infection in 14 infected and 27 uninfected ADTKD-MUC1 individuals was 7.06 ± 4.12 vs. 10.21 ± 4.02 U/mL (P = 0.035). Over three years duration, our longitudinal registry identified 19 COVID-19 deaths in 360 ADTKD-MUC1 individuals (5%) vs. 3 deaths in 478 ADTKD-UMOD individuals (0.6%) (P = 0.0007). Multivariate logistic regression revealed the following odds ratios (95% confidence interval) for COVID-19 deaths: ADTKD-MUC1 8.4 (2.9-29.5), kidney transplant 5.5 (1.6-9.1), body mass index (kg/m2) 1.1 (1.0-1.2), age (y) 1.04 (1.0-1.1). CONCLUSIONS Individuals with ADTKD-MUC1 are at an eight-fold increased risk of COVID-19 mortality vs. ADTKD-UMOD individuals. Haplo-insufficient production of mucin-1 may be responsible.
Collapse
Affiliation(s)
- Kendrah O Kidd
- Wake Forest School of Medicine, Section on Nephrology, Winston-Salem, NC, 27157, USA
- Department of Paediatrics and Inherited Metabolic Disorders, Research Unit of Rare Diseases, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Abbigail Taylor
- Wake Forest School of Medicine, Section on Nephrology, Winston-Salem, NC, 27157, USA
| | - Lauren Martin
- Wake Forest School of Medicine, Section on Nephrology, Winston-Salem, NC, 27157, USA
| | - Victoria Robins
- Wake Forest School of Medicine, Section on Nephrology, Winston-Salem, NC, 27157, USA
| | - John A Sayer
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Renal Services, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Newcastle Biomedical Research Centre, NIHR, Newcastle upon Tyne, UK
| | - Eric Olinger
- Center for Human Genetics, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Holly R Mabillard
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Gregory Papagregoriou
- Department of Biological Sciences, Molecular Medicine Research Center, University of Cyprus, Nicosia, Cyprus
| | - Constantinos Deltas
- Department of Biological Sciences, Molecular Medicine Research Center, University of Cyprus, Nicosia, Cyprus
| | | | - Peter J Conlon
- Department of Nephrology and Transplantation Beaumont Hospital, Dublin, Ireland
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Richard Edmund Hogan
- Department of Nephrology and Transplantation Beaumont Hospital, Dublin, Ireland
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Elhussein A E Elhassan
- Department of Nephrology and Transplantation Beaumont Hospital, Dublin, Ireland
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Drahomíra Springer
- Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and the First Faculty of Medicine of Charles University, Prague, Czech Republic
| | - Tomáš Zima
- Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and the First Faculty of Medicine of Charles University, Prague, Czech Republic
| | - Claudia Izzi
- Clinical Genetics Unit, University of Brescia and Spedali Civili, Brescia, Italy
| | - Alena Vrbacká
- Department of Paediatrics and Inherited Metabolic Disorders, Research Unit of Rare Diseases, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lenka Piherová
- Department of Paediatrics and Inherited Metabolic Disorders, Research Unit of Rare Diseases, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Martin Radina
- Department of Paediatrics and Inherited Metabolic Disorders, Research Unit of Rare Diseases, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Petr Vylet'al
- Department of Paediatrics and Inherited Metabolic Disorders, Research Unit of Rare Diseases, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Katerina Hodanova
- Department of Paediatrics and Inherited Metabolic Disorders, Research Unit of Rare Diseases, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martina Zivna
- Wake Forest School of Medicine, Section on Nephrology, Winston-Salem, NC, 27157, USA
- Department of Paediatrics and Inherited Metabolic Disorders, Research Unit of Rare Diseases, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Stanislav Kmoch
- Wake Forest School of Medicine, Section on Nephrology, Winston-Salem, NC, 27157, USA
- Department of Paediatrics and Inherited Metabolic Disorders, Research Unit of Rare Diseases, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Medirex Group Academy, Trnava, Slovakia
| | - Anthony J Bleyer
- Wake Forest School of Medicine, Section on Nephrology, Winston-Salem, NC, 27157, USA.
- Department of Paediatrics and Inherited Metabolic Disorders, Research Unit of Rare Diseases, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
4
|
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
|
5
|
Chen X, Chen Y, Zhang Y, Zhang Y, Wang Y, Li Y, Sun Y, Meng G, Yang G, Li H. ZG16 impacts gut microbiota-associated intestinal inflammation and pulmonary mucosal function through bacterial metabolites. Int Immunopharmacol 2024; 141:112995. [PMID: 39191121 DOI: 10.1016/j.intimp.2024.112995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 07/04/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Zymogen granule 16 (ZG16) is a secretory glycoprotein found in zymogen granules, which also plays an important role in colorectal inflammation and cancer. Herein, a ZG16 gene knock-out (ZG16-/-) mouse line was established and we found that ZG16 deletion damaged the intestinal mucosal barrier and gut microbiota, which resulted in low-level inflammation and further promoted the development of ulcerative colitis and inflammation-related colorectal cancer. Meanwhile, a metabolomics analysis on mouse feces showed that the metabolites significantly differed between ZG16-/- and WT mice, which were important mediators of host-microbiota communication and may impact the pulmonary inflammation of mice. Indeed, ZG16-/- mice showed more severe inflammation in a bronchial asthma model. Taken together, the results demonstrate that ZG16 plays a pivotal role in inhibiting inflammation and regulating immune responses in colorectum and lung of experimental animals, which may provide a better understanding of the underlying mechanism of human inflammatory diseases associated with ZG16.
Collapse
Affiliation(s)
- Xinping Chen
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Yixin Chen
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Ying Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Yonghuan Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Yao Wang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Yingjia Li
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Yaqi Sun
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Guangxun Meng
- The Center for Microbes, Development, and Health, CAS Key Laboratory of Molecular Virology & Immunology, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Shanghai 200031, PR China.
| | - Guiwen Yang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, PR China.
| | - Hua Li
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, PR China; The Center for Microbes, Development, and Health, CAS Key Laboratory of Molecular Virology & Immunology, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Shanghai 200031, PR China.
| |
Collapse
|
6
|
Munyayi TA, Crous A. Colorimetric Biosensor for Early Detection of MUC1-Positive Cells Using Aptamer-Conjugated Plasmonic Gold Nanostars. ACS APPLIED NANO MATERIALS 2024; 7:24886-24896. [DOI: 10.1021/acsanm.4c04793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Affiliation(s)
- Tozivepi Aaron Munyayi
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| | - Anine Crous
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| |
Collapse
|
7
|
Dyachenko EI, Bel’skaya LV. Salivary Transmembrane Mucins of the MUC1 Family (CA 15-3, CA 27.29, MCA) in Breast Cancer: The Effect of Human Epidermal Growth Factor Receptor 2 (HER2). Cancers (Basel) 2024; 16:3461. [PMID: 39456554 PMCID: PMC11506585 DOI: 10.3390/cancers16203461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/01/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
The MUC1 family of transmembrane glycoproteins (CA 15-3, CA 27.29, MCA) is aberrantly expressed among patients with breast cancer. Objectives: to measure the level of degradation products of MUC1, including CA 15-3, CA 27.29, and MCA, in the saliva of breast cancer patients and to describe the biochemical processes that influence their expression and the regulation of their biological functions. Methods: The case-control study included three groups (breast cancer, fibroadenomas, and healthy controls). All study participants provided saliva samples strictly before starting treatment. The levels of MUC1, including CA 15-3, CA 27.29, and MCA, free progesterone and estradiol, cytokines (MCP-1, VEGF, TNF-α, IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-18), and amino acids (Asp, Gln, Gly, His, Leu + Ile, Orn, Phe, Pro, Tyr) were determined. Results: It was shown that the levels of the MUC1 family in the saliva of patients with HER2-positive breast cancer were significantly lower compared to the control group. The level of pro-inflammatory cytokines and the level of free estradiol affected the expression of MUC1. We obtained a reliable relationship between the aggressive nature of tumor growth, an increased level of pro-inflammatory cytokines, a low level of free estradiol, and the suppressed expression of salivary MUC1. Conclusions: Among patients with aggressive breast cancer, a high level of pro-inflammatory cytokines, and a low level of free estradiol, there was an inhibition of the expression of pathologically unchanged glycoprotein MUC1 in saliva.
Collapse
Affiliation(s)
| | - Lyudmila V. Bel’skaya
- Biochemistry Research Laboratory, Omsk State Pedagogical University, 644099 Omsk, Russia;
| |
Collapse
|
8
|
Tharavecharak S, Fujimoto H, Yasuma T, D'Alessandro-Gabazza CN, Toda M, Tomaru A, Saiki H, Uemura M, Kogue Y, Ito T, Furuhashi K, Okano T, Takeshita A, Nishihama K, Ono R, Hataji O, Nosaka T, Kobayashi T, Gabazza EC. Bleomycin-Induced Pulmonary Fibrosis in Transgenic Mice Carrying the Human MUC5B rs35705950 Variant. Cells 2024; 13:1523. [PMID: 39329706 PMCID: PMC11430646 DOI: 10.3390/cells13181523] [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: 07/27/2024] [Revised: 09/01/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, often fatal lung disease characterized by tissue scarring and declining lung function. The MUC5B promoter polymorphism rs35705950, a significant genetic predisposition for IPF, paradoxically associates with better survival and slower disease progression than other IPF genotypes. This study investigates the potential paradoxical protective effects of this MUC5B variant in lung fibrosis. For this purpose, we developed a transgenic mouse model overexpressing the human MUC5B rs35705950 variant in the proximal large airways. Lung fibrosis was induced through subcutaneous injection of bleomycin. Results demonstrated significantly reduced lung fibrosis severity in transgenic mice compared to wild-type mice, assessed by trichrome staining, Ashcroft scoring, and hydroxyproline levels. Additionally, transgenic mice showed significantly lower levels of inflammatory cells and cytokines (TNFα, IL-6, IFNγ) and growth factors (PDGF, CTGF, IL-13) in the bronchoalveolar lavage fluid and lung tissues. There was also a significant decrease in mRNA expressions of fibrosis-related markers (periostin, fibronectin, Col1a1). In summary, this study reveals that mucin overexpression related to the MUC5B rs35705950 variant in the large airways significantly attenuates lung fibrosis and inflammatory responses in transgenic mice. These findings suggest that the rs35705950 variant modulates inflammatory and fibrotic responses in the proximal airways, which may contribute to the slower disease progression observed in IPF patients carrying this variant. Our study offers a possible explanation for the paradoxical beneficial effects of the MUC5B variant despite its role as a significant predisposing factor for IPF.
Collapse
Affiliation(s)
- Suphachai Tharavecharak
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Hajime Fujimoto
- Department Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Taro Yasuma
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Microbiome Research Center, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Department of Diabetes, Endocrinology and Metabolism, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Corina N D'Alessandro-Gabazza
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Masaaki Toda
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Atsushi Tomaru
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Haruko Saiki
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Mei Uemura
- Department of Diabetes, Endocrinology and Metabolism, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Yurie Kogue
- Department Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Toshiyuki Ito
- Department Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Kazuki Furuhashi
- Department Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Tomohito Okano
- Department Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Atsuro Takeshita
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Department of Diabetes, Endocrinology and Metabolism, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Kota Nishihama
- Department of Diabetes, Endocrinology and Metabolism, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Ryoichi Ono
- Department of Microbiology and Molecular Genetics, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan
| | - Osamu Hataji
- Respiratory Center, Matsusaka Municipal Hospital, Tonomachi1550, Matsusaka 515-8544, Mie, Japan
| | - Tetsuya Nosaka
- Department of Microbiology and Molecular Genetics, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan
| | - Tetsu Kobayashi
- Department Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Microbiome Research Center, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Esteban C Gabazza
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Department Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Microbiome Research Center, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Respiratory Center, Matsusaka Municipal Hospital, Tonomachi1550, Matsusaka 515-8544, Mie, Japan
| |
Collapse
|
9
|
Cao X, Tan J, Zheng R, Wang F, Zhou L, Yi J, Yuan R, Dai Q, Song L, Dai A. Targeting necroptosis: a promising avenue for respiratory disease treatment. Cell Commun Signal 2024; 22:418. [PMID: 39192326 DOI: 10.1186/s12964-024-01804-6] [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: 06/20/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024] Open
Abstract
Respiratory diseases are a growing concern in public health because of their potential to endanger the global community. Cell death contributes critically to the pathophysiology of respiratory diseases. Recent evidence indicates that necroptosis, a unique form of programmed cell death (PCD), plays a vital role in the molecular mechanisms underlying respiratory diseases, distinguishing it from apoptosis and conventional necrosis. Necroptosis is a type of inflammatory cell death governed by receptor-interacting serine/threonine protein kinase 1 (RIPK1), RIPK3, and mixed-lineage kinase domain-like protein (MLKL), resulting in the release of intracellular contents and inflammatory factors capable of initiating an inflammatory response in adjacent tissues. These necroinflammatory conditions can result in significant organ dysfunction and long-lasting tissue damage within the lungs. Despite evidence linking necroptosis to various respiratory diseases, there are currently no specific alternative treatments that target this mechanism. This review provides a comprehensive overview of the most recent advancements in understanding the significance and mechanisms of necroptosis. Specifically, this review emphasizes the intricate association between necroptosis and respiratory diseases, highlighting the potential use of necroptosis as an innovative therapeutic approach for treating these conditions.
Collapse
Affiliation(s)
- Xianya Cao
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Junlan Tan
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Runxiu Zheng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Feiying Wang
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Lingling Zhou
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Jian Yi
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Rong Yuan
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Qin Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Lan Song
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Aiguo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China.
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China.
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China.
| |
Collapse
|
10
|
Kidd KO, Williams AH, Taylor A, Martin L, Robins V, Sayer JA, Olinger E, Mabillard HR, Papagregoriou G, Deltas C, Stavrou C, Conlon PJ, Hogan RE, Elhassan EAE, Springer D, Zima T, Izzi C, Vrbacká A, Piherová L, Pohludka M, Radina M, Vylet'al P, Hodanova K, Zivna M, Kmoch S, Bleyer AJ. Eight-Fold Increased COVID-19 Mortality in Autosomal Dominant Tubulointerstitial Kidney Disease due to MUC1 Mutations: An Observational Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.03.24309887. [PMID: 39006416 PMCID: PMC11245082 DOI: 10.1101/2024.07.03.24309887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Background MUC1 and UMOD pathogenic variants cause autosomal dominant tubulointerstitial kidney disease (ADTKD). MUC1 is expressed in kidney, nasal mucosa and respiratory tract, while UMOD is expressed only in kidney. Due to haplo-insufficiency ADTKD- MUC1 patients produce approximately 50% of normal mucin-1. Methods To determine whether decreased mucin-1 production was associated with an increased COVID-19 risk, we sent a survey to members of an ADTKD registry in September 2021, after the initial, severe wave of COVID-19. We linked results to previously obtained ADTKD genotype and plasma CA15-3 (mucin-1) levels and created a longitudinal registry of COVID-19 related deaths. Results Surveys were emailed to 637 individuals, with responses from 89 ADTKD- MUC1 and 132 ADTKD- UMOD individuals. 19/83 (23%) ADTKD- MUC1 survey respondents reported a prior COVID-19 infection vs. 14/125 (11%) ADTKD- UMOD respondents (odds ratio (OR) 2.35 (95%CI 1.60-3.11, P = 0.0260). Including additional familial cases reported from survey respondents, 10/41 (24%) ADTKD- MUC1 individuals died of COVID-19 vs. 1/30 (3%) with ADTKD- UMOD , with OR 9.21 (95%CI 1.22-69.32), P = 0.03. The mean plasma mucin-1 level prior to infection in 14 infected and 27 uninfected ADTKD- MUC1 individuals was 7.06±4.12 vs. 10.21±4.02 U/mL ( P = 0.035). Over three years duration, our longitudinal registry identified 19 COVID-19 deaths in 360 ADTKD- MUC1 individuals (5%) vs. 3 deaths in 478 ADTKD- UMOD individuals (0.6%) ( P = 0.0007). Multivariate logistic regression revealed the following odds ratios (95% confidence interval) for COVID-19 deaths: ADTKD- MUC1 8.4 (2.9-29.5), kidney transplant 5.5 (1.6-9.1), body mass index (kg/m 2 ) 1.1 (1.0-1.2), age (y) 1.04 (1.0-1.1). Conclusions Individuals with ADTKD- MUC1 are at an eight-fold increased risk of COVID-19 mortality vs. ADTKD- UMOD individuals. Haplo-insufficient production of mucin-1 may be responsible.
Collapse
|
11
|
Fang Y, Zhang Y, Bi S, Peng B, Wang C, Ju H, Liu Y. Securing LYTAC with Logic-Identification System for Cancer Cell-Selective Membrane Protein Degradation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310039. [PMID: 38431928 DOI: 10.1002/smll.202310039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 02/18/2024] [Indexed: 03/05/2024]
Abstract
Lysosome-targeting chimera (LYTAC) links proteins of interest (POIs) with lysosome-targeting receptors (LTRs) to achieve membrane protein degradation, which is becoming a promising therapeutic modality. However, cancer cell-selective membrane protein degradation remains a big challenge considering expressions of POIs in both cancer cells and normal cells, as well as broad tissue distribution of LTRs. Here a logic-identification system is designed, termed Logic-TAC, based on cell membrane-guided DNA calculations to secure LYTAC selectively for cancer cells. Logic-TAC is designed as a duplex DNA structure, with both POI and LTR recognition regions sealed to avoid systematic toxicity during administration. MCF-7 and MCF-10A are chosen as sample cancer cell and normal cell respectively. As input 1 for logic-identification, membrane proteins EpCAM, which is highly expressed by MCF-7 but barely by MCF-10A, reacts with Logic-TAC to expose POI recognition region. As input 2 for logic-identification, Logic-TAC binds to POI, membrane protein MUC1, to expose LTR recognition region. As output, MUC1 is connected to LTR and degraded via lysosome pathway selectively for cancer cell MCF-7 with little side effect on normal cell MCF-10A. The logic-identification system also demonstrated satisfactory in vivo therapeutic results, indicating its promising potential in precise targeted therapy.
Collapse
Affiliation(s)
- Yanyun Fang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yue Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shiyi Bi
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Bo Peng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Caixia Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Ying Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
12
|
Khayer N, Jalessi M, Farhadi M, Azad Z. S100a9 might act as a modulator of the Toll-like receptor 4 transduction pathway in chronic rhinosinusitis with nasal polyps. Sci Rep 2024; 14:9722. [PMID: 38678138 PMCID: PMC11055867 DOI: 10.1038/s41598-024-60205-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/19/2024] [Indexed: 04/29/2024] Open
Abstract
Chronic rhinosinusitis with nasal polyp (CRSwNP) is a highly prevalent disorder characterized by persistent nasal and sinus mucosa inflammation. Despite significant morbidity and decreased quality of life, there are limited effective treatment options for such a disease. Therefore, identifying causal genes and dysregulated pathways paves the way for novel therapeutic interventions. In the current study, a three-way interaction approach was used to detect dynamic co-expression interactions involved in CRSwNP. In this approach, the internal evolution of the co-expression relation between a pair of genes (X, Y) was captured under a change in the expression profile of a third gene (Z), named the switch gene. Subsequently, the biological relevancy of the statistically significant triplets was confirmed using both gene set enrichment analysis and gene regulatory network reconstruction. Finally, the importance of identified switch genes was confirmed using a random forest model. The results suggested four dysregulated pathways in CRSwNP, including "positive regulation of intracellular signal transduction", "arachidonic acid metabolic process", "spermatogenesis" and "negative regulation of cellular protein metabolic process". Additionally, the S100a9 as a switch gene together with the gene pair {Cd14, Tpd52l1} form a biologically relevant triplet. More specifically, we suggested that S100a9 might act as a potential upstream modulator in toll-like receptor 4 transduction pathway in the major CRSwNP pathologies.
Collapse
Affiliation(s)
- Nasibeh Khayer
- Skull Base Research Center, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Maryam Jalessi
- Skull Base Research Center, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, Rasoul Akram Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Farhadi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, Rasoul Akram Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Azad
- Skull Base Research Center, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
da Silva ACG, de Mendonça ICF, Valadares MC. Characterization and applicability of a novel physiologically relevant 3D-tetraculture bronchial model for in vitro assessment of respiratory sensitization. Toxicology 2024; 503:153756. [PMID: 38369009 DOI: 10.1016/j.tox.2024.153756] [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: 12/20/2023] [Revised: 01/29/2024] [Accepted: 02/12/2024] [Indexed: 02/20/2024]
Abstract
Chemical Respiratory Allergy (CRA) is triggered after exposure to Low Molecular Weight (LMW) sensitizers and manifests clinically as asthma and rhinitis. From a risk/toxicity assessment point of view, there are few methods, none of them validated, for evaluating the respiratory sensitization potential of chemicals once the in vivo-based models usually employed for inhalation toxicity addressment do not comprise allergenicity endpoints specifically. Based on that, we developed, characterized, and evaluated the applicability of a 3D-tetraculture airway model reconstructed with bronchial epithelial, fibroblasts, endothelial and monocytic cell lines. Moreover, we exposed the tissue to maleic anhydride (MA) aerosols to challenge the model and subsequently assessed inflammatory and functional aspects of the tissue. The reconstructed tissue presented phenotypic biomarkers compatible with human bronchial epithelium, and MA aerosol exposure triggered an increased IL-8 and IL-6 production, reactive oxygen species (ROS) formation, and apoptosis of epithelial cells. Besides, augmented IL-8 production by monocytic cells was also found, correlating with dendritic cell activation within the co-culture model after MA exposure. Our results demonstrated that the 3D-tetraculture bronchial model presents hallmarks related to human airways' structure and function. Additionally, exposure to a respiratory sensitizer induced inflammatory and functional alterations in the reconstructed tissue, rendering it a valuable tool for exploring the mechanistic framework of chemically induced respiratory sensitization.
Collapse
Affiliation(s)
- Artur Christian Garcia da Silva
- Laboratory of Education and Research in In vitro Toxicology, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | | | - Marize Campos Valadares
- Laboratory of Education and Research in In vitro Toxicology, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| |
Collapse
|
14
|
Huang TH, Wei SH, Kuo HI, Hou HY, Kuo CW, Tseng YL, Lin SH, Wu CL. Baseline Blood Levels of Mucin-1 Are Associated with Crucial On-Treatment Adverse Outcomes in Patients with Idiopathic Pulmonary Fibrosis Receiving Antifibrotic Pirfenidone. Biomedicines 2024; 12:402. [PMID: 38398004 PMCID: PMC10886731 DOI: 10.3390/biomedicines12020402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Mucin-1 is a multi-functional glycoprotein expressed by type II alveolocytes and may be detectable in the circulation following pulmonary fibrosis. The prognostic utility of baseline pre-treatment blood levels of mucin-1 in patients with idiopathic pulmonary fibrosis (IPF) receiving antifibrotics has not yet been fully established. We retrospectively studied a cohort of patients (from two hospitals) with IPF who were receiving pirfenidone for >12 weeks. Baseline blood mucin-1 levels were measured via sandwich enzyme-linked immunosorbent assays. We investigated the performance of mucin-1 levels in longitudinally predicting the risks of acute exacerbation of IPF (AE-IPF) and severe adverse outcomes (SAO), including lung transplantation and death. Seventy patients were included; 20 developed AE-IPF; and 31 had SAO during the follow-up period. Patients with baseline mucin-1 levels ≥2.5 ng/mL had enhanced risks of AE-IPF (adjusted hazard ratio [aHR], 14.07; 95% confidence interval [CI], 4.26-46.49) and SAO within 2 years (aHR, 7.87; 95% CI, 2.86-21.70) and anytime during the follow-up (aHR, 4.68; 95% CI, 2.11-10.39). The risks increased across subgroups with increasing mucin-1 levels. Patients in the "mucin-1 ≥ 2.5" group also exhibited an accelerated decline in DLCO. This study supports baseline blood mucin-1 levels as a biomarker for IPF that predicts adverse outcomes during pirfenidone treatment.
Collapse
Affiliation(s)
- Tang-Hsiu Huang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 35 Siaodong Rd., Tainan 704, Taiwan; (T.-H.H.); (C.-W.K.); (S.-H.L.)
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng Li Rd., Tainan 704, Taiwan; (S.-H.W.); (H.-Y.H.)
| | - Sheng-Huan Wei
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng Li Rd., Tainan 704, Taiwan; (S.-H.W.); (H.-Y.H.)
| | - Hung-I Kuo
- Chest Hospital, Ministry of Health and Welfare, 864 Zhongshan Rd., Rende Dist., Tainan 717, Taiwan;
| | - Hsin-Yu Hou
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng Li Rd., Tainan 704, Taiwan; (S.-H.W.); (H.-Y.H.)
| | - Chin-Wei Kuo
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 35 Siaodong Rd., Tainan 704, Taiwan; (T.-H.H.); (C.-W.K.); (S.-H.L.)
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng Li Rd., Tainan 704, Taiwan; (S.-H.W.); (H.-Y.H.)
| | - Yau-Lin Tseng
- Division of Thoracic Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng Li Rd., Tainan 704, Taiwan;
| | - Sheng-Hsiang Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 35 Siaodong Rd., Tainan 704, Taiwan; (T.-H.H.); (C.-W.K.); (S.-H.L.)
- Department of Public Health, College of Medicine, National Cheng Kung University, 138 Sheng Li Rd., Tainan 704, Taiwan
- Biostatistics Consulting Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng Li Rd., Tainan 704, Taiwan
| | - Chao-Liang Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, 1 Dasyue Road, East District, Tainan 701, Taiwan
- Ditmanson Medical Foundation Chia-Yi Christian Hospital, 539 Chung Hsiao Rd., Chiayi 600, Taiwan
| |
Collapse
|
15
|
Milara J, Morell A, Roger I, Montero P, Cortijo J. Mechanisms underlying corticosteroid resistance in patients with asthma: a review of current knowledge. Expert Rev Respir Med 2023; 17:701-715. [PMID: 37658478 DOI: 10.1080/17476348.2023.2255124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 08/25/2023] [Accepted: 08/31/2023] [Indexed: 09/03/2023]
Abstract
INTRODUCTION Corticosteroids are the most cost-effective anti-inflammatory drugs available for the treatment of asthma. Despite their effectiveness, several asthmatic patients have corticosteroid resistance or insensitivity and exhibit a poor response. Corticosteroid insensitivity implies a poor prognosis due to challenges in finding alternative therapeutic options for asthma. AREAS COVERED In this review, we describe asthma phenotypes and endotypes, as well as their differential responsiveness to corticosteroids. In addition, we describe the mechanism of action of corticosteroids underlying their regulation of the expression of glucocorticoid receptors (GRs) and their anti-inflammatory effects. Furthermore, we summarize the mechanistic evidence underlying corticosteroid-insensitive asthma, which is mainly related to changes in GR gene expression, structure, and post-transcriptional modifications. Finally, various pharmacological strategies designed to reverse corticosteroid insensitivity are discussed. EXPERT OPINION Corticosteroid insensitivity is influenced by the asthma phenotype, endotype, and severity, and serves as an indication for biological therapy. The molecular mechanisms underlying corticosteroid-insensitive asthma have been used to develop targeted therapeutic strategies. However, the lack of clinical trials prevents the clinical application of these treatments.
Collapse
Affiliation(s)
- Javier Milara
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- Pharmacy department, University General Hospital of Valencia, Valencia, Spain
- CIBERES, Health Institute Carlos III, Valencia, Spain
| | - Anselm Morell
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Inés Roger
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- CIBERES, Health Institute Carlos III, Valencia, Spain
| | - Paula Montero
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- Pharmacy department, University General Hospital of Valencia, Valencia, Spain
| | - Julio Cortijo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- CIBERES, Health Institute Carlos III, Valencia, Spain
| |
Collapse
|
16
|
Radziejewska I. Galectin-3 and Epithelial MUC1 Mucin-Interactions Supporting Cancer Development. Cancers (Basel) 2023; 15:2680. [PMID: 37345016 DOI: 10.3390/cancers15102680] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 06/23/2023] Open
Abstract
Aberrant glycosylation of cell surface proteins is a very common feature of many cancers. One of the glycoproteins, which undergoes specific alterations in the glycosylation of tumor cells is epithelial MUC1 mucin, which is highly overexpressed in the malignant state. Such changes lead to the appearance of tumor associated carbohydrate antigens (TACAs) on MUC1, which are rarely seen in healthy cells. One of these structures is the Thomsen-Friedenreich disaccharide Galβ1-3GalNAc (T or TF antigen), which is typical for about 90% of cancers. It was revealed that increased expression of the T antigen has a big impact on promoting cancer progression and metastasis, among others, due to the interaction of this antigen with the β-galactose binding protein galectin-3 (Gal-3). In this review, we summarize current information about the interactions between the T antigen on MUC1 mucin and Gal-3, and their impact on cancer progression and metastasis.
Collapse
Affiliation(s)
- Iwona Radziejewska
- Department of Medical Chemistry, Medical University of Białystok, ul. Mickiewicza 2a, 15-222 Białystok, Poland
| |
Collapse
|
17
|
Yfantis A, Mylonis I, Chachami G, Nikolaidis M, Amoutzias GD, Paraskeva E, Simos G. Transcriptional Response to Hypoxia: The Role of HIF-1-Associated Co-Regulators. Cells 2023; 12:cells12050798. [PMID: 36899934 PMCID: PMC10001186 DOI: 10.3390/cells12050798] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/22/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
The Hypoxia Inducible Factor 1 (HIF-1) plays a major role in the cellular response to hypoxia by regulating the expression of many genes involved in adaptive processes that allow cell survival under low oxygen conditions. Adaptation to the hypoxic tumor micro-environment is also critical for cancer cell proliferation and therefore HIF-1 is also considered a valid therapeutical target. Despite the huge progress in understanding regulation of HIF-1 expression and activity by oxygen levels or oncogenic pathways, the way HIF-1 interacts with chromatin and the transcriptional machinery in order to activate its target genes is still a matter of intense investigation. Recent studies have identified several different HIF-1- and chromatin-associated co-regulators that play important roles in the general transcriptional activity of HIF-1, independent of its expression levels, as well as in the selection of binding sites, promoters and target genes, which, however, often depends on cellular context. We review here these co-regulators and examine their effect on the expression of a compilation of well-characterized HIF-1 direct target genes in order to assess the range of their involvement in the transcriptional response to hypoxia. Delineating the mode and the significance of the interaction between HIF-1 and its associated co-regulators may offer new attractive and specific targets for anticancer therapy.
Collapse
Affiliation(s)
- Angelos Yfantis
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece; (A.Y.); (I.M.); (G.C.)
| | - Ilias Mylonis
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece; (A.Y.); (I.M.); (G.C.)
| | - Georgia Chachami
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece; (A.Y.); (I.M.); (G.C.)
| | - Marios Nikolaidis
- Bioinformatics Laboratory, Department of Biochemistry and Biotechnology, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece; (M.N.); (G.D.A.)
| | - Grigorios D. Amoutzias
- Bioinformatics Laboratory, Department of Biochemistry and Biotechnology, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece; (M.N.); (G.D.A.)
| | - Efrosyni Paraskeva
- Laboratory of Physiology, Faculty of Medicine, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece;
| | - George Simos
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece; (A.Y.); (I.M.); (G.C.)
- Gerald Bronfman Department of Oncology, Faculty of Medicine, McGill University, Montreal, QC H4A 3T2, Canada
- Correspondence:
| |
Collapse
|
18
|
Brown B, Ojha V, Fricke I, Al-Sheboul SA, Imarogbe C, Gravier T, Green M, Peterson L, Koutsaroff IP, Demir A, Andrieu J, Leow CY, Leow CH. Innate and Adaptive Immunity during SARS-CoV-2 Infection: Biomolecular Cellular Markers and Mechanisms. Vaccines (Basel) 2023; 11:408. [PMID: 36851285 PMCID: PMC9962967 DOI: 10.3390/vaccines11020408] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/16/2023] Open
Abstract
The coronavirus 2019 (COVID-19) pandemic was caused by a positive sense single-stranded RNA (ssRNA) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, other human coronaviruses (hCoVs) exist. Historical pandemics include smallpox and influenza, with efficacious therapeutics utilized to reduce overall disease burden through effectively targeting a competent host immune system response. The immune system is composed of primary/secondary lymphoid structures with initially eight types of immune cell types, and many other subtypes, traversing cell membranes utilizing cell signaling cascades that contribute towards clearance of pathogenic proteins. Other proteins discussed include cluster of differentiation (CD) markers, major histocompatibility complexes (MHC), pleiotropic interleukins (IL), and chemokines (CXC). The historical concepts of host immunity are the innate and adaptive immune systems. The adaptive immune system is represented by T cells, B cells, and antibodies. The innate immune system is represented by macrophages, neutrophils, dendritic cells, and the complement system. Other viruses can affect and regulate cell cycle progression for example, in cancers that include human papillomavirus (HPV: cervical carcinoma), Epstein-Barr virus (EBV: lymphoma), Hepatitis B and C (HB/HC: hepatocellular carcinoma) and human T cell Leukemia Virus-1 (T cell leukemia). Bacterial infections also increase the risk of developing cancer (e.g., Helicobacter pylori). Viral and bacterial factors can cause both morbidity and mortality alongside being transmitted within clinical and community settings through affecting a host immune response. Therefore, it is appropriate to contextualize advances in single cell sequencing in conjunction with other laboratory techniques allowing insights into immune cell characterization. These developments offer improved clarity and understanding that overlap with autoimmune conditions that could be affected by innate B cells (B1+ or marginal zone cells) or adaptive T cell responses to SARS-CoV-2 infection and other pathologies. Thus, this review starts with an introduction into host respiratory infection before examining invaluable cellular messenger proteins and then individual immune cell markers.
Collapse
Affiliation(s)
| | | | - Ingo Fricke
- Independent Immunologist and Researcher, 311995 Lamspringe, Germany
| | - Suhaila A Al-Sheboul
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
- Department of Medical Microbiology, International School of Medicine, Medipol University-Istanbul, Istanbul 34810, Turkey
| | | | - Tanya Gravier
- Independent Researcher, MPH, San Francisco, CA 94131, USA
| | | | | | | | - Ayça Demir
- Faculty of Medicine, Afyonkarahisar University, Istanbul 03030, Turkey
| | - Jonatane Andrieu
- Faculté de Médecine, Aix–Marseille University, 13005 Marseille, France
| | - Chiuan Yee Leow
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, USM, Penang 11800, Malaysia
| | - Chiuan Herng Leow
- Institute for Research in Molecular Medicine, (INFORMM), Universiti Sains Malaysia, USM, Penang 11800, Malaysia
| |
Collapse
|
19
|
Kishikawa S, Hayashi T, Takamochi K, Takekawa S, Sasahara N, Handa T, Saito T, Suzuki K, Yao T. Comprehensive clinicopathological characteristics and mucin core protein expression profiles of bronchiolar adenoma. Histopathology 2023; 82:264-275. [PMID: 36180978 DOI: 10.1111/his.14806] [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: 05/14/2022] [Revised: 09/07/2022] [Accepted: 09/25/2022] [Indexed: 12/13/2022]
Abstract
AIMS Bronchiolar adenoma (BA) is a novel entity in the 2021 WHO classification of lung tumours. The expression profile of mucin core proteins in BAs is not clear. The aim of this study was to clarify the expression profiles of mucins and to validate the clinicopathologic and molecular features of BAs. METHODS AND RESULTS We examined the clinicopathological, immunohistochemical, and molecular features of 20 BAs. Our cohort comprised 10 proximal and 10 distal BAs. Only seven of 18 patients (39%) were accurately diagnosed with BA at the time of intraoperative consultation. The frequent genetic alterations were BRAF V600E (35%) and KRAS (30%) mutations, which were mutually exclusive. The expression of MUC1, MUC4, and MUC5B was observed in all cases and that of MUC5AC and MUC6 was observed in nine (45%) and five (25%) cases, respectively. MUC4 was diffusely expressed in 18 cases. In contrast, MUC1, MUC5AC, MUC5B, and MUC6 displayed a patchy expression pattern. These expression patterns were similar to that of bronchiolar epithelium in normal lung tissue. In addition, overexpression of MUC1 and MUC4 on the entire cell surface was not observed in any of the BAs, suggesting their benign nature. CONCLUSION BA commonly exhibits diffuse MUC4 and patchy MUC1 and MUC5B expression. Its unique expression pattern is probably attributed to mucin expression specific to the bronchiolar epithelium. These results confirm the clinicopathologic and molecular characteristics of BA, including the difficulty in intraoperative frozen section diagnosis and the broad morphologic spectrum of BA derived from the bronchiolar epithelium.
Collapse
Affiliation(s)
- Satsuki Kishikawa
- Department of Human Pathology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Takuo Hayashi
- Department of Human Pathology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Kazuya Takamochi
- Department of General Thor, acic Surgery, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Shiori Takekawa
- Department of General Thor, acic Surgery, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Noriko Sasahara
- Department of Human Pathology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Takafumi Handa
- Department of Human Pathology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Tsuyoshi Saito
- Department of Human Pathology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Kenji Suzuki
- Department of General Thor, acic Surgery, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Takashi Yao
- Department of Human Pathology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | | |
Collapse
|
20
|
Wang K, Zhang T, Liu M, Wang D, Zhu H, Wang Z, Yu F, Liu Y, Zhao W. Synthesis and immunological evaluation of Mincle ligands-based antitumor vaccines. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.108065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
21
|
Živná M, Kidd KO, Barešová V, Hůlková H, Kmoch S, Bleyer AJ. Autosomal dominant tubulointerstitial kidney disease: A review. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:309-324. [PMID: 36250282 PMCID: PMC9619361 DOI: 10.1002/ajmg.c.32008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/10/2022] [Accepted: 09/29/2022] [Indexed: 01/11/2023]
Abstract
The clinical characteristics of autosomal dominant tubulointerstitial kidney disease (ADTKD) include bland urinary sediment, slowly progressive chronic kidney disease (CKD) with many patients reaching end stage renal disease (ESRD) between age 20 and 70 years, and autosomal dominant inheritance. Due to advances in genetic diagnosis, ADTKD is becoming increasingly recognized as a cause of CKD. Pathogenic variants in UMOD, MUC1, and REN are the most common causes of ADTKD. ADTKD-UMOD is also associated with hyperuricemia and gout. ADTKD-REN often presents in childhood with mild hypotension, CKD, hyperkalemia, acidosis, and anemia. ADTKD-MUC1 patients present only with CKD. This review describes the pathophysiology, genetics, clinical manifestation, and diagnosis for ADTKD, with an emphasis on genetic testing and genetic counseling suggestions for patients.
Collapse
Affiliation(s)
- Martina Živná
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Kendrah O. Kidd
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles UniversityPragueCzech Republic
- Wake Forest University School of MedicineSection on NephrologyWinston‐SalemNorth CarolinaUSA
| | - Veronika Barešová
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Helena Hůlková
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Stanislav Kmoch
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles UniversityPragueCzech Republic
- Wake Forest University School of MedicineSection on NephrologyWinston‐SalemNorth CarolinaUSA
| | - Anthony J. Bleyer
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles UniversityPragueCzech Republic
- Wake Forest University School of MedicineSection on NephrologyWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
22
|
D’Agnano V, Scialò F, Perna F, Atripaldi L, Sanduzzi S, Allocca V, Vitale M, Pastore L, Bianco A, Perrotta F. Exploring the Role of Krebs von den Lungen-6 in Severe to Critical COVID-19 Patients. Life (Basel) 2022; 12:1141. [PMID: 36013321 PMCID: PMC9409731 DOI: 10.3390/life12081141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/19/2022] [Accepted: 07/26/2022] [Indexed: 01/02/2023] Open
Abstract
COVID-19 encompasses a broad spectrum of clinical conditions caused by SARS-CoV-2 infection. More severe cases experience acute respiratory and/or multiorgan failure. KL-6 is a glycoprotein expressed mainly from type II alveolar cells with pro-fibrotic properties. Serum KL-6 concentrations have been found in patients with COVID-19. However, the relevance of KL-6 in patients with severe and critical COVID-19 has not been fully elucidated. Methods: Retrospective data from consecutive severe to critical COVID-19 patients were collected at UOC Clinica Pnuemologica “Vanvitelli”, A.O. dei Colli, Naples, Italy. The study included patients with a positive rhinopharyngeal swab for SARS-CoV-2 RNA with severe or critical COVID-19. Results: Among 87 patients, 24 had poor outcomes. The median KL-6 value in survivors was significantly lower when compared with dead or intubated patients (530 U/mL versus 1069 U/mL p < 0.001). KL-6 was correlated with body mass index (BMI) (r: 0.279, p: 0.009), lung ultrasound score (LUS) (r: 0.429, p < 0.001), Chung Score (r: 0.390, p < 0.001). KL-6 was associated with the risk of death or oro-tracheal intubation (IOT) after adjusting for gender, BMI, Charlson Index, Chung Score, and PaO2/FIO2 (OR 1.003 95% CI 1.001−1.004, p < 0.001). Serum KL-6 value of 968 has a sensitivity of 79.2%, specificity of 87.1%, PPV 70.4%, NPV 91.5%, AUC: O.85 for risk of death or IOT. Conclusions: The presented research highlights the relevance of serum KL-6 in severe to critical COVID-19 patients in predicting the risk of death or IOT.
Collapse
Affiliation(s)
- Vito D’Agnano
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.D.); (F.S.); (L.A.); (S.S.); (V.A.); (A.B.)
| | - Filippo Scialò
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.D.); (F.S.); (L.A.); (S.S.); (V.A.); (A.B.)
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy; (M.V.); (L.P.)
| | - Francesco Perna
- Section of Respiratory Diseases, Department of Clinical Medicine and Surgery, Monaldi Hospital, University of Naples Federico II, 80131 Naples, Italy;
- Section of Biochemical Clinical Laboratory, Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Lidia Atripaldi
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.D.); (F.S.); (L.A.); (S.S.); (V.A.); (A.B.)
| | - Stefano Sanduzzi
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.D.); (F.S.); (L.A.); (S.S.); (V.A.); (A.B.)
| | - Valentino Allocca
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.D.); (F.S.); (L.A.); (S.S.); (V.A.); (A.B.)
| | - Maria Vitale
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy; (M.V.); (L.P.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Lucio Pastore
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy; (M.V.); (L.P.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Andrea Bianco
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.D.); (F.S.); (L.A.); (S.S.); (V.A.); (A.B.)
| | - Fabio Perrotta
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.D.); (F.S.); (L.A.); (S.S.); (V.A.); (A.B.)
| |
Collapse
|
23
|
Mucin 1 Inhibits Ferroptosis and Sensitizes Vitamin E to Alleviate Sepsis-Induced Acute Lung Injury through GSK3 β/Keap1-Nrf2-GPX4 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2405943. [PMID: 35910848 PMCID: PMC9334047 DOI: 10.1155/2022/2405943] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 12/14/2022]
Abstract
Background Ferroptosis is a nonapoptotic form of programmed cell death, which may be related to the occurrence and development of sepsis-induced acute respiratory distress syndrome (ARDS)/acute lung injury (ALI). Mucin 1 (MUC1) is a kind of macromolecule transmembrane glycoprotein. Previous studies have shown that MUC1 could relieve ALI in sepsis and predict whether sepsis patients would develop into ARDS. However, the role of MUC1 in the ferroptosis of sepsis-induced ALI/ARDS remains unclear. Materials and Methods Sera samples from 50 patients with sepsis/septic shock were used to detect iron metabolism-related markers. Western blot and qRT-PCR were conducted to detect the expression levels of ferroptosis-related genes. Enzyme-linked immunosorbent assay (ELISA) was performed to evaluate inflammatory factors. Transmission electron microscopy (TEM) was used to assess morphological changes of cells. Results The results showed that the iron metabolism-related indicators in sepsis-induced ARDS patients changed significantly, suggesting the iron metabolism disorder. The expression levels of ferroptosis-related genes in lung tissues of sepsis had marked changes, and the lipid peroxidation levels increased, while Ferrostatin-1 (Fer-1) could reverse the above results, which confirmed the occurrence of ferroptosis. In terms of mechanism studies, inhibition of MUC1 dimerization could increase the expression level of Keap1, reduce the phosphorylation level of GSK3β, inhibit the entry of Nrf2 into the nucleus, further inhibit the expression level of GPX4, enhance the lipid peroxidation level of lung tissues, trigger ferroptosis, and aggravate lung injury. Besides, inhibiting MUC1 reversed the alleviating effect of vitamin E on ALI caused by sepsis, increased the aggregation of inflammatory cells in lung tissues, and aggravated alveolar injury and edema. Conclusions Our study was the first to explore the changes of iron metabolism indicators in ALI/ARDS of sepsis, clarify the important role of ferroptosis in ALI/ARDS induced by sepsis, and reveal the effects and specific mechanisms of MUC1 in regulating ferroptosis, as well as the sensitization on vitamin E.
Collapse
|
24
|
Ros-Lucas JA, Pascual-Figal DA, Noguera-Velasco JA, Hernández-Vicente Á, Cebreiros-López I, Arnaldos-Carrillo M, Martínez-Ardil IM, García-Vázquez E, Aparicio-Vicente M, Solana-Martínez E, Ruiz-Martínez SY, Fernández-Mula L, Andujar-Espinosa R, Fernández-Suarez B, Sánchez-Caro MD, Peñalver-Mellado C, Ruiz-López FJ. CA 15-3 prognostic biomarker in SARS-CoV-2 pneumonia. Sci Rep 2022; 12:6738. [PMID: 35469047 PMCID: PMC9037059 DOI: 10.1038/s41598-022-10726-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 04/11/2022] [Indexed: 11/28/2022] Open
Abstract
The severity of lung involvement is the main prognostic factor in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Carbohydrate antigen 15-3 (CA 15-3), a marker of lung damage and fibrosis, could help predict the prognosis of SARS-CoV-2 pneumonia. This was a retrospective and observational study. CA 15-3 was analyzed in the blood samples of patients consecutively admitted for SARS-CoV-2 pneumonia and whose blood samples were available in the biobank. Other prognostic markers were also measured (interleukin 6 [IL6], C-reactive protein [CRP], D-dimer, troponin T, and NT-ProBNP). The occurrence of in-hospital complications was registered, including death, the need for medical intensive care, and oxygen therapy at discharge. In this study, 539 patients were recruited (54.9% men, mean age: 59.6 ± 16.4 years). At admission, the mean concentrations of CA 15-3 was 20.5 ± 15.8 U/mL, and the concentration was correlated with male sex, older age, and other severity markers of coronavirus disease of 2019 (COVID-19) (IL6, CRP, D-dimer, troponine T, and NT-ProBNP). CA 15-3 levels were higher in patients who died (n = 56, 10.4%) (35.33 ± 30.45 vs. 18.8 ± 12.11, p < 0.001), who required intensive medical support (n = 78, 14.4%; 31.17 ± 27.83 vs. 18.68 ± 11.83; p < 0.001), and who were discharged with supplemental oxygen (n = 64, 13.3%; 22.65 ± 14.41 vs. 18.2 ± 11.7; p = 0.011). Elevated CA 15-3 levels (above 34.5 U/mL) were a strong predictor of a complicated in-hospital course, in terms of a higher risk of death (adjusted odds ratio [OR] 3.74, 95% confidence interval [CI]: 1.22–11.9, p = 0.022) and need for intensive care (adjusted OR 4.56, 95% CI: 1.37–15.8) after adjusting for all other risk factors. The degree of lung damage and fibrosis evaluated in terms of CA 15-3 concentrations may allow early identification of the increased risk of complications in patients with SARS-CoV-2 pneumonia.
Collapse
Affiliation(s)
- José Antonio Ros-Lucas
- Pneumology Service, Clinical University Hospital Virgen de La Arrixaca, Murcia, Spain. .,IMIB- Arrixaca, Murcia, Spain. .,Catholic University Murcia (UCAM), Murcia, Spain.
| | - Domingo Andrés Pascual-Figal
- IMIB- Arrixaca, Murcia, Spain.,Cardiology Service, Clinical University Hospital Virgen de La Arrixaca, , Murcia, Spain.,University of Murcia (UMU), Murcia, Spain.,National Center for Cardiovascular Research (CNIC), Madrid, Spain.,CIBER Cardiovascular, Murcia, Spain
| | | | | | - Iria Cebreiros-López
- Clinical Laboratory Service, Clinical University Hospital Virgen de La Arrixaca, Murcia, Spain
| | - María Arnaldos-Carrillo
- Clinical Laboratory Service, Clinical University Hospital Virgen de La Arrixaca, Murcia, Spain
| | | | - Elisa García-Vázquez
- University of Murcia (UMU), Murcia, Spain.,Internal Medicine, Infectious Diseases Section, Clinical University Hospital Virgen de La Arrixaca, Murcia, Spain
| | | | - Elena Solana-Martínez
- Pneumology Service, Clinical University Hospital Virgen de La Arrixaca, Murcia, Spain
| | | | - Laura Fernández-Mula
- Pneumology Service, Clinical University Hospital Virgen de La Arrixaca, Murcia, Spain
| | - Rubén Andujar-Espinosa
- Pneumology Service, Clinical University Hospital Virgen de La Arrixaca, Murcia, Spain.,University of Murcia (UMU), Murcia, Spain
| | | | | | | | - Francisco José Ruiz-López
- Pneumology Service, Clinical University Hospital Virgen de La Arrixaca, Murcia, Spain.,University of Murcia (UMU), Murcia, Spain
| |
Collapse
|
25
|
Lillehoj EP, Luzina IG, Atamas SP. Mammalian Neuraminidases in Immune-Mediated Diseases: Mucins and Beyond. Front Immunol 2022; 13:883079. [PMID: 35479093 PMCID: PMC9035539 DOI: 10.3389/fimmu.2022.883079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/21/2022] [Indexed: 12/28/2022] Open
Abstract
Mammalian neuraminidases (NEUs), also known as sialidases, are enzymes that cleave off the terminal neuraminic, or sialic, acid resides from the carbohydrate moieties of glycolipids and glycoproteins. A rapidly growing body of literature indicates that in addition to their metabolic functions, NEUs also regulate the activity of their glycoprotein targets. The simple post-translational modification of NEU protein targets-removal of the highly electronegative sialic acid-affects protein folding, alters protein interactions with their ligands, and exposes or covers proteolytic sites. Through such effects, NEUs regulate the downstream processes in which their glycoprotein targets participate. A major target of desialylation by NEUs are mucins (MUCs), and such post-translational modification contributes to regulation of disease processes. In this review, we focus on the regulatory roles of NEU-modified MUCs as coordinators of disease pathogenesis in fibrotic, inflammatory, infectious, and autoimmune diseases. Special attention is placed on the most abundant and best studied NEU1, and its recently discovered important target, mucin-1 (MUC1). The role of the NEU1 - MUC1 axis in disease pathogenesis is discussed, along with regulatory contributions from other MUCs and other pathophysiologically important NEU targets.
Collapse
Affiliation(s)
- Erik P. Lillehoj
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Irina G. Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Research Service, Baltimore Veterans Affairs (VA) Medical Center, Baltimore, MD, United States
| | - Sergei P. Atamas
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
26
|
Liu Y, Liu X, Liu H, Wang J, Zhang Y, Zhao W, Zhou J. DNA‐Gated N‐CDs@SiO
2
Nanoparticles‐Based Biosensor for MUC1 Detection. ChemistrySelect 2022. [DOI: 10.1002/slct.202104309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Yuhong Liu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials School of Chemistry and Materials Science Nanjing Normal University Nanjing 210023 China
- College of Life Sciences Key Laboratory of Applied Photochemistry Nanjing Normal University Nanjing 210023 China
| | - Xuan Liu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials School of Chemistry and Materials Science Nanjing Normal University Nanjing 210023 China
| | - Huaxiao Liu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials School of Chemistry and Materials Science Nanjing Normal University Nanjing 210023 China
| | - Jingzhi Wang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials School of Chemistry and Materials Science Nanjing Normal University Nanjing 210023 China
| | - Yawen Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials School of Chemistry and Materials Science Nanjing Normal University Nanjing 210023 China
| | - Wenbo Zhao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials School of Chemistry and Materials Science Nanjing Normal University Nanjing 210023 China
| | - Jiahong Zhou
- College of Life Sciences Key Laboratory of Applied Photochemistry Nanjing Normal University Nanjing 210023 China
| |
Collapse
|
27
|
Li M, Wang K, Zhang Y, Fan M, Li A, Zhou J, Yang T, Shi P, Li D, Zhang G, Chen M, Ren H. Ferroptosis-Related Genes in Bronchoalveolar Lavage Fluid Serves as Prognostic Biomarkers for Idiopathic Pulmonary Fibrosis. Front Med (Lausanne) 2021; 8:693959. [PMID: 34671612 PMCID: PMC8520927 DOI: 10.3389/fmed.2021.693959] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 08/31/2021] [Indexed: 01/02/2023] Open
Abstract
Background: Idiopathic pulmonary fibrosis (IPF) is a chronic progressive disease with unknown etiology and unfavorable prognosis. Ferroptosis is a form of regulated cell death with an iron-dependent way that is involved in the development of various diseases. Whereas the prognostic value of ferroptosis-related genes (FRGs) in IPF remains uncertain and needs to be further elucidated. Methods: The FerrDb database and the previous studies were screened to explore the FRGs. The data of patients with IPF were obtained from the GSE70866 dataset. Wilcoxon's test and univariate Cox regression analysis were applied to identify the FRGs that are differentially expressed between normal and patients with IPF and associated with prognosis. Next, a multigene signature was constructed by the least absolute shrinkage and selection operator (LASSO)-penalized Cox model in the training cohort and evaluated by using calibration and receiver operating characteristic (ROC) curves. Then, 30% of the dataset samples were randomly selected for internal validation. Finally, the potential function and pathways that might be affected by the risk score-related differently expressed genes (DEGs) were further explored. Results: A total of 183 FRGs were identified by the FerrDb database and the previous studies, and 19 of them were differentially expressed in bronchoalveolar lavage fluid (BALF) between IPF and healthy controls and associated with prognosis (p < 0.05). There were five FRGs (aconitase 1 [ACO1], neuroblastoma RAS viral (v-ras) oncogene homolog [NRAS], Ectonucleotide pyrophosphatase/phosphodiesterase 2 [ENPP2], Mucin 1 [MUC1], and ZFP36 ring finger protein [ZFP36]) identified as risk signatures and stratified patients with IPF into the two risk groups. The overall survival rate in patients with high risk was significantly lower than that in patients with low risk (p < 0.001). The calibration and ROC curve analysis confirmed the predictive capacity of this signature, and the results were further verified in the validation group. Risk score-related DEGs were found enriched in ECM-receptor interaction and focal adhesion pathways. Conclusion: The five FRGs in BALF can be used for prognostic prediction in IPF, which may contribute to improving the management strategies of IPF.
Collapse
Affiliation(s)
- Meng Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China.,Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Ke Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Yanpeng Zhang
- Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China.,Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China.,Department of Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Meng Fan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Anqi Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Jiejun Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Tian Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Puyu Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Dan Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Guangjian Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Mingwei Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Hui Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China.,Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| |
Collapse
|
28
|
Chen W, Zhang Z, Zhang S, Zhu P, Ko JKS, Yung KKL. MUC1: Structure, Function, and Clinic Application in Epithelial Cancers. Int J Mol Sci 2021; 22:ijms22126567. [PMID: 34207342 PMCID: PMC8234110 DOI: 10.3390/ijms22126567] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/18/2022] Open
Abstract
The transmembrane glycoprotein mucin 1 (MUC1) is a mucin family member that has different functions in normal and cancer cells. Owing to its structural and biochemical properties, MUC1 can act as a lubricant, moisturizer, and physical barrier in normal cells. However, in cancer cells, MUC1 often undergoes aberrant glycosylation and overexpression. It is involved in cancer invasion, metastasis, angiogenesis, and apoptosis by virtue of its participation in intracellular signaling processes and the regulation of related biomolecules. This review introduces the biological structure and different roles of MUC1 in normal and cancer cells and the regulatory mechanisms governing these roles. It also evaluates current research progress and the clinical applications of MUC1 in cancer therapy based on its characteristics.
Collapse
Affiliation(s)
- Wenqing Chen
- Division of Teaching and Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China;
| | - Zhu Zhang
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong, China; (Z.Z.); (S.Z.); (P.Z.)
| | - Shiqing Zhang
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong, China; (Z.Z.); (S.Z.); (P.Z.)
| | - Peili Zhu
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong, China; (Z.Z.); (S.Z.); (P.Z.)
| | - Joshua Ka-Shun Ko
- Division of Teaching and Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China;
- Correspondence: (J.K.-S.K.); (K.K.-L.Y.); Tel.: +852-3411-2907 (J.K.-S.K.); +852-3411-7060 (K.K.-L.Y.); Fax: +852-3411-2461 (J.K.-S.K.); +852-3411-5995 (K.K.-L.Y.)
| | - Ken Kin-Lam Yung
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong, China; (Z.Z.); (S.Z.); (P.Z.)
- Correspondence: (J.K.-S.K.); (K.K.-L.Y.); Tel.: +852-3411-2907 (J.K.-S.K.); +852-3411-7060 (K.K.-L.Y.); Fax: +852-3411-2461 (J.K.-S.K.); +852-3411-5995 (K.K.-L.Y.)
| |
Collapse
|