Distribution of ACE2, CD147, CD26, and other SARS-CoV-2 associated molecules in tissues and immune cells in health and in asthma, COPD, obesity, hypertension, and COVID-19 risk factors

Urszula Radzikowska, Mei Ding, Ge Tan, Damir Zhakparov, Yaqi Peng, Paulina Wawrzyniak, Ming Wang, Shuo Li, Hideaki Morita, Can Altunbulakli, Matthias Reiger, Avidan U Neumann, Nonhlanhla Lunjani, Claudia Traidl-Hoffmann, Kari C Nadeau, Liam O'Mahony, Cezmi Akdis, Milena Sokolowska, Urszula Radzikowska, Mei Ding, Ge Tan, Damir Zhakparov, Yaqi Peng, Paulina Wawrzyniak, Ming Wang, Shuo Li, Hideaki Morita, Can Altunbulakli, Matthias Reiger, Avidan U Neumann, Nonhlanhla Lunjani, Claudia Traidl-Hoffmann, Kari C Nadeau, Liam O'Mahony, Cezmi Akdis, Milena Sokolowska

Abstract

Background: Morbidity and mortality from COVID-19 caused by novel coronavirus SARS-CoV-2 is accelerating worldwide, and novel clinical presentations of COVID-19 are often reported. The range of human cells and tissues targeted by SARS-CoV-2, its potential receptors and associated regulating factors are still largely unknown. The aim of our study was to analyze the expression of known and potential SARS-CoV-2 receptors and related molecules in the extensive collection of primary human cells and tissues from healthy subjects of different age and from patients with risk factors and known comorbidities of COVID-19.

Methods: We performed RNA sequencing and explored available RNA-Seq databases to study gene expression and co-expression of ACE2, CD147 (BSG), and CD26 (DPP4) and their direct and indirect molecular partners in primary human bronchial epithelial cells, bronchial and skin biopsies, bronchoalveolar lavage fluid, whole blood, peripheral blood mononuclear cells (PBMCs), monocytes, neutrophils, DCs, NK cells, ILC1, ILC2, ILC3, CD4+ and CD8+ T cells, B cells, and plasmablasts. We analyzed the material from healthy children and adults, and from adults in relation to their disease or COVID-19 risk factor status.

Results: ACE2 and TMPRSS2 were coexpressed at the epithelial sites of the lung and skin, whereas CD147 (BSG), cyclophilins (PPIA andPPIB), CD26 (DPP4), and related molecules were expressed in both epithelium and in immune cells. We also observed a distinct age-related expression profile of these genes in the PBMCs and T cells from healthy children and adults. Asthma, COPD, hypertension, smoking, obesity, and male gender status generally led to the higher expression of ACE2- and CD147-related genes in the bronchial biopsy, BAL, or blood. Additionally, CD147-related genes correlated positively with age and BMI. Interestingly, we also observed higher expression of CD147-related genes in the lesional skin of patients with atopic dermatitis.

Conclusions: Our data suggest different receptor repertoire potentially involved in the SARS-CoV-2 infection at the epithelial barriers and in the immune cells. Altered expression of these receptors related to age, gender, obesity and smoking, as well as with the disease status, might contribute to COVID-19 morbidity and severity patterns.

Keywords: COPD; COVID-19; COVID-19 children; SARS receptor; asthma; hypertension; obesity.

Conflict of interest statement

Dr Radzikowska has nothing to disclose. Dr Ding has nothing to disclose. Dr Tan has nothing to disclose. Dr Zhakparov has nothing to disclose. Dr Peng has nothing to disclose. Dr Wawrzyniak has nothing to disclose. Dr Wang has nothing to disclose. Dr LI has nothing to disclose. Dr Morita has nothing to disclose. Dr Altunbulakli has nothing to disclose. Dr Reiger reports personal fees from Bencard, Germany, personal fees from Roche‐Posay, Germany, personal fees from Galderma, Germany, personal fees from Sebapharma, Germany, grants from CLR, Germany, outside the submitted work. Dr Neumann has nothing to disclose. Dr Lunjani has nothing to disclose. Dr Traidl‐Hoffmann reports grants and personal fees from Töpfer, personal fees from Sanofi, personal fees from Novartis, grants and personal fees from Sebapharma, grants and personal fees from Danone Nutricia, personal fees from La Roche Posay, personal fees from Lilly Pharma, personal fees from Mylan, outside the submitted work. Dr Nadeau reports grants and other from NIAID, other from Novartis, personal fees and other from Regeneron, grants and other from FARE, grants from EAT, other from Sanofi, other from Astellas, other from Nestle, other from BeforeBrands, other from Alladapt, other from ForTra, other from Genentech, other from AImmune Therapeutics, other from DBV Technologies, personal fees from Astrazeneca, personal fees from ImmuneWorks, personal fees from Cour Pharmaceuticals, grants from Allergenis, grants from Ukko Pharma, other from AnaptysBio, other from Adare Pharmaceuticals, other from Stallergenes‐Greer, other from NHLBI, other from NIEHS, other from EPA, other from WAO Center of Excellence, other from Iggenix, other from Probio, other from Vedanta, other from Centecor, other from Seed, from Immune Tolerance Network, from NIH, outside the submitted work. In addition, Dr Nadeau has a patent Inhibition of Allergic Reaction to Peanut Allergen using an IL‐33 Inhibitor pending, a patent Special Oral Formula for Decreasing Food Allergy Risk and Treatment for Food Allergy pending, a patent Basophil Activation Based Diagnostic Allergy Test pending, a patent Granulocyte‐based methods for detecting and monitoring immune system disorders pending, a patent Methods and Assays for Detecting and Quantifying Pure Subpopulations of White Blood Cells in Immune System Disorders pending, a patent Mixed Allergen Compositions and Methods for Using the Same pending, and a patent Microfluidic Device and Diagnostic Methods for Allergy Testing Based on Detection of Basophil Activation pending. Dr O'Mahony reports personal fees from AHL, grants from GSK, outside the submitted work. Dr Akdis reports grants from Allergopharma, Idorsia, Swiss National Science Foundation, Christine Kühne‐Center for Allergy Research and Education, European Commission's Horison's 2020 Framework Programme, Cure, Novartis Research Institutes, Astra Zeneca, Scibase, Glakso Smith‐Kline and other from Sanofi & Regeneron. Dr Sokolowska reports grants from SNSF, grants from GSK, outside the submitted work.

© 2020 EAACI and John Wiley and Sons A/S. Published by John Wiley and Sons Ltd.

Figures

Figure 1
Figure 1
ACE2 and TMPRSS2 are expressed at the barrier sites, whereas CD147, cyclophilins, CD26, and their interaction partners are present in immune cells and in epithelium. Expression of A) ACE2, B) TMPRSS2, C) BSG, D) PPIA, E) PPIB, F) S100A9, G) SLC16A7, H) SLC2A1, I) CD44, J) ITGB1, K) NFATC3, L) DPP4 genes in in vitro air liquid interface (ALI)—differentiated human primary bronchial epithelial cells (n = 10) and in ex vivo human primary bronchial biopsies (n = 5), bronchoalveolar fluid cells (n = 5), whole blood (n = 5), neutrophils (n = 4), classical monocytes (n = 4), plasmacytoid dendritic cells (n = 4), group 1, 2, and 3 innate lymphoid cells (n = 6 per group), natural‐killer cells (n = 4), naïve CD4+ T cells (n = 4), terminal effector CD4+ T cells (n = 2), naïve CD8+ T cells (n = 4), effector memory CD8+ T cells (n = 4), naïve B cells (n = 4), plasmablasts (n = 4), and skin biopsies (n = 6) from healthy adults. Data obtained from in vitro approaches are highlighted in red. Names of the proteins encoded by analyzed genes are stated in the brackets. HBECS, human bronchial epithelial cells; Bronch. biop., bronchial biopsy; BAL, bronchoalveolar fluid cells; Class. monocytes, classical monocytes; pDCs, plasmacytoid dendritic cells; ILC1, group 1 innate lymphoid cells; ILC2, group 2 innate lymphoid cells; ILC3, group 3 innate lymphoid cells; NK, natural killer cells; naïve CD4+, naïve CD4+ T cells; term. eff. CD4+, terminal effector CD4+ T cells; naïve CD8+, naïve CD8+ T cells; eff. mem. CD8+, effector memory CD8+ T cells
Figure 2
Figure 2
Distinct expression profile of ACE2‐, CD147‐, and CD26‐related genes in PBMCs and T cells of children and adults. A) Expression of ACE2‐, CD147‐, NFAT‐, and CD‐26‐related genes in the human PBMCs in healthy children aged 5‐17 months (n = 21), 12‐36 month (n = 14), 4‐16 years (n = 16) and healthy adults aged 16‐67 years (n = 19). B) Expression of ACE2‐, CD147‐, NFAT‐, and CD‐26‐related genes in the human naïve CD4+ T cells from 12 months old healthy children (n = 18) and 20‐35 years old healthy adults (n = 4). All heatmaps display normalized gene expression across the groups (rows normalization). Color‐coding represents gene families related to ACE2 (orange), CD147 (green), NF‐ATs (purple) and CD26 (yellow). MO, months old; YO, years old; PBMCs, peripheral blood mononuclear cells
Figure 3
Figure 3
Asthma, COPD, hypertension, smoking, obesity and gender are associated with differential expression of ACE2‐, CD147‐, and CD26‐related genes in immune cells and tissues. A) Differential expression of ACE2, TMPRSS2, BSG, SLC2A1, CD44, and ITGA3 genes in in vitro air liquid interface (ALI)‐differentiated human primary bronchial epithelial cells from non‐diseased controls (n = 5), asthma (n = 6) and COPD (n = 5) patients. B) Differential expression of ACE2, BSG, SLC7A5, ITGA3, ITGA6 genes in bronchial biopsies from non‐diseased controls (n = 16), patients with asthma (n = 22) and COPD (n = 3), or in comparison of smokers (n = 21) with non‐smoking individuals (n = 19). C) Differential expression of BSG, PPIA, S100A9, CD44, SLC16A7, SLC16A3, ITGA3, NFATC1, NFATC2 genes in the bronchoalveolar fluid (BAL) from the control individuals (n = 16), patients with asthma (n = 22) and COPD (n = 2), or in comparison of hypertensive (n = 9) with normotensive (n = 31) individuals; smokers (n = 20) with non‐smokers (n = 19); obese (n = 21) with non‐obese (n = 19); and males (n = 26) with females (n = 14). D) Differential expression of BSG, PPIA, S100A9, CD44, SLC16A7, ITGA6, NFATC2, LGALS3 and NOD2 genes in the whole blood of non‐diseased controls (n = 17), patients with asthma (n = 21) and COPD (n = 3), or in comparison of hypertensive (n = 9) with normotensive (n = 32) individuals; smokers (n = 21), with non‐smokers (n = 19); obese (n = 21) with non‐obese individuals (n = 20); and males (n = 27) with females (n = 14). Names of the proteins encoded by analyzed genes are stated in the brackets. *P < .05, **P < .01, ***P < .001, ****P < .0001. HBECS, human bronchial epithelial cells; Bronch. biop., bronchial biopsy; BAL, bronchoalveolar fluid cells; COPD, chronic obstructive pulmonary disease
Figure 4
Figure 4
Expression of certain CD147‐related genes correlates with BMI and age in the BAL and blood. Correlation of A) SLC16A3 expression and BMI, B) ITGA3 expression and BMI, C) LGALS3 expression and BMI, and D) CD44 expression and age in the bronchoalveolar fluid (BAL). Correlation of E) BSG expression and BMI, F) PPIA expression and BMI, G) S100A9 expression and BMI, H) CD44 expression and BMI, I) LGALS3 expression and BMI, J) SLC16A3 and age in the whole blood. Spearman correlation coefficient (r) was calculated, with the threshold of significance set to P = .05. Names of the proteins encoded by analyzed genes are stated in the brackets. BAL, bronchoalveolar fluid cells; BMI, body mass index
Figure 5
Figure 5
Unique expression profile of ACE2‐ and CD147‐related genes in lesional skin in patients with atopic dermatitis. Expression of ACE2, TMPRSS2, BSG, PPIA, PPIB, S100A9, CD44, SLC16A1, SLC16A3, SLC7A5, SLC3A2, SLC2A1, ITGA3, ITGA6, NFATC3, JUP, NME1, NOD2, SDC1, andDPP4 genes in the skin of healthy controls (n = 6) and in the lesional (n = 11), and non‐lesional (n = 11) skin of atopic dermatitis patients. Names of the proteins encoded by analyzed genes are stated in the margins. *P < .05, **P < .01, ***P < .001, ****P < .0001
Figure 6
Figure 6
Summary of the tissue and cellular expression, and models of A) ACE2, B) CD147, C) CD26, and their interaction partners. Please refer to the text for further details. CypA, Cyclophilin A; CypB, Cyclophilin B; HA, Hyaluronic acid; Gal‐3, Galectin 3; MCTs, Monocarboxylate transporters. Figure created with BioRender.com

References

    1. Zhang JJ, Dong X, Cao YY, et al. Clinical characteristics of 140 patients infected with SARS‐CoV‐2 in Wuhan, China. Allergy 2020;75:1730‐1741.
    1. Wu A, Peng Y, Huang B, et al. Genome Composition and Divergence of the Novel Coronavirus (2019‐nCoV) Originating in China. Cell Host Microbe. 2020;27(3):325‐328.
    1. Hoffmann M, Kleine‐Weber H, Schroeder S, et al. SARS‐CoV‐2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020;181(2):271‐280.e8.
    1. Yan R, Zhang Y, Li Y, Xia L, Guo Y, Zhou Q. Structural basis for the recognition of SARS‐CoV‐2 by full‐length human ACE2. Science 2020;367(6485):1444‐1448.
    1. Ziegler C, Allon S, Nyquist S, et al. SARS‐CoV‐2 receptor ACE2 is an interferon‐stimulated gene in human airway epithelial cells and is enriched in specific cell subsets across tissues. SSRN Electronic Journal. 2020.
    1. Wu C, Zheng M. Single‐cell RNA expression profiling shows that ACE2, the putative receptor for COVID‐2019, has significant expression in nasal and mounth tissue and is co‐expressed with TMPRSS2 and not co‐expressed with SLC6A19 in the tissues. PREPRINT (Version 1) available at Research Square. 12 March 2020.
    1. Chu H, Zhou J, Wong BH, et al. Middle east respiratory syndrome coronavirus efficiently infects human primary T Lymphocytes and activates the extrinsic and intrinsic apoptosis pathways. J Infect Dis. 2016;213(6):904‐914.
    1. Gu J, Gong E, Zhang B, et al. Multiple organ infection and the pathogenesis of SARS. J Exp Med. 2005;202(3):415‐424.
    1. Wang K, Chen W, Zhou Y‐S, et al.SARS‐CoV‐2 invades host cells via a novel route: CD147‐spike protein. bioRxiv. 2020:2020.2003.2014.988345.
    1. Chen Z, Mi L, Xu J, et al. Function of HAb18G/CD147 in invasion of host cells by severe acute respiratory syndrome coronavirus. J Infect Dis. 2005;191(5):755‐760.
    1. Pushkarsky T, Zybarth G, Dubrovsky L, et al. CD147 facilitates HIV‐1 infection by interacting with virus‐associated cyclophilin A. Proc Natl Acad Sci USA. 2001;98(11):6360‐6365.
    1. Watanabe A, Yoneda M, Ikeda F, Terao‐Muto Y, Sato H, Kai C. CD147/EMMPRIN acts as a functional entry receptor for measles virus on epithelial cells. J Virol. 2010;84(9):4183‐4193.
    1. Zhang MY, Zhang Y, Wu XD, et al. Disrupting CD147‐RAP2 interaction abrogates erythrocyte invasion by Plasmodium falciparum. Blood 2018;131(10):1111‐1121.
    1. Bian H, Zheng Z‐H, Wei D, et al. Meplazumab treats COVID‐19 pneumonia: an open‐labelled, concurrent controlled add‐on clinical trial. medRxiv. 2020:2020.2003.2021.20040691.
    1. Muramatsu T. Basigin (CD147), a multifunctional transmembrane glycoprotein with various binding partners. J Biochem. 2016;159(5):481‐490.
    1. Yurchenko V, Constant S, Eisenmesser E, Bukrinsky M. Cyclophilin‐CD147 interactions: a new target for anti‐inflammatory therapeutics. Clin Exp Immunol. 2010;160(3):305‐317.
    1. Hibino T, Sakaguchi M, Miyamoto S, et al. S100A9 is a novel ligand of EMMPRIN that promotes melanoma metastasis. Cancer Res. 2013;73(1):172‐183.
    1. Kato N, Yuzawa Y, Kosugi T, et al. The E‐selectin ligand basigin/CD147 is responsible for neutrophil recruitment in renal ischemia/reperfusion. J Am Soc Nephrol. 2009;20(7):1565‐1576.
    1. Seizer P, Borst O, Langer HF, et al. EMMPRIN (CD147) is a novel receptor for platelet GPVI and mediates platelet rolling via GPVI‐EMMPRIN interaction. Thromb Haemost. 2009;101(4):682‐686.
    1. Huang W, Luo WJ, Zhu P, et al. Modulation of CD147‐induced matrix metalloproteinase activity: role of CD147 N‐glycosylation. Biochem J. 2013;449(2):437‐448.
    1. Vankadari N, Wilce JA. Emerging WuHan (COVID‐19) coronavirus: glycan shield and structure prediction of spike glycoprotein and its interaction with human CD26. Emerg Microbes Infect. 2020;9(1):601‐604.
    1. Goyal P, Choi JJ, Pinheiro LC, et al. Clinical Characteristics of Covid‐19 in New York City. N Engl J Med. 2020.
    1. Wathelet MG, Orr M, Frieman MB, Baric RS. Severe acute respiratory syndrome coronavirus evades antiviral signaling: role of nsp1 and rational design of an attenuated strain. J Virol. 2007;81(21):11620‐11633.
    1. Tanaka Y, Sato Y, Sasaki T. Suppression of coronavirus replication by cyclophilin inhibitors. Viruses 2013;5(5):1250‐1260.
    1. Zhao K, Li J, He W, et al. Cyclophilin B facilitates the replication of Orf virus. Virol J. 2017;14(1):114.
    1. de Wilde AH, Zevenhoven‐Dobbe JC, van der Meer Y, et al. Cyclosporin A inhibits the replication of diverse coronaviruses. J Gen Virol. 2011;92(Pt 11):2542‐2548.
    1. Pfefferle S, Schopf J, Kogl M, et al. The SARS‐coronavirus‐host interactome: identification of cyclophilins as target for pan‐coronavirus inhibitors. PLoS Pathog. 2011;7(10):e1002331.
    1. Pushkarsky T, Yurchenko V, Laborico A, Bukrinsky M. CD147 stimulates HIV‐1 infection in a signal‐independent fashion. Biochem Biophys Res Commun. 2007;363(3):495‐499.
    1. Kirk P, Wilson MC, Heddle C, Brown MH, Barclay AN, Halestrap AP. CD147 is tightly associated with lactate transporters MCT1 and MCT4 and facilitates their cell surface expression. EMBO J. 2000;19(15):3896‐3904.
    1. Ait‐Ali N, Fridlich R, Millet‐Puel G, et al. Rod‐derived cone viability factor promotes cone survival by stimulating aerobic glycolysis. Cell 2015;161(4):817‐832.
    1. Slomiany MG, Grass GD, Robertson AD, et al. Hyaluronan, CD44, and emmprin regulate lactate efflux and membrane localization of monocarboxylate transporters in human breast carcinoma cells. Cancer Res. 2009;69(4):1293‐1301.
    1. Berditchevski F, Chang S, Bodorova J, Hemler ME. Generation of monoclonal antibodies to integrin‐associated proteins. Evidence that alpha3beta1 complexes with EMMPRIN/basigin/OX47/M6. J Biol Chem. 1997;272(46):29174‐29180.
    1. Xu D, Hemler ME. Metabolic activation‐related CD147‐CD98 complex. Mol Cell Proteomics. 2005;4(8):1061‐1071.
    1. Khunkaewla P, Schiller HB, Paster W, et al. LFA‐1‐mediated leukocyte adhesion regulated by interaction of CD43 with LFA‐1 and CD147. Mol Immunol. 2008;45(6):1703‐1711.
    1. Priglinger CS, Szober CM, Priglinger SG, et al. Galectin‐3 induces clustering of CD147 and integrin‐beta1 transmembrane glycoprotein receptors on the RPE cell surface. PLoS One 2013;8(7):e70011.
    1. Till A, Rosenstiel P, Brautigam K, et al. A role for membrane‐bound CD147 in NOD2‐mediated recognition of bacterial cytoinvasion. J Cell Sci. 2008;121(Pt 4):487‐495.
    1. Zhou S, Zhou H, Walian PJ, Jap BK. CD147 is a regulatory subunit of the gamma‐secretase complex in Alzheimer's disease amyloid beta‐peptide production. Proc Natl Acad Sci USA. 2005;102(21):7499‐7504.
    1. Yao H, Teng Y, Sun Q, et al. Important functional roles of basigin in thymocyte development and T cell activation. Int J Biol Sci. 2013;10(1):43‐52.
    1. Ruiz S, Castro‐Castro A, Bustelo XR. CD147 inhibits the nuclear factor of activated T‐cells by impairing Vav1 and Rac1 downstream signaling. J Biol Chem. 2008;283(9):5554‐5566.
    1. Muller MR, Rao A. NFAT, immunity and cancer: a transcription factor comes of age. Nat Rev Immunol. 2010;10(9):645‐656.
    1. Macian F. NFAT proteins: key regulators of T‐cell development and function. Nat Rev Immunol. 2005;5(6):472‐484.
    1. Solstad T, Bains SJ, Landskron J, et al. CD147 (Basigin/Emmprin) identifies FoxP3+CD45RO+CTLA4+‐activated human regulatory T cells. Blood 2011;118(19):5141‐5151.
    1. Kim J, Yang YL, Jeong Y, Jang YS. Middle East Respiratory Syndrome‐Coronavirus Infection into Established hDPP4‐Transgenic Mice Accelerates Lung Damage Via Activation of the Pro‐Inflammatory Response and Pulmonary Fibrosis. J Microbiol Biotechnol. 2020;30(3):427‐438.
    1. van Doremalen N, Miazgowicz KL, Milne‐Price S, et al. Host species restriction of Middle East respiratory syndrome coronavirus through its receptor, dipeptidyl peptidase 4. J Virol. 2014;88(16):9220‐9232.
    1. Kleine‐Weber H, Schroeder S, Kruger N, et al. Polymorphisms in dipeptidyl peptidase 4 reduce host cell entry of Middle East respiratory syndrome coronavirus. Emerg Microbes Infect. 2020;9(1):155‐168.
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139‐140.
    1. Zou X, Chen K, Zou J, Han P, Hao J, Han Z. Single‐cell RNA‐seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to 2019‐nCoV infection. Front Med. 2020;14(2):185‐192.
    1. Uhlen M, Fagerberg L, Hallstrom BM, et al. Tissue‐based map of the human proteome. Science. 2015;347(6220):1260419.
    1. Lu X, Zhang L, Du H, et al. SARS‐CoV‐2 Infection in Children. N Engl J Med. 2020;382(17):1663‐1665.
    1. Wu Z, McGoogan JM. Characteristics of and Important Lessons From the Coronavirus Disease 2019 (COVID‐19) Outbreak in China: Summary of a Report of 72314 Cases From the Chinese Center for Disease Control and Prevention. JAMA 2020;323(13):1239.
    1. Erkeller‐Yuksel FM, Deneys V, Yuksel B, et al. Age‐related changes in human blood lymphocyte subpopulations. J Pediatr. 1992;120(2 Pt 1):216‐222.
    1. Watanabe M, Risi R, Tuccinardi D, Baquero CJ, Manfrini S, Gnessi L. Obesity and SARS‐CoV‐ 2: a population to safeguard. Diabetes Metab Res Rev. 2020;e3325.
    1. Recalcati S. Cutaneous manifestations in COVID‐19: a first perspective. J Eur Acad Dermatol Venereol. 2020.
    1. Kursat Azkur A, Akdis M, Azkur D, et al. Immune response to SARS‐CoV‐2 and mechanisms of immunopathological changes in COVID‐19. Allergy. 2020;75:1564‐1581.
    1. Wang Q, Zhang Y, Wu L, et al. Structural and functional basis of SARS‐CoV‐2 entry by using human ACE2. Cell 2020;181(4):894‐904.e9.
    1. Sungnak W, Huang N, Becavin C, et al. SARS‐CoV‐2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat Med 2020;26(5):681‐687.
    1. Ziegler CGK, Allon SJ, Nyquist SK, et al. SARS‐CoV‐2 receptor ACE2 is an interferon‐stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell 2020.
    1. Bröer S. The role of the neutral amino acid transporter B0AT1 (SLC6A19) in Hartnup disorder and protein nutrition. IUBMB Life 2009;61(6):591‐599.
    1. Leung JM, Yang CX, Tam A, et al. ACE‐2 Expression in the Small Airway Epithelia of Smokers and COPD Patients: Implications for COVID‐19. Eur Respir J. 2020;2000688.
    1. Imai Y, Kuba K, Rao S, et al. Angiotensin‐converting enzyme 2 protects from severe acute lung failure. Nature. 2005;436(7047):112‐116.
    1. Jackson DJ, Busse WW, Bacharier LB, et al. Association of respiratory allergy, asthma and expression of the SARS‐CoV‐2 receptor, ACE2. J Allergy Clin Immunol. 2020.
    1. Peng L, Liu J, Xu W, et al. SARS‐CoV‐2 can be detected in urine, blood, anal swabs and oropharyngeal swabs specimens. J Med Virol. 2020.
    1. Bao W, Min D, Twigg SM, et al. Monocyte CD147 is induced by advanced glycation end products and high glucose concentration: possible role in diabetic complications. Am J Physiol Cell Physiol. 2010;299(5):C1212‐1219.
    1. Zhang H, Fan Q, Xie H, et al. Elevated Serum Cyclophilin B Levels Are Associated with the Prevalence and Severity of Metabolic Syndrome. Front Endocrinol. 2017;8(360).
    1. Hahn JN, Kaushik DK, Yong VW. The role of EMMPRIN in T cell biology and immunological diseases. J Leukoc Biol. 2015;98(1):33‐48.
    1. Halestrap AP. The monocarboxylate transporter family–Structure and functional characterization. IUBMB Life. 2012;64(1):1‐9.
    1. Darmoul D, Voisin T, Couvineau A, et al. Regional expression of epithelial dipeptidyl peptidase IV in the human intestines. Biochem Biophys Res Commun. 1994;203(2):1224‐1229.
    1. Li Y, Jiang D, Liang J, et al. Severe lung fibrosis requires an invasive fibroblast phenotype regulated by hyaluronan and CD44. J Exp Med. 2011;208(7):1459‐1471.
    1. Pure E, Cuff CA. A crucial role for CD44 in inflammation. Trends Mol Med. 2001;7(5):213‐221.
    1. Chen H, Hou J, Jiang X, et al. Response of memory CD8+ T cells to severe acute respiratory syndrome (SARS) coronavirus in recovered SARS patients and healthy individuals. J Immunol (Baltimore, Md: 1950). 2005;175(1):591‐598.
    1. Bouaziz JD, Duong T, Jachiet M, et al. Vascular skin symptoms in COVID‐19: a french observational study. J Eur Acad Dermatol Venereol. 2020.
    1. Wollenberg A, Flohr C, Simon D, et al. European Task Force on Atopic Dermatitis (ETFAD) statement on severe acute respiratory syndrome coronavirus 2 (SARS‐Cov‐2)‐infection and atopic dermatitis. J Eur Acad Dermatol Venereol. 2020.
    1. Seegraber M, Worm M, Werfel T, et al. Recurrent eczema herpeticum ‐ a retrospective European multicenter study evaluating the clinical characteristics of eczema herpeticum cases in atopic dermatitis patients. J Eur Acad Dermatol Venereol. 2019;34(5):1074‐1079.
    1. Vankadari N, Wilce JA. Emerging COVID‐19 coronavirus: glycan shield and structure prediction of spike glycoprotein and its interaction with human CD26. Emerg Microbes Infect. 2020;9(1):601‐604.
    1. Allan DSJ, Cerdeira AS, Ranjan A, et al. Transcriptome analysis reveals similarities between human blood CD3(‐) CD56(bright) cells and mouse CD127(+) innate lymphoid cells. Sci Rep. 2017;7(1):3501.
    1. Peters MJ, Joehanes R, Pilling LC, et al. The transcriptional landscape of age in human peripheral blood. Nat Commun. 2015;6:8570.
    1. Team CC‐R. Coronavirus Disease 2019 in Children ‐ United States, February 12‐April 2, 2020. MMWR Morb Mortal Wkly Rep. 2020;69(14):422‐426.

Source: PubMed

3
Abonnieren