1
|
Gurevich E, Landau D. Tubulointerstitial nephritis in children and adolescents. Pediatr Nephrol 2024:10.1007/s00467-024-06526-y. [PMID: 39320551 DOI: 10.1007/s00467-024-06526-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024]
Abstract
The tubulointerstitial compartment comprises most of the kidney parenchyma. Inflammation in this compartment (tubulointerstitial nephritis-TIN) can be acute and resolves if the offending factor is withdrawn or may enter a chronic process leading to irreversible kidney damage. Etiologic factors differ, including different exposures, infections, and autoimmune and genetic tendency, and the initial damage can be acute, recurrent, or permanent, determining whether the acute inflammatory process will lead to complete healing or to a chronic course of inflammation leading to fibrosis. Clinical and laboratory findings of TIN are often nonspecific, which may lead to delayed diagnosis and a poorer clinical outcome. We provide a general review of TIN, with special mention of the molecular pathophysiological mechanisms of the associated kidney damage.
Collapse
Affiliation(s)
- Evgenia Gurevich
- Pediatrics Department, Barzilai University Medical Center, Ashqelon, Israel.
- Ben Gurion University of Negev, Faculty of Health Sciences, Beer Sheva, Israel.
| | - Daniel Landau
- Department of Nephrology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
2
|
Durán M, Ariceta G, Semidey ME, Castells-Esteve C, Casal-Pardo A, Lu B, Meseguer A, Cantero-Recasens G. Renal antiporter ClC-5 regulates collagen I/IV through the β-catenin pathway and lysosomal degradation. Life Sci Alliance 2024; 7:e202302444. [PMID: 38670633 PMCID: PMC11053357 DOI: 10.26508/lsa.202302444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Mutations in Cl-/H+ antiporter ClC-5 cause Dent's disease type 1 (DD1), a rare tubulopathy that progresses to renal fibrosis and kidney failure. Here, we have used DD1 human cellular models and renal tissue from DD1 mice to unravel the role of ClC-5 in renal fibrosis. Our results in cell systems have shown that ClC-5 deletion causes an increase in collagen I (Col I) and IV (Col IV) intracellular levels by promoting their transcription through the β-catenin pathway and impairing their lysosomal-mediated degradation. Increased production of Col I/IV in ClC-5-depleted cells ends up in higher release to the extracellular medium, which may lead to renal fibrosis. Furthermore, our data have revealed that 3-mo-old mice lacking ClC-5 (Clcn5 +/- and Clcn5 -/- ) present higher renal collagen deposition and fibrosis than WT mice. Altogether, we describe a new regulatory mechanism for collagens' production and release by ClC-5, which is altered in DD1 and provides a better understanding of disease progression to renal fibrosis.
Collapse
Affiliation(s)
- Mònica Durán
- https://ror.org/01d5vx451 Renal Physiopathology Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Gema Ariceta
- https://ror.org/01d5vx451 Renal Physiopathology Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
- Pediatric Nephrology Department, Vall d'Hebron University Hospital, Barcelona, Spain
- Pediatrics Department, School of Medicine, Autonomous University of Barcelona (UAB), Bellaterra, Spain
| | - Maria E Semidey
- Department of Pathology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Carla Castells-Esteve
- https://ror.org/01d5vx451 Renal Physiopathology Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Andrea Casal-Pardo
- https://ror.org/01d5vx451 Renal Physiopathology Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Baisong Lu
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Anna Meseguer
- https://ror.org/01d5vx451 Renal Physiopathology Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
- Biochemistry and Molecular Biology Department, School of Medicine, Autonomous University of Barcelona (UAB), Bellaterra, Spain
| | - Gerard Cantero-Recasens
- https://ror.org/01d5vx451 Renal Physiopathology Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| |
Collapse
|
3
|
Ergunay T, Collino F, Bianchi G, Sedrakyan S, Perin L, Bussolati B. Extracellular vesicles in kidney development and pediatric kidney diseases. Pediatr Nephrol 2024; 39:1967-1975. [PMID: 37775581 PMCID: PMC11147923 DOI: 10.1007/s00467-023-06165-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/24/2023] [Accepted: 09/08/2023] [Indexed: 10/01/2023]
Abstract
Extracellular vesicles (EVs) are membranous cargo particles that mediate intercellular communication. They are heterogeneous in size and mechanism of release, and found in all biological fluids. Since EV content is in relation to the originating cell type and to its physiopathological conditions, EVs are under study to understand organ physiology and pathology. In addition, EV surface cargo, or corona, can be influenced by the microenvironment, leading to the concept that EV-associated molecules can represent useful biomarkers for diseases. Recent studies also focus on the use of natural, engineered, or synthetic EVs for therapeutic purposes. This review highlights the role of EVs in kidney development, pediatric kidney diseases, including inherited disorders, and kidney transplantation. Although few studies exist, they have promising results and may guide researchers in this field. Main limitations, including the influence of age on EV analyses, are also discussed.
Collapse
Affiliation(s)
- Tunahan Ergunay
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Federica Collino
- Laboratory of Translational Research in Paediatric Nephro-Urology, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy
- Paediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gaia Bianchi
- Laboratory of Translational Research in Paediatric Nephro-Urology, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy
| | - Sargis Sedrakyan
- GOFARR Laboratory, Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Laura Perin
- GOFARR Laboratory, Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.
- Molecular Biotechnology Center, University of Turin, via Nizza 52, 10126, Turin, Italy.
| |
Collapse
|
4
|
Eule CJ, Hu J, Hedges D, Jani A, Pshak T, Manley BJ, Sanchez A, Dreicer R, Myint ZW, Zakharia Y, Lam ET. Clinical and Genomic Features of Patients with Renal Cell Carcinoma and Advanced Chronic Kidney Disease: Analysis of a Multi-Institutional Database. Cancers (Basel) 2024; 16:1920. [PMID: 38791999 PMCID: PMC11119962 DOI: 10.3390/cancers16101920] [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: 04/22/2024] [Revised: 04/30/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Patients with advanced chronic kidney disease (ACKD) are at an increased risk of developing renal cell carcinoma (RCC), but molecular alterations in RCC specimens arising from ACKD and overall survival (OS) in affected patients are not well defined. PATIENTS AND METHODS Using the Oncology Research Information Exchange Network (ORIEN) Total Cancer Care® protocol, 296 consented adult patients with RCC and somatic tumor whole exome sequencing were included. Patients with ACKD were defined as those with serum creatinine ≥1.5 mg/dL prior to RCC diagnosis. RESULTS Of 296 patients with RCC, 61 met the criteria for ACKD. The most common somatic mutations in the overall cohort were in VHL (126, 42.6%), PBRM1 (102, 34.5%), and SETD2 (54, 18.2%). BAP1 had a decreased mutational frequency in RCC specimens from patients without ACKD as compared to those with ACKD (10.6% versus 1.6%), but this was not statistically significant in univariable (OR 0.14, p = 0.056) or multivariable (OR 0.15, p = 0.067) analysis. Median OS was not reached in either cohort. CONCLUSIONS Using the clinicogenomic ORIEN database, our study found lower rates of BAP1 mutations in RCC specimens from patients with ACKD, which may reflect a BAP1-independent mutational driver of RCC in patients with ACKD.
Collapse
Affiliation(s)
- Corbin J. Eule
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Junxiao Hu
- Biostatistics Core, University of Colorado Cancer Center, Aurora, CO 80045, USA
| | | | - Alkesh Jani
- Division of Nephrology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Thomas Pshak
- Division of Urology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Brandon J. Manley
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Alejandro Sanchez
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Robert Dreicer
- Division of Medical Oncology, Department of Medicine, University of Virginia Comprehensive Cancer Center, Charlottesville, VA 22908, USA
| | - Zin W. Myint
- Division of Medical Oncology, Department of Internal Medicine, Markey Cancer Center, University of Kentucky, Lexington, KY 40506, USA
| | - Yousef Zakharia
- Division of Hematology, Oncology, and Blood and Bone Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA
| | - Elaine T. Lam
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
5
|
Kopacz A, Klóska D, Cysewski D, Kraszewska I, Przepiórska K, Lenartowicz M, Łoboda A, Grochot-Przęczek A, Nowak W, Józkowicz A, Piechota-Polańczyk A. Co-administration of angiotensin II and simvastatin triggers kidney injury upon heme oxygenase-1 deficiency. Free Radic Biol Med 2023; 205:188-201. [PMID: 37302617 DOI: 10.1016/j.freeradbiomed.2023.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023]
Abstract
Kidneys are pivotal organ in iron redistribution and can be severely damaged in the course of hemolysis. In our previous studies, we observed that induction of hypertension with angiotensin II (Ang II) combined with simvastatin administration results in a high mortality rate or the appearance of signs of kidney failure in heme oxygenase-1 knockout (HO-1 KO) mice. Here, we aimed to address the mechanisms underlying this effect, focusing on heme and iron metabolism. We show that HO-1 deficiency leads to iron accumulation in the renal cortex. Higher mortality of Ang II and simvastatin-treated HO-1 KO mice coincides with increased iron accumulation and the upregulation of mucin-1 in the proximal convoluted tubules. In vitro studies showed that mucin-1 hampers heme- and iron-related oxidative stress through the sialic acid residues. In parallel, knock-down of HO-1 induces the glutathione pathway in an NRF2-depedent manner, which likely protects against heme-induced toxicity. To sum up, we showed that heme degradation during heme overload is not solely dependent on HO-1 enzymatic activity, but can be modulated by the glutathione pathway. We also identified mucin-1 as a novel redox regulator. The results suggest that hypertensive patients with less active HMOX1 alleles may be at higher risk of kidney injury after statin treatment.
Collapse
Affiliation(s)
- Aleksandra Kopacz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| | - Damian Klóska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland; Molecular Mechanisms of Diseases Laboratory, Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Dominik Cysewski
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warszawa, Poland; Clinical Research Centre, Medical University of Białystok, Białystok, Poland
| | - Izabela Kraszewska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Karolina Przepiórska
- Laboratory of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland; Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Małgorzata Lenartowicz
- Laboratory of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Anna Grochot-Przęczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Witold Nowak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Aleksandra Piechota-Polańczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
6
|
Urinary Extracellular Vesicles in Chronic Kidney Disease: From Bench to Bedside? Diagnostics (Basel) 2023; 13:diagnostics13030443. [PMID: 36766548 PMCID: PMC9913975 DOI: 10.3390/diagnostics13030443] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
Extracellular vesicles are a diverse group of particles that include exosomes, microvesicles, and apoptotic bodies and are defined by size, composition, site of origin, and density. They incorporate various bioactive molecules from their cell of origin during formation, such as soluble proteins, membrane receptors, nucleic acids (mRNAs and miRNAs), and lipids, which can then be transferred to target cells. Extracellular vesicles/exosomes have been extensively studied as a critical factor in pathophysiological processes of human diseases. Urinary extracellular vesicles could be a promising liquid biopsy for determining the pattern and/or severity of kidney histologic injury. The signature of urinary extracellular vesicles may pave the way for noninvasive methods to supplement existing testing methods for diagnosing kidney diseases. We discuss the potential role of urinary extracellular vesicles in various chronic kidney diseases in this review, highlighting open questions and discussing the potential for future research.
Collapse
|
7
|
Lausecker F, Lennon R, Randles MJ. The kidney matrisome in health, aging, and disease. Kidney Int 2022; 102:1000-1012. [PMID: 35870643 DOI: 10.1016/j.kint.2022.06.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 06/15/2022] [Accepted: 06/24/2022] [Indexed: 02/06/2023]
Abstract
Dysregulated extracellular matrix is the hallmark of fibrosis, and it has a profound impact on kidney function in disease. Furthermore, perturbation of matrix homeostasis is a feature of aging and is associated with declining kidney function. Understanding these dynamic processes, in the hope of developing therapies to combat matrix dysregulation, requires the integration of data acquired by both well-established and novel technologies. Owing to its complexity, the extracellular proteome, or matrisome, still holds many secrets and has great potential for the identification of clinical biomarkers and drug targets. The molecular resolution of matrix composition during aging and disease has been illuminated by cutting-edge mass spectrometry-based proteomics in recent years, but there remain key questions about the mechanisms that drive altered matrix composition. Basement membrane components are particularly important in the context of kidney function; and data from proteomic studies suggest that switches between basement membrane and interstitial matrix proteins are likely to contribute to organ dysfunction during aging and disease. Understanding the impact of such changes on physical properties of the matrix, and the subsequent cellular response to altered stiffness and viscoelasticity, is of critical importance. Likewise, the comparison of proteomic data sets from multiple organs is required to identify common matrix biomarkers and shared pathways for therapeutic intervention. Coupled with single-cell transcriptomics, there is the potential to identify the cellular origin of matrix changes, which could enable cell-targeted therapy. This review provides a contemporary perspective of the complex kidney matrisome and draws comparison to altered matrix in heart and liver disease.
Collapse
Affiliation(s)
- Franziska Lausecker
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester University Hospitals National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Michael J Randles
- Chester Medical School, Faculty of Medicine and Life Sciences, University of Chester, Chester, UK.
| |
Collapse
|
8
|
Takizawa K, Ueda K, Sekiguchi M, Nakano E, Nishimura T, Kajiho Y, Kanda S, Miura K, Hattori M, Hashimoto J, Hamasaki Y, Hisano M, Omori T, Okamoto T, Kitayama H, Fujita N, Kuramochi H, Ichiki T, Oka A, Harita Y. Urinary extracellular vesicles signature for diagnosis of kidney disease. iScience 2022; 25:105416. [PMID: 36439984 PMCID: PMC9684058 DOI: 10.1016/j.isci.2022.105416] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/07/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022] Open
Abstract
Congenital disorders characterized by the quantitative and qualitative reduction in the number of functional nephrons are the primary cause of chronic kidney disease (CKD) in children. We aimed to describe the alteration of urinary extracellular vesicles (uEVs) associated with decreased renal function during childhood. By nanoparticle tracking analysis and quantitative proteomics, we identified differentially expressed proteins in uEVs in bilateral renal hypoplasia, which is characterized by a congenitally reduced number of nephrons. This expression signature of uEVs reflected decreased renal function in CKD patients by congenital anomalies of the kidney and urinary tract or ciliopathy. As a proof-of-concept, we constructed a prototype ELISA system that enabled the isolation of uEVs and quantitation of expression of molecules representing the signature. The system identified decreased renal function even in its early stage. The uEVs signature could pave the way for non-invasive methods that can complement existing testing methods for diagnosing kidney diseases. Urinary extracellular vesicles (uEVs) are altered in chronic kidney disease (CKD) Characteristic expression signatures associated with childhood CKD are identified An ELISA utilizing the signature detected decreased renal function uEVs signature has potential in diagnosing kidney diseases
Collapse
Affiliation(s)
- Keiichi Takizawa
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Koji Ueda
- Project for Personalized Cancer Medicine, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Masahiro Sekiguchi
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Eiji Nakano
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Tatsuya Nishimura
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yuko Kajiho
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Shoichiro Kanda
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kenichiro Miura
- Department of Pediatric Nephrology, Tokyo Women’s Medical University, Tokyo 162-8666, Japan
| | - Motoshi Hattori
- Department of Pediatric Nephrology, Tokyo Women’s Medical University, Tokyo 162-8666, Japan
| | - Junya Hashimoto
- Department of Nephrology, Faculty of Medicine, Toho University, Tokyo 143-8541, Japan
| | - Yuko Hamasaki
- Department of Nephrology, Faculty of Medicine, Toho University, Tokyo 143-8541, Japan
| | - Masataka Hisano
- Department of Nephrology, Chiba Children’s Hospital, Chiba 266-0007, Japan
| | - Tae Omori
- Department of Pediatrics, Tokyo Metropolitan Bokutoh Hospital, Tokyo 130-8575, Japan
| | - Takayuki Okamoto
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Hokkaido 060-8648, Japan
| | - Hirotsugu Kitayama
- Department of Nephrology, Shizuoka Children’s Hospital, Shizuoka, 420-8660, Japan
| | - Naoya Fujita
- Department of Nephrology, Aichi Children’s Health and Medical Center, Aichi 474-8710, Japan
| | - Hiromi Kuramochi
- Department of Materials Engineering, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Takanori Ichiki
- Department of Materials Engineering, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Akira Oka
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yutaka Harita
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
- Corresponding author
| |
Collapse
|
9
|
Econimo L, Schaeffer C, Zeni L, Cortinovis R, Alberici F, Rampoldi L, Scolari F, Izzi C. Autosomal Dominant Tubulointerstitial Kidney Disease (ADTKD): an emerging cause of genetic chronic kidney disease. Kidney Int Rep 2022; 7:2332-2344. [DOI: 10.1016/j.ekir.2022.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/07/2022] [Accepted: 08/18/2022] [Indexed: 10/15/2022] Open
|
10
|
Abstract
Morphological transitions are typically attributed to the actions of proteins and lipids. Largely overlooked in membrane shape regulation is the glycocalyx, a pericellular membrane coat that resides on all cells in the human body. Comprised of complex sugar polymers known as glycans as well as glycosylated lipids and proteins, the glycocalyx is ideally positioned to impart forces on the plasma membrane. Large, unstructured polysaccharides and glycoproteins in the glycocalyx can generate crowding pressures strong enough to induce membrane curvature. Stress may also originate from glycan chains that convey curvature preference on asymmetrically distributed lipids, which are exploited by binding factors and infectious agents to induce morphological changes. Through such forces, the glycocalyx can have profound effects on the biogenesis of functional cell surface structures as well as the secretion of extracellular vesicles. In this review, we discuss recent evidence and examples of these mechanisms in normal health and disease.
Collapse
Affiliation(s)
- Joe Chin-Hun Kuo
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA; ,
| | - Matthew J Paszek
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA; , .,Field of Biomedical Engineering and Field of Biophysics, Cornell University, Ithaca, New York 14853, USA.,Kavli Institute at Cornell for Nanoscale Science, Ithaca, New York 14853, USA
| |
Collapse
|
11
|
Al-Bataineh MM, Kinlough CL, Mi Z, Jackson EK, Mutchler SM, Emlet DR, Kellum JA, Hughey RP. KIM-1-mediated anti-inflammatory activity is preserved by MUC1 induction in the proximal tubule during ischemia-reperfusion injury. Am J Physiol Renal Physiol 2021; 321:F135-F148. [PMID: 34151589 PMCID: PMC8424662 DOI: 10.1152/ajprenal.00127.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 11/22/2022] Open
Abstract
Cell-associated kidney injury molecule-1 (KIM-1) exerts an anti-inflammatory role following kidney injury by mediating efferocytosis and downregulating the NF-κB pathway. KIM-1 cleavage blunts its anti-inflammatory activities. We reported that mucin 1 (MUC1) is protective in a mouse model of ischemia-reperfusion injury (IRI). As both KIM-1 and MUC1 are induced in the proximal tubule (PT) during IRI and are a disintegrin and metalloprotease 17 (ADAM17) substrates, we tested the hypothesis that MUC1 protects KIM-1 activity. Muc1 knockout (KO) mice and wild-type (WT) littermates were subjected to IRI. KIM-1, MUC1, and ADAM17 levels (and signaling pathways) were assessed by immunoblot analysis. PT localization was assessed by confocal microscopy and an in situ proximity ligation assay. Findings were extended using human kidneys and urine as well as KIM-1-mediated efferocytosis assays in mouse PT cultures. In response to tubular injury in mouse and human kidneys, we observed induction and coexpression of KIM-1 and MUC1 in the PT. Compared with WT mice, Muc1 KO mice had higher urinary KIM-1 and lower kidney KIM-1. KIM-1 was apical in the PT of WT kidneys but predominately with luminal debris in Muc1 KO mice. Efferocytosis was reduced in Muc1 KO PT cultures compared with WT cultures, whereas inflammation was increased in Muc1 KO kidneys compared with WT kidneys. MUC1 was cleaved by ADAM17 in PT cultures and blocked KIM-1 shedding in Madin-Darby canine kidney cells. We conclude that KIM-1-mediated efferocytosis and thus anti-inflammatory activity during IRI is preserved in the injured kidney by MUC1 inhibition of KIM-1 shedding.NEW & NOTEWORTHY KIM-1 plays a key role in the recovery of the tubule epithelium during renal IRI by mediating efferocytosis and associated signaling that suppresses inflammation. Excessive cleavage of KIM-1 by ADAM17 provides a decoy receptor that aggravates efferocytosis and subsequent signaling. Our data from experiments in mice, patients, and cultured cells show that MUC1 is also induced during IRI and competes with KIM-1 for cleavage by ADAM17. Consequently, MUC1 protects KIM-1 anti-inflammatory activity in the damaged kidney.
Collapse
Affiliation(s)
- Mohammad M Al-Bataineh
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Carol L Kinlough
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Zaichuan Mi
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Stephanie M Mutchler
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - David R Emlet
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - John A Kellum
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Rebecca P Hughey
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
12
|
Liang Z, Zhou P, Wang Y, Zhang Y, Li D, Su X, Fan Y, Tang Y, Jiang L, Wang W. Pulmonary Adenofibroma: Clinicopathological and Genetic Analysis of 7 Cases With Literature Review. Front Oncol 2021; 11:667111. [PMID: 34350112 PMCID: PMC8328424 DOI: 10.3389/fonc.2021.667111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/21/2021] [Indexed: 02/05/2023] Open
Abstract
Introduction Pulmonary adenofibroma (PAF), characterized by biphasic differentiation composed of gland-like space lined by respiratory epithelium and stromal spindle cells, is a rare benign tumor of the lung. PAF was reported infrequently and inconsistently with diagnostic criteria and withstood higher risk of misdiagnosis as solitary fibrous tumors (SFTs) due to their morphological resemblance. In this study, we report seven cases of PAF with gene sequencing results and summarize the data of previous literature. Materials and Methods Seven cases of PAF with surgically resection samples were collected from Pathology department of West China Hospital, Sichuan University between 2009 to 2020. Immunohistochemical studies were performed in all cases and 3 cases underwent a 425-gene panel next-generation sequencing (NGS). Results Five female and two male patients were included in this study, with an average age of 51 years. All the patients were asymptomatic, and the lesion was identified on routine chest radiography. The tumor size measured by computed tomography (CT) ranged from 0.5 to 2.7 cm. Gland-like structures were mostly positive for glandular epithelium markers. The spindle cells in stroma expressed Desmin, SMA, ER and PR in 3 of 7 cases. No well-recognized molecular abnormalities can be identified by NGS in the 3 cases. To date, all the patients are alive, with no evidence of recurrence and metastasis. Conclusion PAF is a unique benign pulmonary tumor with low incidence. Biphasic morphology, IHC stains along with molecular detection is of great significance to make a clear diagnosis.
Collapse
Affiliation(s)
- Zuoyu Liang
- Department of Pathology of West China Hospital, Sichuan University, Chengdu, China
| | - Ping Zhou
- Department of Pathology of West China Hospital, Sichuan University, Chengdu, China
| | - Yuxuan Wang
- Department of Pathology of West China Hospital, Sichuan University, Chengdu, China
| | - Ying Zhang
- Department of Pathology of West China Hospital, Sichuan University, Chengdu, China
| | - Dan Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-Related Molecular Network, and Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxing Su
- Bioinformatics Department of Berry Oncology Corporation, Fuzhou, China
| | - Yu Fan
- Bioinformatics Department of Berry Oncology Corporation, Fuzhou, China
| | - Yuan Tang
- Department of Pathology of West China Hospital, Sichuan University, Chengdu, China
| | - Lili Jiang
- Department of Pathology of West China Hospital, Sichuan University, Chengdu, China
| | - Weiya Wang
- Department of Pathology of West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Jamaly S, Tsokos MG, Bhargava R, Brook OR, Hecht JL, Abdi R, Moulton VR, Satyam A, Tsokos GC. Complement activation and increased expression of Syk, mucin-1 and CaMK4 in kidneys of patients with COVID-19. Clin Immunol 2021; 229:108795. [PMID: 34252574 PMCID: PMC8270746 DOI: 10.1016/j.clim.2021.108795] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Acute and chronic kidney failure is common in hospitalized patients with COVID-19, yet the mechanism of injury and predisposing factors remain poorly understood. We investigated the role of complement activation by determining the levels of deposited complement components (C1q, C3, FH, C5b-9) and immunoglobulin along with the expression levels of the injury-associated molecules spleen tyrosine kinase (Syk), mucin-1 (MUC1) and calcium/calmodulin-dependent protein kinase IV (CaMK4) in the kidney tissues of people who succumbed to COVID-19. We report increased deposition of C1q, C3, C5b-9, total immunoglobulin, and high expression levels of Syk, MUC1 and CaMK4 in the kidneys of COVID-19 patients. Our study provides strong rationale for the expansion of trials involving the use of inhibitors of these molecules, in particular C1q, C3, Syk, MUC1 and CaMK4 to treat patients with COVID-19.
Collapse
Affiliation(s)
- Simin Jamaly
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States; Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, N-9037 Tromsø, Norway
| | - Maria G Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Rhea Bhargava
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Olga R Brook
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Jonathan L Hecht
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Vaishali R Moulton
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Abhigyan Satyam
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
14
|
Ahn HS, Kim JH, Jeong H, Yu J, Yeom J, Song SH, Kim SS, Kim IJ, Kim K. Differential Urinary Proteome Analysis for Predicting Prognosis in Type 2 Diabetes Patients with and without Renal Dysfunction. Int J Mol Sci 2020; 21:ijms21124236. [PMID: 32545899 PMCID: PMC7352871 DOI: 10.3390/ijms21124236] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/05/2020] [Accepted: 06/12/2020] [Indexed: 12/28/2022] Open
Abstract
Renal dysfunction, a major complication of type 2 diabetes, can be predicted from estimated glomerular filtration rate (eGFR) and protein markers such as albumin concentration. Urinary protein biomarkers may be used to monitor or predict patient status. Urine samples were selected from patients enrolled in the retrospective diabetic kidney disease (DKD) study, including 35 with good and 19 with poor prognosis. After removal of albumin and immunoglobulin, the remaining proteins were reduced, alkylated, digested, and analyzed qualitatively and quantitatively with a nano LC-MS platform. Each protein was identified, and its concentration normalized to that of creatinine. A prognostic model of DKD was formulated based on the adjusted quantities of each protein in the two groups. Of 1296 proteins identified in the 54 urine samples, 66 were differentially abundant in the two groups (area under the curve (AUC): p-value < 0.05), but none showed significantly better performance than albumin. To improve the predictive power by multivariate analysis, five proteins (ACP2, CTSA, GM2A, MUC1, and SPARCL1) were selected as significant by an AUC-based random forest method. The application of two classifiers—support vector machine and random forest—showed that the multivariate model performed better than univariate analysis of mucin-1 (AUC: 0.935 vs. 0.791) and albumin (AUC: 1.0 vs. 0.722). The urinary proteome can reflect kidney function directly and can predict the prognosis of patients with chronic kidney dysfunction. Classification based on five urinary proteins may better predict the prognosis of DKD patients than urinary albumin concentration or eGFR.
Collapse
Affiliation(s)
- Hee-Sung Ahn
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (H.-S.A.); (J.Y.)
| | - Jong Ho Kim
- Department of Internal Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Korea; (J.H.K.); (S.H.S.); (S.S.K.)
| | - Hwangkyo Jeong
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea;
| | - Jiyoung Yu
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (H.-S.A.); (J.Y.)
| | - Jeonghun Yeom
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Seoul 05505, Korea;
| | - Sang Heon Song
- Department of Internal Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Korea; (J.H.K.); (S.H.S.); (S.S.K.)
| | - Sang Soo Kim
- Department of Internal Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Korea; (J.H.K.); (S.H.S.); (S.S.K.)
| | - In Joo Kim
- Department of Internal Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Korea; (J.H.K.); (S.H.S.); (S.S.K.)
- Correspondence: (I.J.K.); (K.K.); Tel.: +82-51-240-7224 (I.J.K.); +82-2-1688-7575 (K.K.)
| | - Kyunggon Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (H.-S.A.); (J.Y.)
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea;
- Clinical Proteomics Core Laboratory, Convergence Medicine Research Center, Asan Medical Center, Seoul 05505, Korea
- Bio-Medical Institute of Technology, Asan Medical Center, Seoul 05505, Korea
- Correspondence: (I.J.K.); (K.K.); Tel.: +82-51-240-7224 (I.J.K.); +82-2-1688-7575 (K.K.)
| |
Collapse
|
15
|
Wang Z, Cao YJ. Adoptive Cell Therapy Targeting Neoantigens: A Frontier for Cancer Research. Front Immunol 2020; 11:176. [PMID: 32194541 PMCID: PMC7066210 DOI: 10.3389/fimmu.2020.00176] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/23/2020] [Indexed: 12/19/2022] Open
Abstract
Adoptive cell therapy (ACT) is a kind of immunotherapy in which T cells are genetically modified to express a chimeric antigen receptor (CAR) or T cell receptor (TCR), and ACT has made a great difference in treating multiple types of tumors. ACT is not perfect, and it can be followed by severe side effects, which hampers the application of ACT in clinical trials. One of the most promising methods to minimize side effects is to endow adoptive T cells with the ability to target neoantigens, which are specific to tumor cells. With the development of antigen screening technologies, more methods can be applied to discover neoantigens in cancer cells, such as whole-exome sequencing combined with mass spectrometry, neoantigen screening through an inventory-shared neoantigen peptide library, and neoantigen discovery via trogocytosis. In this review, we focus on the side effects of existing antigens and their solutions, illustrate the strategies of finding neoantigens in CAR-T and TCR-T therapies through methods reported by other researchers, and summarize the clinical behavior of these neoantigens.
Collapse
Affiliation(s)
- Zhidong Wang
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yu J Cao
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| |
Collapse
|
16
|
Milara J, Ballester B, Montero P, Escriva J, Artigues E, Alós M, Pastor-Clerigues A, Morcillo E, Cortijo J. MUC1 intracellular bioactivation mediates lung fibrosis. Thorax 2019; 75:132-142. [PMID: 31801904 DOI: 10.1136/thoraxjnl-2018-212735] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/05/2019] [Accepted: 11/15/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Serum KL6/mucin 1 (MUC1) has been identified as a potential biomarker in idiopathic pulmonary fibrosis (IPF), but the role of MUC1 intracellular bioactivation in IPF is unknown. OBJECTIVE To characterise MUC1 intracellular bioactivation in IPF. METHODS AND RESULTS The expression and phosphorylation of Thr41 and Tyr46 on the intracellular MUC1-cytoplasmic tail (CT) was increased in patients with IPF (n=22) compared with healthy subjects (n=21) and localised to fibroblasts and hyperplastic alveolar type II cells. Transforming growth factor (TGF)-β1 phosphorylated SMAD3 and thereby increased the phosphorylation of MUC1-CT Thr41 and Tyr46 in lung fibroblasts and alveolar type II cells, activating β-catenin to form a phospho-Smad3/MUC1-CT and MUC1-CT/β-catenin nuclear complex. This nuclear complex promoted alveolar epithelial type II and fibroblast to myofibroblast transitions, as well as cell senescence and fibroblast proliferation. The inhibition of MUC1-CT nuclear translocation using the inhibitor, GO-201 or silencing MUC1 by siRNA, reduced myofibroblast transition, senescence and proliferation in vitro. Bleomycin-induced lung fibrosis was reduced in mice treated with GO-201 and in MUC1-knockout mice. The profibrotic lectin, galectin-3, directly activated MUC1-CT and served as a bridge between the TGF-β receptor and the MUC1-C domain, indicating TGF-β1-dependent and TGF-β1-independent intracellular bioactivation of MUC1. CONCLUSIONS MUC1 intracellular bioactivation is enhanced in IPF and promotes fibrotic processes that could represent potential druggable targets for IPF.
Collapse
Affiliation(s)
- Javier Milara
- Pharmacy, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Beatriz Ballester
- Pharmacology, Universitat de Valencia Facultat de Medicina i Odontologia, Valencia, Spain
| | - Paula Montero
- Farmacología, Universitat de Valencia Facultat de Medicina i Odontologia, Valencia, Spain
| | - Juan Escriva
- Thoracic Surgery, Hospital Universitari i Politecnic La Fe, Valencia, Spain
| | - Enrique Artigues
- Surgery, Hospital General Universitari de València, Valencia, Spain
| | - Manuel Alós
- Pharmacy, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Alfonso Pastor-Clerigues
- Unidad de Investigación y Docencia, Consorci Hospital General Universitari de Valencia, Valencia, Spain
| | | | - Julio Cortijo
- Pharmacology, University of Valencia, Valencia, Spain
| |
Collapse
|
17
|
Abstract
Autosomal dominant tubulointerstitial kidney disease (ADTKD) is a recently defined entity that includes rare kidney diseases characterized by tubular damage and interstitial fibrosis in the absence of glomerular lesions, with inescapable progression to end-stage renal disease. These diseases have long been neglected and under-recognized, in part due to confusing and inconsistent terminology. The introduction of a gene-based, unifying terminology led to the identification of an increasing number of cases, with recent data suggesting that ADTKD is one of the more common monogenic kidney diseases after autosomal dominant polycystic kidney disease, accounting for ~5% of monogenic disorders causing chronic kidney disease. ADTKD is caused by mutations in at least five different genes, including UMOD, MUC1, REN, HNF1B and, more rarely, SEC61A1. These genes encode various proteins with renal and extra-renal functions. The mundane clinical characteristics and lack of appreciation of family history often result in a failure to diagnose ADTKD. This Primer highlights the different types of ADTKD and discusses the distinct genetic and clinical features as well as the underlying mechanisms.
Collapse
|
18
|
Fini ME, Jeong S, Gong H, Martinez-Carrasco R, Laver NMV, Hijikata M, Keicho N, Argüeso P. Membrane-associated mucins of the ocular surface: New genes, new protein functions and new biological roles in human and mouse. Prog Retin Eye Res 2019; 75:100777. [PMID: 31493487 DOI: 10.1016/j.preteyeres.2019.100777] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/26/2019] [Accepted: 08/31/2019] [Indexed: 01/17/2023]
Abstract
The mucosal glycocalyx of the ocular surface constitutes the point of interaction between the tear film and the apical epithelial cells. Membrane-associated mucins (MAMs) are the defining molecules of the glycocalyx in all mucosal epithelia. Long recognized for their biophysical properties of hydration, lubrication, anti-adhesion and repulsion, MAMs maintain the wet ocular surface, lubricate the blink, stabilize the tear film and create a physical barrier to the outside world. However, it is increasingly appreciated that MAMs also function as cell surface receptors that transduce information from the outside to the inside of the cell. A number of excellent review articles have provided perspective on the field as it has progressed since 1987, when molecular cloning of the first MAM was reported. The current article provides an update for the ocular surface, placing it into the broad context of findings made in other organ systems, and including new genes, new protein functions and new biological roles. We discuss the epithelial tissue-equivalent with mucosal differentiation, the key model system making these advances possible. In addition, we make the first systematic comparison of MAMs in human and mouse, establishing the basis for using knockout mice for investigations with the complexity of an in vivo system. Lastly, we discuss findings from human genetics/genomics, which are providing clues to new MAM roles previously unimagined. Taken together, this information allows us to generate hypotheses for the next stage of investigation to expand our knowledge of MAM function in intracellular signaling and roles unique to the ocular surface.
Collapse
Affiliation(s)
- M Elizabeth Fini
- Department of Ophthalmology, Tufts University School of Medicine, at New England Eye Center, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA.
| | - Shinwu Jeong
- USC Roski Eye Institute and Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, 1975 Zonal Ave, Los Angeles, CA, 90033, USA.
| | - Haiyan Gong
- Department of Ophthalmology, Boston University School of Medicine, 72 E Concord St, Boston, MA, 02118, USA.
| | - Rafael Martinez-Carrasco
- Department of Ophthalmology, Tufts University School of Medicine, at New England Eye Center, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA.
| | - Nora M V Laver
- Department of Ophthalmology, Tufts University School of Medicine, at New England Eye Center, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA.
| | - Minako Hijikata
- Department of Pathophysiology and Host Defense, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, 3-1-24 Matsuyama, Kiyose-shi, Tokyo, 204-8533, Japan.
| | - Naoto Keicho
- Department of Pathophysiology and Host Defense, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, 3-1-24 Matsuyama, Kiyose-shi, Tokyo, 204-8533, Japan.
| | - Pablo Argüeso
- Department of Ophthalmology, Harvard Medical School, at Schepens Eye Research Institute of Mass. Eye and Ear, 20 Staniford St, Boston, MA, 02114, USA.
| |
Collapse
|
19
|
Affiliation(s)
- Daniel P Gale
- Division of Medicine, Department of Nephrology, University College London, London, UK
| | - Robert Kleta
- Division of Medicine, Department of Nephrology, University College London, London, UK
| |
Collapse
|