1
|
Siegerist F, Kliewe F, Hammer E, Schakau P, Chi Soh JE, Weber C, Lindenmeyer M, Reichelt-Wurm S, Drenic V, Chatziantoniou C, Chadjichristos CE, Zhang Y, Simm S, Banas MC, Nauck M, Völker U, Endlich N. The role of the tricellular junction protein ILDR2 in glomerulopathies: Expression patterns and functional insights. iScience 2024; 27:111329. [PMID: 39640577 PMCID: PMC11617378 DOI: 10.1016/j.isci.2024.111329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/05/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
The tricellular tight junctions are crucial for the regulation of paracellular flux at tricellular junctions, where tricellulin (MARVELD2) and angulins (ILDR1, ILDR2, or LSR) are localized. The role of ILDR2 in podocytes, specialized epithelial cells in the kidney, is still unknown. We investigated the role of ILDR2 in glomeruli and its influence on blood filtration. Western blots, single-cell RNA sequencing (scRNA-seq), and superresolution microscopy showed a strong expression of ILDR2 in podocytes that colocalized with the podocyte-specific claudin CLDN5. Co-immunoprecipitation revealed that ILDR2 interacts with CLDN5. In glomerulopathies, induced by nephrotoxic serum and by desoxycorticosterone acetate (DOCA)-salt heminephrectomy, ILDR2 was strongly up-regulated. Furthermore, Ildr2 knockout mice exhibited glomerular hypertrophy and decreased podocyte density. However, they did not develop effacement of podocyte foot processes or proteinuria. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) proteomic analysis of isolated glomeruli showed an increase in matrix proteins, such as fibronectin and collagens. This suggests a protective role of ILDR2 in glomerulopathies.
Collapse
Affiliation(s)
- Florian Siegerist
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Felix Kliewe
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Elke Hammer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Paul Schakau
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Joanne Ern Chi Soh
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Claudia Weber
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Maja Lindenmeyer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Vedran Drenic
- NIPOKA GmbH, Center of High-End Imaging, Greifswald, Germany
| | - Christos Chatziantoniou
- National Institute for Health and Medical Research (INSERM), Unité Mixte de Recherche (UMR)-S1155, Tenon Hospital, Sorbonne Universités, Paris, France
| | - Christos E. Chadjichristos
- National Institute for Health and Medical Research (INSERM), Unité Mixte de Recherche (UMR)-S1155, Tenon Hospital, Sorbonne Universités, Paris, France
| | - Yiying Zhang
- Vagelos College of Physicians and Surgeons, Medical School of Columbia University, New York, NY, USA
| | - Stefan Simm
- Institute of Bioinformatics, University Medicine Greifswald, Greifswald, Germany
- Institute of Bioanalysis, Coburg University of Applied Sciences, Coburg, Germany
| | - Miriam C. Banas
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Matthias Nauck
- Institute for Clinical Chemistry and Laboratory Medicine, University Medicine, Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
- NIPOKA GmbH, Center of High-End Imaging, Greifswald, Germany
| |
Collapse
|
2
|
Gerlach GF, Imseis ZH, Cooper SL, Santos AN, O’Brien LL. Mapping of the podocin proximity-dependent proteome reveals novel components of the kidney podocyte foot process. Front Cell Dev Biol 2023; 11:1195037. [PMID: 37325559 PMCID: PMC10262054 DOI: 10.3389/fcell.2023.1195037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/03/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction: The unique architecture of glomerular podocytes is integral to kidney filtration. Interdigitating foot processes extend from the podocyte cell body, wrap around fenestrated capillaries, and form specialized junctional complexes termed slit diaphragms to create a molecular sieve. However, the full complement of proteins which maintain foot process integrity, and how this localized proteome changes with disease, remain to be elucidated. Methods: Proximity-dependent biotin identification (BioID) enables the identification of spatially localized proteomes. To this end, we developed a novel in vivo BioID knock-in mouse model. We utilized the slit diaphragm protein podocin (Nphs2) to create a podocin-BioID fusion. Podocin-BioID localizes to the slit diaphragm, and biotin injection leads to podocyte-specific protein biotinylation. We isolated the biotinylated proteins and performed mass spectrometry to identify proximal interactors. Results and Discussion: Gene ontology analysis of 54 proteins specifically enriched in our podocin-BioID sample revealed 'cell junctions,' 'actin binding,' and 'cytoskeleton organization' as top terms. Known foot process components were identified, and we further uncovered two novel proteins: the tricellular junctional protein Ildr2 and the CDC42 and N-WASP interactor Fnbp1l. We confirmed that Ildr2 and Fnbp1l are expressed by podocytes and partially colocalize with podocin. Finally, we investigated how this proteome changes with age and uncovered a significant increase in Ildr2. This was confirmed by immunofluorescence on human kidney samples and suggests altered junctional composition may preserve podocyte integrity. Together, these assays have led to new insights into podocyte biology and support the efficacy of utilizing BioID in vivo to interrogate spatially localized proteomes in health, aging, and disease.
Collapse
Affiliation(s)
| | | | | | | | - Lori L. O’Brien
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
3
|
Palmer DS, Zhou W, Abbott L, Wigdor EM, Baya N, Churchhouse C, Seed C, Poterba T, King D, Kanai M, Bloemendal A, Neale BM. Analysis of genetic dominance in the UK Biobank. Science 2023; 379:1341-1348. [PMID: 36996212 PMCID: PMC10345642 DOI: 10.1126/science.abn8455] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/15/2023] [Indexed: 04/01/2023]
Abstract
Classical statistical genetics theory defines dominance as any deviation from a purely additive, or dosage, effect of a genotype on a trait, which is known as the dominance deviation. Dominance is well documented in plant and animal breeding. Outside of rare monogenic traits, however, evidence in humans is limited. We systematically examined common genetic variation across 1060 traits in a large population cohort (UK Biobank, N = 361,194 samples analyzed) for evidence of dominance effects. We then developed a computationally efficient method to rapidly assess the aggregate contribution of dominance deviations to heritability. Lastly, observing that dominance associations are inherently less correlated between sites at a genomic locus than their additive counterparts, we explored whether they may be leveraged to identify causal variants more confidently.
Collapse
Affiliation(s)
- Duncan S. Palmer
- Analytical and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Wei Zhou
- Analytical and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Liam Abbott
- Analytical and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Nikolas Baya
- Analytical and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Claire Churchhouse
- Analytical and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Cotton Seed
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Tim Poterba
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniel King
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Masahiro Kanai
- Analytical and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alex Bloemendal
- Analytical and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Benjamin M. Neale
- Analytical and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
4
|
Kim KS, Koo HY, Bok J. Alternative splicing in shaping the molecular landscape of the cochlea. Front Cell Dev Biol 2023; 11:1143428. [PMID: 36936679 PMCID: PMC10018040 DOI: 10.3389/fcell.2023.1143428] [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: 01/13/2023] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
The cochlea is a complex organ comprising diverse cell types with highly specialized morphology and function. Until now, the molecular underpinnings of its specializations have mostly been studied from a transcriptional perspective, but accumulating evidence points to post-transcriptional regulation as a major source of molecular diversity. Alternative splicing is one of the most prevalent and well-characterized post-transcriptional regulatory mechanisms. Many molecules important for hearing, such as cadherin 23 or harmonin, undergo alternative splicing to produce functionally distinct isoforms. Some isoforms are expressed specifically in the cochlea, while some show differential expression across the various cochlear cell types and anatomical regions. Clinical phenotypes that arise from mutations affecting specific splice variants testify to the functional relevance of these isoforms. All these clues point to an essential role for alternative splicing in shaping the unique molecular landscape of the cochlea. Although the regulatory mechanisms controlling alternative splicing in the cochlea are poorly characterized, there are animal models with defective splicing regulators that demonstrate the importance of RNA-binding proteins in maintaining cochlear function and cell survival. Recent technological breakthroughs offer exciting prospects for overcoming some of the long-standing hurdles that have complicated the analysis of alternative splicing in the cochlea. Efforts toward this end will help clarify how the remarkable diversity of the cochlear transcriptome is both established and maintained.
Collapse
Affiliation(s)
- Kwan Soo Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hei Yeun Koo
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
- *Correspondence: Jinwoong Bok,
| |
Collapse
|
5
|
Wang L, Zhai R, Song G, Wang Y. Analyses of the expression and prognosis of ILDR1 in human gastric cancer. Heliyon 2022; 8:e10253. [PMID: 36091962 PMCID: PMC9450077 DOI: 10.1016/j.heliyon.2022.e10253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/20/2022] [Accepted: 08/05/2022] [Indexed: 11/12/2022] Open
Abstract
The worldwide mortality rate of gastric cancer worldwide remains high. Immunoglobulin-like domain containing receptor 1 (ILDR1) belongs to an evolutionarily conserved protein family, and little is known about this gene in gastric cancer. In this paper, we analyzed the expression of ILDR1 and its relationship with clinical outcomes in gastric cancer using publicly available databases. ONCOMINE, GEPIA2, UALCAN, Kaplan–Meier Plotter, cBioPortal, GeneMANIA and LinkedOmics databases were used to analyze the expression, prognostic values, mutations and functional networks of ILDR1 in gastric cancer. We observed that ILDR1 was overexpressed in gastric cancer than in normal tissues. ILDR1 expression was significantly higher in patients with gastric cancer than in normal controls during subgroup analysis based on cancer stage, patient’s race, sex, age, tumor grade, H. pylori infection, histological subtype, and nodal metastasis status. Survival analysis showed that upregulation of ILDR1 expression was significantly associated with poor prognosis. Genomic alterations in ILDR1 were analyzed using cBioPortal, protein–protein interaction (PPI) networks were constructed using GeneMANIA and the co-expressed genes, gene ontology, and pathways of ILDR1 were determined using the LinkedOmics web tool. ILDR1 showed significant differences in expression between gastric cancer and normal tissues and, thus, may be a promising prognostic biomarker for gastric cancer.
Collapse
|
6
|
Chandra R, Aryal DK, Douros JD, Shahid R, Davis SJ, Campbell JE, Ilkayeya O, White PJ, Rodriguez R, Newgard CB, Wetsel WC, Liddle RA. Ildr1 gene deletion protects against diet-induced obesity and hyperglycemia. PLoS One 2022; 17:e0270329. [PMID: 35749484 PMCID: PMC9231709 DOI: 10.1371/journal.pone.0270329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/09/2022] [Indexed: 11/24/2022] Open
Abstract
Objective Immunoglobulin-like Domain-Containing Receptor 1 (ILDR1) is expressed on nutrient sensing cholecystokinin-positive enteroendocrine cells of the gastrointestinal tract and it has the unique ability to induce fat-mediated CCK secretion. However, the role of ILDR1 in CCK-mediated regulation of satiety is unknown. In this study, we examined the effects of ILDR1 on food intake and metabolic activity using mice with genetically-deleted Ildr1. Methods The expression of ILDR1 in murine tissues and the measurement of adipocyte cell size were evaluated by light and fluorescence confocal microscopy. The effects of Ildr1 deletion on mouse metabolism were quantitated using CLAMS chambers and by targeted metabolomics assays of multiple tissues. Hormone levels were measured by ELISA. The effects of Ildr1 gene deletion on glucose and insulin levels were determined using in vivo oral glucose tolerance, meal tolerance, and insulin tolerance tests, as well as ex vivo islet perifusion. Results ILDR1 is expressed in a wide range of tissues. Analysis of metabolic data revealed that although Ildr1-/- mice consumed more food than wild-type littermates, they gained less weight on a high fat diet and exhibited increased metabolic activity. Adipocytes in Ildr1-/- mice were significantly smaller than in wild-type mice fed either low or high fat diets. ILDR1 was expressed in both alpha and beta cells of pancreatic islets. Based on oral glucose and mixed meal tolerance tests, Ildr1-/- mice were more effective at lowering post-prandial glucose levels, had improved insulin sensitivity, and glucose-regulated insulin secretion was enhanced in mice lacking ILDR1. Conclusion Ildr1 loss significantly modified metabolic activity in these mutant mice. While Ildr1 gene deletion increased high fat food intake, it reduced weight gain and improved glucose tolerance. These findings indicate that ILDR1 modulates metabolic responses to feeding in mice.
Collapse
Affiliation(s)
- Rashmi Chandra
- Department of Medicine, Division of Gastroenterology, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail: (RC); (RAL)
| | - Dipendra K. Aryal
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Jonathan D. Douros
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Rafiq Shahid
- Department of Medicine, Division of Gastroenterology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Supriya J. Davis
- Department of Medicine, Division of Gastroenterology, Duke University Medical Center, Durham, North Carolina, United States of America
- Swarthmore College, Swarthmore, Pennsylvania, United States of America
| | - Jonathan E. Campbell
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Olga Ilkayeya
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, North Carolina, United States of America
| | - Phillip J. White
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, North Carolina, United States of America
| | - Ramona Rodriguez
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina, United States of America
- Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University, Durham, North Carolina, United States of America
| | - Christopher B. Newgard
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, North Carolina, United States of America
| | - William C. Wetsel
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina, United States of America
- Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University, Durham, North Carolina, United States of America
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Rodger A. Liddle
- Department of Medicine, Division of Gastroenterology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Veterans Affairs Medical Center, Durham, North Carolina, United States of America
- * E-mail: (RC); (RAL)
| |
Collapse
|
7
|
ILDR1 promotes influenza A virus replication through binding to PLSCR1. Sci Rep 2022; 12:8515. [PMID: 35595813 PMCID: PMC9122930 DOI: 10.1038/s41598-022-12598-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 05/05/2022] [Indexed: 12/13/2022] Open
Abstract
As a natural antiviral regulator, phospholipid scramblase 1 (PLSCR1) has been shown to inhibit influenza virus replication in infected cells through interacting with NP of influenza A virus (IAV). But its antiviral function as well as the underlying regulatory mechanism has not been examined in vivo. In the present work, we show that PLSCR1 expression is decreased in H1N1 SIV-infected mice, and Plscr1−/− mice are more susceptible to H1N1 SIV infection. By performing yeast two-hybrid screening, we identified immunoglobulin-like domain-containing receptor 1 (ILDR1) as a novel PLSCR1-binding partner. ILDR1 is highly expressed in the lungs, and its expression level is increased after virus infection. Interestingly, ILDR1 could not directly interact with virus NP protein, but could combine with PLSCR1 competitively. Our data indicates that there is a previously unidentified PLSCR1-ILDR1-NP regulatory pathway playing a vital role in limiting IAV infection, which provides novel insights into IAV-host interactions.
Collapse
|
8
|
Cao W, Lei S, Zeng Z, Xiao C, Sun B, Xie P, Li Y, Luo D, Yu W. Transformer 2 alpha homolog is a downstream gene of hypoxia-inducible factor 1 subunit alpha and is involved in the progression of pancreatic cancer. Bioengineered 2022; 13:13238-13251. [PMID: 35635094 PMCID: PMC9275993 DOI: 10.1080/21655979.2022.2079243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 11/06/2022] Open
Abstract
Intratumoral hypoxia is a common feature of pancreatic cancer (PC) and also plays a role in its progression. However, hypoxia-regulated signatures in PC are still not completely understood. This study aimed to identify core hypoxia-associated genes and determine their underlying molecular mechanisms in PC cells. Transformer 2 alpha homolog (TRA2A) was found to be an important hypoxia-associated gene, which was upregulated in PC tissues and in PC cells cultured under hypoxia. High TRA2A expression was associated with advanced stage, poor differentiation, and lymph node metastasis. Under normoxic and hypoxic conditions, knockdown of TRA2A both markedly suppressed PC cell proliferation and motility in vitro and in vivo, as well as activation of the AKT pathway. Hypoxia-inducible factor 1 subunit alpha (HIF1α) upregulated the transcription of TRA2A by directly binding to its promoter. TRA2A showed a co-expression relationship with HIF1α in PC tissues. Overexpression of TRA2A alleviated the pro-inhibitive functions of HIF1α-inhibition on PC cell proliferation and motility under hypoxia. In conclusion, TRA2A is a crucial downstream gene of HIF1α that accelerates the proliferation and motility of PC cells. TRA2A may be a novel and practical molecular target for investigating the hypoxic response of PC cells.Abbreviations: TRA2A, transformer 2A protein; PC, pancreatic cancer; HIF1α, hypoxia-inducible factor 1-alpha; GEO, Gene Expression Omnibus; IHC, immunohistochemical staining.
Collapse
Affiliation(s)
- Wenpeng Cao
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Shan Lei
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhirui Zeng
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Chaolun Xiao
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Baofei Sun
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Peng Xie
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical, Guizhou Medical University, Guiyang, Guizhou, China
- Key Laboratory of Medical Molecular Biology, School of Basic Medical, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yumei Li
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Daopeng Luo
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Wenfeng Yu
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical, Guizhou Medical University, Guiyang, Guizhou, China
- Key Laboratory of Medical Molecular Biology, School of Basic Medical, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
9
|
Liu G, Liu X, Yang Y. Comparative transcriptome analysis of miRNA in hydronephrosis male children caused by ureteropelvic junction obstruction with or without renal functional injury. PeerJ 2022; 10:e12962. [PMID: 35237468 PMCID: PMC8884061 DOI: 10.7717/peerj.12962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 01/27/2022] [Indexed: 01/11/2023] Open
Abstract
MicroRNAs (miRNAs or miRs) are non-coding RNAs that contribute to pathological processes of various kidney diseases. Renal function injury represents a final common outcome of congenital obstructive nephropathy and has attracted a great deal of attention. However the molecular mechanisms are still not fully established. In this study, we compared transcriptome sequencing data of miRNAs of renal tissues from congenital hydronephrosis children with or without renal functional injury, in order to better understand whether microRNAs could play important roles in renal functional injury after ureteropelvic junction obstruction. A total of 22 microRNAs with significant changes in their expression were identified. Five microRNAs were up-regulated and 17 microRNAs were down-regulated in the renal tissues of the hydronephrosis patients with renal function injury compared with those without renal function injury. MicroRNA target genes were predicted by three major online miRNA target prediction algorithms, and all these mRNAs were used to perform the gene ontology analysis and Kyoto Encyclopedia of Gene and Genomes pathway analysis. Then, twelve candidate human and rat homologous miRNAs were selected for validation using RT-qPCR in vitro and in vivo; only miR-187-3p had a trend identical to that detected by the sequencing results among the human tissues, in vivo and in vitro experimental models. In addition, we found that the change of miR-187-3p in vivo was consistent with results in vitro models and showed a decrease trend in time dependence. These results provided a detailed catalog of candidate miRNAs to investigate their regulatory role in renal injury of congenital hydronephrosis, indicating that they may serve as candidate biomarkers or therapeutic targets in the future.
Collapse
Affiliation(s)
- Ge Liu
- Urology Division, Pediatric Surgery Department, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Xin Liu
- Urology Division, Pediatric Surgery Department, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Yi Yang
- Urology Division, Pediatric Surgery Department, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of China
| |
Collapse
|
10
|
Jiang XW, Yu WH, Wang Y, Xiong ZL, Ma XL, Zhou C, Huo MH. Acetyl-11-keto-beta-boswellic acid promotes sciatic nerve repair after injury: molecular mechanism. Neural Regen Res 2022; 17:2778-2784. [PMID: 35662229 PMCID: PMC9165397 DOI: 10.4103/1673-5374.339494] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Previous studies showed that acetyl-11-keto-beta-boswellic acid (AKBA), the active ingredient in the natural Chinese medicine Boswellia, can stimulate sciatic nerve injury repair via promoting Schwann cell proliferation. However, the underlying molecular mechanism remains poorly understood. In this study, we performed genomic sequencing in a rat model of sciatic nerve crush injury after gastric AKBA administration for 30 days. We found that the phagosome pathway was related to AKBA treatment, and brain-derived neurotrophic factor expression in the neurotrophic factor signaling pathway was also highly up-regulated. We further investigated gene and protein expression changes in the phagosome pathway and neurotrophic factor signaling pathway. Myeloperoxidase expression in the phagosome pathway was markedly decreased, and brain-derived neurotrophic factor, nerve growth factor, and nerve growth factor receptor expression levels in the neurotrophic factor signaling pathway were greatly increased. Additionally, expression levels of the inflammatory factors CD68, interleukin-1β, pro-interleukin-1β, and tumor necrosis factor-α were also decreased. Myelin basic protein- and β3-tubulin-positive expression as well as the axon diameter-to-total nerve diameter ratio in the injured sciatic nerve were also increased. These findings suggest that, at the molecular level, AKBA can increase neurotrophic factor expression through inhibiting myeloperoxidase expression and reducing inflammatory reactions, which could promote myelin sheath and axon regeneration in the injured sciatic nerve.
Collapse
|
11
|
Alternative Splicing of Cdh23 Exon 68 Is Regulated by RBM24, RBM38, and PTBP1. Neural Plast 2020; 2020:8898811. [PMID: 32774357 PMCID: PMC7397384 DOI: 10.1155/2020/8898811] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/19/2020] [Accepted: 07/01/2020] [Indexed: 01/08/2023] Open
Abstract
Alternative splicing plays a pivotal role in modulating the function of eukaryotic proteins. In the inner ear, many genes undergo alternative splicing, and errors in this process lead to hearing loss. Cadherin 23 (CDH23) forms part of the so-called tip links, which are indispensable for mechanoelectrical transduction (MET) in the hair cells. Cdh23 gene contains 69 exons, and exon 68 is subjected to alternative splicing. Exon 68 of the Cdh23 gene is spliced into its mRNA only in a few cell types including hair cells. The mechanism responsible for the alternative splicing of Cdh23 exon 68 remains elusive. In the present work, we performed a cell-based screening to look for splicing factors that regulate the splicing of Cdh23 exon 68. RBM24 and RBM38 were identified to enhance the inclusion of Cdh23 exon 68. The splicing of Cdh23 exon 68 is affected in Rbm24 knockdown or knockout cells. Moreover, we also found that PTBP1 inhibits the inclusion of Cdh23 exon 68. Taken together, we show here that alternative splicing of Cdh23 exon 68 is regulated by RBM24, RBM38, and PTBP1.
Collapse
|
12
|
Liu Q, Zhu L, Liu X, Zheng J, Liu Y, Ruan X, Cao S, Cai H, Li Z, Xue Y. TRA2A-induced upregulation of LINC00662 regulates blood-brain barrier permeability by affecting ELK4 mRNA stability in Alzheimer's microenvironment. RNA Biol 2020; 17:1293-1308. [PMID: 32372707 DOI: 10.1080/15476286.2020.1756055] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The blood-brain barrier (BBB) plays a pivotal role in the maintenance and regulation of the neural microenvironment. The BBB breakdown is a pathological change in early Alzheimer's disease (AD). RNA-binding proteins (RBPs) and long non-coding RNAs (lncRNAs) are involved in the regulation of BBB permeability. Our study demonstrates the role of TRA2A/LINC00662/ELK4 axis in regulating BBB permeability in AD microenvironment. In Aβ1-42-incubated microvascular endothelial cells (ECs) of the BBB model in vitro, TRA2A and LINC00662 were enriched. TRA2A increased the stability of LINC00662 by binding with it. The knockdown of either TRA2A or LINC00662 decreased BBB permeability due to increased expression of tight junction-related proteins. ELK4 was less expressed in the BBB model in AD microenvironment in vitro. LINC00662 mediated the degradation of ELK4 mRNA by SMD pathway. Downregulation of ELK4 increased BBB permeability by increasing the tight junction-related protein expression.TRA2A/LINC00662/ELK4 axis plays a crucial role in the regulation of BBB permeability in AD microenvironment, which may provide a novel target for the therapy of AD.
Collapse
Affiliation(s)
- Qianshuo Liu
- Department of Neurobiology, School of Life Sciences, China Medical University , Shenyang, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University , Shenyang, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University , Shenyang, People's Republic of China
| | - Lu Zhu
- Department of Neurobiology, School of Life Sciences, China Medical University , Shenyang, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University , Shenyang, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University , Shenyang, People's Republic of China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University , Shenyang, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease , Shenyang, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province , Shenyang, People's Republic of China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University , Shenyang, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease , Shenyang, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province , Shenyang, People's Republic of China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University , Shenyang, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease , Shenyang, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province , Shenyang, People's Republic of China
| | - Xuelei Ruan
- Department of Neurobiology, School of Life Sciences, China Medical University , Shenyang, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University , Shenyang, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University , Shenyang, People's Republic of China
| | - Shuo Cao
- Department of Neurobiology, School of Life Sciences, China Medical University , Shenyang, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University , Shenyang, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University , Shenyang, People's Republic of China
| | - Heng Cai
- Department of Neurosurgery, Shengjing Hospital of China Medical University , Shenyang, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease , Shenyang, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province , Shenyang, People's Republic of China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University , Shenyang, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease , Shenyang, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province , Shenyang, People's Republic of China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University , Shenyang, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University , Shenyang, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University , Shenyang, People's Republic of China
| |
Collapse
|
13
|
Huetter J, Gritzan U, Gutcher I, Doecke WD, Luetke-Eversloh MV, Golfier S, Roider HG, Frisk AL, Hunter J, Pow A, Drake A, Levine Z, Levy O, Azulay M, Barbiro I, Cojocaru G, Vaknin I, Kreft B, Roese L. Characterization of BAY 1905254, an Immune Checkpoint Inhibitor Targeting the Immunoglobulin-Like Domain Containing Receptor 2 (ILDR2). Cancer Immunol Res 2020; 8:895-911. [DOI: 10.1158/2326-6066.cir-19-0321] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 11/15/2019] [Accepted: 04/13/2020] [Indexed: 11/16/2022]
|
14
|
González-Mariscal L, Miranda J, Gallego-Gutiérrez H, Cano-Cortina M, Amaya E. Relationship between apical junction proteins, gene expression and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183278. [PMID: 32240623 DOI: 10.1016/j.bbamem.2020.183278] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/09/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022]
Abstract
The apical junctional complex (AJC) is a cell-cell adhesion system present at the upper portion of the lateral membrane of epithelial cells integrated by the tight junction (TJ) and the adherens junction (AJ). This complex is crucial to initiate and stabilize cell-cell adhesion, to regulate the paracellular transit of ions and molecules and to maintain cell polarity. Moreover, we now consider the AJC as a hub of signal transduction that regulates cell-cell adhesion, gene transcription and cell proliferation and differentiation. The molecular components of the AJC are multiple and diverse and depending on the cellular context some of the proteins in this complex act as tumor suppressors or as promoters of cell transformation, migration and metastasis outgrowth. Here, we describe these new roles played by TJ and AJ proteins and their potential use in cancer diagnostics and as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Lorenza González-Mariscal
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico.
| | - Jael Miranda
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Helios Gallego-Gutiérrez
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Misael Cano-Cortina
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Elida Amaya
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| |
Collapse
|
15
|
Huang K, Zhou L, Alanis K, Hou J, Baker LA. Imaging effects of hyperosmolality on individual tricellular junctions. Chem Sci 2020; 11:1307-1315. [PMID: 33209250 PMCID: PMC7643560 DOI: 10.1039/c9sc05114g] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 12/10/2019] [Indexed: 01/26/2023] Open
Abstract
The use of hyperosmolar agents (osmotherapy) has been a major treatment for intracranial hypertension, which occurs frequently in brain diseases or trauma. However, side-effects of osmotherapy on the brain, especially on the blood-brain barrier (BBB) are still not fully understood. Hyperosmolar conditions, termed hyperosmolality here, are known to transiently disrupt the tight junctions (TJs) at the endothelium of the BBB resulting in loss of BBB function. Present techniques for evaluation of BBB transport typically reveal aggregated responses from the entirety of BBB transport components, with little or no opportunity to evaluate heterogeneity present in the system. In this study, we utilized potentiometric-scanning ion conductance microscopy (P-SICM) to acquire nanometer-scale conductance maps of Madin-Darby Canine Kidney strain II (MDCKII) cells under hyperosmolality, from which two types of TJs, bicellular tight junctions (bTJs) and tricellular tight junctions (tTJs), can be visualized and differentiated. We discovered that hyperosmolality leads to increased conductance at tTJs without significant alteration in conductance at bTJs. To quantify this effect, an automated computer vision algorithm was designed to extract and calculate conductance components at both tTJs and bTJs. Additionally, lowering Ca2+ concentration in the bath facilitates tTJ disruption under hyperosmolality. Strengthening tTJ structure by overexpressing immunoglobulin-like domain-containing receptor 1 (ILDR1) protein abrogates the effect of hyperosmolality. We posit that osmotic stress physically disrupts tTJ structure, as evidenced by super-resolution microscopy. Findings from this study not only provide a high-resolution view of TJ structure and function, but also can inform current osmotherapy and drug delivery strategies for brain diseases.
Collapse
Affiliation(s)
- Kaixiang Huang
- Department of Chemistry , Indiana University , 800 E. Kirkwood Avenue , Bloomington , Indiana 47405 , USA .
| | - Lushan Zhou
- Department of Chemistry , Indiana University , 800 E. Kirkwood Avenue , Bloomington , Indiana 47405 , USA .
| | - Kristen Alanis
- Department of Chemistry , Indiana University , 800 E. Kirkwood Avenue , Bloomington , Indiana 47405 , USA .
| | - Jianghui Hou
- Renal Division , Washington University Medical School , 660 S. Euclid Avenue , St. Louis , Missouri 63110 , USA
| | - Lane A Baker
- Department of Chemistry , Indiana University , 800 E. Kirkwood Avenue , Bloomington , Indiana 47405 , USA .
| |
Collapse
|
16
|
Identification of Binding Partners of Deafness-Related Protein PDZD7. Neural Plast 2018; 2018:2062346. [PMID: 29796015 PMCID: PMC5896214 DOI: 10.1155/2018/2062346] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 01/24/2018] [Accepted: 02/14/2018] [Indexed: 12/20/2022] Open
Abstract
PDZD7 is an important deafness gene, whose mutations are associated with syndromic and nonsyndromic hearing loss. PDZD7 contains multiple PDZ domains that are essential for organizing various proteins into protein complex. Several PDZD7-binding proteins have been identified, including usherin, ADGRV1, whirlin, harmonin, SANS, and MYO7A, all belonging to USH proteins. Here, we report the identification of novel PDZD7-binding partners through yeast two-hybrid screening using the first two PDZ domains of PDZD7 as bait. Eleven proteins were identified, most of which have not been reported as PDZD7-binding partners before. Among the identified proteins, ADGRV1, gelsolin, and β-catenin have been shown to play important roles in hearing, whereas the functions of other proteins in the inner ear remain elusive. We confirmed the expression of one candidate PDZD7-binding protein, CADM1, in the mouse inner ear and evaluated the auditory function of Cadm1 knockout mice by performing auditory brainstem response (ABR) measurement. Unexpectedly, Cadm1 knockout mice show normal hearing threshold, which might be explained by the possible compensation by its homologs that are also expressed in the inner ear. Taken together, our work identified several novel PDZD7-binding proteins, which will help us to further understand the role of PDZD7 in hearing transduction.
Collapse
|