1
|
Zhang X, Li M, Mao X, Yao Z, Zhu W, Yuan Z, Gao X, Pan S, Zhang Y, Zhao J, Mao H. Small Intestinal Submucosa Hydrogel Loaded With Gastrodin for the Repair of Achilles Tendinopathy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401886. [PMID: 39185812 DOI: 10.1002/smll.202401886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 08/05/2024] [Indexed: 08/27/2024]
Abstract
Achilles tendinopathy (AT) is an injury caused by overuse of the Achilles tendon or sudden force on the Achilles tendon, with a considerable inflammatory infiltrate. As Achilles tendinopathy progresses, inflammation and inflammatory factors affect the remodeling of the extracellular matrix (ECM) of the tendon. Gastrodin(Gas), the main active ingredient of Astrodia has anti-inflammatory, antioxidant, and anti-apoptotic properties. The small intestinal submucosa (SIS) is a naturally decellularized extracellular matrix(dECM)material and has a high content of growth factors as well as good biocompatibility. However, the reparative effects of SIS and Gas on Achilles tendinopathy and their underlying mechanisms remain unknown. Here, it is found that SIS hydrogel loaded with gastrodin restored the mechanical strength of the Achilles tendon, facilitated ECM remodeling, and restored ordered collagen arrangement by promoting the translocation of protein synthesis. It also decreases the expression of inflammatory factors and reduces the infiltration of inflammatory cells by inhibiting the NF-κB signaling pathway. It is believed that through further research, Gas + SIS may be used in the future for the treatment of Achilles tendinopathy and other Achilles tendon injury disorders.
Collapse
Affiliation(s)
- Xiqian Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, P. R. China
| | - Mei Li
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, the First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, P. R. China
| | - Xufeng Mao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, P. R. China
| | - Zheyu Yao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, P. R. China
| | - Weilai Zhu
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Zheyang Yuan
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, P. R. China
| | - Xiang Gao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, P. R. China
| | - Senghao Pan
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Yijun Zhang
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Jiyuan Zhao
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Haijiao Mao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, P. R. China
| |
Collapse
|
2
|
Matoo S, Graves MJ, Choi MS, Idris RAES, Acharya P, Thapa G, Nguyen T, Atallah SY, Tipirneni AK, Stevenson PJ, Crawley SW. The microvillar protocadherin CDHR5 associates with EBP50 to promote brush border assembly. Mol Biol Cell 2024; 35:ar36. [PMID: 38170579 PMCID: PMC10916864 DOI: 10.1091/mbc.e23-02-0065] [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: 02/23/2023] [Revised: 10/27/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024] Open
Abstract
Transporting epithelial cells of the gut and kidney interact with their luminal environment through a densely packed collection of apical microvilli known as a brush border (BB). Proper brush border assembly depends on the intermicrovillar adhesion complex (IMAC), a protocadherin-based adhesion complex found at the distal tips of microvilli that mediates adhesion between neighboring protrusions to promote their organized packing. Loss of the IMAC adhesion molecule Cadherin-related family member 5 (CDHR5) results in significant brush border defects, though the functional properties of this protocadherin have not been thoroughly explored. Here, we show that the cytoplasmic tail of CDHR5 contributes to its correct apical targeting and functional properties in an isoform-specific manner. Library screening identified the Ezrin-associated scaffolds EBP50 and E3KARP as cytoplasmic binding partners for CDHR5. Consistent with this, loss of EBP50 disrupted proper brush border assembly with cells exhibiting markedly reduced apical IMAC levels. Together, our results shed light on the apical targeting determinants of CDHR5 and further define the interactome of the IMAC involved in brush border assembly.
Collapse
Affiliation(s)
- Samaneh Matoo
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Maura J. Graves
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Myoung Soo Choi
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | | | - Prashun Acharya
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Garima Thapa
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Tram Nguyen
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Sarah Y. Atallah
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Ashna K. Tipirneni
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | | | - Scott W. Crawley
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| |
Collapse
|
3
|
Arntz OJ, Thurlings RM, Blaney Davidson EN, Jansen PWTC, Vermeulen M, Koenders MI, van der Kraan PM, van de Loo FAJ. Profiling of plasma extracellular vesicles identifies proteins that strongly associate with patient's global assessment of disease activity in rheumatoid arthritis. Front Med (Lausanne) 2024; 10:1247778. [PMID: 38274452 PMCID: PMC10808582 DOI: 10.3389/fmed.2023.1247778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/30/2023] [Indexed: 01/27/2024] Open
Abstract
Background Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic synovial inflammation and cartilage/bone damage. Intercellular messengers such as IL-1 and TNF play a crucial role in the pathophysiology of RA but have limited diagnostic and prognostic values. Therefore, we assessed whether the protein content of the recently discovered extracellular vesicles (EVs), which have gained attention in the pathogenesis of RA, correlates with disease activity parameters in RA patients. Methods We identified and quantified proteins in plasma-derived EVs (pEVs), isolated by size exclusion chromatography from 17 RA patients by mass spectrophotometry (MS). Quantified protein levels were correlated with laboratory and clinical parameters and the patient's own global assessment of their disease activity (PGA-VAS). In a second MS run, the pEV proteins of nine other RA patients were quantified and compared to those from nine healthy controls (HC). Results No differences were observed in the concentration, size, and protein content of pEVs from RA patients. Proteomics revealed >95% overlapping proteins in RA-pEVs, compared to HC-pEVs (data are available via ProteomeXchange with identifier PXD046058). Remarkably, in both runs, the level of far more RA-pEV proteins correlated positively to PGA-VAS than to either clinical or laboratory parameters. Interestingly, all observed PGA-VAS positively correlated RA-pEV proteins were associated with the actin-cytoskeleton linker proteins, ezrin, and moesin. Conclusion Our observation suggests that PGA-VAS (loss of vitality) may have a different underlying pathological mechanism in RA, possibly related to enhanced muscle actin-cytoskeleton activity. Furthermore, our study contributes to the growing awareness and evidence that pEVs contain valuable biomarkers for diseases, with added value for RA patients.
Collapse
Affiliation(s)
- Onno J. Arntz
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rogier M. Thurlings
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Pascal W. T. C. Jansen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen, Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen, Netherlands
| | - Marije I. Koenders
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Fons A. J. van de Loo
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
4
|
Lin Q, Cao J, Yu J, Zhu Y, Shen Y, Wang S, Wang Y, Liu Z, Chang Y. YAP-mediated trophoblast dysfunction: the common pathway underlying pregnancy complications. Cell Commun Signal 2023; 21:353. [PMID: 38098027 PMCID: PMC10722737 DOI: 10.1186/s12964-023-01371-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/29/2023] [Indexed: 12/17/2023] Open
Abstract
Yes-associated protein (YAP) is a pivotal regulator in cellular proliferation, survival, differentiation, and migration, with significant roles in embryonic development, tissue repair, and tumorigenesis. At the maternal-fetal interface, emerging evidence underscores the importance of precisely regulated YAP activity in ensuring successful pregnancy initiation and progression. However, despite the established association between YAP dysregulation and adverse pregnancy outcomes, insights into the impact of aberrant YAP levels in fetal-derived, particularly trophoblast cells, and the ensuing dysfunction at the maternal-fetal interface remain limited. This review comprehensively examines YAP expression and its regulatory mechanisms in trophoblast cells throughout pregnancy. We emphasize its integral role in placental development and maternal-fetal interactions and delve into the correlations between YAP dysregulation and pregnancy complications. A nuanced understanding of YAP's functions during pregnancy could illuminate intricate molecular mechanisms and pave the way for innovative prevention and treatment strategies for pregnancy complications. Video Abstract.
Collapse
Affiliation(s)
- Qimei Lin
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Maternity Hospital, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Jiasong Cao
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Maternity Hospital, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Jing Yu
- School of Clinical Medicine, Tianjin Medical University, Tianjin, 300070, China
| | - Yu Zhu
- School of Clinical Medicine, Tianjin Medical University, Tianjin, 300070, China
| | - Yongmei Shen
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Maternity Hospital, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Shuqi Wang
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Maternity Hospital, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Yixin Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Zhen Liu
- Academy of Clinical Medicine, Medical College, Tianjin University, Tianjin, 300072, China
| | - Ying Chang
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Maternity Hospital, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, China.
- Academy of Clinical Medicine, Medical College, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
5
|
Kammala A, Benson M, Ganguly E, Richardson L, Menon R. Functional role and regulation of permeability-glycoprotein (P-gp) in the fetal membrane during drug transportation. Am J Reprod Immunol 2022; 87:e13515. [PMID: 34873775 PMCID: PMC8776608 DOI: 10.1111/aji.13515] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/09/2021] [Accepted: 11/26/2021] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE Na+ /H+ exchange regulatory factor-1 (NHERF-1) is a class I PDZ (PSD95/Discs-large/ZO-1) binding protein involved in cell-surface expression and stabilization of transporter proteins, including permeability-glycoprotein (P-gp) in various cell types. P-gp, expressed in placental trophoblasts, is an efflux transporter protein that influences the pharmacokinetics of various drugs used during pregnancy. Previously we have reported that NHERF-1 regulates fetal membrane inflammation. However, the role of NHERF-1 in regulating P-gp in the fetal membrane during drug transportation remains unclear. This study determined the interplay between NHERF-1 and P-gp in human fetal membrane cells. METHODS Fetal membranes from normal, term cesareans were screened for P-gp by immunohistochemistry (IHC). Chorionic trophoblast (CTC), with the highest expression of P-gp among fetal membrane cells, was further used to test interactive properties between NHERF-1 and P-gp. BeWo (placental trophoblast cell line) cells were used as a control. Immunoprecipitation (IP) of CTC lysates using the P-gp antibody followed by western blot determined co-precipitation of NHERF-1. Silencing NHERF-1 using small interfering RNA further tested the relevance of NHERF-1 in P-gp expression and function in CTC and BeWo cells. NHERF-1 regulation of P-gp's efflux function (drug resistance) was further tested using the ENZOTM efflux dye kit. RESULTS Immunohistochemistry localized, and western blot confirmed P-gp in human fetal membranes, primarily in the CTC with limited expression in the amnion epithelial layer. P-gp expression in the membranes was similar to that seen in the placenta. IP data showed P-gp co-precipitating with NHERF1. Silencing of NHERF-1 resulted in significant drug resistance suggesting P-gp function mediated through NHERF1 in CTCs. CONCLUSION Proinflammatory mediator NHERF-1 regulates P-gp and control drug transportation across the fetal membranes. Our data suggest a novel functional role for fetal membranes during pregnancy. Besides the placenta, fetal membranes may also regulate efflux of materials at the feto-maternal interface and control drug transport during pregnancy.
Collapse
Affiliation(s)
| | | | | | | | - Ramkumar Menon
- Corresponding author: Ramkumar Menon, Professor, Department of Obstetrics & Gynecology, Director of the Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, Texas 77555-1062, USA
| |
Collapse
|
6
|
Cecil A, Sambashivaiah S, Bilichodmath S, John RS. mRNA Expression of Ezrin in Gingival Crevicular Fluid and Whole Blood of Gingivitis and Chronic Periodontitis Patients - A Polymerase Chain Reaction Study. Contemp Clin Dent 2022; 13:267-273. [PMID: 36213856 PMCID: PMC9533393 DOI: 10.4103/ccd.ccd_6_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/04/2021] [Accepted: 03/21/2021] [Indexed: 11/15/2022] Open
Abstract
Background A comparative analysis of protein expression of gingival crevicular fluid (GCF) obtained from healthy individuals and individuals with periodontal diseases would help to identify proteins involved in periodontal disease progression. Among the identified proteins, Moesin which is a disease-associated protein belongs to the ezrin-radixin-moesin protein family and was proved to play an important role in the recognition of oral bacteria contributing to the consequent development of inflammatory immune responses involved in periodontal disease development. Aim The aim of the study is to quantify and compare mRNA expression levels of ezrin in GCF and whole blood of gingivitis and chronic periodontitis patients. Materials and Methods A total of 60 patients were selected for the study and were divided into three groups as follows: Group 1 (20 participants with healthy gingiva), Group 2 (20 participants with gingivitis), and Group 3 (20 participants with chronic periodontitis). Clinical parameters such as gingival index, periodontal index, probing pocket depth, and clinical attachment level were assessed. GCF and blood samples were taken from these patients and assessed for the mRNA expression of ezrin using real-time polymerase chain reaction. Results The expression and mean relative quantification of mRNA expression of ezrin in GCF and blood were higher for periodontitis (18.32 ± 8.398, 19.34 ± 9.487) when compared to that of gingivitis (5.34 ± 3.609, 5.48 ± 4.428) and healthy individuals (2.33 ± 0.643, 3.47 ± 1.923) and they positively correlated with the clinical parameters. Conclusion The increased expression of ezrin can be considered as a good indicator to assess the inflammatory activity in periodontitis and gingivitis.
Collapse
Affiliation(s)
- Anju Cecil
- Department of Periodontology, Saveetha Dental College and Hospital, Chennai, Tamil Nadu, India,Address for correspondence:Dr. Anju Cecil, Senior Lecturer, Department of Periodontology, Saveetha Dental College and Hospital, Chennai - 600 077, Tamil Nadu, India. E-mail:
| | - Savita Sambashivaiah
- Department of Periodontology, Rajarajeswari Dental College and Hospital, Bengaluru, Karnataka, India
| | - Shivaprasad Bilichodmath
- Department of Periodontology, Rajarajeswari Dental College and Hospital, Bengaluru, Karnataka, India
| | - Rubin S. John
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospital, Chennai, Tamil Nadu, India
| |
Collapse
|
7
|
Kammala AK, Bahal D, Yang C, Panettieri RA, Das R, Subramanian H. Na +/H + Exchanger Regulatory Factor 1 Mediates the Pathogenesis of Airway Inflammation in a Murine Model of House Dust Mite-Induced Asthma. THE JOURNAL OF IMMUNOLOGY 2021; 206:2301-2311. [PMID: 33952618 DOI: 10.4049/jimmunol.2001199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/16/2021] [Indexed: 11/19/2022]
Abstract
Na+/H+ exchanger regulatory factor 1 (NHERF1), a class I PDZ-binding protein, regulates G protein-coupled receptor signaling in some cell types. NHERF1 also functions as a scaffolding protein and activates non-G protein-coupled receptor signaling pathways, thereby contributing to the pathogenesis of various diseases. Although we have previously shown that NHERF1 regulates mast cell functions, there is little information regarding the role of NHERF1 in other immune cells. How NHERF1 regulates the pathogenesis of allergic disease such as asthma also remains unknown. In the current study, we show that NHERF1 promotes allergic airway inflammation in a house dust mite extract (HDME)-induced mouse model of asthma. Specifically, HDME-specific serum IgE levels, airway leukocyte numbers, and goblet cell hyperplasia were reduced in NHERF1+/- mice as compared with NHERF1+/+ mice. Interestingly, the gene expression of inflammatory (IL-17a, IL-25, and IL-33) as well as T helper 2 (Th2) cytokines (IL-4, IL-5, and IL-13) and several chemokines that recruit eosinophils, neutrophils, and lymphocytes were also decreased in the lungs of NHERF1+/- mice exposed to HDME. Consistent with these observations, microRNAs regulating mucus production, inflammation, Th2 effector functions, and IL-13 expression were increased in the lungs of HDME-treated NHERF1+/- mice. Overall, our studies reveal a unique role for NHERF1 in regulating asthma pathogenesis, and further elucidation of the mechanisms through which NHERF1 modulates allergic inflammation will lead to the development of new therapeutic strategies for asthma.
Collapse
Affiliation(s)
- Ananth K Kammala
- Department of Physiology, Michigan State University, East Lansing, MI
| | - Devika Bahal
- Department of Physiology, Michigan State University, East Lansing, MI
| | - Canchai Yang
- Department of Physiology, Michigan State University, East Lansing, MI
| | | | - Rupali Das
- Department of Physiology, Michigan State University, East Lansing, MI
| | | |
Collapse
|
8
|
Álvarez-Carrión L, Gutiérrez-Rojas I, Rodríguez-Ramos MR, Ardura JA, Alonso V. MINDIN Exerts Protumorigenic Actions on Primary Prostate Tumors via Downregulation of the Scaffold Protein NHERF-1. Cancers (Basel) 2021; 13:436. [PMID: 33498862 PMCID: PMC7865820 DOI: 10.3390/cancers13030436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 11/17/2022] Open
Abstract
Advanced prostate cancer preferential metastasis to bone is associated with osteomimicry. MINDIN is a secreted matrix protein upregulated in prostate tumors that overexpresses bone-related genes during prostate cancer progression. Na+/H+ exchanger regulatory factor (NHERF-1) is a scaffold protein that has been involved both in tumor regulation and osteogenesis. We hypothesize that NHERF-1 modulation is a mechanism used by MINDIN to promote prostate cancer progression. We analyzed the expression of NHERF-1 and MINDIN in human prostate samples and in a premetastatic prostate cancer mouse model, based on the implantation of prostate adenocarcinoma TRAMP-C1 (transgenic adenocarcinoma of the mouse prostate) cells in immunocompetent C57BL/6 mice. The relationship between NHERF-1 and MINDIN and their effects on cell proliferation, migration, survival and osteomimicry were evaluated. Upregulation of MINDIN and downregulation of NHERF-1 expression were observed both in human prostate cancer samples and in the TRAMP-C1 model. MINDIN silencing restored NHERF-1 expression to control levels in the mouse model. Stimulation with MINDIN reduced NHERF-1 expression and triggered its mobilization from the plasma membrane to the cytoplasm in TRAMP-C1 cells. MINDIN-dependent downregulation of NHERF-1 promoted tumor cell migration and proliferation without affecting osteomimicry and adhesion. We propose that MINDIN downregulates NHERF-1 expression leading to promotion of processes involved in prostate cancer progression.
Collapse
Affiliation(s)
- Luis Álvarez-Carrión
- Bone Physiopathology Laboratory, Applied Molecular Medicine Institute (IMMA), Universidad San Pablo-CEU, CEU Universities, Campus Monteprincipe, 28925 Alcorcón, Spain; (L.Á.-C.); (I.G.-R.); (M.R.R.-R.)
| | - Irene Gutiérrez-Rojas
- Bone Physiopathology Laboratory, Applied Molecular Medicine Institute (IMMA), Universidad San Pablo-CEU, CEU Universities, Campus Monteprincipe, 28925 Alcorcón, Spain; (L.Á.-C.); (I.G.-R.); (M.R.R.-R.)
| | - María Rosario Rodríguez-Ramos
- Bone Physiopathology Laboratory, Applied Molecular Medicine Institute (IMMA), Universidad San Pablo-CEU, CEU Universities, Campus Monteprincipe, 28925 Alcorcón, Spain; (L.Á.-C.); (I.G.-R.); (M.R.R.-R.)
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Campus Monteprincipe, 28925 Alcorcón, Spain
| | - Juan A. Ardura
- Bone Physiopathology Laboratory, Applied Molecular Medicine Institute (IMMA), Universidad San Pablo-CEU, CEU Universities, Campus Monteprincipe, 28925 Alcorcón, Spain; (L.Á.-C.); (I.G.-R.); (M.R.R.-R.)
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Campus Monteprincipe, 28925 Alcorcón, Spain
| | - Verónica Alonso
- Bone Physiopathology Laboratory, Applied Molecular Medicine Institute (IMMA), Universidad San Pablo-CEU, CEU Universities, Campus Monteprincipe, 28925 Alcorcón, Spain; (L.Á.-C.); (I.G.-R.); (M.R.R.-R.)
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Campus Monteprincipe, 28925 Alcorcón, Spain
| |
Collapse
|
9
|
Kammala AK, Syed M, Yang C, Occhiuto CJ, Subramanian H. A Critical Role for Na +/H + Exchanger Regulatory Factor 1 in Modulating FcεRI-Mediated Mast Cell Activation. THE JOURNAL OF IMMUNOLOGY 2020; 206:471-480. [PMID: 33361207 DOI: 10.4049/jimmunol.2000671] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/23/2020] [Indexed: 01/01/2023]
Abstract
Mast cells are tissue-resident immune cells that play pivotal roles in initiating and amplifying allergic/anaphylactic reactions in humans. Their activation occurs via multiple mechanisms, which include cross-linking of the IgE-bound, high-affinity IgE receptors (FcεRI) by allergens or Ags and the binding of anaphylatoxins such as C3a to its receptor, C3aR. We have previously demonstrated that the Na+/H+ exchanger regulatory factor 1 (NHERF1) promotes C3aR functions in human mast cells. In the current study, we show that NHERF1 regulates mast cell response following FcεRI stimulation. Specifically, intracellular Ca2+ mobilization, activation of the MAPKs (ERK1/2 and P38), and production of cytokines (IL-13 and IL-6) following exposure to IgE/Ag were significantly reduced in mast cells from NHERF1+/‒ mice. In agreement with our in vitro data, mast cell-mediated passive cutaneous anaphylaxis and passive systemic anaphylaxis were reduced in NHERF1+/‒ mice and mast cell-deficient KitW-sh/W-sh mice engrafted with NHERF1+/‒ mast cells. Mechanistically, the levels of microRNAs (miRNAs) that regulate mast cell responses, miRNA 155-3p and miRNA 155-5p, were altered in mast cells from NHERF1+/‒ mice. Moreover, NHERF1 rapidly localized to the nucleus of mast cells following FcεRI stimulation. In summary, our results suggest that the NHERF1 acts as an adapter molecule and promotes IgE/Ag-induced mast cell activation. Further elucidating the mechanisms through which NHERF1 modulates mast cell responses will lend insights into the development of new therapeutic strategies to target mast cells during anaphylaxis or other allergic diseases.
Collapse
Affiliation(s)
- Ananth K Kammala
- Department of Physiology, Michigan State University, East Lansing, MI 48824
| | - Meesum Syed
- Department of Physiology, Michigan State University, East Lansing, MI 48824
| | - Canchai Yang
- Department of Physiology, Michigan State University, East Lansing, MI 48824
| | | | | |
Collapse
|
10
|
Sodium Hydrogen Exchanger Regulatory Factor-1 (NHERF1) Regulates Fetal Membrane Inflammation. Int J Mol Sci 2020; 21:ijms21207747. [PMID: 33092043 PMCID: PMC7589612 DOI: 10.3390/ijms21207747] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/13/2020] [Accepted: 10/16/2020] [Indexed: 11/20/2022] Open
Abstract
The fetal inflammatory response, a key contributor of infection-associated preterm birth (PTB), is mediated by nuclear factor kappa B (NF-kB) activation. Na+/H+ exchanger regulatory factor-1 (NHERF1) is an adapter protein that can regulate intracellular signal transduction and thus influence NF-kB activation. Accordingly, NHERF1 has been reported to enhance proinflammatory cytokine release and amplify inflammation in a NF-kB-dependent fashion in different cell types. The objective of this study was to examine the role of NHERF1 in regulating fetal membrane inflammation during PTB. We evaluated the levels of NHERF1 in human fetal membranes from term labor (TL), term not in labor (TNIL), and PTB and in a CD1 mouse model of PTB induced by lipopolysaccharide (LPS). Additionally, primary cultures of fetal membrane cells were treated with LPS, and NHERF1 expression and cytokine production were evaluated. Gene silencing methods using small interfering RNA targeting NHERF1 were used to determine the functional relevance of NHERF1 in primary cultures. NHERF1 expression was significantly (p < 0.001) higher in TL and PTB membranes compared to TNIL membranes, and this coincided with enhanced (p < 0.01) interleukin (IL)-6 and IL-8 expression levels. LPS-treated animals delivering PTB had increased levels of NHERF1, IL-6, and IL-8 compared to phosphate-buffered saline (PBS; control) animals. Silencing of NHERF1 expression resulted in a significant reduction in NF-kB activation and IL-6 and IL-8 production as well as increased IL-10 production. In conclusion, downregulation of NHERF1 increased anti-inflammatory IL-10, and reducing NHERF1 expression could be a potential therapeutic strategy to reduce the risk of infection/inflammation associated with PTB.
Collapse
|
11
|
NHERF1 Loss Upregulates Enzymes of the Pentose Phosphate Pathway in Kidney Cortex. Antioxidants (Basel) 2020; 9:antiox9090862. [PMID: 32937931 PMCID: PMC7554817 DOI: 10.3390/antiox9090862] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
(1) Background: We previously showed Na/H exchange regulatory factor 1 (NHERF1) loss resulted in increased susceptibility to cisplatin nephrotoxicity. NHERF1-deficient cultured proximal tubule cells and proximal tubules from NHERF1 knockout (KO) mice exhibit altered mitochondrial protein expression and poor survival. We hypothesized that NHERF1 loss results in changes in metabolic pathways and/or mitochondrial dysfunction, leading to increased sensitivity to cisplatin nephrotoxicity. (2) Methods: Two to 4-month-old male wildtype (WT) and KO mice were treated with vehicle or cisplatin (20 mg/kg dose IP). After 72 h, kidney cortex homogenates were utilized for metabolic enzyme activities. Non-treated kidneys were used to isolate mitochondria for mitochondrial respiration via the Seahorse XF24 analyzer. Non-treated kidneys were also used for LC-MS analysis to evaluate kidney ATP abundance, and electron microscopy (EM) was utilized to evaluate mitochondrial morphology and number. (3) Results: KO mouse kidneys exhibit significant increases in malic enzyme and glucose-6 phosphate dehydrogenase activity under baseline conditions but in no other gluconeogenic or glycolytic enzymes. NHERF1 loss does not decrease kidney ATP content. Mitochondrial morphology, number, and area appeared normal. Isolated mitochondria function was similar between WT and KO. Conclusions: KO kidneys experience a shift in metabolism to the pentose phosphate pathway, which may sensitize them to the oxidative stress imposed by cisplatin.
Collapse
|
12
|
Effect of Thymoquinone on Acute Kidney Injury Induced by Sepsis in BALB/c Mice. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1594726. [PMID: 32626733 PMCID: PMC7315249 DOI: 10.1155/2020/1594726] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/01/2020] [Indexed: 01/26/2023]
Abstract
Acute kidney injury (AKI) is a common complication of sepsis and has also been observed in some patients suffering from the new coronavirus pneumonia COVID-19, which is currently a major global concern. Thymoquinone (TQ) is one of the most active ingredients in Nigella sativa seeds. It has a variety of beneficial properties including anti-inflammatory and antioxidative activities. Here, we investigated the possible protective effects of TQ against kidney damage in septic BALB/c mice. Eight-week-old male BALB/c mice were divided into four groups: control, TQ, cecal ligation and puncture (CLP), and TQ+CLP. CLP was performed after 2 weeks of TQ gavage. After 48 h, we measured the histopathological alterations in the kidney tissue and the serum levels of creatinine (CRE) and blood urea nitrogen (BUN). We also evaluated pyroptosis (NLRP3, caspase-1), apoptosis (caspase-3, caspase-8), proinflammatory (TNF-α, IL-1β, and IL-6)-related protein and gene expression levels. Our results demonstrated that TQ inhibited CLP-induced increased serum CRE and BUN levels. It also significantly inhibited the high levels of NLRP3, caspase-1, caspase-3, caspase-8, TNF-α, IL-1β, and IL-6 induced by CLP. Furthermore, NF-κB protein level was significantly decreased in the TQ+CLP group than in the CLP group. Together, our results indicate that TQ may be a potential therapeutic agent for sepsis-induced AKI.
Collapse
|
13
|
Yang F, Hu M, Chang S, Huang J, Si Y, Wang J, Cheng S, Jiang WG. Alteration in the sensitivity to crizotinib by Na +/H + exchanger regulatory factor 1 is dependent to its subcellular localization in ALK-positive lung cancers. BMC Cancer 2020; 20:202. [PMID: 32164629 PMCID: PMC7068933 DOI: 10.1186/s12885-020-6687-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 02/27/2020] [Indexed: 12/17/2022] Open
Abstract
Background Na+/H+ exchanger regulatory factor 1 (NHERF1) is an important scaffold protein participates in the modulation of a variety of intracellular signal pathways. NHERF1 was able to enhance the effects of chemo-drugs in breast and cervical cancer cells. Anaplastic lymphoma kinase (ALK) fusion mutations are validated molecules targeted therapy in lung cancers, where crizotinib can be used as the specific inhibitor to suppress tumor progression. However, due to the less frequent occurrence of ALK mutations and the complexity for factors to determine drug responses, the genes that could alter crizotinib sensitivity are unclear. Methods Both ALK-translocated and ALK-negative lung adenocarcinoma specimens in tissue sections were collected for immunohistochemistry. The possible mechanisms of NHERF1 and its role in the cell sensitivity to crizotinib were investigated using an ALK-positive and crizotinib-sensitive lung adenocarcinoma cell line H3122. Either a NHERF1 overexpression vector or agents for NHERF1 knockdown was used for crizotinib sensitivity measures, in association with cell viability and apoptosis assays. Results The expression level of NHERF1 in ALK-translocated NSCLC was significantly higher than that in other lung cancer tissues. NHERF1 expression in ALK positive lung cancer cells was regulated by ALK activities, and was in return able to alter the sensitivity to crizotinib. The function of NHERF1 to influence crizotinib sensitivity was depending on its subcellular distribution in cytosol instead of its nucleus localized form. Conclusion Ectopically overexpressed NHERF1 could be a functional protein for consideration to suppress lung cancers. The determination of NHERF1 levels in ALK positive NSCLC tissues might be useful to predict crizotinib resistance, especially by distinguishing cytosolic or nuclear localized NHERF1 for the overexpressed molecules.
Collapse
Affiliation(s)
- Fenglian Yang
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China.,Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing, China
| | - Mu Hu
- Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing, China.,Department of Thoracic Surgery, Beijing Xuanwu Hospital, Beijing, 100053, P.R. China
| | - Siyuan Chang
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China.,Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing, China
| | - Jing Huang
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China.,Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing, China
| | - Yang Si
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China.,Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing, China
| | - Jinghui Wang
- Department of Medical Oncology, Beijing Chest Hospital, Beijing, 101149, P.R. China
| | - Shan Cheng
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China. .,Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing, China.
| | - Wen G Jiang
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China.,Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing, China.,Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| |
Collapse
|
14
|
Bushau-Sprinkle AM, Lederer ED. New roles of the Na +/H + exchange regulatory factor 1 scaffolding protein: a review. Am J Physiol Renal Physiol 2020; 318:F804-F808. [PMID: 31984791 DOI: 10.1152/ajprenal.00467.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Na+/H+ exchange regulatory factor 1 (NHERF1), a member of a PDZ scaffolding protein family, was first identified as an organizer of membrane-bound protein complexes composed of hormone receptors, signal transduction pathways, and electrolyte and mineral transporters and channels. NHERF1 is involved in the regulation of Na+/H+ exchanger 3, Na+-dependent phosphate transporter 2a, and Na+-K+-ATPase through its ability to scaffold these transporters to the plasma membrane, allowing regulation of these protein complexes with their associated hormone receptors. Recently, NHERF1 has received increased interest in its involvement in a variety of functions, including cell structure and trafficking, tumorigenesis and tumor behavior, inflammatory responses, and tissue injury. In this review, we highlight the evidence for the expansive role of NHERF1 in cell biology and speculate on the implications for renal physiology and pathophysiology.
Collapse
Affiliation(s)
- Adrienne M Bushau-Sprinkle
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky.,Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky
| | - Eleanor D Lederer
- Division of Nephrology, Department of Medicine, University of Louisville, Louisville, Kentucky.,Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky
| |
Collapse
|
15
|
Lin CM, Titchenell PM, Keil JM, Garcia-Ocaña A, Bolinger MT, Abcouwer SF, Antonetti DA. Inhibition of Atypical Protein Kinase C Reduces Inflammation-Induced Retinal Vascular Permeability. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2392-2405. [PMID: 30220554 DOI: 10.1016/j.ajpath.2018.06.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 06/01/2018] [Accepted: 06/13/2018] [Indexed: 01/14/2023]
Abstract
Changes in permeability of retinal blood vessels contribute to macular edema and the pathophysiology of numerous ocular diseases, including diabetic retinopathy, retinal vein occlusions, and macular degeneration. Vascular endothelial growth factor (VEGF) induces retinal permeability and macular thickening in these diseases. However, inflammatory agents, such as tumor necrosis factor-α (TNF-α), also may drive vascular permeability, specifically in patients unresponsive to anti-VEGF therapy. Recent evidence suggests VEGF and TNF-α induce permeability through distinct mechanisms; however, both require the activation of atypical protein kinase C (aPKC). We provide evidence, using genetic mouse models and therapeutic intervention with small molecules, that inhibition of aPKC prevented or reduced vascular permeability in animal models of retinal inflammation. Expression of a kinase-dead aPKC transgene, driven by a vascular and hematopoietic restricted promoter, reduced retinal vascular permeability in an ischemia-reperfusion model of retinal injury. This effect was recapitulated with a small-molecule inhibitor of aPKC. Expression of the kinase-dead aPKC transgene dramatically reduced the expression of inflammatory factors and blocked the attraction of inflammatory monocytes and granulocytes after ischemic injury. Coinjection of VEGF with TNF-α was sufficient to induce permeability, edema, and retinal inflammation, and treatment with an aPKC inhibitor prevented VEGF/TNF-α-induced permeability. These data suggest that aPKC contributes to inflammation-driven retinal vascular pathology and may be an attractive target for therapeutic intervention.
Collapse
Affiliation(s)
- Cheng-Mao Lin
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan
| | - Paul M Titchenell
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jason M Keil
- Molecular and Behavioral Neuroscience Institute, Department of Human Genetics and Medical Scientist Training Program, University of Michigan, Ann Arbor, Michigan
| | - Adolfo Garcia-Ocaña
- Diabetes, Obesity and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Diseases, The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mark T Bolinger
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan
| | - Steven F Abcouwer
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan
| | - David A Antonetti
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
16
|
Zhang Z, Zhang H, Chen R, Wang Z. Oral supplementation with ursolic acid ameliorates sepsis-induced acute kidney injury in a mouse model by inhibiting oxidative stress and inflammatory responses. Mol Med Rep 2018; 17:7142-7148. [PMID: 29568928 PMCID: PMC5928665 DOI: 10.3892/mmr.2018.8767] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 08/16/2017] [Indexed: 02/07/2023] Open
Abstract
Ursolic acid (UA) as a multiple bioactive native compound has recently been demonstrated to treat sepsis in animal models. However, the beneficial effects of UA in sepsis‑induced acute kidney injury (AKI) are not completely understood. In the present study, the effect of UA on sepsis‑induced AKI in cecal ligation and puncture (CLP) surgery mice was investigated. Renal histomorphological analysis was performed by hematoxylin and eosin staining. The expression of inflammatory markers in the kidney of septic mice was measured by reverse transcription‑quantitative polymerase chain reaction and western blotting. The results demonstrated that UA administration improved survival in septic mice induced by CLP surgery. The treatment with UA revealed protection against AKI induced by CLP surgery, including the alleviation of glomerular damage and vacuolization in the proximal tubules. In addition, the effects of UA on oxidative stress and inflammation in septic mice were determined. The findings suggested that UA may protect against sepsis‑induced AKI by inhibiting reactive oxygen species and inflammatory cytokines, including tumor necrosis factor‑α, interleukin (IL)‑1β and IL‑6, in the kidney from septic mice. Finally, UA inhibited CLP‑induced activation of nuclear factor‑κB signaling in the kidney from septic mice. The findings of the present study demonstrated that UA may be used as a potential therapeutic agent for complications of sepsis, especially for sepsis-induced AKI.
Collapse
Affiliation(s)
- Zhenyu Zhang
- Department of Critical Care Medicine, Beijing Tsinghua Changgung Hospital, Beijing 102218, P.R. China
| | - Hong Zhang
- Department of Applied Chemistry, Peking University, Beijing 100871, P.R. China
| | - Rui Chen
- Department of Critical Care Medicine, Beijing Tsinghua Changgung Hospital, Beijing 102218, P.R. China
| | - Zhong Wang
- Department of Critical Care Medicine, Beijing Tsinghua Changgung Hospital, Beijing 102218, P.R. China
| |
Collapse
|
17
|
Centonze M, Saponaro C, Mangia A. NHERF1 Between Promises and Hopes: Overview on Cancer and Prospective Openings. Transl Oncol 2018; 11:374-390. [PMID: 29455084 PMCID: PMC5852411 DOI: 10.1016/j.tranon.2018.01.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/02/2018] [Accepted: 01/05/2018] [Indexed: 02/07/2023] Open
Abstract
Na+/H+ exchanger regulatory factor 1 (NHERF1) is a scaffold protein, with two tandem PDZ domains and a carboxyl-terminal ezrin-binding (EB) region. This particular sticky structure is responsible for its interaction with different molecules to form multi-complexes that have a pivotal role in a lot of diseases. In particular, its involvement during carcinogenesis and cancer progression has been deeply analyzed in different tumors. The role of NHERF1 is not unique in cancer; its activity is connected to its subcellular localization. The literature data suggest that NHERF1 could be a new prognostic/predictive biomarker from breast cancer to hematological cancers. Furthermore, the high potential of this molecule as therapeutical target in different carcinomas is a new challenge for precision medicine. These evidences are part of a future view to improving patient clinical management, which should allow different tumor phenotypes to be treated with tailored therapies. This article reviews the biology of NHERF1, its engagement in different signal pathways and its involvement in different cancers, with a specific focus on breast cancer. It also considers NHERF1 potential role during inflammation related to most human cancers, designating new perspectives in the study of this "Janus-like" protein.
Collapse
Affiliation(s)
- Matteo Centonze
- Functional Biomorphology Laboratory, IRCCS-Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Concetta Saponaro
- Functional Biomorphology Laboratory, IRCCS-Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Anita Mangia
- Functional Biomorphology Laboratory, IRCCS-Istituto Tumori "Giovanni Paolo II", Bari, Italy.
| |
Collapse
|
18
|
Gu Y, Yu H, Hao C, Martin TA, Hargest R, He J, Cheng S, Jiang WG. NHERF1 regulates the progression of colorectal cancer through the interplay with VEGFR2 pathway. Oncotarget 2018; 8:7753-7765. [PMID: 27999191 PMCID: PMC5352358 DOI: 10.18632/oncotarget.13949] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/01/2016] [Indexed: 01/05/2023] Open
Abstract
The oncogenic role of ectopic expression of Na+/H+ exchanger regulatory factor 1 (NHERF1) was recently suggested in colorectal cancer, where it was implicated in playing a role in the tumor hypoxia microenvironment. Here we showed that a high level expression of NHERF1 was found in colorectal cancer tissues and that the expression of NHERF1 was positively correlated with VEGFR2 expression. The prognostic value of VEGFR2 expression in colorectal cancer relied on the expression of NHERF1. The up-regulation of NHERF1 induced by the exposure to hypoxia in colon cancer cells depended on the activation of VEGFR2 signaling. NHERF1 in turn inhibited the activation of VEGFR2 signaling which could be regulated by the interaction between NHERF1 and VEGFR2, resulting in the reduction of migration and invasion of colon cancer cells. These results suggest a dynamic interplay between NHERF1 and VEGFR2 signaling in colorectal cancer, which could explain the contribution of NHERF1 to the regulation of tumor cell responses to the hypoxia microenvironment.
Collapse
Affiliation(s)
- Yanan Gu
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, China.,Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing 100069, China
| | - Hefen Yu
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, China.,Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing 100069, China
| | - Chengcheng Hao
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, China.,Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing 100069, China
| | - Tracey A Martin
- Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing 100069, China.,Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Rachel Hargest
- Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing 100069, China.,Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Junqi He
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, China.,Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing 100069, China
| | - Shan Cheng
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, China.,Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing 100069, China
| | - Wen G Jiang
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, China.,Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing 100069, China.,Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
19
|
Vaquero J, Nguyen Ho-Bouldoires TH, Clapéron A, Fouassier L. Role of the PDZ-scaffold protein NHERF1/EBP50 in cancer biology: from signaling regulation to clinical relevance. Oncogene 2017; 36:3067-3079. [PMID: 28068322 DOI: 10.1038/onc.2016.462] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/26/2016] [Accepted: 10/31/2016] [Indexed: 12/14/2022]
Abstract
The transmission of cellular information requires fine and subtle regulation of proteins that need to interact in a coordinated and specific way to form efficient signaling networks. The spatial and temporal coordination relies on scaffold proteins. Thanks to protein interaction domains such as PDZ domains, scaffold proteins organize multiprotein complexes enabling the proper transmission of cellular information through intracellular networks. NHERF1/EBP50 is a PDZ-scaffold protein that was initially identified as an organizer and regulator of transporters and channels at the apical side of epithelia through actin-binding ezrin-moesin-radixin proteins. Since, NHERF1/EBP50 has emerged as a major regulator of cancer signaling network by assembling cancer-related proteins. The PDZ-scaffold EBP50 carries either anti-tumor or pro-tumor functions, two antinomic functions dictated by EBP50 expression or subcellular localization. The dual function of NHERF1/EBP50 encompasses the regulation of several major signaling pathways engaged in cancer, including the receptor tyrosine kinases PDGFR and EGFR, PI3K/PTEN/AKT and Wnt-β-catenin pathways.
Collapse
Affiliation(s)
- J Vaquero
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,FONDATION ARC, Villejuif, France
| | - T H Nguyen Ho-Bouldoires
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,FONDATION ARC, Villejuif, France
| | - A Clapéron
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | - L Fouassier
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| |
Collapse
|
20
|
Nguyen Ho-Bouldoires TH, Clapéron A, Mergey M, Wendum D, Desbois-Mouthon C, Tahraoui S, Fartoux L, Chettouh H, Merabtene F, Scatton O, Gaestel M, Praz F, Housset C, Fouassier L. Mitogen-activated protein kinase-activated protein kinase 2 mediates resistance to hydrogen peroxide-induced oxidative stress in human hepatobiliary cancer cells. Free Radic Biol Med 2015; 89:34-46. [PMID: 26169728 DOI: 10.1016/j.freeradbiomed.2015.07.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 06/25/2015] [Accepted: 07/08/2015] [Indexed: 12/21/2022]
Abstract
The development and progression of liver cancer are characterized by increased levels of reactive oxygen species (ROS). ROS-induced oxidative stress impairs cell proliferation and ultimately leads to cell death. Although liver cancer cells are especially resistant to oxidative stress, mechanisms of such resistance remain understudied. We identified the MAPK-activated protein kinase 2 (MK2)/heat shock protein 27 (Hsp27) signaling pathway mediating defenses against oxidative stress. In addition to MK2 and Hsp27 overexpression in primary liver tumors compared to adjacent nontumorous tissues, the MK2/Hsp27 pathway is activated by hydrogen peroxide-induced oxidative stress in hepatobiliary cancer cells. MK2 inactivation or inhibition of MK2 or Hsp27 expression increases caspase-3 and PARP cleavage and DNA breaks and therefore cell death. Interestingly, MK2/Hsp27 inhibition decreases antioxidant defenses such as heme oxygenase 1 through downregulation of the transcription factor nuclear factor erythroid-derived 2-like 2. Moreover, MK2/Hsp27 inhibition decreases both phosphorylation of epidermal growth factor receptor (EGFR) and expression of its ligand, heparin-binding EGF-like growth factor. A new identified partner of MK2, the scaffold PDZ protein EBP50, could facilitate these effects through MK2/Hsp27 pathway regulation. These findings demonstrate that the MK2/Hsp27 pathway actively participates in resistance to oxidative stress and may contribute to liver cancer progression.
Collapse
Affiliation(s)
- Thanh Huong Nguyen Ho-Bouldoires
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Audrey Clapéron
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Martine Mergey
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Dominique Wendum
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Service d'Anatomie et Cytologie Pathologiques, AP-HP, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Christèle Desbois-Mouthon
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Sylvana Tahraoui
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Laetitia Fartoux
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Service d'Hépatologie, AP-HP, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Hamza Chettouh
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Fatiha Merabtene
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Olivier Scatton
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Service de Chirurgie Hépato-Biliaire et Transplantation Hépatique, AP-HP, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Matthias Gaestel
- Institute of Physiological Chemistry, Hannover Medical School, D-30625 Hannover, Germany
| | - Françoise Praz
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Chantal Housset
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Service d'Hépatologie, AP-HP, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Laura Fouassier
- INSERM UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France.
| |
Collapse
|
21
|
Li M, Mennone A, Soroka CJ, Hagey LR, Ouyang X, Weinman EJ, Boyer JL. Na(+) /H(+) exchanger regulatory factor 1 knockout mice have an attenuated hepatic inflammatory response and are protected from cholestatic liver injury. Hepatology 2015; 62:1227-36. [PMID: 26108984 PMCID: PMC4589453 DOI: 10.1002/hep.27956] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 06/17/2015] [Indexed: 12/31/2022]
Abstract
UNLABELLED The intercellular adhesion molecule 1 (ICAM-1) is induced in mouse liver after bile duct ligation (BDL) and plays a key role in neutrophil-mediated liver injury in BDL mice. ICAM-1 has been shown to interact with cytoskeletal ezrin-radixin-moesin (ERM) proteins that also interact with the PDZ protein, Na(+) /H(+) exchanger regulatory factor 1 (NHERF-1/EBP50). In NHERF-1(-/-) mice, ERM proteins are significantly reduced in brush-border membranes from kidney and small intestine. ERM knockdown reduces ICAM-1 expression in response to tumor necrosis factor alpha. Here we show that NHERF-1 assembles ERM proteins, ICAM-1 and F-actin into a macromolecule complex that is increased in mouse liver after BDL. Compared to wild-type (WT) mice, both sham-operated and BDL NHERF-1(-/-) mice have lower levels of activated ERM and ICAM-1 protein in the liver accompanied by significantly reduced hepatic neutrophil accumulation, serum alanine aminotransferase, and attenuated liver injury after BDL. However, total bile acid concentrations in serum and liver of sham and BDL NHERF-1(-/-) mice were not significantly different from WT controls, although hepatic tetrahydroxylated bile acids and Cyp3a11 messenger RNA levels were higher in NHERF-1(-/-) BDL mice. CONCLUSION NHERF-1 participates in the inflammatory response that is associated with BDL-induced liver injury. Deletion of NHERF-1 in mice leads to disruption of the formation of ICAM-1/ERM/NHERF-1 complex and reduction of hepatic ERM proteins and ICAM-1, molecules that are up-regulated and are essential for neutrophil-mediated liver injury in cholestasis. Further study of the role of NHERF-1 in the inflammatory response in cholestasis and other forms of liver injury should lead to discovery of new therapeutic targets in hepatic inflammatory diseases.
Collapse
Affiliation(s)
- Man Li
- Yale Liver Center, Yale University School of Medicine, New Haven, CT
| | - Albert Mennone
- Yale Liver Center, Yale University School of Medicine, New Haven, CT
| | - Carol J. Soroka
- Yale Liver Center, Yale University School of Medicine, New Haven, CT
| | - Lee R. Hagey
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Xinshou Ouyang
- Yale Liver Center, Yale University School of Medicine, New Haven, CT
| | - Edward J. Weinman
- Department of Medicine, Baltimore, MD,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - James L. Boyer
- Yale Liver Center, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
22
|
Song GJ, Leslie KL, Barrick S, Mamonova T, Fitzpatrick JM, Drombosky KW, Peyser N, Wang B, Pellegrini M, Bauer PM, Friedman PA, Mierke DF, Bisello A. Phosphorylation of ezrin-radixin-moesin-binding phosphoprotein 50 (EBP50) by Akt promotes stability and mitogenic function of S-phase kinase-associated protein-2 (Skp2). J Biol Chem 2014; 290:2879-87. [PMID: 25492869 DOI: 10.1074/jbc.m114.609768] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The regulation of the cell cycle by the ubiquitin-proteasome system is dependent on the activity of E3 ligases. Skp2 (S-phase kinase associated protein-2) is the substrate recognition subunit of the E3 ligase that ubiquitylates the cell cycle inhibitors p21(cip1) and p27(kip1) thus promoting cell cycle progression. Increased expression of Skp2 is frequently observed in diseases characterized by excessive cell proliferation, such as cancer and neointima hyperplasia. The stability and cellular localization of Skp2 are regulated by Akt, but the molecular mechanisms underlying these effects remain only partly understood. The scaffolding protein Ezrin-Binding Phosphoprotein of 50 kDa (EBP50) contains two PDZ domains and plays a critical role in the development of neointimal hyperplasia. Here we report that EBP50 directly binds Skp2 via its first PDZ domain. Moreover, EBP50 is phosphorylated by Akt on Thr-156 within the second PDZ domain, an event that allosterically promotes binding to Skp2. The interaction with EBP50 causes cytoplasmic localization of Skp2, increases Skp2 stability and promotes proliferation of primary vascular smooth muscle cells. Collectively, these studies define a novel regulatory mechanism contributing to aberrant cell growth and highlight the importance of scaffolding function of EBP50 in Akt-dependent cell proliferation.
Collapse
Affiliation(s)
- Gyun Jee Song
- From the Department of Pharmacology and Chemical Biology, Department of Pharmacology, Brain Science and Engineering Institute, School of Medicine, Kungpook National University, Daegu 702-701, Korea, and
| | | | - Stacey Barrick
- From the Department of Pharmacology and Chemical Biology
| | | | | | | | - Noah Peyser
- From the Department of Pharmacology and Chemical Biology
| | - Bin Wang
- From the Department of Pharmacology and Chemical Biology
| | - Maria Pellegrini
- Department of Chemistry, Dartmouth College, Hanover, New Hampshire 03755
| | - Philip M Bauer
- Vascular Medicine Institute, and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| | | | - Dale F Mierke
- Department of Chemistry, Dartmouth College, Hanover, New Hampshire 03755
| | - Alessandro Bisello
- From the Department of Pharmacology and Chemical Biology, Vascular Medicine Institute, and
| |
Collapse
|
23
|
Zheng F, Jewell H, Fitzpatrick J, Zhang J, Mierke DF, Grigoryan G. Computational design of selective peptides to discriminate between similar PDZ domains in an oncogenic pathway. J Mol Biol 2014; 427:491-510. [PMID: 25451599 DOI: 10.1016/j.jmb.2014.10.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 10/21/2014] [Accepted: 10/23/2014] [Indexed: 11/25/2022]
Abstract
Reagents that target protein-protein interactions to rewire signaling are of great relevance in biological research. Computational protein design may offer a means of creating such reagents on demand, but methods for encoding targeting selectivity are sorely needed. This is especially challenging when targeting interactions with ubiquitous recognition modules--for example, PDZ domains, which bind C-terminal sequences of partner proteins. Here we consider the problem of designing selective PDZ inhibitor peptides in the context of an oncogenic signaling pathway, in which two PDZ domains (NHERF-2 PDZ2-N2P2 and MAGI-3 PDZ6-M3P6) compete for a receptor C-terminus to differentially modulate oncogenic activities. Because N2P2 has been shown to increase tumorigenicity and M3P6 to decreases it, we sought to design peptides that inhibit N2P2 without affecting M3P6. We developed a structure-based computational design framework that models peptide flexibility in binding yet is efficient enough to rapidly analyze tradeoffs between affinity and selectivity. Designed peptides showed low-micromolar inhibition constants for N2P2 and no detectable M3P6 binding. Peptides designed for reverse discrimination bound M3P6 tighter than N2P2, further testing our technology. Experimental and computational analysis of selectivity determinants revealed significant indirect energetic coupling in the binding site. Successful discrimination between N2P2 and M3P6, despite their overlapping binding preferences, is highly encouraging for computational approaches to selective PDZ targeting, especially because design relied on a homology model of M3P6. Still, we demonstrate specific deficiencies of structural modeling that must be addressed to enable truly robust design. The presented framework is general and can be applied in many scenarios to engineer selective targeting.
Collapse
Affiliation(s)
- Fan Zheng
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Heather Jewell
- Department of Computer Science, Dartmouth College, Hanover, NH 03755, USA
| | | | - Jian Zhang
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Dale F Mierke
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA
| | - Gevorg Grigoryan
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA; Department of Computer Science, Dartmouth College, Hanover, NH 03755, USA.
| |
Collapse
|