1
|
Kawabata K, Takahashi T, Tanaka K, Kurokawa Y, Yamamoto K, Saito T, Momose K, Yamashita K, Makino T, Yokouchi T, Kawai K, Serada S, Fujimoto M, Nakajima K, Naka T, Eguchi H, Doki Y. Lipolysis-stimulated lipoprotein receptor promote lipid uptake and fatty acid oxidation in gastric cancer. Gastric Cancer 2024; 27:1258-1272. [PMID: 39294388 DOI: 10.1007/s10120-024-01552-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 09/02/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND Lipolysis-stimulated lipoprotein receptor (LSR), a lipid receptor, is associated with cancer progression. However, detailed effects on intracellular metabolism are unclear. We aimed to elucidate the mechanism of LSR-mediated lipid metabolism in gastric cancer. METHODS We investigated lipid metabolic changes induced by lipoprotein administration in gastric cancer cells and evaluated the significance of LSR expression and lipid droplets formation in gastric cancer patients. The efficacy of inhibiting β-oxidation in gastric cancer cells was also examined in vitro and vivo. RESULTS In gastric cancer cells, LSR promoted cellular uptake of lipoprotein and cell proliferation. Furthermore, the inhibition of LSR in gastric cancer cells expressing high levels of LSR counteracted both effects. Immunohistochemical analysis of human gastric cancer tissues showed that the increase in lipid droplets via LSR is a factor that influences prognosis. Lipidomics analysis of LSR-high-expressing gastric cancer cells revealed an increase in β-oxidation. Based on these results, we used etomoxir, a β-oxidation inhibitor, and found that it inhibited cell proliferation as well as the suppression of LSR. Similarly, in a mouse xenograft model of LSR-highly expressing gastric cancer cells, the tumor growth effect of high-fat diet feeding was counteracted by etomoxir, consistent with the Ki-67 labeling index. CONCLUSIONS We demonstrated that lipids are taken up into gastric cancer cells via LSR and cause an increase in β-oxidation, resulting in the promotion of cancer progression. Controlling LSR-mediated lipid metabolism may be a novel therapeutic strategy for gastric cancer.
Collapse
Affiliation(s)
- Kota Kawabata
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita Suita, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita Suita, Japan.
| | - Koji Tanaka
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita Suita, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita Suita, Japan
| | - Kazuyoshi Yamamoto
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita Suita, Japan
| | - Takuro Saito
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita Suita, Japan
| | - Kota Momose
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita Suita, Japan
| | - Kotaro Yamashita
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita Suita, Japan
| | - Tomoki Makino
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita Suita, Japan
| | - Takashi Yokouchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita Suita, Japan
| | - Kunihiko Kawai
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita Suita, Japan
| | - Satoshi Serada
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University School of Medicine, Yahaba, Japan
| | - Minoru Fujimoto
- Division of Allergy and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Yahaba, Japan
| | - Kiyokazu Nakajima
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita Suita, Japan
| | - Tetsuji Naka
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University School of Medicine, Yahaba, Japan
- Division of Allergy and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Yahaba, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita Suita, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita Suita, Japan
| |
Collapse
|
2
|
Jiang M, Wang X, Chen Z, Wang X, An Y, Ding L, Xu M, Fan B, Jiao P, Wang C, Wang M, Sun H, Zhao S, Gong Y. Lipolysis-Stimulated Lipoprotein Receptor in Proximal Tubule, BMP-SMAD Signaling, and Kidney Disease. J Am Soc Nephrol 2024; 35:1016-1033. [PMID: 38809616 PMCID: PMC11377808 DOI: 10.1681/asn.0000000000000382] [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: 12/17/2023] [Accepted: 05/14/2024] [Indexed: 05/31/2024] Open
Abstract
Key Points
We identify that lipolysis-stimulated lipoprotein receptor is highly enriched in the nucleus of mouse and human kidney.This study provides new insights into the role of lipolysis-stimulated lipoprotein receptor in kidney disease.
Background
Lipolysis-stimulated lipoprotein receptor (LSR) is a single-pass membrane protein that plays essential roles in tricellular tight junction organization in the epithelium and endothelium, but its function in kidney physiology and disease development remains unknown.
Methods
Conditional Lsr deletion mice were generated and analyzed to investigate the function of LSR in proximal tubule. Unilateral ischemia-reperfusion was used as an injury model to investigate the role of LSR in AKI and the progression to CKD. Detailed mechanistic analyses were conducted using whole-transcriptome RNA sequencing, immunofluorescence, dual-luciferase reporter gene assay, coimmunoprecipitation, RNA immunoprecipitation, and adeno-associated virus-mediated gene overexpression and knockdown.
Results
The nuclear localization of LSR was found in the kidney. Proximal tubule–specific Lsr knockout mice exhibited alleviated kidney damage and fibrosis compared with those in wild-type mice in response unilateral ischemia-reperfusion injury. Loss of LSR resulted in downregulation of Chrdl1 and activation of bone morphogenetic protein (BMP)-mothers against decapentaplegic homolog (SMAD) signaling in proximal tubules. Treatment with CHRDL1 counteracted the protective effect of LSR deletion in the unilaterally ischemic injured kidney. In addition, the systemic delivery of Chrdl1 short hairpin RNA attenuated injury-induced kidney fibrosis. LSR formed a complex with 14-3-3θ in the nucleus of proximal tubular cells, thereby reducing the interaction between human antigen R and 14-3-3θ, consequently leading to the translocation of unbound human antigen R to the cytoplasm. The absence of LSR promoted the association of 14-3-3θ with human antigen R, potentially resulting in decreased human antigen R levels in the cytoplasm. Reduced human antigen R levels impaired Chrdl1 mRNA stability, subsequently leading to the activation of BMP-SMAD signaling.
Conclusions
Deletion of LSR in proximal tubule deregulated Chrdl1 to activate BMP-SMAD signaling and ameliorated kidney disease.
Collapse
Affiliation(s)
- Min Jiang
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, China
- Department of Pharmacology, Binzhou Medical University, Yantai, China
| | - Xiangdong Wang
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Zhenni Chen
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Xin Wang
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Yanan An
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, China
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Lixia Ding
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Mengyuan Xu
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Baozhen Fan
- Department of Urology, Binzhou Medical University Hospital, Binzhou, China
| | - Peng Jiao
- Department of Urology, Binzhou Medical University Hospital, Binzhou, China
| | - Chao Wang
- Department of Urology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Mingxia Wang
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, China
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Hui Sun
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, China
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Shengtian Zhao
- Department of Urology, Binzhou Medical University Hospital, Binzhou, China
- Shandong Provincial Engineering Laboratory of Urologic Tissue Reconstruction, Jinan, China
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yongfeng Gong
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, China
- Department of Physiology, Binzhou Medical University, Yantai, China
| |
Collapse
|
3
|
Papatheodorou P, Minton NP, Aktories K, Barth H. An Updated View on the Cellular Uptake and Mode-of-Action of Clostridioides difficile Toxins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1435:219-247. [PMID: 38175478 DOI: 10.1007/978-3-031-42108-2_11] [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] [Indexed: 01/05/2024]
Abstract
Research on the human gut pathogen Clostridioides (C.) difficile and its toxins continues to attract much attention as a consequence of the threat to human health posed by hypervirulent strains. Toxin A (TcdA) and Toxin B (TcdB) are the two major virulence determinants of C. difficile. Both are single-chain proteins with a similar multidomain architecture. Certain hypervirulent C. difficile strains also produce a third toxin, namely binary toxin CDT (C. difficile transferase). C. difficile toxins are the causative agents of C. difficile-associated diseases (CDADs), such as antibiotics-associated diarrhea and pseudomembranous colitis. For that reason, considerable efforts have been expended to unravel their molecular mode-of-action and the cellular mechanisms responsible for their uptake. Many of these studies have been conducted in European laboratories. Here, we provide an update on our previous review (Papatheodorou et al. Adv Exp Med Biol, 2018) on important advances in C. difficile toxins research.
Collapse
Affiliation(s)
- Panagiotis Papatheodorou
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany.
| | - Nigel P Minton
- BBSRC/EPSRC Synthetic Biology Research Centre, University of Nottingham, Nottingham, UK
| | - Klaus Aktories
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Holger Barth
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
4
|
An Y, Wang C, Fan B, Wang Z, Li Y, Kong F, Zhou C, Cao Z, Wang M, Sun H, Zhao S, Gong Y. LSR targets YAP to modulate intestinal Paneth cell differentiation. Cell Rep 2023; 42:113118. [PMID: 37703178 DOI: 10.1016/j.celrep.2023.113118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 07/26/2023] [Accepted: 08/25/2023] [Indexed: 09/15/2023] Open
Abstract
Lipolysis-stimulated lipoprotein receptor (LSR) is a multi-functional protein that is best known for its roles in assembly of epithelial tricellular tight junctions and hepatic clearance of lipoproteins. Here, we investigated whether LSR contributes to intestinal epithelium homeostasis and pathogenesis of intestinal disease. By using multiple conditional deletion mouse models and ex vivo cultured organoids, we find that LSR elimination in intestinal stem cells results in the disappearance of Paneth cells without affecting the differentiation of other cell lineages. Mechanistic studies reveal that LSR deficiency increases abundance of YAP by modulating its phosphorylation and proteasomal degradation. Using gain- and loss-of-function studies, we show that LSR protects against necrotizing enterocolitis through enhancement of Paneth cell differentiation in small-intestinal epithelium. Thus, this study identifies LSR as an upstream negative regulator of YAP activity, an essential factor for Paneth cell differentiation, and a potential therapeutic target for necrotizing enterocolitis.
Collapse
Affiliation(s)
- Yanan An
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China; Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
| | - Chao Wang
- Department of Urology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Baozhen Fan
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Ziqi Wang
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Ying Li
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Feng Kong
- Shandong Provincial Engineering Laboratory of Urologic Tissue Reconstruction, Jinan, Shandong, China; Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chengjun Zhou
- Department of Pathology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhang Cao
- Department of Pathology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Mingxia Wang
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Hui Sun
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Shengtian Zhao
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China; Shandong Provincial Engineering Laboratory of Urologic Tissue Reconstruction, Jinan, Shandong, China; Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Yongfeng Gong
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China; Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China.
| |
Collapse
|
5
|
Lipolysis-stimulated lipoprotein receptor-targeted antibody-drug conjugate demonstrates potent antitumor activity against epithelial ovarian cancer. Neoplasia 2022; 35:100853. [PMID: 36413881 PMCID: PMC9679668 DOI: 10.1016/j.neo.2022.100853] [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: 06/08/2022] [Revised: 11/05/2022] [Accepted: 11/08/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) is a lethal malignant tumor, for which new treatment options are urgently required. Lipolysis-stimulated lipoprotein receptor (LSR) is widely expressed in EOC, and it is associated with poor prognosis. In this study, we developed an antibody-drug conjugate (ADC) targeting LSR as a new therapeutic approach to EOC. METHODS We, herein, developed novel anti-LSR monoclonal antibodies (mAbs) and an LSR-ADC by conjugating monomethyl auristatin E as a payload. We subsequently evaluated the in vitro and in vivo (on xenograft models) antitumor effect of the LSR-ADC. RESULTS An overexpression of LSR was observed not only in the primary EOC tumor but also in its lymph node and omental metastases. The EOC cell lines NOVC7-C and OVCAR3 strongly expressed LSR (as compared to ES2 cells). Both the anti-LSR mAb and the LSR-ADC were able to specifically bind to LSR-positive cells and were rapidly internalized and trafficked to the lysosomes. The LSR-ADC demonstrated a potent antitumor effect against NOVC-7C and OVCAR3, but little activity against ES2 cells. In vitro, the LSR-ADC exhibited a potent antitumor effect against NOVC-7C and OVCAR3. Moreover, in the OVCAR3 xenograft models as well as in the patient-derived xenograft models of LSR-positive EOC, the LSR-ADC significantly inhibited tumor growth. The LSR-ADC also suppressed the omental/bowel metastases in OVCAR3-Luc xenografts and improved the median survival. CONCLUSION The developed LSR-ADC demonstrated a significant antitumor activity against LSR-positive EOC cell lines and tumors. Our preclinical data support the use of the LSR-ADC as a novel therapy for patients with LSR-positive ovarian cancer.
Collapse
|
6
|
Voggel J, Fink G, Zelck M, Wohlfarth M, Post JM, Bindila L, Rauh M, Amann K, Alejandre Alcázar MA, Dötsch J, Nüsken KD, Nüsken E. Elevated n-3/n-6 PUFA ratio in early life diet reverses adverse intrauterine kidney programming in female rats. J Lipid Res 2022; 63:100283. [PMID: 36152882 PMCID: PMC9619183 DOI: 10.1016/j.jlr.2022.100283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/30/2022] [Accepted: 09/11/2022] [Indexed: 11/27/2022] Open
Abstract
Intrauterine growth restriction (IUGR) predisposes to chronic kidney disease via activation of proinflammatory pathways, and omega-3 PUFAs (n-3 PUFAs) have anti-inflammatory properties. In female rats, we investigated 1) how an elevated dietary n-3/n-6 PUFA ratio (1:1) during postnatal kidney development modifies kidney phospholipid (PL) and arachidonic acid (AA) metabolite content and 2) whether the diet counteracts adverse molecular protein signatures expected in IUGR kidneys. IUGR was induced by bilateral uterine vessel ligation or intrauterine stress through sham operation 3.5 days before term. Control (C) offspring were born after uncompromised pregnancy. On postnatal (P) days P2–P39, rats were fed control (n-3/n-6 PUFA ratio 1:20) or n-3 PUFA intervention diet (N3PUFA; ratio 1:1). Plasma parameters (P33), kidney cortex lipidomics and proteomics, as well as histology (P39) were studied. We found that the intervention diet tripled PL-DHA content (PC 40:6; P < 0.01) and lowered both PL-AA content (PC 38:4 and lyso-phosphatidylcholine 20:4; P < 0.05) and AA metabolites (HETEs, dihydroxyeicosatrienoic acids, and epoxyeicosatrienoic acids) to 25% in all offspring groups. After ligation, our network analysis of differentially expressed proteins identified an adverse molecular signature indicating inflammation and hypercoagulability. N3PUFA diet reversed 61 protein alterations (P < 0.05), thus mitigating adverse IUGR signatures. In conclusion, an elevated n-3/n-6 PUFA ratio in early diet strongly reduces proinflammatory PLs and mediators while increasing DHA-containing PLs regardless of prior intrauterine conditions. Counteracting a proinflammatory hypercoagulable protein signature in young adult IUGR individuals through early diet intervention may be a feasible strategy to prevent developmentally programmed kidney damage in later life.
Collapse
Affiliation(s)
- Jenny Voggel
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Gregor Fink
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Magdalena Zelck
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Maria Wohlfarth
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Julia M Post
- Clinical Lipidomics Unit, Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Laura Bindila
- Clinical Lipidomics Unit, Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Erlangen, Germany
| | - Kerstin Amann
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-University Erlangen, Erlangen, Germany
| | - Miguel A Alejandre Alcázar
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Institute for Lung Health, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Gießen, Germany
| | - Jörg Dötsch
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Kai-Dietrich Nüsken
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Eva Nüsken
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany.
| |
Collapse
|
7
|
Lipolysis-Stimulated Lipoprotein Receptor Acts as Sensor to Regulate ApoE Release in Astrocytes. Int J Mol Sci 2022; 23:ijms23158630. [PMID: 35955777 PMCID: PMC9368974 DOI: 10.3390/ijms23158630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 11/17/2022] Open
Abstract
Astroglia play an important role, providing de novo synthesized cholesterol to neurons in the form of ApoE-lipidated particles; disruption of this process can increase the risk of Alzheimer’s disease. We recently reported that glia-specific suppression of the lipolysis-stimulated lipoprotein receptor (LSR) gene leads to Alzheimer’s disease-like memory deficits. Since LSR is an Apo-E lipoprotein receptor, our objective of this study was to determine the effect of LSR expression modulation on cholesterol and ApoE output in mouse astrocytes expressing human ApoE3. qPCR analysis showed that siRNA-mediated lsr knockdown significantly increased expression of the genes involved in cholesterol synthesis, secretion, and metabolism. Analysis of media and lipoprotein fractions showed increased cholesterol and lipidated ApoE output in HDL-like particles. Further, lsr expression could be upregulated when astrocytes were incubated 5 days in media containing high levels (two-fold) of lipoprotein, or after 8 h treatment with 1 µM LXR agonist T0901317 in lipoprotein-deficient media. In both conditions of increased lsr expression, the ApoE output was repressed or unchanged despite increased abca1 mRNA levels and cholesterol production. We conclude that LSR acts as a sensor of lipoprotein content in the medium and repressor of ApoE release, while ABCA1 drives cholesterol efflux, thereby potentially affecting cholesterol load, ApoE lipidation, and limiting cholesterol trafficking towards the neuron.
Collapse
|
8
|
Nagase Y, Hiramatsu K, Funauchi M, Shiomi M, Masuda T, Kakuda M, Nakagawa S, Miyoshi A, Matsuzaki S, Kobayashi E, Kimura T, Serada S, Ueda Y, Naka T, Kimura T. Anti-lipolysis-stimulated lipoprotein receptor monoclonal antibody as a novel therapeutic agent for endometrial cancer. BMC Cancer 2022; 22:679. [PMID: 35729527 PMCID: PMC9210735 DOI: 10.1186/s12885-022-09789-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 06/16/2022] [Indexed: 12/29/2022] Open
Abstract
Background Endometrial cancer (EC) is a common gynecologic malignancy and patients with advanced and recurrent EC have a poor prognosis. Although chemotherapy is administered for those patients, the efficacy of current chemotherapy is limited. Therefore, it is necessary to develop novel therapeutic agents for EC. In this study, we focused on lipolysis-stimulated lipoprotein receptor (LSR), a membrane protein highly expressed in EC cells, and developed a chimeric chicken–mouse anti-LSR monoclonal antibody (mAb). This study investigated the antitumor effect of an anti-LSR mAb and the function of LSR in EC. Methods We examined the expression of LSR in 228 patients with EC using immunohistochemistry and divided them into two groups: high-LSR (n = 153) and low-LSR groups (n = 75). We developed a novel anti-LSR mAb and assessed its antitumor activity in an EC cell xenograft mouse model. Pathway enrichment analysis was performed using protein expression data of EC samples. LSR-knockdown EC cell lines (HEC1 and HEC116) were generated by transfected with small interfering RNA and used for assays in vitro. Results High expression of LSR was associated with poor overall survival (hazard ratio: 3.53, 95% confidence interval: 1.35–9.24, p = 0.01), advanced stage disease (p = 0.045), deep myometrial invasion (p = 0.045), and distant metastasis (p < 0.01). In EC with deep myometrial invasion, matrix metalloproteinase (MMP) 2 was highly expressed along with LSR. Anti-LSR mAb significantly inhibited the tumor growth in EC cell xenograft mouse model (tumor volume, 407.1 mm3versus 726.3 mm3, p = 0.019). Pathway enrichment analysis identified the mitogen-activated protein kinase (MAPK) pathway as a signaling pathway associated with LSR expression. Anti-LSR mAb suppressed the activity of MAPK in vivo. In vitro assays using EC cell lines demonstrated that LSR regulated cell proliferation, invasion, and migration through MAPK signaling, particularly MEK/ERK signaling and membrane-type 1 MMP (MT1-MMP) and MMP2. Moreover, ERK1/2-knockdown suppressed cell proliferation, invasion, migration, and the expression of MT1-MMP and MMP2. Conclusions Our results suggest that LSR contributes to tumor growth, invasion, metastasis, and poor prognosis of EC through MAPK signaling. Anti-LSR mAb is a potential therapeutic agent for EC. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09789-6.
Collapse
Affiliation(s)
- Yoshikazu Nagase
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kosuke Hiramatsu
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Masashi Funauchi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Division of Clinical Immunology, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan.,Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Iwate, Japan.,Department of Clinical Immunology, Kochi Medical School, Kochi University, Kochi, Japan
| | - Mayu Shiomi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tatsuo Masuda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Mamoru Kakuda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Satoshi Nakagawa
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ai Miyoshi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shinya Matsuzaki
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Gynecology, Osaka International Cancer Institute, Osaka, Japan
| | - Eiji Kobayashi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Toshihiro Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Satoshi Serada
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Iwate, Japan.,Department of Clinical Immunology, Kochi Medical School, Kochi University, Kochi, Japan
| | - Yutaka Ueda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tetsuji Naka
- Division of Clinical Immunology, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan.,Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Iwate, Japan.,Department of Clinical Immunology, Kochi Medical School, Kochi University, Kochi, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
9
|
Weiß F, Holthaus D, Kraft M, Klotz C, Schneemann M, Schulzke JD, Krug SM. Human duodenal organoid-derived monolayers serve as a suitable barrier model for duodenal tissue. Ann N Y Acad Sci 2022; 1515:155-167. [PMID: 35666953 DOI: 10.1111/nyas.14804] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Usually, duodenal barriers are investigated using intestinal cell lines like Caco-2, which in contrast to native tissue are limited in cell-type representation. Organoids can consist of all intestinal cell types and are supposed to better reflect the in vivo situation. Growing three-dimensionally, with the apical side facing the lumen, application of typical physiological techniques to analyze the barrier is difficult. Organoid-derived monolayers (ODMs) were developed to overcome this. After optimizing culturing conditions, ODMs were characterized and compared to Caco-2 and duodenal tissue. Tight junction composition and appearance were analyzed, and electrophysiological barrier properties, like paracellular and transcellular barrier function and macromolecule permeability, were evaluated. Furthermore, transcriptomic data were analyzed. ODMs had tight junction protein expression and paracellular barrier properties much more resembling the originating tissue than Caco-2. Transcellular barrier was similar between ODMs and native tissue but was increased in Caco-2. Transcriptomic data showed that Caco-2 expressed fewer solute carriers than ODMs and native tissue. In conclusion, while Caco-2 cells differ mostly in transcellular properties, ODMs reflect trans- and paracellular properties of the originating tissue. If cultured under optimized conditions, ODMs possess reproducible functionality, and the variety of different cell types makes them a suitable model for human tissue-specific investigations.
Collapse
Affiliation(s)
- Franziska Weiß
- Clinical Physiology/Nutritional Medicine, Charité - Universitätsmedizin Berlin, CBF, Berlin, Germany
| | - David Holthaus
- Department of Infectious Diseases, Unit 16 Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institute, Berlin, Germany
| | - Martin Kraft
- Department of Infectious Diseases, Unit 16 Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institute, Berlin, Germany
| | - Christian Klotz
- Department of Infectious Diseases, Unit 16 Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institute, Berlin, Germany
| | - Martina Schneemann
- Clinical Physiology/Nutritional Medicine, Charité - Universitätsmedizin Berlin, CBF, Berlin, Germany
| | - Jörg D Schulzke
- Clinical Physiology/Nutritional Medicine, Charité - Universitätsmedizin Berlin, CBF, Berlin, Germany
| | - Susanne M Krug
- Clinical Physiology/Nutritional Medicine, Charité - Universitätsmedizin Berlin, CBF, Berlin, Germany
| |
Collapse
|
10
|
SAINI SIMMI, WALIA GAGANDEEPKAUR, SACHDEVA MOHINDERPAL, GUPTA VIPIN. Genomics of body fat distribution. J Genet 2021. [DOI: 10.1007/s12041-021-01281-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
11
|
Tight Junction Modulating Bioprobes for Drug Delivery System to the Brain: A Review. Pharmaceutics 2020; 12:pharmaceutics12121236. [PMID: 33352631 PMCID: PMC7767277 DOI: 10.3390/pharmaceutics12121236] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/19/2022] Open
Abstract
The blood-brain barrier (BBB), which is composed of endothelial cells, pericytes, astrocytes, and neurons, separates the brain extracellular fluid from the circulating blood, and maintains the homeostasis of the central nervous system (CNS). The BBB endothelial cells have well-developed tight junctions (TJs) and express specific polarized transport systems to tightly control the paracellular movements of solutes, ions, and water. There are two types of TJs: bicellular TJs (bTJs), which is a structure at the contact of two cells, and tricellular TJs (tTJs), which is a structure at the contact of three cells. Claudin-5 and angulin-1 are important components of bTJs and tTJs in the brain, respectively. Here, we review TJ-modulating bioprobes that enable drug delivery to the brain across the BBB, focusing on claudin-5 and angulin-1.
Collapse
|
12
|
Ehsani S. COVID-19 and iron dysregulation: distant sequence similarity between hepcidin and the novel coronavirus spike glycoprotein. Biol Direct 2020; 15:19. [PMID: 33066821 PMCID: PMC7563913 DOI: 10.1186/s13062-020-00275-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022] Open
Abstract
The spike glycoprotein of the SARS-CoV-2 virus, which causes COVID-19, has attracted attention for its vaccine potential and binding capacity to host cell surface receptors. Much of this research focus has centered on the ectodomain of the spike protein. The ectodomain is anchored to a transmembrane region, followed by a cytoplasmic tail. Here we report a distant sequence similarity between the cysteine-rich cytoplasmic tail of the coronavirus spike protein and the hepcidin protein that is found in humans and other vertebrates. Hepcidin is thought to be the key regulator of iron metabolism in humans through its inhibition of the iron-exporting protein ferroportin. An implication of this preliminary observation is to suggest a potential route of investigation in the coronavirus research field making use of an already-established literature on the interplay of local and systemic iron regulation, cytokine-mediated inflammatory processes, respiratory infections and the hepcidin protein. The question of possible homology and an evolutionary connection between the viral spike protein and hepcidin is not assessed in this report, but some scenarios for its study are discussed.
Collapse
Affiliation(s)
- Sepehr Ehsani
- Theoretical and Philosophical Biology, Department of Philosophy, University College London, Bloomsbury, London, WC1E 6BT, UK.
- Ronin Institute for Independent Scholarship, Montclair, NJ, 07043, USA.
| |
Collapse
|
13
|
Tachibana K, Kondoh M. A Method to Prepare a Bioprobe for Regulatory Science of the Drug Delivery System to the Brain: An Angulin Binder to Modulate Tricellular Tight Junction-Seal. Methods Mol Biol 2020; 2367:291-304. [PMID: 32789775 DOI: 10.1007/7651_2020_317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epithelium acts as a barrier separating the interior and exterior of the body, and the epithelial and endothelial cells form tight junctions (TJs) by sealing the paracellular space. The blood-brain barrier (BBB) endothelial cells have well-developed TJs and express specific polarized transport systems to tightly control paracellular movements of solutes, ions, and water. Thus, more than 98% of small-molecular-weight drugs cannot pass the BBB. The tricellular TJ (tTJ) is a structure at contacts of three cells. Angulin-1, also known as lipolysis-stimulated lipoprotein receptor (LSR), is one of angulin family and is abundantly expressed in brain endothelial cells, which plays an important role in barrier function of the BBB. The C-terminal domain of a receptor-binding component of Clostridium perfringens iota-toxin (Ib421-664), also named as angubindin-1, binds to its receptors angulin-1 and angulin-3. This angubindin-1 modulates the tTJ barrier and is able to deliver a 16-mer gapmer antisense oligonucleotide (5.3 kDa) without adverse effects. Thus, angulin binders, such as angubindin-1, are useful tools for studying the safety assessment of tTJ-targeted drug delivery and BBB permeability modulation. Here, we provide a protocol for the expression and purification of recombinant angubindin-1 protein as angulin binders, an analysis method for angubindin-1 binding affinity, and a procedure for assessing the effect of modulating tight junction integrity.
Collapse
Affiliation(s)
- Keisuke Tachibana
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
| | - Masuo Kondoh
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
| |
Collapse
|
14
|
Stiles BG. Clostridial Binary Toxins: Basic Understandings that Include Cell Surface Binding and an Internal "Coup de Grâce". Curr Top Microbiol Immunol 2019; 406:135-162. [PMID: 27380267 DOI: 10.1007/82_2016_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Clostridium species can make a remarkable number of different protein toxins, causing many diverse diseases in humans and animals. The binary toxins of Clostridium botulinum, C. difficile, C. perfringens, and C. spiroforme are one group of enteric-acting toxins that attack the actin cytoskeleton of various cell types. These enterotoxins consist of A (enzymatic) and B (cell binding/membrane translocation) components that assemble on the targeted cell surface or in solution, forming a multimeric complex. Once translocated into the cytosol via endosomal trafficking and acidification, the A component dismantles the filamentous actin-based cytoskeleton via mono-ADP-ribosylation of globular actin. Knowledge of cell surface receptors and how these usurped, host-derived molecules facilitate intoxication can lead to novel ways of defending against these clostridial binary toxins. A molecular-based understanding of the various steps involved in toxin internalization can also unveil therapeutic intervention points that stop the intoxication process. Furthermore, using these bacterial proteins as medicinal shuttle systems into cells provides intriguing possibilities in the future. The pertinent past and state-of-the-art present, regarding clostridial binary toxins, will be evident in this chapter.
Collapse
Affiliation(s)
- Bradley G Stiles
- Biology Department, Wilson College, Chambersburg, PA, 17201, USA.
| |
Collapse
|
15
|
Receptor-Binding and Uptake of Binary Actin-ADP-Ribosylating Toxins. Curr Top Microbiol Immunol 2019; 406:119-133. [PMID: 27817176 DOI: 10.1007/82_2016_46] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Binary actin-ADP-ribosylating toxins (e.g., Clostridium botulinum C2 toxin or Clostridium perfringens iota toxin ) consist of two separate proteins: An ADP-ribosyltransferase, which modifies actin thereby inhibiting actin polymerization, and a binding component that forms heptamers after proteolytic activation. While C2 toxin interacts with carbohydrate structures on host cells, the group of iota-like toxins binds to lipolysis-stimulated lipoprotein receptor (LSR). Here, we review LSR and discuss the role and function of LSR in interaction of iota-like toxins with host cells.
Collapse
|
16
|
El Hajj A, Yen FT, Oster T, Malaplate C, Pauron L, Corbier C, Lanhers MC, Claudepierre T. Age-related changes in regiospecific expression of Lipolysis Stimulated Receptor (LSR) in mice brain. PLoS One 2019; 14:e0218812. [PMID: 31233547 PMCID: PMC6590887 DOI: 10.1371/journal.pone.0218812] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/10/2019] [Indexed: 11/18/2022] Open
Abstract
The regulation of cholesterol, an essential brain lipid, ensures proper neuronal development and function, as demonstrated by links between perturbations of cholesterol metabolism and neurodegenerative diseases, including Alzheimer’s disease. The central nervous system (CNS) acquires cholesterol via de novo synthesis, where glial cells provide cholesterol to neurons. Both lipoproteins and lipoprotein receptors are key elements in this intercellular transport, where the latter recognize, bind and endocytose cholesterol containing glia-produced lipoproteins. CNS lipoprotein receptors are like those in the periphery, among which include the ApoB, E binding lipolysis stimulated lipoprotein receptor (LSR). LSR is a multimeric protein complex that has multiple isoforms including α and α’, which are seen as a doublet at 68 kDa, and β at 56 kDa. While complete inactivation of murine lsr gene is embryonic lethal, studies on lsr +/- mice revealed altered brain cholesterol distribution and cognitive functions. In the present study, LSR profiling in different CNS regions revealed regiospecific expression of LSR at both RNA and protein levels. At the RNA level, the hippocampus, hypothalamus, cerebellum, and olfactory bulb, all showed high levels of total lsr compared to whole brain tissues, whereas at the protein level, only the hypothalamus, olfactory bulb, and retina showed the highest levels of total LSR. Interestingly, major regional changes in LSR expression were observed in aged mice which suggests changes in cholesterol homeostasis in specific structures in the aging brain. Immunocytostaining of primary cultures of mature murine neurons and glial cells isolated from different CNS regions showed that LSR is expressed in both neurons and glial cells. However, lsr RNA expression in the cerebellum was predominantly higher in glial cells, which was confirmed by the immunocytostaining profile of cerebellar neurons and glia. Based on this observation, we would propose that LSR in glial cells may play a key role in glia-neuron cross talk, particularly in the feedback control of cholesterol synthesis to avoid cholesterol overload in neurons and to maintain proper functioning of the brain throughout life.
Collapse
Affiliation(s)
- Aseel El Hajj
- Qualivie, UR AFPA laboratory, ENSAIA, University of Lorraine, Vandoeuvre-les-Nancy, Lorraine, France
| | - Frances T. Yen
- Qualivie, UR AFPA laboratory, ENSAIA, University of Lorraine, Vandoeuvre-les-Nancy, Lorraine, France
- * E-mail: (TC); (FTY)
| | - Thierry Oster
- Qualivie, UR AFPA laboratory, ENSAIA, University of Lorraine, Vandoeuvre-les-Nancy, Lorraine, France
| | - Catherine Malaplate
- Qualivie, UR AFPA laboratory, ENSAIA, University of Lorraine, Vandoeuvre-les-Nancy, Lorraine, France
| | - Lynn Pauron
- Qualivie, UR AFPA laboratory, ENSAIA, University of Lorraine, Vandoeuvre-les-Nancy, Lorraine, France
| | - Catherine Corbier
- Qualivie, UR AFPA laboratory, ENSAIA, University of Lorraine, Vandoeuvre-les-Nancy, Lorraine, France
| | - Marie-Claire Lanhers
- Qualivie, UR AFPA laboratory, ENSAIA, University of Lorraine, Vandoeuvre-les-Nancy, Lorraine, France
| | - Thomas Claudepierre
- Qualivie, UR AFPA laboratory, ENSAIA, University of Lorraine, Vandoeuvre-les-Nancy, Lorraine, France
- * E-mail: (TC); (FTY)
| |
Collapse
|
17
|
Nagahama M, Takehara M, Kobayashi K. Interaction of Clostridium perfringens Iota Toxin and Lipolysis-Stimulated Lipoprotein Receptor (LSR). Toxins (Basel) 2018; 10:toxins10100405. [PMID: 30297616 PMCID: PMC6215307 DOI: 10.3390/toxins10100405] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 09/29/2018] [Accepted: 10/04/2018] [Indexed: 01/05/2023] Open
Abstract
Iota toxin produced by Clostridium perfringens is a binary, actin ADP-ribosylating toxin that is organized into the enzymatically active component Ia and the binding component Ib. Lipolysis-stimulated lipoprotein receptor (LSR) has been identified as a cellular receptor of Ib. Here, we investigated the functional interaction between Ib and LSR, where siRNA for LSR blocked the toxin-mediated cytotoxicity and the binding of Ib. The addition of Ib to LSR-green fluorescence protein (GFP)-transfected cells at 4 °C resulted in colocalization with LSR and Ib on the cell surface. Upon transfer of the cells from 4 °C to 37 °C, LSR and Ib were internalized and observed in cytoplasmic vesicles. When the cells were incubated with Ib at 37 °C and fractionated using the Triton-insoluble membrane, Ib oligomer was localized in insoluble factions that fulfilled the criteria of lipid rafts, and LSR was clustered in lipid rafts. To examine the interaction between N-terminal extracellular region of LSR and Ib, we constructed a series of LSR N-terminal deletions. Ten amino acids residues can be deleted from this end without any reduction of Ib binding. However, deletion of 15 N-terminal residues drastically reduces its ability to bind Ib. These results demonstrate that Ib binds to the LSR N-terminal 10 to 15 residues and endocytoses into trafficking endosomes together with LSR.
Collapse
Affiliation(s)
- Masahiro Nagahama
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan.
| | - Masaya Takehara
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan.
| | - Keiko Kobayashi
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan.
| |
Collapse
|
18
|
Sugase T, Takahashi T, Serada S, Fujimoto M, Ohkawara T, Hiramatsu K, Koh M, Saito Y, Tanaka K, Miyazaki Y, Makino T, Kurokawa Y, Yamasaki M, Nakajima K, Hanazaki K, Mori M, Doki Y, Naka T. Lipolysis-stimulated lipoprotein receptor overexpression is a novel predictor of poor clinical prognosis and a potential therapeutic target in gastric cancer. Oncotarget 2018; 9:32917-32928. [PMID: 30250639 PMCID: PMC6152476 DOI: 10.18632/oncotarget.25952] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/28/2018] [Indexed: 01/01/2023] Open
Abstract
The prognosis of patients with advanced gastric cancer (GC) remains poor despite the recent advances in molecular targeted therapies, and the search for biomarkers that can predict prognosis and additional new agents with acceptable toxicity profiles are needed. Lipolysis-stimulated lipoprotein receptor (LSR) is a lipoprotein receptor that binds to triglyceride-rich lipoproteins and related to some malignancies. Herein, we examined the association between LSR expression and the prognosis of patients with GC, and investigated the antitumor effect of a previously developed anti-human LSR monoclonal antibody (#1-25). We first performed immunohistochemical analysis of LSR protein expression in GC and normal tissues, and then examined its association with the prognosis of 110 patients with GC. LSR was overexpressed in most of primary GC and metastatic tumors, but not in normal tissues. Patients with strong LSR expression (N = 80, 72.7%) had significantly poorer overall survival (OS) than those with weak expression (P = 0.017). Multivariate analysis identified strong LSR (as well as pT) as independent and significant prognostic factors for OS. Next, we demonstrated that very low density lipoprotein (VLDL) treatment increases cell proliferation in LSR-expressing GC cell lines in vitro; LSR inhibition using #1-25 inhibited VLDL-induced proliferation by suppressing JAK/STAT and PI3K signaling. In vivo, we demonstrated a marked antitumor effect of #1-25 in 2 distinct GC cell line xenograft mice models. Our findings suggest that LSR plays a key functional role in GC development, and that this antigen can be therapeutically targeted to improve GC treatment.
Collapse
Affiliation(s)
- Takahito Sugase
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan.,Center for Intractable Immune Disease, Kochi University, Nankoku, Japan.,Department of Surgery, Kochi University, Nankoku, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Satoshi Serada
- Center for Intractable Immune Disease, Kochi University, Nankoku, Japan
| | - Minoru Fujimoto
- Center for Intractable Immune Disease, Kochi University, Nankoku, Japan
| | - Tomoharu Ohkawara
- Center for Intractable Immune Disease, Kochi University, Nankoku, Japan
| | - Kosuke Hiramatsu
- Center for Intractable Immune Disease, Kochi University, Nankoku, Japan
| | - Masahiro Koh
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yurina Saito
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Koji Tanaka
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yasuhiro Miyazaki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tomoki Makino
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Makoto Yamasaki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kiyokazu Nakajima
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | | | - Masaki Mori
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tetsuji Naka
- Center for Intractable Immune Disease, Kochi University, Nankoku, Japan
| |
Collapse
|
19
|
Czulkies BA, Mastroianni J, Lutz L, Lang S, Schwan C, Schmidt G, Lassmann S, Zeiser R, Aktories K, Papatheodorou P. Loss of LSR affects epithelial barrier integrity and tumor xenograft growth of CaCo-2 cells. Oncotarget 2018; 8:37009-37022. [PMID: 27391068 PMCID: PMC5514888 DOI: 10.18632/oncotarget.10425] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/13/2016] [Indexed: 11/25/2022] Open
Abstract
The lipolysis-stimulated lipoprotein receptor (LSR) is a lipoprotein receptor, serves as host receptor for clostridial iota-like toxins and is involved in the formation of tricellular contacts. Of particular interest is the role of LSR in progression of various cancers. Here we aimed to study the tumor growth of LSR-deficient colon carcinoma-derived cell lines HCT116 and CaCo-2 in a mouse xenograft model. Whereas knockout of LSR had no effect on tumor growth of HCT116 cells, we observed that CaCo-2 LSR knockout tumors grew to a smaller size than their wild-type counterparts. Histological analysis revealed increased apoptotic and necrotic cell death in a tumor originating from LSR-deficient CaCo-2 cells. LSR-deficient CaCo-2 cells exhibited increased cell proliferation in vitro and an altered epithelial morphology with impaired targeting of tricellulin to tricellular contacts. In addition, loss of LSR reduced the transepithelial electrical resistance of CaCo-2 cell monolayers and increased permeability for small molecules. Moreover, LSR-deficient CaCo-2 cells formed larger cysts in 3D culture than their wild-type counterparts. Our study provides evidence that LSR affects epithelial morphology and barrier formation in CaCo-2 cells and examines for the first time the effects of LSR deficiency on the tumor growth properties of colon carcinoma-derived cell lines.
Collapse
Affiliation(s)
- Bernd A Czulkies
- Institute of Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-University (ALU), Freiburg, Germany
| | - Justin Mastroianni
- Department of Hematology and Oncology, University Medical Center, ALU, Freiburg, Germany
| | - Lisa Lutz
- Department of Pathology, University Medical Center, ALU, Freiburg, Germany
| | - Sarah Lang
- Institute of Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-University (ALU), Freiburg, Germany
| | - Carsten Schwan
- Institute of Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-University (ALU), Freiburg, Germany
| | - Gudula Schmidt
- Institute of Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-University (ALU), Freiburg, Germany
| | - Silke Lassmann
- Department of Pathology, University Medical Center, ALU, Freiburg, Germany.,German Consortium for Translational Cancer Research (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Centre for Biological Signalling Studies (BIOSS), ALU, Freiburg, Germany
| | - Robert Zeiser
- Department of Hematology and Oncology, University Medical Center, ALU, Freiburg, Germany.,Centre for Biological Signalling Studies (BIOSS), ALU, Freiburg, Germany
| | - Klaus Aktories
- Institute of Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Centre for Biological Signalling Studies (BIOSS), ALU, Freiburg, Germany.,Freiburg Institute for Advanced Studies (FRIAS), ALU, Freiburg, Germany
| | - Panagiotis Papatheodorou
- Institute of Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Present address: Institute of Pharmaceutical Biotechnology. University of Ulm, Ulm, Germany.,Present address: Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| |
Collapse
|
20
|
Cellular Uptake and Mode-of-Action of Clostridium difficile Toxins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1050:77-96. [DOI: 10.1007/978-3-319-72799-8_6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
21
|
Shimada H, Satohisa S, Kohno T, Takahashi S, Hatakeyama T, Konno T, Tsujiwaki M, Saito T, Kojima T. The roles of tricellular tight junction protein lipolysis-stimulated lipoprotein receptor in malignancy of human endometrial cancer cells. Oncotarget 2017; 7:27735-52. [PMID: 27036040 PMCID: PMC5053684 DOI: 10.18632/oncotarget.8408] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 03/16/2016] [Indexed: 02/07/2023] Open
Abstract
Lipolysis-stimulated lipoprotein receptor (LSR) has been identified as a novel molecular constituent of tricellular contacts that have a barrier function for the cellular sheet. LSR recruits tricellulin (TRIC), which is the first molecular component of tricellular tight junctions. Knockdown of LSR increases cell motility and invasion of certain cancer cells. However, the behavior and the roles of LSR in endometrial cancer remain unknown. In the present study, we investigated the behavior and roles of LSR in normal and endometrial cancer cells in vivo and in vitro. In endometriosis and endometrial cancer, LSR was observed not only in the subapical region but also throughout the lateral region as well as in normal endometrial epithelial cells in the secretory phase, and LSR in the cancer was reduced in correlation with the malignancy. Knockdown of LSR by the siRNA in cells of the endometrial cancer cell line Sawano, induced cell migration, invasion and proliferation, while TRIC relocalized from the tricellular region to the bicellular region at the membrane. In Sawano cells and normal HEEs, a decrease of LSR induced by leptin and an increase of LSR induced by adiponectin and the drugs for type 2 diabetes metformin and berberine were observed via distinct signaling pathways including JAK2/STAT. In Sawano cells, metformin and berberine prevented cell migration and invasion induced by downregulation of LSR by the siRNA and leptin treatment. The dissection of the mechanism in the downregulation of endometrial LSR during obesity is important in developing new diagnostic and therapy for endometrial cancer.
Collapse
Affiliation(s)
- Hiroshi Shimada
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Seiro Satohisa
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takayuki Kohno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Syunta Takahashi
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tsubasa Hatakeyama
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takumi Konno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mitsuhiro Tsujiwaki
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tsuyoshi Saito
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takashi Kojima
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
22
|
Hiramatsu K, Serada S, Enomoto T, Takahashi Y, Nakagawa S, Nojima S, Morimoto A, Matsuzaki S, Yokoyama T, Takahashi T, Fujimoto M, Takemori H, Ueda Y, Yoshino K, Morii E, Kimura T, Naka T. LSR Antibody Therapy Inhibits Ovarian Epithelial Tumor Growth by Inhibiting Lipid Uptake. Cancer Res 2017; 78:516-527. [DOI: 10.1158/0008-5472.can-17-0910] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 09/29/2017] [Accepted: 11/15/2017] [Indexed: 11/16/2022]
|
23
|
Higashi T, Miller AL. Tricellular junctions: how to build junctions at the TRICkiest points of epithelial cells. Mol Biol Cell 2017; 28:2023-2034. [PMID: 28705832 PMCID: PMC5509417 DOI: 10.1091/mbc.e16-10-0697] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/05/2017] [Accepted: 05/10/2017] [Indexed: 01/07/2023] Open
Abstract
Tricellular contacts are the places where three cells meet. In vertebrate epithelial cells, specialized structures called tricellular tight junctions (tTJs) and tricellular adherens junctions (tAJs) have been identified. tTJs are important for the maintenance of barrier function, and disruption of tTJ proteins contributes to familial deafness. tAJs have recently been attracting the attention of mechanobiologists because these sites are hot spots of epithelial tension. Although the molecular components, regulation, and function of tTJs and tAJs, as well as of invertebrate tricellular junctions, are beginning to be characterized, many questions remain. Here we broadly cover what is known about tricellular junctions, propose a new model for tension transmission at tAJs, and discuss key open questions.
Collapse
Affiliation(s)
- Tomohito Higashi
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Ann L Miller
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
24
|
Reaves DK, Hoadley KA, Fagan-Solis KD, Jima DD, Bereman M, Thorpe L, Hicks J, McDonald D, Troester MA, Perou CM, Fleming JM. Nuclear Localized LSR: A Novel Regulator of Breast Cancer Behavior and Tumorigenesis. Mol Cancer Res 2017; 15:165-178. [PMID: 27856957 PMCID: PMC5290211 DOI: 10.1158/1541-7786.mcr-16-0085-t] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 09/28/2016] [Accepted: 10/23/2016] [Indexed: 01/17/2023]
Abstract
Lipolysis-stimulated lipoprotein receptor (LSR) has been found in the plasma membrane and is believed to function in lipoprotein endocytosis and tight junctions. Given the impact of cellular metabolism and junction signaling pathways on tumor phenotypes and patient outcome, it is important to understand how LSR cellular localization mediates its functions. We conducted localization studies, evaluated DNA binding, and examined the effects of nuclear LSR in cells, xenografts, and clinical specimens. We found LSR within the membrane, cytoplasm, and the nucleus of breast cancer cells representing multiple intrinsic subtypes. Chromatin immunoprecipitation (ChIP) showed direct binding of LSR to DNA, and sequence analysis identified putative functional motifs and post-translational modifications of the LSR protein. While neither overexpression of transcript variants, nor pharmacologic manipulation of post-translational modification significantly altered localization, inhibition of nuclear export enhanced nuclear localization, suggesting a mechanism for nuclear retention. Coimmunoprecipitation and proximal ligation assays indicated LSR-pericentrin interactions, presenting potential mechanisms for nuclear-localized LSR. The clinical significance of LSR was evaluated using data from over 1,100 primary breast tumors, which showed high LSR levels in basal-like tumors and tumors from African-Americans. In tumors histosections, nuclear localization was significantly associated with poor outcomes. Finally, in vivo xenograft studies revealed that basal-like breast cancer cells that overexpress LSR exhibited both membrane and nuclear localization, and developed tumors with 100% penetrance, while control cells lacking LSR developed no tumors. These results show that nuclear LSR alters gene expression and may promote aggressive cancer phenotypes. IMPLICATIONS LSR functions in the promotion of aggressive breast cancer phenotypes and poor patient outcome via differential subcellular localization to alter cell signaling, bioenergetics, and gene expression. Mol Cancer Res; 15(2); 165-78. ©2016 AACR.
Collapse
Affiliation(s)
- Denise K Reaves
- Department of Biology, North Carolina Central University, Durham, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Katherine A Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Katerina D Fagan-Solis
- Department of Biology, North Carolina Central University, Durham, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Dereje D Jima
- Center for Human Health and the Environment, Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina
| | - Michael Bereman
- Center for Human Health and the Environment, Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina
| | - Lynnelle Thorpe
- Department of Biology, North Carolina Central University, Durham, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jyla Hicks
- Department of Biology, North Carolina Central University, Durham, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - David McDonald
- Department of Biology, North Carolina Central University, Durham, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Melissa A Troester
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Jodie M Fleming
- Department of Biology, North Carolina Central University, Durham, North Carolina.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Center for Human Health and the Environment, Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
25
|
Mruk DD, Cheng CY. The Mammalian Blood-Testis Barrier: Its Biology and Regulation. Endocr Rev 2015; 36:564-91. [PMID: 26357922 PMCID: PMC4591527 DOI: 10.1210/er.2014-1101] [Citation(s) in RCA: 405] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 09/03/2015] [Indexed: 12/31/2022]
Abstract
Spermatogenesis is the cellular process by which spermatogonia develop into mature spermatids within seminiferous tubules, the functional unit of the mammalian testis, under the structural and nutritional support of Sertoli cells and the precise regulation of endocrine factors. As germ cells develop, they traverse the seminiferous epithelium, a process that involves restructuring of Sertoli-germ cell junctions, as well as Sertoli-Sertoli cell junctions at the blood-testis barrier. The blood-testis barrier, one of the tightest tissue barriers in the mammalian body, divides the seminiferous epithelium into 2 compartments, basal and adluminal. The blood-testis barrier is different from most other tissue barriers in that it is not only comprised of tight junctions. Instead, tight junctions coexist and cofunction with ectoplasmic specializations, desmosomes, and gap junctions to create a unique microenvironment for the completion of meiosis and the subsequent development of spermatids into spermatozoa via spermiogenesis. Studies from the past decade or so have identified the key structural, scaffolding, and signaling proteins of the blood-testis barrier. More recent studies have defined the regulatory mechanisms that underlie blood-testis barrier function. We review here the biology and regulation of the mammalian blood-testis barrier and highlight research areas that should be expanded in future studies.
Collapse
Affiliation(s)
- Dolores D Mruk
- Center for Biomedical Research, Population Council, New York, New York 10065
| | - C Yan Cheng
- Center for Biomedical Research, Population Council, New York, New York 10065
| |
Collapse
|
26
|
Hemmasi S, Czulkies BA, Schorch B, Veit A, Aktories K, Papatheodorou P. Interaction of the Clostridium difficile Binary Toxin CDT and Its Host Cell Receptor, Lipolysis-stimulated Lipoprotein Receptor (LSR). J Biol Chem 2015; 290:14031-44. [PMID: 25882847 DOI: 10.1074/jbc.m115.650523] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Indexed: 12/17/2022] Open
Abstract
CDT (Clostridium difficile transferase) is a binary, actin ADP-ribosylating toxin frequently associated with hypervirulent strains of the human enteric pathogen C. difficile, the most serious cause of antibiotic-associated diarrhea and pseudomembranous colitis. CDT leads to the collapse of the actin cytoskeleton and, eventually, to cell death. Low doses of CDT result in the formation of microtubule-based protrusions on the cell surface that increase the adherence and colonization of C. difficile. The lipolysis-stimulated lipoprotein receptor (LSR) is the host cell receptor for CDT, and our aim was to gain a deeper insight into the interplay between both proteins. We show that CDT interacts with the extracellular, Ig-like domain of LSR with an affinity in the nanomolar range. We identified LSR splice variants in the colon carcinoma cell line HCT116 and disrupted the LSR gene in these cells by applying the CRISPR-Cas9 technology. LSR truncations ectopically expressed in LSR knock-out cells indicated that intracellular parts of LSR are not essential for plasma membrane targeting of the receptor and cellular uptake of CDT. By generating a series of N- and C-terminal truncations of the binding component of CDT (CDTb), we found that amino acids 757-866 of CDTb are sufficient for binding to LSR. With a transposon-based, random mutagenesis approach, we identified potential LSR-interacting epitopes in CDTb. This study increases our understanding about the interaction between CDT and its receptor LSR, which is key to the development of anti-toxin strategies for preventing cell entry of the toxin.
Collapse
Affiliation(s)
- Sarah Hemmasi
- From the Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, the Fakultät für Biologie
| | - Bernd A Czulkies
- From the Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, the Hermann Staudinger Graduate School
| | - Björn Schorch
- From the Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, the Fakultät für Biologie, the Spemann Graduate School of Biology and Medicine, and
| | - Antonia Veit
- From the Institut für Experimentelle und Klinische Pharmakologie und Toxikologie
| | - Klaus Aktories
- From the Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, the Centre for Biological Signalling Studies (BIOSS), Albert-Ludwigs-Universität Freiburg, D-79104 Freiburg, Germany
| | | |
Collapse
|
27
|
Sohet F, Lin C, Munji RN, Lee SY, Ruderisch N, Soung A, Arnold TD, Derugin N, Vexler ZS, Yen FT, Daneman R. LSR/angulin-1 is a tricellular tight junction protein involved in blood-brain barrier formation. ACTA ACUST UNITED AC 2015; 208:703-11. [PMID: 25753034 PMCID: PMC4362448 DOI: 10.1083/jcb.201410131] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Lipolysis-stimulated lipoprotein receptor, a component of the paracellular barrier at tricellular junctions, is necessary for proper blood–brain barrier sealing during embryogenesis. The blood–brain barrier (BBB) is a term used to describe the unique properties of central nervous system (CNS) blood vessels. One important BBB property is the formation of a paracellular barrier made by tight junctions (TJs) between CNS endothelial cells (ECs). Here, we show that Lipolysis-stimulated lipoprotein receptor (LSR), a component of paracellular junctions at points in which three cell membranes meet, is greatly enriched in CNS ECs compared with ECs in other nonneural tissues. We demonstrate that LSR is specifically expressed at tricellular junctions and that its expression correlates with the onset of BBB formation during embryogenesis. We further demonstrate that the BBB does not seal during embryogenesis in Lsr knockout mice with a leakage to small molecules. Finally, in mouse models in which BBB was disrupted, including an experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis and a middle cerebral artery occlusion (MCAO) model of stroke, LSR was down-regulated, linking loss of LSR and pathological BBB leakage.
Collapse
Affiliation(s)
- Fabien Sohet
- Department of Pharmacology and Department of Neuroscience, University of California, San Diego, La Jolla, CA 92093 Department of Pharmacology and Department of Neuroscience, University of California, San Diego, La Jolla, CA 92093
| | - Christina Lin
- Department of Anatomy, Department of Pediatrics, and Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | - Roeben N Munji
- Department of Pharmacology and Department of Neuroscience, University of California, San Diego, La Jolla, CA 92093 Department of Pharmacology and Department of Neuroscience, University of California, San Diego, La Jolla, CA 92093
| | - Seo Yeon Lee
- Department of Anatomy, Department of Pediatrics, and Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | - Nadine Ruderisch
- Department of Anatomy, Department of Pediatrics, and Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | - Allison Soung
- Department of Pharmacology and Department of Neuroscience, University of California, San Diego, La Jolla, CA 92093 Department of Pharmacology and Department of Neuroscience, University of California, San Diego, La Jolla, CA 92093
| | - Thomas D Arnold
- Department of Anatomy, Department of Pediatrics, and Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | - Nikita Derugin
- Department of Anatomy, Department of Pediatrics, and Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | - Zinaida S Vexler
- Department of Anatomy, Department of Pediatrics, and Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | - Frances T Yen
- Unité de Recherche Animal et Fonctionnalités des Produits Animaux (URAFPA), EA3998, Université de Lorraine, 54000 Nancy, France
| | - Richard Daneman
- Department of Pharmacology and Department of Neuroscience, University of California, San Diego, La Jolla, CA 92093 Department of Pharmacology and Department of Neuroscience, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
28
|
Layeghkhavidaki H, Lanhers MC, Akbar S, Gregory-Pauron L, Oster T, Grova N, Appenzeller B, Jasniewski J, Feidt C, Corbier C, Yen FT. Inhibitory action of benzo[α]pyrene on hepatic lipoprotein receptors in vitro and on liver lipid homeostasis in mice. PLoS One 2014; 9:e102991. [PMID: 25054229 PMCID: PMC4108373 DOI: 10.1371/journal.pone.0102991] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 06/25/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Dyslipidemia associated with obesity often manifests as increased plasma LDL and triglyceride-rich lipoprotein levels suggesting changes in hepatic lipoprotein receptor status. Persistent organic pollutants have been recently postulated to contribute to the obesity etiology by increasing adipogenesis, but little information is available on their potential effect on hepatic lipoprotein metabolism. OBJECTIVE The objective of this study was to investigate the effect of the common environmental pollutant, benzo[α]pyrene (B[α]P) on two lipoprotein receptors, the LDL-receptor and the lipolysis-stimulated lipoprotein receptor (LSR) as well as the ATP-binding cassette transporter A1 (ABCA1) using cell and animal models. RESULTS LSR, LDL-receptor as well as ABCA1 protein levels were significantly decreased by 26-48% in Hepa1-6 cells incubated (<2 h) in the presence of B[α]P (≤1 µM). Real-time PCR analysis and lactacystin studies revealed that this effect was due primarily to increased proteasome, and not lysosomal-mediated degradation rather than decreased transcription. Furthermore, ligand blots revealed that lipoproteins exposed to 1 or 5 µM B[α]P displayed markedly decreased (42-86%) binding to LSR or LDL-receptor. B[α]P-treated (0.5 mg/kg/48 h, i.p. 15 days) C57BL/6J mice displayed higher weight gain, associated with significant increases in plasma cholesterol, triglycerides, and liver cholesterol content, and decreased hepatic LDL-receptor and ABCA1 levels. Furthermore, correlational analysis revealed that B[α]P abolished the positive association observed in control mice between the LSR and LDL-receptor. Interestingly, levels of other proteins involved in liver cholesterol metabolism, ATP-binding cassette transporter G1 and scavenger receptor-BI, were decreased, while those of acyl-CoA:cholesterol acyltransferase 1 and 2 were increased in B[α]P-treated mice. CONCLUSIONS B[α]P demonstrates inhibitory action on LSR and LDL-R, as well as ABCA1, which we propose leads to modified lipid status in B[α]P-treated mice, thus providing new insight into mechanisms underlying the involvement of pollutants in the disruption of lipid homeostasis, potentially contributing to dyslipidemia associated with obesity.
Collapse
Affiliation(s)
- Hamed Layeghkhavidaki
- Unité de Recherche Animal et Fonctionnalités des Produits Animaux EA3998, Université de Lorraine, Vandœuvre-lès-Nancy, France
- Institut National de Recherche Agronomique USC 0340, Vandœuvre-lès-Nancy, France
| | - Marie-Claire Lanhers
- Unité de Recherche Animal et Fonctionnalités des Produits Animaux EA3998, Université de Lorraine, Vandœuvre-lès-Nancy, France
- Institut National de Recherche Agronomique USC 0340, Vandœuvre-lès-Nancy, France
| | - Samina Akbar
- Unité de Recherche Animal et Fonctionnalités des Produits Animaux EA3998, Université de Lorraine, Vandœuvre-lès-Nancy, France
- Institut National de Recherche Agronomique USC 0340, Vandœuvre-lès-Nancy, France
| | - Lynn Gregory-Pauron
- Unité de Recherche Animal et Fonctionnalités des Produits Animaux EA3998, Université de Lorraine, Vandœuvre-lès-Nancy, France
- Institut National de Recherche Agronomique USC 0340, Vandœuvre-lès-Nancy, France
| | - Thierry Oster
- Unité de Recherche Animal et Fonctionnalités des Produits Animaux EA3998, Université de Lorraine, Vandœuvre-lès-Nancy, France
- Institut National de Recherche Agronomique USC 0340, Vandœuvre-lès-Nancy, France
| | - Nathalie Grova
- Laboratory of Analytical Human Biomonitoring, Centre de Recherche Public de la Santé, Luxembourg, Luxembourg
| | - Brice Appenzeller
- Laboratory of Analytical Human Biomonitoring, Centre de Recherche Public de la Santé, Luxembourg, Luxembourg
| | - Jordane Jasniewski
- Laboratoire d'Ingenérie des Biomolécules, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Cyril Feidt
- Unité de Recherche Animal et Fonctionnalités des Produits Animaux EA3998, Université de Lorraine, Vandœuvre-lès-Nancy, France
- Institut National de Recherche Agronomique USC 0340, Vandœuvre-lès-Nancy, France
| | - Catherine Corbier
- Unité de Recherche Animal et Fonctionnalités des Produits Animaux EA3998, Université de Lorraine, Vandœuvre-lès-Nancy, France
- Institut National de Recherche Agronomique USC 0340, Vandœuvre-lès-Nancy, France
| | - Frances T. Yen
- Unité de Recherche Animal et Fonctionnalités des Produits Animaux EA3998, Université de Lorraine, Vandœuvre-lès-Nancy, France
- Institut National de Recherche Agronomique USC 0340, Vandœuvre-lès-Nancy, France
- Institut National de la Santé et de la Recherche Médicale, Vandœuvre-lès-Nancy, France
| |
Collapse
|
29
|
Fagan-Solis KD, Reaves DK, Rangel MC, Popoff MR, Stiles BG, Fleming JM. Challenging the roles of CD44 and lipolysis stimulated lipoprotein receptor in conveying Clostridium perfringens iota toxin cytotoxicity in breast cancer. Mol Cancer 2014; 13:163. [PMID: 24990559 PMCID: PMC4086999 DOI: 10.1186/1476-4598-13-163] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/24/2014] [Indexed: 01/01/2023] Open
Abstract
Background Translational exploration of bacterial toxins has come to the forefront of research given their potential as a chemotherapeutic tool. Studies in select tissues have demonstrated that Clostridium perfringens iota toxin binds to CD44 and lipolysis stimulated lipoprotein receptor (LSR) cell-surface proteins. We recently demonstrated that LSR expression correlates with estrogen receptor positive breast cancers and that LSR signaling directs aggressive, tumor-initiating cell behaviors. Herein, we identify the mechanisms of iota toxin cytotoxicity in a tissue-specific, breast cancer model with the ultimate goal of laying the foundation for using iota toxin as a targeted breast cancer therapy. Methods In vitro model systems were used to determine the cytotoxic effect of iota toxin on breast cancer intrinsic subtypes. The use of overexpression and knockdown technologies confirmed the roles of LSR and CD44 in regulating iota toxin endocytosis and induction of cell death. Lastly, cytotoxicity assays were used to demonstrate the effect of iota toxin on a validated set of tamoxifen resistant breast cancer cell lines. Results Treatment of 14 breast cancer cell lines revealed that LSR+/CD44- lines were highly sensitive, LSR+/CD44+ lines were slightly sensitive, and LSR-/CD44+ lines were resistant to iota cytotoxicity. Reduction in LSR expression resulted in a significant decrease in toxin sensitivity; however, overexpression of CD44 conveyed toxin resistance. CD44 overexpression was correlated with decreased toxin-stimulated lysosome formation and decreased cytosolic levels of iota toxin. These findings indicated that expression of CD44 drives iota toxin resistance through inhibition of endocytosis in breast cancer cells, a role not previously defined for CD44. Moreover, tamoxifen-resistant breast cancer cells exhibited robust expression of LSR and were highly sensitive to iota-induced cytotoxicity. Conclusions Collectively, these data are the first to show that iota toxin has the potential to be an effective, targeted therapy for breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Jodie M Fleming
- Department of Biology, North Carolina Central University, Durham, NC, USA.
| |
Collapse
|
30
|
Furuse M, Izumi Y, Oda Y, Higashi T, Iwamoto N. Molecular organization of tricellular tight junctions. Tissue Barriers 2014; 2:e28960. [PMID: 25097825 PMCID: PMC4117683 DOI: 10.4161/tisb.28960] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/19/2014] [Accepted: 04/21/2014] [Indexed: 01/22/2023] Open
Abstract
When the apicolateral border of epithelial cells is compared with a polygon, its sides correspond to the apical junctional complex, where cell adhesion molecules assemble from the plasma membranes of two adjacent cells. On the other hand, its vertices correspond to tricellular contacts, where the corners of three cells meet. Vertebrate tricellular contacts have specialized structures of tight junctions, termed tricellular tight junctions (tTJs). tTJs were identified by electron microscopic observations more than 40 years ago, but have been largely forgotten in epithelial cell biology since then. The identification of tricellulin and angulin family proteins as tTJ-associated membrane proteins has enabled us to study tTJs in terms of not only the paracellular barrier function but also unknown characteristics of epithelial cell corners via molecular biological approaches.
Collapse
Affiliation(s)
- Mikio Furuse
- Division of Cell Biology; Department of Physiology and Cell Biology; Kobe University Graduate School of Medicine; Kobe, Japan ; Division of Cerebral Structure; National Institute for Physiological Sciences; Okazaki, Aichi Japan
| | - Yasushi Izumi
- Division of Cell Biology; Department of Physiology and Cell Biology; Kobe University Graduate School of Medicine; Kobe, Japan
| | - Yukako Oda
- Division of Cell Biology; Department of Physiology and Cell Biology; Kobe University Graduate School of Medicine; Kobe, Japan
| | - Tomohito Higashi
- Division of Cell Biology; Department of Physiology and Cell Biology; Kobe University Graduate School of Medicine; Kobe, Japan
| | - Noriko Iwamoto
- Division of Cell Biology; Department of Physiology and Cell Biology; Kobe University Graduate School of Medicine; Kobe, Japan
| |
Collapse
|
31
|
Reaves DK, Fagan-Solis KD, Dunphy K, Oliver SD, Scott DW, Fleming JM. The role of lipolysis stimulated lipoprotein receptor in breast cancer and directing breast cancer cell behavior. PLoS One 2014; 9:e91747. [PMID: 24637461 PMCID: PMC3956714 DOI: 10.1371/journal.pone.0091747] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 02/14/2014] [Indexed: 01/23/2023] Open
Abstract
The claudin-low molecular subtype of breast cancer is of particular interest for clinically the majority of these tumors are poor prognosis, triple negative, invasive ductal carcinomas. Claudin-low tumors are characterized by cancer stem cell-like features and low expression of cell junction and adhesion proteins. Herein, we sought to define the role of lipolysis stimulated lipoprotein receptor (LSR) in breast cancer and cancer cell behavior as LSR was recently correlated with tumor-initiating features. We show that LSR was expressed in epithelium, endothelium, and stromal cells within the healthy breast tissue, as well as in tumor epithelium. In primary breast tumor bioposies, LSR expression was significantly correlated with invasive ductal carcinomas compared to invasive lobular carcinomas, as well as ERα positive tumors and breast cancer cell lines. LSR levels were significantly reduced in claudin-low breast cancer cell lines and functional studies illustrated that re-introduction of LSR into a claudin-low cell line suppressed the EMT phenotype and reduced individual cell migration. However, our data suggest that LSR may promote collective cell migration. Re-introduction of LSR in claudin-low breast cancer cell lines reestablished tight junction protein expression and correlated with transepithelial electrical resistance, thereby reverting claudin-low lines to other intrinsic molecular subtypes. Moreover, overexpression of LSR altered gene expression of pathways involved in transformation and tumorigenesis as well as enhanced proliferation and survival in anchorage independent conditions, highlighting that reestablishment of LSR signaling promotes aggressive/tumor initiating cell behaviors. Collectively, these data highlight a direct role for LSR in driving aggressive breast cancer behavior.
Collapse
Affiliation(s)
- Denise K. Reaves
- Department of Biology, North Carolina Central University, Durham, North Carolina, United States of America
| | - Katerina D. Fagan-Solis
- Department of Biology, North Carolina Central University, Durham, North Carolina, United States of America
| | - Karen Dunphy
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, United States of America
| | - Shannon D. Oliver
- Department of Biology, North Carolina Central University, Durham, North Carolina, United States of America
| | - David W. Scott
- Department of Cell Physiology and Cell Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jodie M. Fleming
- Department of Biology, North Carolina Central University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
32
|
Development and optimization of a high-throughput assay to measure neutralizing antibodies against Clostridium difficile binary toxin. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:689-97. [PMID: 24623624 DOI: 10.1128/cvi.00038-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Clostridium difficile strains producing binary toxin, in addition to toxin A (TcdA) and toxin B (TcdB), have been associated with more severe disease and increased recurrence of C. difficile infection in recent outbreaks. Binary toxin comprises two subunits (CDTa and CDTb) and catalyzes the ADP-ribosylation of globular actin (G-actin), which leads to the depolymerization of filamentous actin (F-actin) filaments. A robust assay is highly desirable for detecting the cytotoxic effect of the toxin and the presence of neutralizing antibodies in animal and human sera to evaluate vaccine efficacy. We describe here the optimization, using design-of-experiment (DOE) methodology, of a high-throughput assay to measure the toxin potency and neutralizing antibodies (NAb) against binary toxin. Vero cells were chosen from a panel of cells screened for sensitivity and specificity. We have successfully optimized the CDTa-to-CDTb molar ratio, toxin concentration, cell-seeding density, and sera-toxin preincubation time in the NAb assay using DOE methodology. This assay is robust, produces linear results across serial dilutions of hyperimmune serum, and can be used to quantify neutralizing antibodies in sera from hamsters and monkeys immunized with C. difficile binary toxin-containing vaccines. The assay will be useful for C. difficile diagnosis, for epidemiology studies, and for selecting and optimizing vaccine candidates.
Collapse
|
33
|
Gerding DN, Johnson S, Rupnik M, Aktories K. Clostridium difficile binary toxin CDT: mechanism, epidemiology, and potential clinical importance. Gut Microbes 2014; 5:15-27. [PMID: 24253566 PMCID: PMC4049931 DOI: 10.4161/gmic.26854] [Citation(s) in RCA: 302] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Binary toxin (CDT) is frequently observed in Clostridium difficile strains associated with increased severity of C. difficile infection (CDI). CDT belongs to the family of binary ADP-ribosylating toxins consisting of two separate toxin components: CDTa, the enzymatic ADP-ribosyltransferase which modifies actin, and CDTb which binds to host cells and translocates CDTa into the cytosol. CDTb is activated by serine proteases and binds to lipolysis stimulated lipoprotein receptor. ADP-ribosylation induces depolymerization of the actin cytoskeleton. Toxin-induced actin depolymerization also produces microtubule-based membrane protrusions which form a network on epithelial cells and increase bacterial adherence. Multiple clinical studies indicate an association between binary toxin genes in C. difficile and increased 30-d CDI mortality independent of PCR ribotype. Further studies including measures of binary toxin in stool, analyses of CDI mortality caused by CDT-producing strains, and examination of the relationship of CDT expression to TcdA and TcdB toxin variants and PCR ribotypes are needed.
Collapse
Affiliation(s)
- Dale N Gerding
- Loyola University Chicago Stritch School of Medicine; Hines Veterans Affairs Hospital; Hines, IL USA,Correspondence to: Dale N Gerding,
| | - Stuart Johnson
- Loyola University Chicago Stritch School of Medicine; Hines Veterans Affairs Hospital; Hines, IL USA
| | - Maja Rupnik
- Institute of Public Health Maribor; University of Maribor, Medical Faculty, and Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins; Ljubljana, Slovenia
| | - Klaus Aktories
- Institute of Experimental and Clinical Pharmacology and Toxicology; Albert-Ludwigs-University Freiburg; Freiburg, Germany
| |
Collapse
|
34
|
|
35
|
Haas ME, Attie AD, Biddinger SB. The regulation of ApoB metabolism by insulin. Trends Endocrinol Metab 2013; 24:391-7. [PMID: 23721961 PMCID: PMC3810413 DOI: 10.1016/j.tem.2013.04.001] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 04/10/2013] [Accepted: 04/12/2013] [Indexed: 01/07/2023]
Abstract
The leading cause of death in diabetic patients is cardiovascular disease. Apolipoprotein B (ApoB)-containing lipoprotein particles, which are secreted and cleared by the liver, are essential for the development of atherosclerosis. Insulin plays a key role in the regulation of ApoB. Insulin decreases ApoB secretion by promoting ApoB degradation in the hepatocyte. In parallel, insulin promotes clearance of circulating ApoB particles by the liver via the low-density lipoprotein receptor (LDLR), LDLR-related protein 1 (LRP1), and heparan sulfate proteoglycans (HSPGs). Consequently, the insulin-resistant state of type 2 diabetes (T2D) is associated with increased secretion and decreased clearance of ApoB. Here, we review the mechanisms by which insulin controls the secretion and uptake of ApoB in normal and diabetic livers.
Collapse
Affiliation(s)
- Mary E Haas
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | |
Collapse
|
36
|
Chandra R, Wang Y, Shahid RA, Vigna SR, Freedman NJ, Liddle RA. Immunoglobulin-like domain containing receptor 1 mediates fat-stimulated cholecystokinin secretion. J Clin Invest 2013; 123:3343-52. [PMID: 23863714 DOI: 10.1172/jci68587] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 05/23/2013] [Indexed: 12/29/2022] Open
Abstract
Cholecystokinin (CCK) is a satiety hormone produced by discrete enteroendocrine cells scattered among absorptive cells of the small intestine. CCK is released into blood following a meal; however, the mechanisms inducing hormone secretion are largely unknown. Ingested fat is the major stimulant of CCK secretion. We recently identified a novel member of the lipoprotein remnant receptor family known as immunoglobulin-like domain containing receptor 1 (ILDR1) in intestinal CCK cells and postulated that this receptor conveyed the signal for fat-stimulated CCK secretion. In the intestine, ILDR1 is expressed exclusively in CCK cells. Orogastric administration of fatty acids elevated blood levels of CCK in wild-type mice but not Ildr1-deficient mice, although the CCK secretory response to trypsin inhibitor was retained. The uptake of fluorescently labeled lipoproteins in ILDR1-transfected CHO cells and release of CCK from isolated intestinal cells required a unique combination of fatty acid plus HDL. CCK secretion secondary to ILDR1 activation was associated with increased [Ca2+]i, consistent with regulated hormone release. These findings demonstrate that ILDR1 regulates CCK release through a mechanism dependent on fatty acids and lipoproteins and that absorbed fatty acids regulate gastrointestinal hormone secretion.
Collapse
Affiliation(s)
- Rashmi Chandra
- Department of Medicine, Duke University, Durham, North Carolina 27710, USA
| | | | | | | | | | | |
Collapse
|
37
|
Watanabe K, Watson E, Cremona ML, Millings EJ, Lefkowitch JH, Fischer SG, LeDuc CA, Leibel RL. ILDR2: an endoplasmic reticulum resident molecule mediating hepatic lipid homeostasis. PLoS One 2013; 8:e67234. [PMID: 23826244 PMCID: PMC3691114 DOI: 10.1371/journal.pone.0067234] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 05/15/2013] [Indexed: 11/18/2022] Open
Abstract
Ildr2, a modifier of diabetes susceptibility in obese mice, is expressed in most organs, including islets and hypothalamus, with reduced levels in livers of diabetes-susceptible B6.DBA mice congenic for a 1.8 Mb interval of Chromosome 1. In hepatoma and neuronal cells, ILDR2 is primarily located in the endoplasmic reticulum membrane. We used adenovirus vectors that express shRNA or are driven by the CMV promoter, respectively, to knockdown or overexpress Ildr2 in livers of wild type and ob/ob mice. Livers in knockdown mice were steatotic, with increased hepatic and circulating triglycerides and total cholesterol. Increased circulating VLDL, without reduction in triglyceride clearance suggests an effect of reduced hepatic ILDR2 on hepatic cholesterol clearance. In animals that overexpress Ildr2, hepatic triglyceride and total cholesterol levels were reduced, and strikingly so in ob/ob mice. There were no significant changes in body weight, energy expenditure or glucose/insulin homeostasis in knockdown or overexpressing mice. Knockdown mice showed reduced expression of genes mediating synthesis and oxidation of hepatic lipids, suggesting secondary suppression in response to increased hepatic lipid content. In Ildr2-overexpressing ob/ob mice, in association with reduced liver fat content, levels of transcripts related to neutral lipid synthesis and cholesterol were increased, suggesting “relief” of the secondary suppression imposed by lipid accumulation. Considering the fixed location of ILDR2 in the endoplasmic reticulum, we investigated the possible participation of ILDR2 in ER stress responses. In general, Ildr2 overexpression was associated with increases, and knockdown with decreases in levels of expression of molecular components of canonical ER stress pathways. We conclude that manipulation of Ildr2 expression in liver affects both lipid homeostasis and ER stress pathways. Given these reciprocal interactions, and the relatively extended time-course over which these studies were conducted, we cannot assign causal primacy to either the effects on hepatic lipid homeostasis or ER stress responses.
Collapse
Affiliation(s)
- Kazuhisa Watanabe
- Naomi Berrie Diabetes Center and Department of Pediatrics, Columbia University, New York, New York, United States of America
| | - Elizabeth Watson
- Naomi Berrie Diabetes Center and Department of Pediatrics, Columbia University, New York, New York, United States of America
| | - Maria Laura Cremona
- Naomi Berrie Diabetes Center and Department of Pediatrics, Columbia University, New York, New York, United States of America
| | - Elizabeth J. Millings
- Naomi Berrie Diabetes Center and Department of Pediatrics, Columbia University, New York, New York, United States of America
| | - Jay H. Lefkowitch
- Department of Pathology and Cell Biology, Columbia University, New York, New York, United States of America
| | - Stuart G. Fischer
- Naomi Berrie Diabetes Center and Department of Pediatrics, Columbia University, New York, New York, United States of America
| | - Charles A. LeDuc
- Naomi Berrie Diabetes Center and Department of Pediatrics, Columbia University, New York, New York, United States of America
| | - Rudolph L. Leibel
- Naomi Berrie Diabetes Center and Department of Pediatrics, Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
38
|
Clostridium difficile binary toxin CDT induces clustering of the lipolysis-stimulated lipoprotein receptor into lipid rafts. mBio 2013; 4:e00244-13. [PMID: 23631918 PMCID: PMC3648903 DOI: 10.1128/mbio.00244-13] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Clostridium difficile is the leading cause of antibiotics-associated diarrhea and pseudomembranous colitis. Hypervirulent C. difficile strains produce the binary actin-ADP-ribosylating toxin CDT (C. difficile transferase), in addition to the Rho-glucosylating toxins A and B. We recently identified the lipolysis-stimulated lipoprotein receptor (LSR) as the host receptor that mediates uptake of CDT into target cells. Here we investigated in H1-HeLa cells, which ectopically express LSR, the influence of CDT on the plasma membrane distribution of the receptor. We found by fluorescence microscopy that the binding component of CDT (CDTb) induces clustering of LSR into subcompartments of the plasma membrane. Detergent extraction of cells treated with CDTb, followed by sucrose gradient fractionation, uncovered accumulation of LSR in detergent-resistant membranes (DRMs) that contained typical marker proteins of lipid rafts. Membrane cholesterol depletion with methyl-β-cyclodextrin inhibited the association of LSR with DRMs upon addition of CDTb. The receptor-binding domain of CDTb also triggered LSR clustering into DRMs. CDTb-triggered clustering of LSR into DRMs could be confirmed in Caco-2 cells. Our data suggest that CDT forces its receptor to cluster into lipid rafts and that oligomerization of the B component might enhance but is not essential for this process. C. difficile binary toxin CDT is a member of the iota-like, actin ADP-ribosylating toxin family. The mechanism that mediates endocytic uptake of these toxins still remains elusive. Previous studies highlighted the importance of lipid rafts for oligomerization of the binding component of these toxins and for cell entry. Recently, the host cell receptor for this toxin family, namely, the lipolysis-stimulated lipoprotein receptor (LSR), has been identified. Our study now demonstrates that the binding component of CDT (CDTb) induces clustering of LSR into lipid rafts. Importantly, LSR clustering is efficiently induced also by the receptor-binding domain of CDTb, suggesting that oligomerization of the B component of CDT is not the main trigger of this process. The current work extends our knowledge on the cooperative play between iota-like toxins and their receptor.
Collapse
|
39
|
Furuse M, Oda Y, Higashi T, Iwamoto N, Masuda S. Lipolysis-stimulated lipoprotein receptor: a novel membrane protein of tricellular tight junctions. Ann N Y Acad Sci 2012; 1257:54-8. [PMID: 22671589 DOI: 10.1111/j.1749-6632.2012.06486.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Tricellular tight junctions (tTJs) are specialized structural variants of tight junctions that restrict the free diffusion of solutes at the extracellular space of tricellular contacts. Their presence at cell corners, situated in the angles between three adjacent epithelial cells, was identified early by electron microscopy, but despite their potential importance, tTJs have been generally ignored in epithelial cell biology. Tricellulin was the first molecular component of tTJs shown to be involved in their formation and in epithelial barrier function. However, the precise molecular organization and function of tTJs are still largely unknown. Recently, we identified the lipolysis-stimulated lipoprotein receptor (LSR) as a tTJ-associated membrane protein. LSR recruits tricellulin to tTJs, suggesting that the LSR-tricellulin system plays a key role in tTJ formation. In this paper, we summarize the identification and characterization of LSR as a molecular component of tTJs.
Collapse
Affiliation(s)
- Mikio Furuse
- Division of Cell Biology, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Japan.
| | | | | | | | | |
Collapse
|
40
|
Ahmad N, Girardet JM, Akbar S, Lanhers MC, Paris C, Yen FT, Corbier C. Lactoferrin and its hydrolysate bind directly to the oleate-activated form of the lipolysis stimulated lipoprotein receptor. FEBS J 2012; 279:4361-73. [PMID: 23050782 DOI: 10.1111/febs.12026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 09/14/2012] [Accepted: 10/02/2012] [Indexed: 11/29/2022]
Abstract
The hepatic removal of triglyceride-rich chylomicrons during the postprandial phase represents an important step towards determining the bioavailability of dietary lipids amongst the peripheral tissues. Indeed, elevated postprandial lipemia is often associated with obesity and increased risk of coronary heart disease. The milk protein, lactoferrin, has been shown to inhibit hepatic chylomicron remnant removal by the liver, resulting in increased postprandial lipemia. Despite numerous studies on potential targets for lactoferrin, the molecular mechanisms underlying the effect of lactoferrin remain unclear. We recently demonstrated that the lipolysis stimulated lipoprotein receptor (LSR) contributes to the removal of triglyceride-rich lipoproteins during the postprandial phase. Here, we report that while lactoferrin does not have any significant effect on LSR protein levels in mouse Hepa1-6 cells, this protein colocalizes with LSR in cells but only in the presence of oleate, which is needed to obtain LSR in its active form as lipoprotein receptor. Ligand blotting using purified LSR revealed that lactoferrin binds directly to the receptor in the presence of oleate and prevents the binding of triglyceride-rich lipoproteins. Both C- and N-lobes of lactoferrin as well as a mixture of peptides derived from its hydrolysis retained the ability to bind LSR in its active form. We propose then that the elevated postprandial lipemia observed upon lactoferrin treatment in vivo is mediated in part by its direct interaction with free fatty acid activated LSR, thus preventing clearance of chylomicrons and their remnants through the LSR pathway.
Collapse
Affiliation(s)
- Nazir Ahmad
- Université de Lorraine, Unité de Recherche Animal et Fonctionnalités des Produits Animaux, Vandœuvre-lès-Nancy Cedex, France
| | | | | | | | | | | | | |
Collapse
|
41
|
Takasawa A, Kojima T, Ninomiya T, Tsujiwaki M, Murata M, Tanaka S, Sawada N. Behavior of tricellulin during destruction and formation of tight junctions under various extracellular calcium conditions. Cell Tissue Res 2012; 351:73-84. [PMID: 23073616 PMCID: PMC3536962 DOI: 10.1007/s00441-012-1512-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 09/27/2012] [Indexed: 12/31/2022]
Abstract
Tricellulin is an important component of tricellular tight junctions (TJs) and is involved in the formation of tricellular contacts. However, little is known about its regulation during the assembly and disassembly of tricellular TJs. By using the well-differentiated pancreatic cancer cell line HPAC, which highly expresses tricellulin at tricellular contacts, we have investigated changes in the localization, expression and phosphorylation of tricellulin and in its TJ functions as a barrier and fence during the destruction and formation of TJs induced by changes in the extracellular calcium concentration. During both extracellular Ca2+ depletion caused by EGTA treatment and Ca2+ repletion after Ca2+ starvation, the expression of tricellulin increased in whole lysates and in Triton-X-100-insoluble fractions without any change in its mRNA. The increases in immunoreactivity revealed by Western blotting were prevented by alkaline phosphatase treatment. Immunoprecipitation assays showed that tricellulin was phosphorylated on threonine residues when it increased after Ca2+ depletion and repletion. In the early stage after Ca2+ repletion, tricellulin was expressed not only at tricellular contacts but also in the cytoplasm and at bicellular borders. In confocal laser microscopy, tricellulin was observed at the apical-most regions and basolateral membranes of tricellular contacts after Ca2+ repletion. Knockdown of tricellulin delayed the recovery of the barrier and fence functions after Ca2+ repletion. Thus, the dynamic behavior of tricellulin during the destruction and formation of TJs under various extracellular calcium conditions seems to be closely associated with the barrier and fence functions of TJs.
Collapse
Affiliation(s)
- Akira Takasawa
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
Stenger C, Pinçon A, Hanse M, Royer L, Comte A, Koziel V, Olivier JL, Pillot T, Yen FT. Brain region-specific immunolocalization of the lipolysis-stimulated lipoprotein receptor (LSR) and altered cholesterol distribution in aged LSR+/- mice. J Neurochem 2012; 123:467-76. [PMID: 22909011 DOI: 10.1111/j.1471-4159.2012.07922.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 08/14/2012] [Accepted: 08/14/2012] [Indexed: 11/29/2022]
Abstract
Brain lipid homeostasis is important for maintenance of brain cell function and synaptic communications, and is intimately linked to age-related cognitive decline. Because of the blood-brain barrier's limiting nature, this tissue relies on a complex system for the synthesis and receptor-mediated uptake of lipids between the different networks of neurons and glial cells. Using immunofluorescence, we describe the region-specific expression of the lipolysis-stimulated lipoprotein receptor (LSR), in the mouse hippocampus, cerebellum Purkinje cells, the ependymal cell interface between brain parenchyma and cerebrospinal fluid, and the choroid plexus. Colocalization with cell-specific markers revealed that LSR was expressed in neurons, but not astrocytes. Latency in arms of the Y-maze exhibited by young heterozygote LSR(+/-) mice was significantly different as compared to control LSR(+/+), and increased in older LSR(+/-) mice. Filipin and Nile red staining revealed membrane cholesterol content accumulation accompanied by significantly altered distribution of LSR in the membrane, and decreased intracellular lipid droplets in the cerebellum and hippocampus of old LSR(+/-) mice, as compared to control littermates as well as young LSR(+/-) animals. These data therefore suggest a potential role of LSR in brain cholesterol distribution, which is particularly important in preserving neuronal integrity and thereby cognitive functions during aging.
Collapse
|
43
|
Aktories K, Schwan C, Papatheodorou P, Lang AE. Bidirectional attack on the actin cytoskeleton. Bacterial protein toxins causing polymerization or depolymerization of actin. Toxicon 2012; 60:572-81. [DOI: 10.1016/j.toxicon.2012.04.338] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 04/10/2012] [Indexed: 10/28/2022]
|
44
|
Abstract
Clostridium spiroforme produces the binary actin-ADP-ribosylating toxin CST (C. spiroforme toxin), which has been proposed to be responsible for diarrhea, enterocolitis, and eventually death, especially in rabbits. Here we report on the recombinant production of the enzyme component (CSTa) and the binding component (CSTb) of C. spiroforme toxin in Bacillus megaterium. By using the recombinant toxin components, we show that CST enters target cells via the lipolysis-stimulated lipoprotein receptor (LSR), which has been recently identified as the host cell receptor of the binary toxins Clostridium difficile transferase (CDT) and Clostridium perfringens iota toxin. Microscopic studies revealed that CST, but not the related Clostridium botulinum C2 toxin, colocalized with LSR during toxin uptake and traffic to endosomal compartments. Our findings indicate that CST shares LSR with C. difficile CDT and C. perfringens iota toxin as a host cell surface receptor.
Collapse
|
45
|
Higashi T, Tokuda S, Kitajiri SI, Masuda S, Nakamura H, Oda Y, Furuse M. Analysis of the angulin family consisting of LSR, ILDR1 and ILDR2: tricellulin recruitment, epithelial barrier function and implication in deafness pathogenesis. J Cell Sci 2012; 126:966-77. [DOI: 10.1242/jcs.116442] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tricellular tight junctions (tTJs) seal the extracellular space at tricellular contacts (TCs), where the corners of three epithelial cells meet. To date, the transmembrane proteins tricellulin and lipolysis-stimulated lipoprotein receptor (LSR) are known to be molecular components of tTJs. LSR recruits tricellulin to tTJs, and both proteins are required for the full barrier function of epithelial cellular sheets. Here, we show that two LSR-related proteins, immunoglobulin-like domain-containing receptor (ILDR)1 and ILDR2, are also localized at TCs and recruit tricellulin. The expressions of LSR, ILDR1 and ILDR2 were complementary in various epithelial cell types, although LSR and ILDR1 were coexpressed in some epithelia. ILDR1 was required for the establishment of a strong barrier of the epithelium, similar to LSR, when introduced into cultured epithelial cells, while ILDR2 provided a much weaker barrier. We further analyzed human ILDR1, whose mutations cause a familial deafness, DFNB42, and found that most DFNB42-associated ILDR1 mutant proteins were defective in recruitment of tricellulin. We also found that tricellulin mutant proteins associated with another familial deafness, DFNB49, were not recruited to TCs by ILDR1. These findings show the heterogeneity of the molecular organization of tTJs in terms of the content of LSR, ILDR1 or ILDR2, and suggest that ILDR1-mediated recruitment of tricellulin to TCs is required for hearing. Given their common localization at epithelial cell corners and recruitment of tricellulin, we propose to designate LSR, ILDR1 and ILDR2 as angulin family proteins.
Collapse
|
46
|
Lipolysis-stimulated lipoprotein receptor (LSR) is the host receptor for the binary toxin Clostridium difficile transferase (CDT). Proc Natl Acad Sci U S A 2011; 108:16422-7. [PMID: 21930894 DOI: 10.1073/pnas.1109772108] [Citation(s) in RCA: 150] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Clostridium difficile infection (CDI) causes antibiotic-associated diarrhea and pseudomembranous colitis. Hypervirulent strains of the pathogen, which are responsible for increased morbidity and mortality of CDI, produce the binary actin-ADP ribosylating toxin Clostridium difficile transferase (CDT) in addition to the Rho-glucosylating toxins A and B. CDT depolymerizes the actin cytoskeleton, increases adherence and colonization of Clostridia by induction of microtubule-based cell protrusions and, eventually, causes death of target cells. Using a haploid genetic screen, we identified the lipolysis-stimulated lipoprotein receptor as the membrane receptor for CDT uptake by target cells. Moreover, we show that Clostridium perfringens iota toxin, which is a related binary actin-ADP ribosylating toxin, enters target cells via the lipolysis-stimulated lipoprotein receptor. Identification of the toxin receptors is essential for understanding of the toxin uptake and provides a most valuable basis for antitoxin strategies.
Collapse
|
47
|
Voruganti VS, Diego VP, Haack K, Cole SA, Blangero J, Göring HHH, Laston S, Wenger CR, Ebbesson SOE, Fabsitz RR, Devereux RB, Howard BV, Umans JG, MacCluer JW, Comuzzie AG. A QTL for genotype by sex interaction for anthropometric measurements in Alaskan Eskimos (GOCADAN Study) on chromosome 19q12-13. Obesity (Silver Spring) 2011; 19:1840-6. [PMID: 21527897 PMCID: PMC3525327 DOI: 10.1038/oby.2011.78] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Variation in anthropometric measurements due to sexual dimorphism can be the result of genotype by sex interactions (G×S). The purpose of this study was to examine the sex-specific genetic architecture in anthropometric measurements in Alaskan Eskimos from the Genetics of Coronary Artery Disease in Alaska Natives (GOCADAN) study. Maximum likelihood-based variance components decomposition methods, implemented in SOLAR, were used for G×S analyses. Anthropometric measurements included BMI, waist circumference (WC), waist/height ratio, percent body fat (%BF), and subscapular and triceps skinfolds. Except for WC, mean values of all phenotypes were significantly different in men and women (P < 0.05). All anthropometric measures were significantly heritable (P < 0.001). In a preliminary analysis not allowing for G×S interaction, evidence of linkage was detected between markers D19S414 and D19S220 on chromosome 19 for WC (logarithm of odds (lod) = 3.5), %BF (lod = 1.7), BMI (lod = 2.4), waist/height ratio (lod = 2.5), subscapular (lod = 2.1), and triceps skinfolds (lod = 1.9). In subsequent analyses which allowed for G×S interaction, linkage was again found between these traits and the same two markers on chromosome 19 with significantly improved lod scores for: WC (lod = 4.5), %BF (lod = 3.8), BMI (lod = 3.5), waist/height ratio (lod = 3.2), subscapular (lod = 3.0), and triceps skinfolds (lod = 2.9). These results support the evidence of a G×S interaction in the expression of genetic effects resulting in sexual dimorphism in anthropometric phenotypes and identify the chromosome 19q12-13 region as important for adiposity-related traits in Alaskan Eskimos.
Collapse
Affiliation(s)
- V Saroja Voruganti
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Masuda S, Oda Y, Sasaki H, Ikenouchi J, Higashi T, Akashi M, Nishi E, Furuse M. LSR defines cell corners for tricellular tight junction formation in epithelial cells. J Cell Sci 2011; 124:548-55. [DOI: 10.1242/jcs.072058] [Citation(s) in RCA: 176] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Epithelial cell contacts consist of not only bicellular contacts but also tricellular contacts, where the corners of three cells meet. At tricellular contacts, tight junctions (TJs) generate specialized structures termed tricellular TJs (tTJs) to seal the intercellular space. Tricellulin is the only known molecular component of tTJs and is involved in the formation of tTJs, as well as in the normal epithelial barrier function. However, the detailed molecular mechanism of how tTJs are formed and maintained remains elusive. Using a localization-based expression cloning method, we identified a novel tTJ-associated protein known as lipolysis-stimulated lipoprotein receptor (LSR). Upon LSR knockdown in epithelial cells, tTJ formation was affected and the epithelial barrier function was diminished. Tricellulin accumulation at the tricellular contacts was also diminished in these cells. By contrast, LSR still accumulated at the tricellular contacts upon tricellulin knockdown. Analyses of deletion mutants revealed that the cytoplasmic domain of LSR was responsible for the recruitment of tricellulin. On the basis of these observations, we propose that LSR defines tricellular contacts in epithelial cellular sheets by acting as a landmark to recruit tricellulin for tTJ formation.
Collapse
Affiliation(s)
- Sayuri Masuda
- Division of Cell Biology, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Yukako Oda
- Division of Cell Biology, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Hiroyuki Sasaki
- Department of Molecular and Cell Biology, Institute of DNA Medicine, Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Junichi Ikenouchi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
- Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Tomohito Higashi
- Division of Cell Biology, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Masaya Akashi
- Division of Cell Biology, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Eiichiro Nishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Mikio Furuse
- Division of Cell Biology, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| |
Collapse
|
49
|
Dubois F, Vandermoere F, Gernez A, Murphy J, Toth R, Chen S, Geraghty KM, Morrice NA, MacKintosh C. Differential 14-3-3 affinity capture reveals new downstream targets of phosphatidylinositol 3-kinase signaling. Mol Cell Proteomics 2009; 8:2487-99. [PMID: 19648646 PMCID: PMC2773716 DOI: 10.1074/mcp.m800544-mcp200] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We devised a strategy of 14-3-3 affinity capture and release, isotope differential (d0/d4) dimethyl labeling of tryptic digests, and phosphopeptide characterization to identify novel targets of insulin/IGF1/phosphatidylinositol 3-kinase signaling. Notably four known insulin-regulated proteins (PFK-2, PRAS40, AS160, and MYO1C) had high d0/d4 values meaning that they were more highly represented among 14-3-3-binding proteins from insulin-stimulated than unstimulated cells. Among novel candidates, insulin receptor substrate 2, the proapoptotic CCDC6, E3 ubiquitin ligase ZNRF2, and signaling adapter SASH1 were confirmed to bind to 14-3-3s in response to IGF1/phosphatidylinositol 3-kinase signaling. Insulin receptor substrate 2, ZNRF2, and SASH1 were also regulated by phorbol ester via p90RSK, whereas CCDC6 and PRAS40 were not. In contrast, the actin-associated protein vasodilator-stimulated phosphoprotein and lipolysis-stimulated lipoprotein receptor, which had low d0/d4 scores, bound 14-3-3s irrespective of IGF1 and phorbol ester. Phosphorylated Ser19 of ZNRF2 (RTRAYpS19GS), phospho-Ser90 of SASH1 (RKRRVpS90QD), and phospho- Ser493 of lipolysis-stimulated lipoprotein receptor (RPRARpS493LD) provide one of the 14-3-3-binding sites on each of these proteins. Differential 14-3-3 capture provides a powerful approach to defining downstream regulatory mechanisms for specific signaling pathways.
Collapse
Affiliation(s)
- Fanny Dubois
- Medical Research Council Protein Phosphorylation Unit, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Narvekar P, Berriel Diaz M, Krones-Herzig A, Hardeland U, Strzoda D, Stöhr S, Frohme M, Herzig S. Liver-specific loss of lipolysis-stimulated lipoprotein receptor triggers systemic hyperlipidemia in mice. Diabetes 2009; 58:1040-9. [PMID: 19188430 PMCID: PMC2671043 DOI: 10.2337/db08-1184] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE In mammals, proper storage and distribution of lipids in and between tissues is essential for the maintenance of energy homeostasis. In contrast, aberrantly high levels of triglycerides in the blood ("hypertriglyceridemia") represent a hallmark of the metabolic syndrome and type 2 diabetes. As hypertriglyceridemia has been identified as an important risk factor for cardiovascular complications, in this study we aimed to identify molecular mechanisms in aberrant triglyceride elevation under these conditions. RESEARCH DESIGN AND METHODS To determine the importance of hepatic lipid handling for systemic dyslipidemia, we profiled the expression patterns of various hepatic lipid transporters and receptors under healthy and type 2 diabetic conditions. A differentially expressed lipoprotein receptor was functionally characterized by generating acute, liver-specific loss- and gain-of-function animal models. RESULTS We show that the hepatic expression of lipid transporter lipolysis-stimulated lipoprotein receptor (LSR) is specifically impaired in mouse models of obesity and type 2 diabetes and can be restored by leptin replacement. Experimental imitation of this pathophysiological situation by liver-specific knockdown of LSR promotes hypertriglyceridemia and elevated apolipoprotein (Apo)B and E serum levels in lean wild-type and ApoE knockout mice. In contrast, genetic restoration of LSR expression in obese animals to wild-type levels improves serum triglyceride levels and serum profiles in these mice. CONCLUSIONS The dysregulation of hepatic LSR under obese and diabetic conditions may provide a molecular rationale for systemic dyslipidemia in type 2 diabetes and the metabolic syndrome and represent a novel target for alternative treatment strategies in these patients.
Collapse
MESH Headings
- Animals
- Apolipoproteins E/deficiency
- Blood Glucose/metabolism
- Cholesterol/blood
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 2/physiopathology
- Disease Models, Animal
- Fatty Acids, Nonesterified/blood
- Hyperlipidemias/physiopathology
- Ketone Bodies/blood
- Lipolysis
- Lipoproteins, VLDL/metabolism
- Liver/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Obese
- RNA Interference
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Receptors, LDL/physiology
- Receptors, Lipoprotein/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Triglycerides/blood
Collapse
Affiliation(s)
- Prachiti Narvekar
- Emmy Noether and Marie Curie Research Group “Molecular Metabolic Control,” DKFZ-ZMBH Alliance, German Cancer Research Center Heidelberg, Heidelberg, Germany
| | - Mauricio Berriel Diaz
- Emmy Noether and Marie Curie Research Group “Molecular Metabolic Control,” DKFZ-ZMBH Alliance, German Cancer Research Center Heidelberg, Heidelberg, Germany
| | - Anja Krones-Herzig
- Emmy Noether and Marie Curie Research Group “Molecular Metabolic Control,” DKFZ-ZMBH Alliance, German Cancer Research Center Heidelberg, Heidelberg, Germany
| | - Ulrike Hardeland
- Emmy Noether and Marie Curie Research Group “Molecular Metabolic Control,” DKFZ-ZMBH Alliance, German Cancer Research Center Heidelberg, Heidelberg, Germany
| | - Daniela Strzoda
- Emmy Noether and Marie Curie Research Group “Molecular Metabolic Control,” DKFZ-ZMBH Alliance, German Cancer Research Center Heidelberg, Heidelberg, Germany
| | - Sigrid Stöhr
- Department of Animal Physiology, Philipps University Marburg, Marburg, Germany; and
| | - Marcus Frohme
- Technische und forschende Hochschule Wildau, Wildau, Germany
| | - Stephan Herzig
- Emmy Noether and Marie Curie Research Group “Molecular Metabolic Control,” DKFZ-ZMBH Alliance, German Cancer Research Center Heidelberg, Heidelberg, Germany
- Corresponding author: Stephan Herzig,
| |
Collapse
|