1
|
Kennedy C, Doyle R, Gough O, Mcevoy C, McAnallen S, Hughes M, Sheng X, Crifo B, Andrews D, Gaffney A, Rodriguez J, Kennedy S, Dillon E, Crean D, Zhang W, Yi Z, Nair V, Susztak K, Hirschhorn J, Florez J, Groop PH, Sandholm N, Kretzler M, McKay GJ, McKnight AJ, Maxwell AP, Matallanas D, Dorman A, Martin F, Conlon PJ, Sadlier DM, Brennan E, Godson C. A Novel Role for FERM Domain-Containing Protein 3 in CKD. KIDNEY360 2024; 5:1799-1812. [PMID: 39450948 PMCID: PMC11687992 DOI: 10.34067/kid.0000000602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024]
Abstract
Key Points We have identified a transcriptional signature of 93 genes associated with CKD severity and progression. Protein 4.1, ezrin, radixin, moesin domain-containing protein 3 gene expression is reduced in the context of more severe kidney disease and in individuals who go on to develop progressive disease. Protein 4.1, ezrin, radixin, moesin domain-containing protein 3 interacts with proteins of the cell cytoskeleton and cell-cell junctions in proximal tubule epithelial cells. Background Currently, there are limited methods to link disease severity and risk of disease progression in CKD. To better understand this potential relationship, we interrogated the renal transcriptomic profile of individuals with CKD with measures of CKD severity and identified protein 4.1, ezrin, radixin, moesin-domain containing protein 3 (FRMD3 ) as a candidate gene for follow-up study. Methods RNA-sequencing was used to profile the transcriptome of CKD biopsies from the North Dublin Renal BioBank, the results of which were correlated with clinical parameters. The potential function of FRMD3 was explored by interrogating the FRMD3 interactome and assessing the effect of lentiviral mediated FRMD3 knock down on human renal proximal tubule epithelial cells by assessing cell viability, metabolic activity, and structural markers. Results We identified a subset of 93 genes which are significantly correlated with eGFR and percentage tubulointerstitial fibrosis at time of biopsy and with CKD progression 5 years postbiopsy. These results were validated against transcriptomic data from an external cohort of 432 nephrectomy samples. One of the top-ranking genes from this subset, FRMD3, has previously been associated with the risk of developing diabetic kidney disease. Interrogating the interactome of FRMD3 in tubule epithelial cells revealed interactions with cytoskeletal components of cell-cell junctions. Knockdown of FRMD3 expression in tubule epithelial cells resulted in increased proapoptotic activity within the cells, as well as dysregulation of E-Cadherin. Conclusions We have identified a panel of kidney-specific transcripts correlated with severity and progression of kidney disease, and from this, we have identified a possible role for FRMD3 in tubule cell structure and health.
Collapse
Affiliation(s)
- Ciarán Kennedy
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Ross Doyle
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
- School of Medicine, Mater Misericordiae University Hospital, University College Dublin, Dublin, Ireland
| | - Oisin Gough
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Caitriona Mcevoy
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
- Tallaght University Hospital, Dublin; and Trinity Kidney Centre, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Susan McAnallen
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Maria Hughes
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Xin Sheng
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Bianca Crifo
- School of Biomolecular and Biomedical Science, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Darrell Andrews
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Andrew Gaffney
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Javier Rodriguez
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Susan Kennedy
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
- TriviumVet, Waterford, Ireland
| | - Eugene Dillon
- UCD Conway Institute Core Technologies, University College Dublin, Dublin, Ireland
| | - Daniel Crean
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Weijia Zhang
- Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zhengzi Yi
- Icahn School of Medicine at Mount Sinai, New York, New York
| | - Viji Nair
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Katalin Susztak
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joel Hirschhorn
- Endocrine Division and Diabetes Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jose Florez
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Gareth J. McKay
- Centre for Public Health, Queens University of Belfast, Northern Ireland, United Kingdom
| | - Amy Jayne McKnight
- Centre for Public Health, Queens University of Belfast, Northern Ireland, United Kingdom
| | - Alexander P. Maxwell
- Centre for Public Health, Queens University of Belfast, Northern Ireland, United Kingdom
| | - David Matallanas
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Anthony Dorman
- Department of Pathology, Beaumont Hospital, Dublin, Ireland
| | - Finian Martin
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Peter J. Conlon
- National Kidney Transplant Service, Department of Nephrology and Kidney Transplantation, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Denise M. Sadlier
- School of Medicine, Mater Misericordiae University Hospital, University College Dublin, Dublin, Ireland
| | - Eoin Brennan
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
2
|
Evin F, Kırkgöz T, Atik T, Ak G, Köse T, Kabasakal C, Özkan B, Özen S, Darcan Ş, Gökşen D. "Predicting diabetic kidney disease in youth with type 1 diabetes: Insights from genetic risk assessment". J Diabetes Complications 2024; 38:108833. [PMID: 39293150 DOI: 10.1016/j.jdiacomp.2024.108833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 09/20/2024]
Abstract
OBJECTIVE Diabetic kidney disease (DKD) is influenced by multiple factors, yet its precise progression mechanisms remain largely unclear. This study aimed to create a clinical risk-scoring system based on genetic polymorphisms in the AFF3, CARS, CERS2, ERBB4, GLRA3, RAET1L, TMPO, and ZMIZ1 genes. METHODS The study included a DKD group diagnosed with diabetic kidney disease before age 18 and a WDC group matched by age, gender, and age at diabetes diagnosis. Genetic data and clinical data from diabetes diagnosis to moderately increased albuminuria (MIA) detection were compared between the groups. RESULTS Among 43 DKD cases, 22 were girls and 21 were boys. At MIA diagnosis, mean body weight SDS was -0.24 ± 0.94, height SDS was 0.34 ± 1.15, and BMI SDS was -0.26 ± 0.94. Systolic blood pressure was at the 72nd percentile (2-99), and diastolic blood pressure was at the 74th percentile (33-99). Significant differences in rs267734, rs267738, and rs942263 polymorphisms were found between DKD and non-complication diabetic groups (13[30.2 %] vs 5[11.6 %], p = 0.034; 14[32.6 %] vs 5[11.6 %], p = 0.019; 26[60.5 %] vs 40[93 %], p < 0.001). CONCLUSION Several factors were identified as significant in DKD onset, including low follow-up weight SDS, elevated diastolic blood pressure, presence of rs267734, and absence of rs942263 polymorphisms. The model demonstrated a specificity of 81.4 % and a sensitivity of 74.4 %.
Collapse
Affiliation(s)
- Ferda Evin
- Division of Pediatric Endocrinology, Department of Pediatrics, School of Medicine, Ege University, Izmir, Turkey.
| | - Tarık Kırkgöz
- Division of Pediatric Endocrinology, Department of Pediatrics, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Tahir Atik
- Division of Pediatric Genetics, Department of Pediatrics, School of Medicine, Ege University, Izmir, Turkey
| | - Güneş Ak
- Department of Biochemistry, School of Medicine, Ege University, Izmir, Turkey
| | - Timur Köse
- Department of Biostatistics and Medical Informatics, School of Medicine, Ege University, Izmir, Turkey
| | - Caner Kabasakal
- Division of Pediatric Nephrology, Department of Pediatrics, School of Medicine, Ege University, Izmir, Turkey
| | - Behzat Özkan
- Division of Pediatric Endocrinology, Department of Pediatrics, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Samim Özen
- Division of Pediatric Endocrinology, Department of Pediatrics, School of Medicine, Ege University, Izmir, Turkey
| | - Şükran Darcan
- Division of Pediatric Endocrinology, Department of Pediatrics, School of Medicine, Ege University, Izmir, Turkey
| | - Damla Gökşen
- Division of Pediatric Endocrinology, Department of Pediatrics, School of Medicine, Ege University, Izmir, Turkey
| |
Collapse
|
3
|
Sang SY, Wang YJ, Liang T, Liu Y, Liu JJ, Li H, Liu X, Kang QZ, Wang T. Protein 4.1R regulates M1 macrophages polarization via glycolysis, alleviating sepsis-induced liver injury in mice. Int Immunopharmacol 2024; 128:111546. [PMID: 38237224 DOI: 10.1016/j.intimp.2024.111546] [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: 11/13/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 02/08/2024]
Abstract
Acute liver injury (ALI) is a common clinical disease caused by sepsis, metabolic syndrome, hepatitis virus. Macrophage plays an important role in the development of ALI, which is characterized by polarization and inflammatory regulation. The polarization process of macrophages is related to membrane binding proteins and adaptors. Protein 4.1R acts as an adaptor, linking membrane proteins to the cytoskeleton, and is involved in cell activation and cytokine secretion. However, whether protein 4.1R is involved in regulating macrophage polarization and inflammation-induced liver injury remains unknown. In this study, protein 4.1R is identified with the special effect on macrophage M1 polarization. And it is further demonstrated that protein 4.1R deficiency significantly enhance glycolytic metabolism. Mechanistically, the regulation of protein 4.1R on pyruvate kinase M2 (PKM2) plays a key role in glycolysis metabolism. In addition, we found that protein 4.1R directly interacts with toll-like receptor 4 (TLR4), inhibits the activation of the AKT/HIF-1α signaling pathway. In conclusion, protein 4.1R targets HIF-1α mediated glycolysis regulates M1 macrophage polarization, indicating that protein 4.1R is a candidate for regulating macrophage mediated inflammatory response. In conclusion, we have revealed a novel function of protein 4.1R in macrophage polarization and ALI, providing important insights into the metabolic reprogramming, which is important for ALI therapy. We have revealed a novel function of protein 4.1R in macrophage polarization and ALI, providing important insights into the metabolic reprogramming, which is important for ALI therapy.
Collapse
Affiliation(s)
- Si-Yao Sang
- MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yuan-Jiao Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Taotao Liang
- Department of Hematology, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, China
| | - Yan Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Jiao-Jiao Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Hui Li
- MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xin Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Qiao-Zhen Kang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China.
| | - Ting Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China.
| |
Collapse
|
4
|
The Role of Cytoskeleton Protein 4.1 in Immunotherapy. Int J Mol Sci 2023; 24:ijms24043777. [PMID: 36835189 PMCID: PMC9961941 DOI: 10.3390/ijms24043777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/18/2023] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
Cytoskeleton protein 4.1 is an essential class of skeletal membrane protein, initially found in red blood cells, and can be classified into four types: 4.1R (red blood cell type), 4.1N (neuronal type), 4.1G (general type), and 4.1B (brain type). As research progressed, it was discovered that cytoskeleton protein 4.1 plays a vital role in cancer as a tumor suppressor. Many studies have also demonstrated that cytoskeleton protein 4.1 acts as a diagnostic and prognostic biomarker for tumors. Moreover, with the rise of immunotherapy, the tumor microenvironment as a treatment target in cancer has attracted great interest. Increasing evidence has shown the immunoregulatory potential of cytoskeleton protein 4.1 in the tumor microenvironment and treatment. In this review, we discuss the role of cytoskeleton protein 4.1 within the tumor microenvironment in immunoregulation and cancer development, with the intention of providing a new approach and new ideas for future cancer diagnosis and treatment.
Collapse
|
5
|
Saito M, Hirano M, Izumi T, Mori Y, Ito K, Saitoh Y, Terada N, Sato T, Sukegawa J. Cytoskeletal Protein 4.1G Is Essential for the Primary Ciliogenesis and Osteoblast Differentiation in Bone Formation. Int J Mol Sci 2022; 23:ijms23042094. [PMID: 35216233 PMCID: PMC8878336 DOI: 10.3390/ijms23042094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/06/2022] [Accepted: 02/10/2022] [Indexed: 12/04/2022] Open
Abstract
The primary cilium is a hair-like immotile organelle with specific membrane receptors, including the receptor of Hedgehog signaling, smoothened. The cilium organized in preosteoblasts promotes differentiation of the cells into osteoblasts (osteoblast differentiation) by mediating Hedgehog signaling to achieve bone formation. Notably, 4.1G is a plasma membrane-associated cytoskeletal protein that plays essential roles in various tissues, including the peripheral nervous system, testis, and retina. However, its function in the bone remains unexplored. In this study, we identified 4.1G expression in the bone. We found that, in the 4.1G-knockout mice, calcium deposits and primary cilium formation were suppressed in the trabecular bone, which is preosteoblast-rich region of the newborn tibia, indicating that 4.1G is a prerequisite for osteoblast differentiation by organizing the primary cilia in preosteoblasts. Next, we found that the primary cilium was elongated in the differentiating mouse preosteoblast cell line MC3T3-E1, whereas the knockdown of 4.1G suppressed its elongation. Moreover, 4.1G-knockdown suppressed the induction of the cilia-mediated Hedgehog signaling and subsequent osteoblast differentiation. These results demonstrate a new regulatory mechanism of 4.1G in bone formation that promotes the primary ciliogenesis in the differentiating preosteoblasts and induction of cilia-mediated osteoblast differentiation, resulting in bone formation at the newborn stage.
Collapse
Affiliation(s)
- Masaki Saito
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai 980-8575, Japan; (M.H.); (T.I.); (T.S.)
- Correspondence: ; Tel.: +81-22-717-8207
| | - Marina Hirano
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai 980-8575, Japan; (M.H.); (T.I.); (T.S.)
- Department of Human Health and Nutrition, Shokei Gakuin University, Natori 981-1295, Japan;
| | - Tomohiro Izumi
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai 980-8575, Japan; (M.H.); (T.I.); (T.S.)
| | - Yu Mori
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan; (Y.M.); (K.I.)
| | - Kentaro Ito
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan; (Y.M.); (K.I.)
| | - Yurika Saitoh
- Center for Medical Education, Teikyo University of Science, Adachi-ku, Tokyo 120-0045, Japan;
| | - Nobuo Terada
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto 390-0802, Japan;
| | - Takeya Sato
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai 980-8575, Japan; (M.H.); (T.I.); (T.S.)
| | - Jun Sukegawa
- Department of Human Health and Nutrition, Shokei Gakuin University, Natori 981-1295, Japan;
| |
Collapse
|
6
|
Ning S, Hua L, Ji Z, Fan D, Meng X, Li Z, Wang Q, Guo Z. Protein 4.1 family and ion channel proteins interact to regulate the process of heart failure in rats. Acta Histochem 2021; 123:151748. [PMID: 34271280 DOI: 10.1016/j.acthis.2021.151748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023]
Abstract
Heart failure (HF) is a major cause of death in cardiovascular diseases worldwide, and its molecular mechanisms and effective prevention strategies remain to be further studied. The myocardial cytoskeleton plays a pivotal role in many heart diseases. However, little is known about the function of the membrane cytoskeleton 4.1 protein family and related regulatory mechanisms in the pathogenesis of HF. In this study, we detected the localization and expression of the protein 4.1 family and ion channel proteins in a rat HF model induced by doxorubicin (DOX), and studied the interactions between them. Our results showed that compared with the control group, the HF group displayed an increased expression level of protein 4.1R and decreased levels of protein 4.1 G and 4.1 N. The Nav1.5 protein levels were significantly increased, while the SERCA2a and Cav1.2 protein levels were significantly decreased in the HF group. Furthermore, there is co-localization and interaction between protein 4.1R and Nav1.5, protein 4.1 G and SERCA2a, protein 4.1 N and Cav1.2, respectively. Taken together, the results indicated that the protein 4.1 family might be involved in the occurrence and development of HF through its interaction with ion channel proteins, suggesting that 4.1 proteins may serve as a novel therapeutic target for HF.
Collapse
Affiliation(s)
- Shuwei Ning
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Lei Hua
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Zhenyu Ji
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Dandan Fan
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Xiangguang Meng
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Zhiying Li
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Qian Wang
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Zhikun Guo
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China; Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
7
|
Saito M, Cui L, Hirano M, Li G, Yanagisawa T, Sato T, Sukegawa J. Activity of Adenylyl Cyclase Type 6 Is Suppressed by Direct Binding of the Cytoskeletal Protein 4.1G. Mol Pharmacol 2019; 96:441-451. [PMID: 31383768 DOI: 10.1124/mol.119.116426] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/30/2019] [Indexed: 11/22/2022] Open
Abstract
The G protein-coupled receptor (GPCR) signaling pathways mediated by trimeric G proteins have been extensively elucidated, but their associated regulatory mechanisms remain unclear. Parathyroid hormone (PTH)/PTH-related protein receptor (PTHR) is a GPCR coupled with Gs and Gq Gs activates adenylyl cyclases (ACs), which produces cAMP to regulate various cell fates. We previously showed that cell surface expression of PTHR was increased by its direct interaction with a subcortical cytoskeletal protein, 4.1G, whereas PTHR-mediated Gs/AC/cAMP signaling was suppressed by 4.1G through an unknown mechanism in human embryonic kidney (HEK)293 cells. In the present study, we found that AC type 6 (AC6), one of the major ACs activated downstream of PTHR, interacts with 4.1G in HEK293 cells, and the N-terminus of AC6 (AC6-N) directly and selectively binds to the 4.1/ezrin/radixin/moesin (FERM) domain of 4.1G (4.1G-FERM) in vitro. AC6-N was distributed at the plasma membrane, which was disturbed by knockdown of 4.1G. An AC6-N mutant, AC6-N-3A, in which three consecutive arginine residues are mutated to alanine residues, altered both binding to 4.1G-FERM and its plasma membrane distribution in vivo. Further, we overexpressed AC6-N to competitively inhibit the interaction of endogenous AC6 and 4.1G in cells. cAMP production induced by forskolin, an adenylyl cyclase activator, and PTH-(1-34) was enhanced by AC6-N expression and 4.1G-knockdown. In contrast, AC6-N-3A had no impact on forskolin- and PTH-(1-34)-induced cAMP productions. These data provide a novel regulatory mechanism that AC6 activity is suppressed by the direct binding of 4.1G to AC6-N, resulting in attenuation of PTHR-mediated Gs/AC6/cAMP signaling.
Collapse
Affiliation(s)
- Masaki Saito
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai, Miyagi, Japan (M.S., L.C., M.H., G.L., T.Y., T.S., J.S.); Department of Human Health and Nutrition, Shokei Gakuin University, Natori, Miyagi, Japan (M.H., J.S.); and Faculty of Health Sciences, Tohoku Fukushi University, Sendai, Miyagi, Japan (T.Y.)
| | - Linran Cui
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai, Miyagi, Japan (M.S., L.C., M.H., G.L., T.Y., T.S., J.S.); Department of Human Health and Nutrition, Shokei Gakuin University, Natori, Miyagi, Japan (M.H., J.S.); and Faculty of Health Sciences, Tohoku Fukushi University, Sendai, Miyagi, Japan (T.Y.)
| | - Marina Hirano
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai, Miyagi, Japan (M.S., L.C., M.H., G.L., T.Y., T.S., J.S.); Department of Human Health and Nutrition, Shokei Gakuin University, Natori, Miyagi, Japan (M.H., J.S.); and Faculty of Health Sciences, Tohoku Fukushi University, Sendai, Miyagi, Japan (T.Y.)
| | - Guanjie Li
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai, Miyagi, Japan (M.S., L.C., M.H., G.L., T.Y., T.S., J.S.); Department of Human Health and Nutrition, Shokei Gakuin University, Natori, Miyagi, Japan (M.H., J.S.); and Faculty of Health Sciences, Tohoku Fukushi University, Sendai, Miyagi, Japan (T.Y.)
| | - Teruyuki Yanagisawa
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai, Miyagi, Japan (M.S., L.C., M.H., G.L., T.Y., T.S., J.S.); Department of Human Health and Nutrition, Shokei Gakuin University, Natori, Miyagi, Japan (M.H., J.S.); and Faculty of Health Sciences, Tohoku Fukushi University, Sendai, Miyagi, Japan (T.Y.)
| | - Takeya Sato
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai, Miyagi, Japan (M.S., L.C., M.H., G.L., T.Y., T.S., J.S.); Department of Human Health and Nutrition, Shokei Gakuin University, Natori, Miyagi, Japan (M.H., J.S.); and Faculty of Health Sciences, Tohoku Fukushi University, Sendai, Miyagi, Japan (T.Y.)
| | - Jun Sukegawa
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai, Miyagi, Japan (M.S., L.C., M.H., G.L., T.Y., T.S., J.S.); Department of Human Health and Nutrition, Shokei Gakuin University, Natori, Miyagi, Japan (M.H., J.S.); and Faculty of Health Sciences, Tohoku Fukushi University, Sendai, Miyagi, Japan (T.Y.)
| |
Collapse
|
8
|
A/L B Vasanth Rao VR, Tan SH, Candasamy M, Bhattamisra SK. Diabetic nephropathy: An update on pathogenesis and drug development. Diabetes Metab Syndr 2019; 13:754-762. [PMID: 30641802 DOI: 10.1016/j.dsx.2018.11.054] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 11/29/2018] [Indexed: 01/08/2023]
Abstract
Diabetic nephropathy (DN) is a major cause of end-stage renal disease and affects a large number of individuals with diabetes. However, the development of specific treatments for DN has not yet been identified. Hence, this review is concisely designed to understand the molecular pathways leading to DN in order to develop suitable therapeutic strategies. Extensive literature search have been carried in regard with the pathogenesis and pathophysiology of DN, drug targets and updates on clinical trials, the consequences associated with DN and the potential biomarkers for diagnosis and prediction of DN are discussed in this review. DN is characterised by microalbuminuria and macroalbuminuria, and morphological changes such as glomerular thickening, interstitial fibrosis, formation of nodular glomerulosclerosis and decreased endothelial cell fenestration. Besides, the involvement of renin-angiotensin-aldosterone system, inflammation and genetic factors are the key pathways in the progression of DN. In regard with drug development drugs targeted to epidermal growth factor, inflammatory cytokines, ACTH receptor and TGFβ1 receptors are in pipeline for clinical trials whereas, several drugs have also failed in phase III and phase IV of clinical trials due to lack of efficacy and severe adverse effect. The research on DN is limited with respect to its pathogenesis and drug development. Thus, a more detailed understanding of the pathogenesis of DN is very essential to progress in the drug development process.
Collapse
Affiliation(s)
- Vikram Rao A/L B Vasanth Rao
- School of Postgraduate Studies, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| | - Sean Hong Tan
- School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| | - Mayuren Candasamy
- Department of Life Sciences, School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| | - Subrat Kumar Bhattamisra
- Department of Life Sciences, School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
9
|
Protein 4.1N is required for the formation of the lateral membrane domain in human bronchial epithelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1143-1151. [PMID: 29428502 DOI: 10.1016/j.bbamem.2018.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/16/2018] [Accepted: 02/07/2018] [Indexed: 12/13/2022]
Abstract
The membrane skeleton forms a scaffold on the cytoplasmic side of the plasma membrane. The erythrocyte membrane represents an archetype of such structural organization. It has been documented that a similar membrane skeleton also exits in the Golgi complex. It has been previously shown that βII spectrin and ankyrin G are localized at the lateral membrane of human bronchial epithelial cells. Here we show that protein 4.1N is also located at the lateral membrane where it associates E-cadherin, β-catenin and βII spectrin. Importantly, depletion of 4.1N by RNAi in human bronchial epithelial cells resulted in decreased height of lateral membrane, which was reversed following re-expression of mouse 4.1N. Furthermore, although the initial phase of lateral membrane biogenesis proceeded normally in 4.1N-depleted cells, the final height of the lateral membrane of 4.1N-depleted cells was shorter compared to that of control cells. Our findings together with previous findings imply that 4.1N, βII spectrin and ankyrin G are structural components of the lateral membrane skeleton and that this skeleton plays an essential role in the assembly of a fully functional lateral membrane.
Collapse
|
10
|
王 成, 康 巧, 丁 聪, 李 雅, 梁 桃, 张 成, 王 文, 王 婷. [Construction of a stable 4.1R gene knockout cell model in RAW264.7 cells using CRISPR/Cas9 technique]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:1609-1614. [PMID: 29292253 PMCID: PMC6744011 DOI: 10.3969/j.issn.1673-4254.2017.12.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To construct a cell model of 4.1R gene knockout in murine macrophage cell line RAW264.7 using CRISPR/Cas9 technique. METHODS Three high?grade small?guide RNAs (sgRNAs) that could specifically identify 4.1R gene were synthesized and inserted into lentiCRISPRv2 plasmid. RAW264.7 cells were infected with sgRNA?Cas9 lentivirus from 293T cells transfected with the recombinant sgRNA?lentiCRISPRv2 plasmid, and the positive cells were screened using puromycin and the monoclonal cells were obtained. The expression of 4.1R protein in the monoclonal cells was measured by Western blotting, and the mutation site was confirmed by sequence analysis. Result A 4.1R gene knockout RAW264.7 cell line was obtained, which showed a 19?bp deletion mutation in the 4.1R gene sequence and obviously enhanced proliferation. CONCLUSION We successfully constructed a 4.1R gene knockout macrophage cell line using CRISPR/Cas9 technique, which may facilitate further investigation of the function of 4.1R in macrophages.
Collapse
Affiliation(s)
- 成博 王
- />郑州大学生命科学学院,河南 郑州 450000School of Life Sciences, Zhengzhou University, Zhengzhou 45000, China
| | - 巧珍 康
- />郑州大学生命科学学院,河南 郑州 450000School of Life Sciences, Zhengzhou University, Zhengzhou 45000, China
| | - 聪 丁
- />郑州大学生命科学学院,河南 郑州 450000School of Life Sciences, Zhengzhou University, Zhengzhou 45000, China
| | - 雅雯 李
- />郑州大学生命科学学院,河南 郑州 450000School of Life Sciences, Zhengzhou University, Zhengzhou 45000, China
| | - 桃桃 梁
- />郑州大学生命科学学院,河南 郑州 450000School of Life Sciences, Zhengzhou University, Zhengzhou 45000, China
| | - 成龙 张
- />郑州大学生命科学学院,河南 郑州 450000School of Life Sciences, Zhengzhou University, Zhengzhou 45000, China
| | - 文 王
- />郑州大学生命科学学院,河南 郑州 450000School of Life Sciences, Zhengzhou University, Zhengzhou 45000, China
| | - 婷 王
- />郑州大学生命科学学院,河南 郑州 450000School of Life Sciences, Zhengzhou University, Zhengzhou 45000, China
| |
Collapse
|
11
|
Stankewich MC, Moeckel GW, Ji L, Ardito T, Morrow JS. Isoforms of Spectrin and Ankyrin Reflect the Functional Topography of the Mouse Kidney. PLoS One 2016; 11:e0142687. [PMID: 26727517 PMCID: PMC4703142 DOI: 10.1371/journal.pone.0142687] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 10/26/2015] [Indexed: 11/24/2022] Open
Abstract
The kidney displays specialized regions devoted to filtration, selective reabsorption, and electrolyte and metabolite trafficking. The polarized membrane pumps, channels, and transporters responsible for these functions have been exhaustively studied. Less examined are the contributions of spectrin and its adapter ankyrin to this exquisite functional topography, despite their established contributions in other tissues to cellular organization. We have examined in the rodent kidney the expression and distribution of all spectrins and ankyrins by qPCR, Western blotting, immunofluorescent and immuno electron microscopy. Four of the seven spectrins (αΙΙ, βΙ, βΙΙ, and βΙΙΙ) are expressed in the kidney, as are two of the three ankyrins (G and B). The levels and distribution of these proteins vary widely over the nephron. αΙΙ/βΙΙ is the most abundant spectrin, found in glomerular endothelial cells; on the basolateral membrane and cytoplasmic vesicles in proximal tubule cells and in the thick ascending loop of Henle; and less so in the distal nephron. βΙΙΙ spectrin largely replaces βΙΙ spectrin in podocytes, Bowman’s capsule, and throughout the distal tubule and collecting ducts. βΙ spectrin is only marginally expressed; its low abundance hinders a reliable determination of its distribution. Ankyrin G is the most abundant ankyrin, found in capillary endothelial cells and all tubular segments. Ankyrin B populates Bowman’s capsule, podocytes, the ascending thick loop of Henle, and the distal convoluted tubule. Comparison to the distribution of renal protein 4.1 isoforms and various membrane proteins indicates a complex relationship between the spectrin scaffold, its adapters, and various membrane proteins. While some proteins (e.g. ankyrin B, βΙΙΙ spectrin, and aquaporin 2) tend to share a similar distribution, there is no simple mapping of different spectrins or ankyrins to most membrane proteins. The implications of this data are discussed.
Collapse
Affiliation(s)
- Michael C. Stankewich
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States of America
- * E-mail:
| | - Gilbert W. Moeckel
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States of America
| | - Lan Ji
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States of America
| | - Thomas Ardito
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States of America
| | - Jon S. Morrow
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States of America
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, United States of America
| |
Collapse
|
12
|
Buffon MP, Carpena MP, Sortica DA, Santer A, Carlessi R, de Souza BM, Edelweiss MI, Berger M, Crispim D, Canani LH. rs1888747 polymorphism in the FRMD3 gene, gene and protein expression: role in diabetic kidney disease. Diabetol Metab Syndr 2016; 8:3. [PMID: 26753002 PMCID: PMC4706705 DOI: 10.1186/s13098-015-0121-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 12/23/2015] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND We carried out a case-control study in patients with type 2 diabetes mellitus (T2DM) to evaluate the association between seven single nucleotide polymorphisms (SNPs) previously described to be linked to diabetic kidney disease (DKD) in type 1 diabetes mellitus (T1DM). Additionally, we evaluated gene and protein expression related to the polymorphism associated with DKD. METHODS The association study included 1098 T2DM patients (718 with DKD and 380 without DKD). Out of the 13 polymorphisms associated with DKD in a previous study with T1DM, seven were chosen for evaluation in this sample: rs1888747, rs9521445, rs39075, rs451041, rs1041466, rs1411766 and rs6492208. The expression study included 91 patients who underwent nephrectomy. Gene expression was assessed by RT-qPCR and protein expression in kidney samples was quantified by western blot and it localization by immunohistochemistry. RESULTS The C/C genotype of rs1888747 SNP was associated with protection for DKD (OR = 0.6, 95 % CI 0.3-0.9; P = 0.022). None of the other SNPs were associated with DKD. rs1888747 is located near FRMD3 gene. Therefore, FRMD3 gene and protein expression were evaluated in human kidney tissue according to rs1888747 genotypes. Gene and protein expression were similar in subjects homozygous for the C allele and in those carrying the G allele. CONCLUSIONS Replication of the association between rs1888747 SNP and DKD in a different population suggests that this link is not the result of chance. rs1888747 SNP is located at the FRMD3 gene, which is expressed in human kidney. Therefore, this gene is a candidate gene for DKD. However, in this study, no rs1888747 genotype or specific allele effect on gene and/or protein expression of the FRMD3 gene was demonstrated.
Collapse
Affiliation(s)
- Marjoriê P. Buffon
- />Endocrine Division, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, prédio 12, 4° andar, Porto Alegre, RS 90035-003 Brazil
- />Post-Graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS Brazil
| | - Mariana P. Carpena
- />Post-Graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS Brazil
| | - Denise A. Sortica
- />Endocrine Division, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, prédio 12, 4° andar, Porto Alegre, RS 90035-003 Brazil
- />Post-Graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS Brazil
| | - Andressa Santer
- />Endocrine Division, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, prédio 12, 4° andar, Porto Alegre, RS 90035-003 Brazil
| | - Rodrigo Carlessi
- />Endocrine Division, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, prédio 12, 4° andar, Porto Alegre, RS 90035-003 Brazil
- />Post-Graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS Brazil
| | - Bianca M. de Souza
- />Endocrine Division, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, prédio 12, 4° andar, Porto Alegre, RS 90035-003 Brazil
- />Post-Graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS Brazil
| | - Maria I. Edelweiss
- />Pathology Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS Brazil
- />Post-Graduation Program in Medical Sciences: Ginecology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS Brazil
| | - Milton Berger
- />Urology Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS Brazil
| | - Daisy Crispim
- />Endocrine Division, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, prédio 12, 4° andar, Porto Alegre, RS 90035-003 Brazil
- />Post-Graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS Brazil
| | - Luís H. Canani
- />Endocrine Division, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, prédio 12, 4° andar, Porto Alegre, RS 90035-003 Brazil
- />Post-Graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS Brazil
| |
Collapse
|
13
|
Na+-H+ exchanger-1 (NHE1) regulation in kidney proximal tubule. Cell Mol Life Sci 2015; 72:2061-74. [PMID: 25680790 DOI: 10.1007/s00018-015-1848-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 01/28/2015] [Accepted: 01/29/2015] [Indexed: 01/17/2023]
Abstract
The ubiquitously expressed plasma membrane Na(+)-H(+) exchanger NHE1 is a 12 transmembrane-spanning protein that directs important cell functions such as homeostatic intracellular volume and pH control. The 315 amino acid cytosolic tail of NHE1 binds plasma membrane phospholipids and multiple proteins that regulate additional, ion-translocation independent functions. This review focuses on NHE1 structure/function relationships, as well as the role of NHE1 in kidney proximal tubule functions, including pH regulation, vectorial Na(+) transport, cell volume control and cell survival. The implications of these functions are particularly critical in the setting of progressive, albuminuric kidney diseases, where the accumulation of reabsorbed fatty acids leads to disruption of NHE1-membrane phospholipid interactions and tubular atrophy, which is a poor prognostic factor for progression to end stage renal disease. This review amplifies the vital role of the proximal tubule NHE1 Na(+)-H(+) exchanger as a kidney cell survival factor.
Collapse
|
14
|
Buffon MP, Sortica DA, Gerchman F, Crispim D, Canani LH. FRMD3 gene: its role in diabetic kidney disease. A narrative review. Diabetol Metab Syndr 2015; 7:118. [PMID: 26719775 PMCID: PMC4696171 DOI: 10.1186/s13098-015-0114-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 12/19/2015] [Indexed: 01/15/2023] Open
Abstract
Diabetic kidney disease (DKD) is a chronic complication of diabetes mellitus, which is considered a worldwide epidemic. Several studies have been developed in order to elucidate possible genetic factors involved in this disease. The FRMD3 gene, a strong candidate selected from genome wide association studies (GWAS), encodes the structural protein 4.1O involved in maintaining cell shape and integrity. Some single nucleotide polymorphisms (SNPs) located in FRMD3 have been associated with DKD in different ethnicities. However, despite these findings, the matter is still controversial. The aim of this narrative review is to summarize the evidence regarding the role of FRMD3 in DKD.
Collapse
Affiliation(s)
- Marjoriê Piuco Buffon
- />Endocrine Division, Hospital de Clínicas de Porto Alegre, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos 2350, prédio 12, 4° andar, Porto Alegre, RS 90035-003 Brazil
- />Endocrinology, Federal University of Rio Grande do Sul, Porto Alegre, RS Brazil
- />Federal University of Rio Grande do Sul, Porto Alegre, RS Brazil
| | - Denise Alves Sortica
- />Endocrine Division, Hospital de Clínicas de Porto Alegre, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos 2350, prédio 12, 4° andar, Porto Alegre, RS 90035-003 Brazil
- />Endocrinology, Federal University of Rio Grande do Sul, Porto Alegre, RS Brazil
- />Federal University of Rio Grande do Sul, Porto Alegre, RS Brazil
| | - Fernando Gerchman
- />Endocrine Division, Hospital de Clínicas de Porto Alegre, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos 2350, prédio 12, 4° andar, Porto Alegre, RS 90035-003 Brazil
- />Endocrinology, Federal University of Rio Grande do Sul, Porto Alegre, RS Brazil
- />Federal University of Rio Grande do Sul, Porto Alegre, RS Brazil
| | - Daisy Crispim
- />Endocrine Division, Hospital de Clínicas de Porto Alegre, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos 2350, prédio 12, 4° andar, Porto Alegre, RS 90035-003 Brazil
- />Endocrinology, Federal University of Rio Grande do Sul, Porto Alegre, RS Brazil
| | - Luís Henrique Canani
- />Endocrine Division, Hospital de Clínicas de Porto Alegre, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos 2350, prédio 12, 4° andar, Porto Alegre, RS 90035-003 Brazil
- />Endocrinology, Federal University of Rio Grande do Sul, Porto Alegre, RS Brazil
- />Federal University of Rio Grande do Sul, Porto Alegre, RS Brazil
| |
Collapse
|
15
|
Wang Z, Zhang J, Ye M, Zhu M, Zhang B, Roy M, Liu J, An X. Tumor suppressor role of protein 4.1B/DAL-1. Cell Mol Life Sci 2014; 71:4815-30. [PMID: 25183197 PMCID: PMC11113756 DOI: 10.1007/s00018-014-1707-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 07/21/2014] [Accepted: 08/15/2014] [Indexed: 12/14/2022]
Abstract
Protein 4.1B/DAL-1 is a membrane skeletal protein that belongs to the protein 4.1 family. Protein 4.1B/DAL-1 is localized to sites of cell-cell contact and functions as an adapter protein, linking the plasma membrane to the cytoskeleton or associated cytoplasmic signaling effectors and facilitating their activities in various pathways. Protein 4.1B/DAL-1 is involved in various cytoskeleton-associated processes, such as cell motility and adhesion. Moreover, protein 4.1B/DAL-1 also plays a regulatory role in cell growth, differentiation, and the establishment of epithelial-like cell structures. Protein 4.1B/DAL-1 is normally expressed in multiple human tissues, but loss of its expression or prominent down-regulation of its expression is frequently observed in corresponding tumor tissues and tumor cell lines, suggesting that protein 4.1B/DAL-1 is involved in the molecular pathogenesis of these tumors and acts as a potential tumor suppressor. This review will focus on the structure of protein 4.1B/DAL-1, 4.1B/DAL-1-interacting molecules, 4.1B/DAL-1 inactivation and tumor progression, and anti-tumor activity of the 4.1B/DAL-1.
Collapse
Affiliation(s)
- Zi Wang
- Molecular Biology Research Center, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, 410078 China
| | - Ji Zhang
- Molecular Biology Research Center, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, 410078 China
- Department of Hematology, The First Affiliated Hospital, University of South China, Hengyang, 421001 China
| | - Mao Ye
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, 410082 China
| | - Min Zhu
- Molecular Biology Research Center, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, 410078 China
| | - Bin Zhang
- Department of Histology and Embryology, Xiangya School Medicine, Central South University, Changsha, 410083 China
| | - Mridul Roy
- Molecular Biology Research Center, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, 410078 China
| | - Jing Liu
- Molecular Biology Research Center, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, 410078 China
- State Key Laboratory of Medical Genetics, Central South University, 110 Xiangya Road, Changsha, 410078 China
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, 310 E 67th Street, New York, 10065 USA
| |
Collapse
|
16
|
Zhang J, Yang S, An C, Wang J, Yan H, Huang Y, Song J, Yin C, Baines AJ, Mohandas N, An X. Comprehensive characterization of protein 4.1 expression in epithelium of large intestine. Histochem Cell Biol 2014; 142:529-39. [PMID: 24912669 DOI: 10.1007/s00418-014-1224-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2014] [Indexed: 11/24/2022]
Abstract
The protein 4.1 family consists of four members, 4.1R, 4.1N, 4.1B and 4.1G, each encoded by a distinct gene. All 4.1 mRNAs undergo extensive alternative splicing. Functionally, they usually serve as adapters that link actin-based cytoskeleton to plasma membrane proteins. It has been reported that 4.1 proteins are expressed in most animal cell types and tissues including epithelial cells and epithelial tissues. However, the expression of 4.1 proteins in large intestine has not been well characterized. In the present study, we performed RT-PCR, western blot and immunohistochemistry analysis to characterize the transcripts, the protein expression and cellular localization of 4.1 proteins in the epithelia of mouse large intestine. We show that multiple transcripts derive from each gene, including eight 4.1R isoforms, four 4.1N isoforms, four 4.1B isoforms and six 4.1G isoforms. However, at the protein level, only one or two major bands were detected, implying that not all transcripts are translated and/or the proteins do not accumulate at detectable levels. Immunohistochemistry revealed that 4.1R, 4.1N and 4.1B are all expressed at the lateral membrane as well as cytoplasm of epithelial cells, suggesting a potentially redundant role of these proteins. Our findings not only provide new insights into the structure of protein 4.1 genes but also lay the foundation for future functional studies.
Collapse
Affiliation(s)
- Jingxin Zhang
- Department of Biophysics, Peking University Health Science Center, Xueyuan Road, Haidian District, Beijing, 100191, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Baines AJ, Lu HC, Bennett PM. The Protein 4.1 family: hub proteins in animals for organizing membrane proteins. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1838:605-19. [PMID: 23747363 DOI: 10.1016/j.bbamem.2013.05.030] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 05/22/2013] [Accepted: 05/28/2013] [Indexed: 01/10/2023]
Abstract
Proteins of the 4.1 family are characteristic of eumetazoan organisms. Invertebrates contain single 4.1 genes and the Drosophila model suggests that 4.1 is essential for animal life. Vertebrates have four paralogues, known as 4.1R, 4.1N, 4.1G and 4.1B, which are additionally duplicated in the ray-finned fish. Protein 4.1R was the first to be discovered: it is a major mammalian erythrocyte cytoskeletal protein, essential to the mechanochemical properties of red cell membranes because it promotes the interaction between spectrin and actin in the membrane cytoskeleton. 4.1R also binds certain phospholipids and is required for the stable cell surface accumulation of a number of erythrocyte transmembrane proteins that span multiple functional classes; these include cell adhesion molecules, transporters and a chemokine receptor. The vertebrate 4.1 proteins are expressed in most tissues, and they are required for the correct cell surface accumulation of a very wide variety of membrane proteins including G-Protein coupled receptors, voltage-gated and ligand-gated channels, as well as the classes identified in erythrocytes. Indeed, such large numbers of protein interactions have been mapped for mammalian 4.1 proteins, most especially 4.1R, that it appears that they can act as hubs for membrane protein organization. The range of critical interactions of 4.1 proteins is reflected in disease relationships that include hereditary anaemias, tumour suppression, control of heartbeat and nervous system function. The 4.1 proteins are defined by their domain structure: apart from the spectrin/actin-binding domain they have FERM and FERM-adjacent domains and a unique C-terminal domain. Both the FERM and C-terminal domains can bind transmembrane proteins, thus they have the potential to be cross-linkers for membrane proteins. The activity of the FERM domain is subject to multiple modes of regulation via binding of regulatory ligands, phosphorylation of the FERM associated domain and differential mRNA splicing. Finally, the spectrum of interactions of the 4.1 proteins overlaps with that of another membrane-cytoskeleton linker, ankyrin. Both ankyrin and 4.1 link to the actin cytoskeleton via spectrin, and we hypothesize that differential regulation of 4.1 proteins and ankyrins allows highly selective control of cell surface protein accumulation and, hence, function. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé
Collapse
Affiliation(s)
| | - Hui-Chun Lu
- Randall Division of Cell and Molecular Biophysics, King's College London, UK
| | - Pauline M Bennett
- Randall Division of Cell and Molecular Biophysics, King's College London, UK.
| |
Collapse
|
18
|
Cheng CL, Molday RS. Interaction of 4.1G and cGMP-gated channels in rod photoreceptor outer segments. J Cell Sci 2013; 126:5725-34. [PMID: 24144699 DOI: 10.1242/jcs.137679] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
In photoreceptors, the assembly of signaling molecules into macromolecular complexes is important for phototransduction and maintaining the structural integrity of rod outer segments (ROSs). However, the molecular composition and formation of these complexes are poorly understood. Using immunoprecipitation and mass spectrometry, 4.1G was identified as a new interacting partner for the cyclic-nucleotide gated (CNG) channels in ROSs. 4.1G is a widely expressed multifunctional protein that plays a role in the assembly and stability of membrane protein complexes. Multiple splice variants of 4.1G were cloned from bovine retina. A smaller splice variant of 4.1G selectively interacted with CNG channels not associated with peripherin-2-CNG channel complex. A combination of truncation studies and domain-binding assays demonstrated that CNG channels selectively interacted with 4.1G through their FERM and CTD domains. Using immunofluorescence, labeling of 4.1G was seen to be punctate and partially colocalized with CNG channels in the ROS. Our studies indicate that 4.1G interacts with a subset of CNG channels in the ROS and implicate this protein-protein interaction in organizing the spatial arrangement of CNG channels in the plasma membrane of outer segments.
Collapse
Affiliation(s)
- Christiana L Cheng
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | | |
Collapse
|
19
|
Xi C, Ren C, Hu A, Lin J, Yao Q, Wang Y, Gao Z, An X, Liu C. Defective expression of Protein 4.1N is correlated to tumor progression, aggressive behaviors and chemotherapy resistance in epithelial ovarian cancer. Gynecol Oncol 2013; 131:764-71. [PMID: 23994105 DOI: 10.1016/j.ygyno.2013.08.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 07/16/2013] [Accepted: 08/08/2013] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Protein 4.1N (4.1N) is a member of the Protein 4.1 family that is involved in cellular processes such as cell adhesion, migration and signaling. In this study, we evaluated the expression of 4.1N protein and its potential roles in epithelial ovarian cancer (EOC) tumorigenesis and progression. METHODS 4.1N protein expression was investigated in a total of 280 samples including 74 normal tissues, 35 benign, 30 borderline and 141 malignant epithelial ovarian tumors by immunohistochemistry. Correlation between 4.1N expression levels and clinicopathologic features was statistically analyzed. The expression of 4.1N in EOC cell lines was examined by western blotting. RESULTS Immunohistochemistry analysis revealed that, although there was no loss of 4.1N expression in normal tissues and benign tumors, absence of Protein 4.1N was significantly more common in EOCs (44.0%) than in borderline tumors (3.3%) (p<0.001). Furthermore, loss or decreased expression of 4.1N protein expression was correlated with malignant potential of the tumors (14.3% in benign tumors, 56.7% in borderline tumors and 92.9% in malignancy) (p<0.001). In EOC samples, loss of 4.1N protein was significantly associated with advanced-stage (p=0.004), ascites (p=0.009), omental metastasis (p=0.018), suboptimal debulking (p=0.024), poorly histological differentiation (p=0.009), high-grade serous carcinoma (p=0.001), short progression-free-survival (p=0.018) and poor chemosensitivity to first-line chemotherapy (p=0.029). Moreover, western blotting analysis revealed that expression of 4.1N protein was lost in 4/8 (50%) EOC cell lines. CONCLUSIONS 4.1N protein expression level was significantly decreased during malignant transformation of epithelial ovarian tumors and that loss of 4.1N expression was closely correlated to poorly differentiated and biologically aggressive EOCs.
Collapse
Affiliation(s)
- Chenguang Xi
- Department of Pathology, Peking University Health Science Center, Beijing 100191, China
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Liu C, Weng H, Chen L, Yang S, Wang H, Debnath G, Guo X, Wu L, Mohandas N, An X. Impaired intestinal calcium absorption in protein 4.1R-deficient mice due to altered expression of plasma membrane calcium ATPase 1b (PMCA1b). J Biol Chem 2013; 288:11407-15. [PMID: 23460639 DOI: 10.1074/jbc.m112.436659] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Protein 4.1R was first identified in the erythrocyte membrane skeleton. It is now known that the protein is expressed in a variety of epithelial cell lines and in the epithelia of many tissues, including the small intestine. However, the physiological function of 4.1R in the epithelial cells of the small intestine has not so far been explored. Here, we show that 4.1R knock-out mice exhibited a significantly impaired small intestinal calcium absorption that resulted in secondary hyperparathyroidism as evidenced by increased serum 1,25-(OH)2-vitamin D3 and parathyroid hormone levels, decreased serum calcium levels, hyperplasia of the parathyroid, and demineralization of the bones. 4.1R is located on the basolateral membrane of enterocytes, where it co-localizes with PMCA1b (plasma membrane calcium ATPase 1b). Expression of PMCA1b in enterocytes was decreased in 4.1(-/-) mice. 4.1R directly associated with PMCA1b, and the association involved the membrane-binding domain of 4.1R and the second intracellular loop and C terminus of PMCA1b. Our findings have enabled us to define a functional role for 4.1R in small intestinal calcium absorption through regulation of membrane expression of PMCA1b.
Collapse
Affiliation(s)
- Congrong Liu
- Red Cell Physiology Laboratory, New York Blood Center, New York, New York 10065, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Jung Y, McCarty JH. Band 4.1 proteins regulate integrin-dependent cell spreading. Biochem Biophys Res Commun 2012; 426:578-84. [PMID: 22982319 DOI: 10.1016/j.bbrc.2012.08.129] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 08/25/2012] [Indexed: 11/30/2022]
Abstract
Integrins link the extracellular matrix (ECM) to the cytoskeleton to control cell behaviors including adhesion, spreading and migration. Band 4.1 proteins contain 4.1, ezrin, radixin, moesin (FERM) domains that likely mediate signaling events and cytoskeletal reorganization via integrins. However, the mechanisms by which Band 4.1 proteins and integrins are functionally interconnected remain enigmatic. Here we have investigated roles for Band 4.1 proteins in integrin-mediated cell spreading using primary astrocytes as a model system. We demonstrate that Proteins 4.1B and 4.1G show dynamic patterns of sub-cellular localization in astrocytes spreading on fibronectin. During early stages of cell spreading Proteins 4.1B and 4.1G are enriched in ECM adhesion sites but become more diffusely localized at later stages of spreading. Combinatorial inactivation of Protein 4.1B and 4.1G expression leads to impaired astrocyte spreading. Furthermore, in exogenous expression systems we show that the isolated Protein 4.1 FERM domain significantly enhances integrin-mediated cell spreading. Protein 4.1B is dispensable for reactive astrogliosis in experimental models of cortical injury, likely due to functional compensation by related Protein 4.1 family members. Collectively, these findings reveal that Band 4.1 proteins are important intracellular components for integrin-mediated cell spreading.
Collapse
Affiliation(s)
- Youngsin Jung
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston TX 77030, United States
| | | |
Collapse
|
22
|
Pinder JC, Taylor-Harris PM, Bennett PM, Carter E, Hayes NVL, King MDA, Holt MR, Maggs AM, Gascard P, Baines AJ. Isoforms of protein 4.1 are differentially distributed in heart muscle cells: relation of 4.1R and 4.1G to components of the Ca2+ homeostasis system. Exp Cell Res 2012; 318:1467-79. [PMID: 22429617 DOI: 10.1016/j.yexcr.2012.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 03/01/2012] [Accepted: 03/02/2012] [Indexed: 11/30/2022]
Abstract
The 4.1 proteins are cytoskeletal adaptor proteins that are linked to the control of mechanical stability of certain membranes and to the cellular accumulation and cell surface display of diverse transmembrane proteins. One of the four mammalian 4.1 proteins, 4.1R (80 kDa/120 kDa isoforms), has recently been shown to be required for the normal operation of several ion transporters in the heart (Stagg MA et al. Circ Res, 2008; 103: 855-863). The other three (4.1G, 4.1N and 4.1B) are largely uncharacterised in the heart. Here, we use specific antibodies to characterise their expression, distribution and novel activities in the left ventricle. We detected 4.1R, 4.1G and 4.1N by immunofluorescence and immunoblotting, but not 4.1B. Only one splice variant of 4.1N and 4.1G was seen whereas there are several forms of 4.1R. 4.1N, like 4.1R, was present in intercalated discs, but unlike 4.1R, it was not localised at the lateral plasma membrane. Both 4.1R and 4.1N were in internal structures that, at the level of resolution of the light microscope, were close to the Z-disc (possibly T-tubules). 4.1G was also in intracellular structures, some of which were coincident with sarcoplasmic reticulum. 4.1G existed in an immunoprecipitable complex with spectrin and SERCA2. 80 kDa 4.1R was present in subcellular fractions enriched in intercalated discs, in a complex resistant to solubilization under non-denaturing conditions. At the intercalated disc 4.1R does not colocalise with the adherens junction protein, β-catenin, but does overlap with the other plasma membrane signalling proteins, the Na/K-ATPase and the Na/Ca exchanger NCX1. We conclude that isoforms of 4.1 proteins are differentially compartmentalised in the heart, and that they form specific complexes with proteins central to cardiomyocyte Ca(2+) metabolism.
Collapse
Affiliation(s)
- Jennifer C Pinder
- King's College London, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Nagata M, Sakurai-Yageta M, Yamada D, Goto A, Ito A, Fukuhara H, Kume H, Morikawa T, Fukayama M, Homma Y, Murakami Y. Aberrations of a cell adhesion molecule CADM4 in renal clear cell carcinoma. Int J Cancer 2011; 130:1329-37. [DOI: 10.1002/ijc.26160] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 01/27/2011] [Indexed: 11/07/2022]
|
24
|
Freedman BI, Langefeld CD, Lu L, Divers J, Comeau ME, Kopp JB, Winkler CA, Nelson GW, Johnson RC, Palmer ND, Hicks PJ, Bostrom MA, Cooke JN, McDonough CW, Bowden DW. Differential effects of MYH9 and APOL1 risk variants on FRMD3 Association with Diabetic ESRD in African Americans. PLoS Genet 2011; 7:e1002150. [PMID: 21698141 PMCID: PMC3116917 DOI: 10.1371/journal.pgen.1002150] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 05/07/2011] [Indexed: 12/30/2022] Open
Abstract
Single nucleotide polymorphisms (SNPs) in MYH9 and APOL1 on chromosome 22 (c22) are powerfully associated with non-diabetic end-stage renal disease (ESRD) in African Americans (AAs). Many AAs diagnosed with type 2 diabetic nephropathy (T2DN) have non-diabetic kidney disease, potentially masking detection of DN genes. Therefore, genome-wide association analyses were performed using the Affymetrix SNP Array 6.0 in 966 AA with T2DN and 1,032 non-diabetic, non-nephropathy (NDNN) controls, with and without adjustment for c22 nephropathy risk variants. No associations were seen between FRMD3 SNPs and T2DN before adjusting for c22 variants. However, logistic regression analysis revealed seven FRMD3 SNPs significantly interacting with MYH9-a finding replicated in 640 additional AA T2DN cases and 683 NDNN controls. Contrasting all 1,592 T2DN cases with all 1,671 NDNN controls, FRMD3 SNPs appeared to interact with the MYH9 E1 haplotype (e.g., rs942280 interaction p-value = 9.3E⁻⁷ additive; odds ratio [OR] 0.67). FRMD3 alleles were associated with increased risk of T2DN only in subjects lacking two MYH9 E1 risk haplotypes (rs942280 OR = 1.28), not in MYH9 E1 risk allele homozygotes (rs942280 OR = 0.80; homogeneity p-value = 4.3E⁻⁴). Effects were weaker stratifying on APOL1. FRMD3 SNPS were associated with T2DN, not type 2 diabetes per se, comparing AAs with T2DN to those with diabetes lacking nephropathy. T2DN-associated FRMD3 SNPs were detectable in AAs only after accounting for MYH9, with differential effects for APOL1. These analyses reveal a role for FRMD3 in AA T2DN susceptibility and accounting for c22 nephropathy risk variants can assist in detecting DN susceptibility genes.
Collapse
Affiliation(s)
- Barry I. Freedman
- Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Carl D. Langefeld
- Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Lingyi Lu
- Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Jasmin Divers
- Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Mary E. Comeau
- Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Jeffrey B. Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Cheryl A. Winkler
- Basic Research Laboratory, SAIC-Frederick, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - George W. Nelson
- BSP CCR Genetics Core, SAIC-Frederick, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Randall C. Johnson
- BSP CCR Genetics Core, SAIC-Frederick, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
- Chaire de Bioinformatique, Conservatoire National des Arts et Metiers, Paris, France
| | - Nicholette D. Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Pamela J. Hicks
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Meredith A. Bostrom
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Jessica N. Cooke
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Program in Molecular Medicine and Translational Science, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Caitrin W. McDonough
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Program in Molecular Medicine and Translational Science, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Donald W. Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Section on Endocrinology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| |
Collapse
|
25
|
Meyer AJ, Almendrala DK, Go MM, Krauss SW. Structural protein 4.1R is integrally involved in nuclear envelope protein localization, centrosome-nucleus association and transcriptional signaling. J Cell Sci 2011; 124:1433-44. [PMID: 21486941 DOI: 10.1242/jcs.077883] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The multifunctional structural protein 4.1R is required for assembly and maintenance of functional nuclei but its nuclear roles are unidentified. 4.1R localizes within nuclei, at the nuclear envelope, and in cytoplasm. Here we show that 4.1R, the nuclear envelope protein emerin and the intermediate filament protein lamin A/C co-immunoprecipitate, and that 4.1R-specific depletion in human cells by RNA interference produces nuclear dysmorphology and selective mislocalization of proteins from several nuclear subcompartments. Such 4.1R-deficiency causes emerin to partially redistribute into the cytoplasm, whereas lamin A/C is disorganized at nuclear rims and displaced from nucleoplasmic foci. The nuclear envelope protein MAN1, nuclear pore proteins Tpr and Nup62, and nucleoplasmic proteins NuMA and LAP2α also have aberrant distributions, but lamin B and LAP2β have normal localizations. 4.1R-deficient mouse embryonic fibroblasts show a similar phenotype. We determined the functional effects of 4.1R-deficiency that reflect disruption of the association of 4.1R with emerin and A-type lamin: increased nucleus-centrosome distances, increased β-catenin signaling, and relocalization of β-catenin from the plasma membrane to the nucleus. Furthermore, emerin- and lamin-A/C-null cells have decreased nuclear 4.1R. Our data provide evidence that 4.1R has important functional interactions with emerin and A-type lamin that impact upon nuclear architecture, centrosome-nuclear envelope association and the regulation of β-catenin transcriptional co-activator activity that is dependent on β-catenin nuclear export.
Collapse
Affiliation(s)
- Adam J Meyer
- Department of Genome Dynamics, University of California, Berkeley, CA 94720, USA
| | | | | | | |
Collapse
|
26
|
Comprehensive characterization of expression patterns of protein 4.1 family members in mouse adrenal gland: implications for functions. Histochem Cell Biol 2010; 134:411-20. [PMID: 20890708 DOI: 10.1007/s00418-010-0749-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2010] [Indexed: 01/22/2023]
Abstract
The members of the protein 4.1 family, 4.1R, 4.1G, 4.1N, and 4.1B, are encoded by four genes, all of which undergo complex alternative splicing. It is well established that 4.1R, the prototypical member of the family, serves as an adapter that links the spectrin-actin based cytoskeleton to the plasma membrane in red cells. It is required for mechanical resilience of the membrane, and it ensures the cell surface accumulation of selected membrane proteins. However, the function of 4.1 proteins outside erythrocytes remains under-explored, especially in endocrine tissues. Transcripts of all 4.1 homologs have previously been documented to be abundantly expressed in adrenal gland. In order to begin to decipher the function of 4.1 proteins in adrenal gland, we performed a detailed characterization of the expression pattern of various 4.1 proteins and their cellular localization. We show that 4.1R (~80 and ~135 kDa) splice forms are expressed on the membrane of all cells, while a ~160 kDa 4.1G splice form is distributed in the cytoplasm and the membrane of zona glomerulosa and of medullary cells. Two 4.1N splice forms, ~135 and ~95 kDa, are present in the peri-nuclear region of both zona glomerulosa and medullary cells, while a single ~130 kDa 4.1B splice form, is detected in all layers of adrenal gland in both the cytoplasm and the membrane. The characterization of distinct splice forms of various 4.1 proteins with diverse cellular and sub-cellular localization indicates multiple functions for this family of proteins in endocrine functions of adrenal gland.
Collapse
|
27
|
Baines AJ. The spectrin-ankyrin-4.1-adducin membrane skeleton: adapting eukaryotic cells to the demands of animal life. PROTOPLASMA 2010; 244:99-131. [PMID: 20668894 DOI: 10.1007/s00709-010-0181-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 07/05/2010] [Indexed: 05/29/2023]
Abstract
The cells in animals face unique demands beyond those encountered by their unicellular eukaryotic ancestors. For example, the forces engendered by the movement of animals places stresses on membranes of a different nature than those confronting free-living cells. The integration of cells into tissues, as well as the integration of tissue function into whole animal physiology, requires specialisation of membrane domains and the formation of signalling complexes. With the evolution of mammals, the specialisation of cell types has been taken to an extreme with the advent of the non-nucleated mammalian red blood cell. These and other adaptations to animal life seem to require four proteins--spectrin, ankyrin, 4.1 and adducin--which emerged during eumetazoan evolution. Spectrin, an actin cross-linking protein, was probably the earliest of these, with ankyrin, adducin and 4.1 only appearing as tissues evolved. The interaction of spectrin with ankyrin is probably a prerequisite for the formation of tissues; only with the advent of vertebrates did 4.1 acquires the ability to bind spectrin and actin. The latter activity seems to allow the spectrin complex to regulate the cell surface accumulation of a wide variety of proteins. Functionally, the spectrin-ankyrin-4.1-adducin complex is implicated in the formation of apical and basolateral domains, in aspects of membrane trafficking, in assembly of certain signalling and cell adhesion complexes and in providing stability to otherwise mechanically fragile cell membranes. Defects in this complex are manifest in a variety of hereditary diseases, including deafness, cardiac arrhythmia, spinocerebellar ataxia, as well as hereditary haemolytic anaemias. Some of these proteins also function as tumor suppressors. The spectrin-ankyrin-4.1-adducin complex represents a remarkable system that underpins animal life; it has been adapted to many different functions at different times during animal evolution.
Collapse
Affiliation(s)
- Anthony J Baines
- School of Biosciences and Centre for Biomedical Informatics, University of Kent, Canterbury, CT2 7NJ, UK.
| |
Collapse
|
28
|
|
29
|
Okumura K, Mochizuki E, Yokohama M, Yamakawa H, Shitara H, Mburu P, Yonekawa H, Brown SD, Kikkawa Y. Protein 4.1 expression in the developing hair cells of the mouse inner ear. Brain Res 2010; 1307:53-62. [DOI: 10.1016/j.brainres.2009.10.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Revised: 10/14/2009] [Accepted: 10/14/2009] [Indexed: 01/11/2023]
|
30
|
Pezzolesi MG, Poznik GD, Mychaleckyj JC, Paterson AD, Barati MT, Klein JB, Ng DP, Placha G, Canani LH, Bochenski J, Waggott D, Merchant ML, Krolewski B, Mirea L, Wanic K, Katavetin P, Kure M, Wolkow P, Dunn JS, Smiles A, Walker WH, Boright AP, Bull SB, Doria A, Rogus JJ, Rich SS, Warram JH, Krolewski AS. Genome-wide association scan for diabetic nephropathy susceptibility genes in type 1 diabetes. Diabetes 2009; 58:1403-10. [PMID: 19252134 PMCID: PMC2682673 DOI: 10.2337/db08-1514] [Citation(s) in RCA: 211] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Despite extensive evidence for genetic susceptibility to diabetic nephropathy, the identification of susceptibility genes and their variants has had limited success. To search for genes that contribute to diabetic nephropathy, a genome-wide association scan was implemented on the Genetics of Kidneys in Diabetes collection. RESEARCH DESIGN AND METHODS We genotyped approximately 360,000 single nucleotide polymorphisms (SNPs) in 820 case subjects (284 with proteinuria and 536 with end-stage renal disease) and 885 control subjects with type 1 diabetes. Confirmation of implicated SNPs was sought in 1,304 participants of the Diabetes Control and Complications Trial (DCCT)/Epidemiology of Diabetes Interventions and Complications (EDIC) study, a long-term, prospective investigation of the development of diabetes-associated complications. RESULTS A total of 13 SNPs located in four genomic loci were associated with diabetic nephropathy with P < 1 x 10(-5). The strongest association was at the FRMD3 (4.1 protein ezrin, radixin, moesin [FERM] domain containing 3) locus (odds ratio [OR] = 1.45, P = 5.0 x 10(-7)). A strong association was also identified at the CARS (cysteinyl-tRNA synthetase) locus (OR = 1.36, P = 3.1 x 10(-6)). Associations between both loci and time to onset of diabetic nephropathy were supported in the DCCT/EDIC study (hazard ratio [HR] = 1.33, P = 0.02, and HR = 1.32, P = 0.01, respectively). We demonstratedexpression of both FRMD3 and CARS in human kidney. CONCLUSIONS We identified genetic associations for susceptibility to diabetic nephropathy at two novel candidate loci near the FRMD3 and CARS genes. Their identification implicates previously unsuspected pathways in the pathogenesis of this important late complication of type 1 diabetes.
Collapse
Affiliation(s)
- Marcus G. Pezzolesi
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - G. David Poznik
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Josyf C. Mychaleckyj
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Andrew D. Paterson
- Program in Genetics and Genome Biology, Hospital for Sick Children, University of Toronto, Toronto, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | | | - Jon B. Klein
- Kidney Disease Program, University of Louisville, Louisville, Kentucky
| | - Daniel P.K. Ng
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Community, Occupational and Family Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Grzegorz Placha
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Hypertension, Medical University of Warsaw, Warsaw, Poland
| | - Luis H. Canani
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Endocrinology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Jacek Bochenski
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Daryl Waggott
- Samuel Lunenfeld Research Institute of Mount Sinai Hospital, Prosserman Centre for Health Research, Toronto, Canada
| | | | - Bozena Krolewski
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Lucia Mirea
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Samuel Lunenfeld Research Institute of Mount Sinai Hospital, Prosserman Centre for Health Research, Toronto, Canada
| | - Krzysztof Wanic
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Pisut Katavetin
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Masahiko Kure
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Pawel Wolkow
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Pharmacology, Jagiellonian University, School of Medicine, Krakow, Poland
| | - Jonathon S. Dunn
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Adam Smiles
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - William H. Walker
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Andrew P. Boright
- Department of Medicine, University Health Network, University of Toronto, Toronto, Canada
| | - Shelley B. Bull
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Samuel Lunenfeld Research Institute of Mount Sinai Hospital, Prosserman Centre for Health Research, Toronto, Canada
| | | | - Alessandro Doria
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - John J. Rogus
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - James H. Warram
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Andrzej S. Krolewski
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Corresponding author: Andrzej S. Krolewski,
| |
Collapse
|
31
|
Protein 4.1R links E-cadherin/beta-catenin complex to the cytoskeleton through its direct interaction with beta-catenin and modulates adherens junction integrity. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1788:1458-65. [PMID: 19376086 DOI: 10.1016/j.bbamem.2009.03.022] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Revised: 03/20/2009] [Accepted: 03/23/2009] [Indexed: 11/21/2022]
Abstract
Protein 4.1R (4.1R) is the prototypical member of the protein 4.1 superfamily comprising of the protein 4.1 family (4.1R, 4.1B, 4.1G and 4.1N) and ERM family (ezrin, radixin and meosin). These proteins in general serve as adaptors between the membrane and the cytoskeleton. Here we show that 4.1R expressed in the gastric epithelial cells associates with adherens junction protein beta-catenin. Biochemical examination of 4.1R-deficient stomach epithelia revealed a selective reduction of beta-catenin which is accompanied by a weaker linkage of E-cadherin to the cytoskeleton. In addition, organization of actin cytoskeleton was altered in 4.1R-deficient cells. Moreover, histological examination revealed that cell-cell contacts are impaired and gastric glands are disorganized in 4.1R null stomach epithelia. These results demonstrate an important and previously unidentified role of 4.1R in linking the cadherin/catenin complex to the cytoskeleton through its direct interaction with beta-catenin and in regulating the integrity of adherens junction.
Collapse
|
32
|
Ohno N, Terada N, Komada M, Saitoh S, Costantini F, Pace V, Germann PG, Weber K, Yamakawa H, Ohara O, Ohno S. Dispensable role of protein 4.1B/DAL-1 in rodent adrenal medulla regarding generation of pheochromocytoma and plasmalemmal localization of TSLC1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:506-15. [PMID: 19321127 DOI: 10.1016/j.bbamcr.2009.01.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2008] [Revised: 12/06/2008] [Accepted: 01/06/2009] [Indexed: 02/07/2023]
Abstract
Protein 4.1B is a membrane skeletal protein expressed in various organs, and is associated with tumor suppressor in lung cancer-1 (TSLC1) in vitro. Although involvement of 4.1B in the intercellular junctions and tumor-suppression was suggested, some controversial results posed questions to the general tumor-suppressive function of 4.1B and its relation to TSLC1 in vivo. In this study, the expression of 4.1B and its interaction with TSLC1 were examined in rodent adrenal gland, and the involvement of 4.1B in tumorigenesis and the effect of 4.1B deficiency on TSLC1 distribution were also investigated using rodent pheochromocytoma and 4.1B-knockout mice. Although plasmalemmal immunolocalization of 4.1B was shown in chromaffin cells of rodent adrenal medulla, expression of 4.1B was maintained in developed pheochromocytoma, and morphological abnormality or pheochromocytoma generation could not be found in 4.1B-deficient mice. Furthermore, molecular interaction and colocalization of 4.1B and TSLC1 were observed in mouse adrenal gland, but the immunolocalization of TSLC1 along chromaffin cell membranes was not affected in the 4.1B-deficient mice. These results suggest that the function of 4.1B as tumor suppressor might significantly differ among organs and species, and that plasmalemmal retention of TSLC1 would be maintained by molecules other than 4.1B interacting in rodent chromaffin cells.
Collapse
Affiliation(s)
- Nobuhiko Ohno
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi 409-3898, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Bernkopf DB, Williams ED. Potential role of EPB41L3 (protein 4.1B/Dal-1) as a target for treatment of advanced prostate cancer. Expert Opin Ther Targets 2008; 12:845-53. [PMID: 18554153 DOI: 10.1517/14728222.12.7.845] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Loss of erythrocyte membrane protein band 4.1-like 3 (EPB41L3; aliases: protein 4.1B, differentially expressed in adenocarcinoma of the lung-1 (Dal-1)) expression has been implicated in tumor progression. OBJECTIVE To evaluate literature describing the role of EPB41L3 in tumorigenesis and metastasis, and to consider whether targeting this gene would be useful in the treatment of prostate cancer. METHODS A literature review of studies describing EPB41L3 and its aliases was conducted. Online databases (NCBI, SwissProt) were also interrogated to collect further data. RESULTS/CONCLUSION A growing body of evidence supports a role for loss of EPB41L3 in tumor progression, including in prostate cancer. Therapeutic strategies that could be harnessed to upregulate EPB41L3 gene expression in prostate cancer cells are currently being developed.
Collapse
Affiliation(s)
- Dominic B Bernkopf
- Monash University, Centre for Cancer Research, Monash Institute of Medical Research, Monash Medical Centre, 246 Clayton Road, Clayton, Victoria, 3168, Australia
| | | |
Collapse
|
34
|
Krauss SW, Spence JR, Bahmanyar S, Barth AIM, Go MM, Czerwinski D, Meyer AJ. Downregulation of protein 4.1R, a mature centriole protein, disrupts centrosomes, alters cell cycle progression, and perturbs mitotic spindles and anaphase. Mol Cell Biol 2008; 28:2283-94. [PMID: 18212055 PMCID: PMC2268423 DOI: 10.1128/mcb.02021-07] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2007] [Accepted: 01/08/2008] [Indexed: 01/11/2023] Open
Abstract
Centrosomes nucleate and organize interphase microtubules and are instrumental in mitotic bipolar spindle assembly, ensuring orderly cell cycle progression with accurate chromosome segregation. We report that the multifunctional structural protein 4.1R localizes at centrosomes to distal/subdistal regions of mature centrioles in a cell cycle-dependent pattern. Significantly, 4.1R-specific depletion mediated by RNA interference perturbs subdistal appendage proteins ninein and outer dense fiber 2/cenexin at mature centrosomes and concomitantly reduces interphase microtubule anchoring and organization. 4.1R depletion causes G(1) accumulation in p53-proficient cells, similar to depletion of many other proteins that compromise centrosome integrity. In p53-deficient cells, 4.1R depletion delays S phase, but aberrant ninein distribution is not dependent on the S-phase delay. In 4.1R-depleted mitotic cells, efficient centrosome separation is reduced, resulting in monopolar spindle formation. Multipolar spindles and bipolar spindles with misaligned chromatin are also induced by 4.1R depletion. Notably, all types of defective spindles have mislocalized NuMA (nuclear mitotic apparatus protein), a 4.1R binding partner essential for spindle pole focusing. These disruptions contribute to lagging chromosomes and aberrant microtubule bridges during anaphase/telophase. Our data provide functional evidence that 4.1R makes crucial contributions to the structural integrity of centrosomes and mitotic spindles which normally enable mitosis and anaphase to proceed with the coordinated precision required to avoid pathological events.
Collapse
Affiliation(s)
- Sharon Wald Krauss
- Department of Cell Biology and Imaging, University of California-LBNL, 1 Cyclotron Road, MS 74-157, Berkeley, CA 94720, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Terada N, Ohno N, Saitoh S, Seki G, Komada M, Suzuki T, Yamakawa H, Soleimani M, Ohno S. Interaction of Membrane Skeletal Protein, Protein 4.1B and p55, and Sodium Bicarbonate Cotransporter1 in Mouse Renal S1-S2 Proximal Tubules. J Histochem Cytochem 2007; 55:1199-206. [PMID: 17712176 DOI: 10.1369/jhc.7a7266.2007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Our recent studies demonstrated the localization of protein 4.1B, a member of the 4.1 skeletal membrane proteins, to the basolateral membranes of the S1-S2 renal proximal tubules. In the present studies, we investigated the presence of binding partners that could form a molecular complex with the 4.1B protein. Immunohistochemistry revealed the localization of p55, a membrane-associated guanylate kinase, and the sodium bicarbonate cotransporter1 (NBC1), to the basolateral membrane domain of S1-S2 in mouse renal proximal tubules. Using immunoprecipitation of kidney lysates with anti-p55 antibody, a positive band was blotted with anti-4.1B antibody. GST fusion proteins including the NBC1 and 4.1B regions were confirmed to bind with each other by electrophoresis after mixing. Both NBC1- and 4.1B-specific bands were detected in renal protein mixtures immunoprecipated by either anti-4.1B- or NBC1-specific antibodies. It is likely that NBC1, 4.1B, and p55 form a molecular complex in the basolateral membrane of the kidney S1-S2 proximal tubules. We propose that the 4.1B-containing membrane skeleton may play a role in regulating the Na+ and HCO3- reabsorption in S1-S2 proximal tubules.
Collapse
Affiliation(s)
- Nobuo Terada
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo City, Yamanashi 409-3898, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ohno N, Terada N, Yamakawa H, Komada M, Ohara O, Trapp BD, Ohno S. Expression of protein 4.1G in Schwann cells of the peripheral nervous system. J Neurosci Res 2006; 84:568-77. [PMID: 16752423 DOI: 10.1002/jnr.20949] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The membrane-associated cytoskeletal proteins, including protein 4.1 family, play important roles in membrane integrity, protein targeting, and signal transduction. Although protein 4.1G (4.1G) is expressed ubiquitously in mammalian tissues, it can have very discrete distributions within cells. The present study investigated the expression and distributions of 4.1G in rodent sciatic nerve. Northern and Western blot analysis detected abundant 4.1G mRNA and protein in rat sciatic nerve extracts. Immunohistochemical staining with a 4.1G-specific antibody and double immunolabeling with E-cadherin, betaIV spectrin, and connexin 32 detected 4.1G in paranodal loops, Schmidt-Lanterman incisures, and periaxonal, mesaxonal, and abaxonal membranes of rodent sciatic nerve. Immunoelectron microscopy confirmed the immunodistribution of 4.1G in Schwann cells. In developing mouse sciatic nerves, 4.1G was diffusely distributed in immature Schwann cells and gradually localized at paranodes, incisures, and periaxonal and mesaxonal membranes during their maturation. These data support the concept that 4.1G plays an important role in the membrane expansion and specialization that occurs during formation and maintenance of myelin internodes in the peripheral nervous system.
Collapse
Affiliation(s)
- Nobuhiko Ohno
- Department of Anatomy, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo-City, Yamanashi, Japan
| | | | | | | | | | | | | |
Collapse
|
37
|
Kramer-Zucker AG, Wiessner S, Jensen AM, Drummond IA. Organization of the pronephric filtration apparatus in zebrafish requires Nephrin, Podocin and the FERM domain protein Mosaic eyes. Dev Biol 2006; 285:316-29. [PMID: 16102746 PMCID: PMC2836015 DOI: 10.1016/j.ydbio.2005.06.038] [Citation(s) in RCA: 180] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2005] [Revised: 05/24/2005] [Accepted: 06/17/2005] [Indexed: 02/07/2023]
Abstract
Podocytes are specialized cells of the kidney that form the blood filtration barrier in the kidney glomerulus. The barrier function of podocytes depends upon the development of specialized cell-cell adhesion complexes called slit-diaphragms that form between podocyte foot processes surrounding glomerular blood vessels. Failure of the slit-diaphragm to form results in leakage of high molecular weight proteins into the blood filtrate and urine, a condition called proteinuria. In this work, we test whether the zebrafish pronephros can be used as an assay system for the development of glomerular function with the goal of identifying novel components of the slit-diaphragm. We first characterized the function of the zebrafish homolog of Nephrin, the disease gene associated with the congenital nephritic syndrome of the Finnish type, and Podocin, the gene mutated in autosomal recessive steroid-resistant nephrotic syndrome. Zebrafish nephrin and podocin were specifically expressed in pronephric podocytes and required for the development of pronephric podocyte cell structure. Ultrastructurally, disruption of nephrin or podocin expression resulted in a loss of slit-diaphragms at 72 and 96 h post-fertilization and failure to form normal podocyte foot processes. We also find that expression of the band 4.1/FERM domain gene mosaic eyes in podocytes is required for proper formation of slit-diaphragm cell-cell junctions. A functional assay of glomerular filtration barrier revealed that absence of normal nephrin, podocin or mosaic eyes expression results in loss of glomerular filtration discrimination and aberrant passage of high molecular weight substances into the glomerular filtrate.
Collapse
Affiliation(s)
| | - Stephanie Wiessner
- Renal Unit, Massachusetts General Hospital 149-8000, 149 13th Street, Charlestown, MA 02129, USA
| | - Abbie M. Jensen
- Biology Department, 221 Morrill South, University of Massachusetts, Amherst, MA 01003, USA
| | - Iain A. Drummond
- Renal Unit, Massachusetts General Hospital 149-8000, 149 13th Street, Charlestown, MA 02129, USA
- Corresponding author. Fax: +1 617 726 5669. (I.A. Drummond)
| |
Collapse
|
38
|
Ponthier JL, Schluepen C, Chen W, Lersch RA, Gee SL, Hou VC, Lo AJ, Short SA, Chasis JA, Winkelmann JC, Conboy JG. Fox-2 splicing factor binds to a conserved intron motif to promote inclusion of protein 4.1R alternative exon 16. J Biol Chem 2006; 281:12468-74. [PMID: 16537540 DOI: 10.1074/jbc.m511556200] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of protein 4.1R exon 16 (E16) inclusion during erythropoiesis represents a physiologically important splicing switch that increases 4.1R affinity for spectrin and actin. Previous studies showed that negative regulation of E16 splicing is mediated by the binding of heterogeneous nuclear ribonucleoprotein (hnRNP) A/B proteins to silencer elements in the exon and that down-regulation of hnRNP A/B proteins in erythroblasts leads to activation of E16 inclusion. This article demonstrates that positive regulation of E16 splicing can be mediated by Fox-2 or Fox-1, two closely related splicing factors that possess identical RNA recognition motifs. SELEX experiments with human Fox-1 revealed highly selective binding to the hexamer UGCAUG. Both Fox-1 and Fox-2 were able to bind the conserved UGCAUG elements in the proximal intron downstream of E16, and both could activate E16 splicing in HeLa cell co-transfection assays in a UGCAUG-dependent manner. Conversely, knockdown of Fox-2 expression, achieved with two different siRNA sequences resulted in decreased E16 splicing. Moreover, immunoblot experiments demonstrate mouse erythroblasts express Fox-2. These findings suggest that Fox-2 is a physiological activator of E16 splicing in differentiating erythroid cells in vivo. Recent experiments show that UGCAUG is present in the proximal intron sequence of many tissue-specific alternative exons, and we propose that the Fox family of splicing enhancers plays an important role in alternative splicing switches during differentiation in metazoan organisms.
Collapse
Affiliation(s)
- Julie L Ponthier
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Terada N, Ohno N, Yamakawa H, Ohara O, Ohno S. Topographical significance of membrane skeletal component protein 4.1 B in mammalian organs. Anat Sci Int 2005; 80:61-70. [PMID: 15960311 DOI: 10.1111/j.1447-073x.2005.00094.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The polarized architecture of epithelial cells is a fundamental determinant of cell structures and functions. Both formation and orientation of proper epithelial polarity are needed for cell-cell or cell-matrix adhesion, signal transduction and cytoskeletal interactions of multimolecular complexes at apical, lateral and basal cell membranes. These cell membrane domains are usually segregated by some junctional complexes. Recent molecular genetic studies on the anchor structure between myelin sheaths and axons have indicated the specific molecular organization for polarization of axolemma and the myelin sheaths at paranodes, termed 'septate-like junctions'. It was also speculated that other mammalian organs may use a similar junctional system. The protein 4.1 B was originally found to be localized in paranodes and juxtaparanodes of myelinated nerve fibers. Our recent immunohistochemical studies on protein 4.1B have indicated its significance for the cell-cell and/or cell-matrix adhesion in various rodent organs. The protein 4.1 family of proteins have been supposed to possess variable molecular domains relating to cell adhesion, ion balance, receptor responses and signal transduction. Therefore, more precise studies on the molecular structure and the functional domains of protein 4.1B, as well as on its changes under physiological and pathological conditions, may provide a clue for organogenesis in various mammalian organs.
Collapse
Affiliation(s)
- Nobuo Terada
- Department of Anatomy, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Tamaho, Japan.
| | | | | | | | | |
Collapse
|
40
|
Terada N, Ohno N, Yamakawa H, Baba T, Fujii Y, Ohara O, Ohno S. Immunolocalization of protein 4.1B in the rat digestive system. J Mol Histol 2005; 35:347-53. [PMID: 15503808 DOI: 10.1023/b:hijo.0000039848.86488.74] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Protein 4.1 family proteins are thought to interact with membrane proteins and also membrane skeletons. In this study, immunohistochemical studies by light and electron microscopy were performed with a specific antibody against protein 4.1B. Specific protein 4.1B immunolabeling was observed in simple columnar epithelium in the adult rat large intestine, small intestine and stomach. Protein 4.1B immunolabeling was localized along the membranes facing the adjacent cells (lateral portion) and also facing the extracellular matrix (basal portion). Moreover, a spatial protein 4.1B expression gradient was observed along the crypt-villus axis of the rat small and large intestinal epithelium: strong protein 4.1B expression was present within the villus, with the crypt showing barely any detectable protein 4.1B. The expression of protein 4.1B was not detected in the stratified squamous epithelium in the forestomach or the esophagus. By immunoelectron microscopy, the immunolabeling of the cells was observed to be restricted to the cytoplasmic side just beneath the plasma membrane, including the membranes adjacent to the next cells, except for the tight junctions. We conclude that the protein 4.1B expression pattern is related to the maturation of simple columnar epithelium in the rat digestive system, probably by the effect of adhesion.
Collapse
Affiliation(s)
- Nobuo Terada
- Department of Anatomy, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Tamaho, Japan.
| | | | | | | | | | | | | |
Collapse
|
41
|
Taylor-Harris PM, Keating LA, Maggs AM, Phillips GW, Birks EJ, Franklin RCG, Yacoub MH, Baines AJ, Pinder JC. Cardiac muscle cell cytoskeletal protein 4.1: Analysis of transcripts and subcellular location?relevance to membrane integrity, microstructure, and possible role in heart failure. Mamm Genome 2005; 16:137-51. [PMID: 15834631 DOI: 10.1007/s00335-004-2436-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2004] [Accepted: 10/26/2004] [Indexed: 11/27/2022]
Abstract
The spectrin-based cytoskeleton assembly has emerged as a major player in heart functioning; however, cardiac protein 4.1, a key constituent, is uncharacterized. Protein 4.1 evolved to protect cell membranes against mechanical stresses and to organize membrane microstructure. 4.1 Proteins are multifunctional and, among other activities, link integral/signaling proteins on the plasma and internal membranes with the spectrin-based cytoskeleton. Four genes, EPB41, EPB41L1, EPB41L2, and EPB41L3 encode proteins 4.1R, 4.1N, 4.1G, and 4.1B, respectively. All are extensively spliced. Different isoforms are expressed according to tissue and developmental state, individual function being controlled through inclusion/exclusion of interactive domains. We have defined mouse and human cardiac 4.1 transcripts; other than 4. 1B in humans, all genes show activity. Cardiac transcripts constitutively include conserved FERM and C-terminal domains; both interact with membrane-bound signaling/transport/cell adhesion molecules. Variable splicing within and adjacent to the central spectrin/actin-binding domain enables regulation of cytoskeleton-binding activity. A novel heart-specific exon occurs in human 4.1G, but not in mouse. Immunofluorescence reveals 4.1 staining within mouse cardiomyocytes; thus, both at the plasma membrane and, interdigitated with sarcomeric myosin, across myofibrils in regions close to the sarcoplasmic reticulum. These are all regions to which spectrin locates. 4.1R in human heart shows similar distribution; however, there is limited plasma membrane staining. We conclude that cardiac 4.1s are highly regulated in their ability to crosslink plasma/integral cell membranes with the spectrin-actin cytoskeleton. We speculate that over the repetitive cycles of heart muscle contraction and relaxation, 4.1s are likely to locate, support, and coordinate functioning of key membrane-bound macromolecular assemblies.
Collapse
Affiliation(s)
- Pamela M Taylor-Harris
- Randall Division of Cell and Molecular Biophysics, King's College London, Guy's Campus, London SE1 1UL, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Gascard P, Parra MK, Zhao Z, Calinisan VR, Nunomura W, Rivkees SA, Mohandas N, Conboy JG. Putative tumor suppressor protein 4.1B is differentially expressed in kidney and brain via alternative promoters and 5' alternative splicing. ACTA ACUST UNITED AC 2004; 1680:71-82. [PMID: 15488987 DOI: 10.1016/j.bbaexp.2004.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2004] [Revised: 08/03/2004] [Accepted: 08/10/2004] [Indexed: 10/26/2022]
Abstract
Protein 4.1B has been reported as a tumor suppressor in brain, but not in kidney, despite high expression in both tissues. Here we demonstrate that N-terminal variability in kidney and brain 4.1B isoforms arises through an unusual coupling of RNA processing events in the 5' region of the gene. We describe two transcriptional promoters at far upstream alternative exons 1A and 1B, and show that their respective transcripts splice differentially to exon 2'/2 in a manner that determines mRNA coding capacity. The consequence of this unique processing is that exon 1B transcripts initiate translation at AUG1 (in exon 2') and encode larger 4.1B isoforms with an N-terminal extension; exon 1A transcripts initiate translation at AUG2 (in exon 4) and encode smaller 4.1B isoforms. Tissue-specific differences in promoter utilization may thus explain the abundance of larger 4.1B isoforms in brain but not in kidney. In cell studies, differentiation of PC12 cells was accompanied by translocation of large protein 4.1B isoforms into the nucleus. We propose that first exon specification is coupled to downstream splicing events, generating 4.1B isoforms with diverse roles in kidney and brain physiology, and potentially unique functions in cell proliferation and tumor suppression.
Collapse
Affiliation(s)
- Philippe Gascard
- Life Sciences Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Mail stop 74-157, Berkeley CA 94720, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Huang SC, Liu ES, Chan SH, Munagala ID, Cho HT, Jagadeeswaran R, Benz EJ. Mitotic regulation of protein 4.1R involves phosphorylation by cdc2 kinase. Mol Biol Cell 2004; 16:117-27. [PMID: 15525677 PMCID: PMC539157 DOI: 10.1091/mbc.e04-05-0426] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The nonerythrocyte isoform of the cytoskeletal protein 4.1R (4.1R) is associated with morphologically dynamic structures during cell division and has been implicated in mitotic spindle function. In this study, we define important 4.1R isoforms expressed in interphase and mitotic cells by RT-PCR and mini-cDNA library construction. Moreover, we show that 4.1R is phosphorylated by p34cdc2 kinase on residues Thr60 and Ser679 in a mitosis-specific manner. Phosphorylated 4.1R135 isoform(s) associate with tubulin and Nuclear Mitotic Apparatus protein (NuMA) in intact HeLa cells in vivo as well as with the microtubule-associated proteins in mitotic asters assembled in vitro. Recombinant 4.1R135 is readily phosphorylated in mitotic extracts and reconstitutes mitotic aster assemblies in 4.1R-immunodepleted extracts in vitro. Furthermore, phosphorylation of these residues appears to be essential for the targeting of 4.1R to the spindle poles and for mitotic microtubule aster assembly in vitro. Phosphorylation of 4.1R also enhances its association with NuMA and tubulin. Finally, we used siRNA inhibition to deplete 4.1R from HeLa cells and provide the first direct genetic evidence that 4.1R is required to efficiently focus mitotic spindle poles. Thus, we suggest that 4.1R is a member of the suite of direct cdc2 substrates that are required for the establishment of a bipolar spindle.
Collapse
Affiliation(s)
- Shu-Ching Huang
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Ohno N, Terada N, Murata SI, Yamakawa H, Newsham IF, Katoh R, Ohara O, Ohno S. Immunolocalization of protein 4.1B/DAL-1 during neoplastic transformation of mouse and human intestinal epithelium. Histochem Cell Biol 2004; 122:579-86. [PMID: 15517334 DOI: 10.1007/s00418-004-0716-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2004] [Indexed: 01/27/2023]
Abstract
Recently, we have reported that the protein 4.1B immunolocalization occurred only in matured columnar epithelial cells of normal rat intestines. This finding suggested that protein 4.1B expression could be examined for a possible change during neoplastic transformation of the intestinal mucosa. In the present study, we first present the distribution of mouse protein 4.1B in normal intestinal epithelial cells and tumor cells using the adenomatous polyposis coli (Apc) mutant mouse model. A low level of protein 4.1B expression coincided with the phenotypic transition to carcinoma. To examine the protein 4.1B expression in human intestinal mucosa, we used another antibody against an isoform of the human protein 4.1B, DAL-1 (differentially expressed adenocarcinoma of the lung). Human DAL-1 was also expressed in matured epithelial cells in human colons, with a definite expression gradient along the crypt axis. In human colorectal cancer cells, however, DAL-1 expression was not detected. These results suggest that mouse protein 4.1B and human DAL-1 might have a striking analogy of functions, which may be integrally involved in epithelial proliferation. We propose that loss of protein 4.1B/DAL-1 expression might be a marker of intestinal tumors, indicative of a tumor suppressor function in the intestinal mucosa.
Collapse
Affiliation(s)
- Nobuhiko Ohno
- Department of Anatomy, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Tamaho, 409-3898 Yamanashi, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Parra M, Gee S, Chan N, Ryaboy D, Dubchak I, Mohandas N, Gascard PD, Conboy JG. Differential domain evolution and complex RNA processing in a family of paralogous EPB41 (protein 4.1) genes facilitate expression of diverse tissue-specific isoforms. Genomics 2004; 84:637-46. [PMID: 15475241 DOI: 10.1016/j.ygeno.2004.06.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2004] [Accepted: 06/08/2004] [Indexed: 11/30/2022]
Abstract
The EPB41 (protein 4.1) genes epitomize the resourcefulness of the mammalian genome to encode a complex proteome from a small number of genes. By utilizing alternative transcriptional promoters and tissue-specific alternative pre-mRNA splicing, EPB41, EPB41L2, EPB41L3, and EPB41L1 encode a diverse array of structural adapter proteins. Comparative genomic and transcript analysis of these 140- to 240-kb genes indicates several unusual features: differential evolution of highly conserved exons encoding known functional domains interspersed with unique exons whose size and sequence variations contribute substantially to intergenic diversity; alternative first exons, most of which map far upstream of the coding regions; and complex tissue-specific alternative pre-mRNA splicing that facilitates synthesis of functionally different complements of 4.1 proteins in various cells. Understanding the splicing regulatory networks that control protein 4.1 expression will be critical to a full appreciation of the many roles of 4.1 proteins in normal cell biology and their proposed roles in human cancer.
Collapse
Affiliation(s)
- Marilyn Parra
- Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Huang SC, Jagadeeswaran R, Liu ES, Benz EJ. Protein 4.1R, a Microtubule-associated Protein Involved in Microtubule Aster Assembly in Mammalian Mitotic Extract. J Biol Chem 2004; 279:34595-602. [PMID: 15184364 DOI: 10.1074/jbc.m404051200] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Non-erythroid protein 4.1R (4.1R) consists of a complex family of isoforms. We have shown that 4.1R isoforms localize at the mitotic spindle/spindle poles and associate in a complex with the mitotic-spindle organization proteins Nuclear Mitotic Apparatus protein (NuMA), dynein, and dynactin. We addressed the mitotic function of 4.1R by investigating its association with microtubules, the main component of the mitotic spindles, and its role in mitotic aster assembly in vitro. 4.1R appears to partially co-localize with microtubules throughout the mitotic stages of the cell cycle. In vitro sedimentation assays showed that 4.1R isoforms directly interact with microtubules. Glutathione S-transferase (GST) pull-down assays using GST-4.1R fusions and mitotic cell extracts further showed that the association of 4.1R with tubulin results from both the membrane-binding domain and C-terminal domain of 4.1R. Moreover, 4.1R, but not actin, is a mitotic microtubule-associated protein; 4.1R associates with microtubules in the microtubule pellet of the mitotic asters assembled in mammalian cell-free mitotic extract. The organization of microtubules into asters depends on 4.1R in that immunodepletion of 4.1R from the extract resulted in randomly dispersed microtubules. Furthermore, adding a 135-kDa recombinant 4.1R reconstituted the mitotic asters. Finally, we demonstrated that a mitotic 4.1R isoform appears to form a complex in vivo with tubulin and NuMA in highly synchronized mitotic HeLa extracts. Our results suggest that a 135-kDa non-erythroid 4.1R is important to cell division, because it participates in the formation of mitotic spindles and spindle poles through its interaction with mitotic microtubules.
Collapse
Affiliation(s)
- Shu-Ching Huang
- Department of Medical Oncology, Dana Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
47
|
Terada N, Ohno N, Yamakawa H, Baba T, Fujii Y, Zea Z, Ohara O, Ohno S. Immunohistochemical study of protein 4.1B in the normal and W/W(v) mouse seminiferous epithelium. J Histochem Cytochem 2004; 52:769-77. [PMID: 15150285 DOI: 10.1369/jhc.3a6192.2004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cell-cell adhesion is crucial not only for mechanical adhesion but also for tissue morphogenesis. Protein 4.1B, a member of the protein 4.1 family named from an erythrocyte membrane protein, is a potential organizer of an adherens system. In adult mouse seminiferous tubules, protein 4.1B localized in the basal compartment, especially in the attaching region of spermatogonia and Sertoli cells. Protein 4.1B localization and appearance were not different in each spermatogenic stage. Developmentally, protein 4.1B was not detected at postnatal day 3 (P3), was diffusely localized at P15, and was found in the basal compartment during the third week. By double staining for protein 4.1B and F-actin, their localizations were shown to be different, indicating that protein 4.1B was localized in a region lower than the basal ectoplasmic specialization that formed the Sertoli-Sertoli junction. By electron microscopy, immunoreactive products were seen mainly on the membranes of Sertoli cells. In the W/W(v) mutant mouse, the seminiferous epithelium had few germ cells. Protein 4.1B and beta-catenin were not detected, although the basal ectoplasmic specialization was retained. These results indicate that protein 4.1B may be related to the adhesion between Sertoli cells and germ cells, especially the spermatogonium.
Collapse
Affiliation(s)
- Nobuo Terada
- Department of Anatomy, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Tamaho, Japan.
| | | | | | | | | | | | | | | |
Collapse
|