1
|
Pilling D, Consalvo KM, Kirolos SA, Gomer RH. Differences Between Unstimulated and Stimulated Human Male and Female Neutrophils in Protein and Phosphoprotein Profiles. Proteomics 2025; 25:e202400232. [PMID: 39937132 DOI: 10.1002/pmic.202400232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025]
Abstract
Human males and females show differences in the incidence of neutrophil-associated diseases and differences in neutrophil responses such as a faster response to the chemorepellent Ser-Leu-Ile-Gly-Lys-Val-NH2 (SLIGKV) in males. Little is known about the basis of sex-based differences in human neutrophils. We used mass spectrometry to identify proteins and phosphoproteins in unstimulated human neutrophils and in neutrophils incubated with the SLIGKV, a protease-activated receptor 2 agonist. There were 132 proteins with higher levels in unstimulated male neutrophils; these proteins tended to be associated with RNA regulation, ribosome, and phosphoinositide signaling pathways, whereas 30 proteins with higher levels in unstimulated female neutrophils were associated with metabolic processes, proteasomes, and phosphatase regulatory proteins. Unstimulated male neutrophils had increased phosphorylation of 32 proteins compared to females. After exposure to SLIGKV, male neutrophils showed a faster response in terms of protein phosphorylation compared to female neutrophils. Male neutrophils have higher levels of proteins and higher phosphorylation of proteins associated with RNA processing and signaling pathways. Female neutrophils have higher levels of proteins associated with metabolism and proteolytic pathways. This suggests that male neutrophils might be more ready to adapt to a new environment, and female neutrophils might be more effective at responding to pathogens.
Collapse
Affiliation(s)
- Darrell Pilling
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Kristen M Consalvo
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Sara A Kirolos
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Richard H Gomer
- Department of Biology, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
2
|
Romanet P, Coppin L, Molin A, Santucci N, Le Bras M, Odou MF. Chapter 5: The roles of genetics in primary hyperparathyroidism. ANNALES D'ENDOCRINOLOGIE 2025; 86:101694. [PMID: 39818301 DOI: 10.1016/j.ando.2025.101694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Around 10% of cases of primary hyperparathyroidism are thought to be genetic in origin, some of which are part of a syndromic form such as multiple endocrine neoplasia types 1, 2A or 4 or hyperparathyroidism-jaw tumor syndrome, while the remainder are cases of isolated familial primary hyperparathyroidism. Recognition of these genetic forms is important to ensure appropriate management according to the gene and type of variant involved, but screening for a genetic cause is not justified in all patients presenting primary hyperparathyroidism. The indications for genetic analysis have made it possible to propose a decision tree that takes into account whether the presentation is familial or sporadic, syndromic or isolated, patient age, and histopathological type of parathyroid lesion. Thus, the first consensus recommendation is to propose genetic screening to any patient with a familial form of primary hyperparathyroidism (≥2 1st or 2nd degree relatives) or in syndromic presentation or a sporadic isolated presentation if the patient is under 50 years of age, or over 50 with a recurrent or multi-glandular form, carcinoma, atypical parathyroid tumor and/or loss of parafibromin expression. The panel of genes currently recommended for first-line treatment comprises MEN1, CDKN1B, CDC73, CASR, GNA11, AP2S1 and GCM2. Other genes may also be involved in familial primary hyperparathyroidism, but in a much more rarely and less consistently. The second recommendation is to propose genetic screening, up to and including whole-genome sequencing in the event of inconclusive panel analysis, to patients with proven familial primary hyperparathyroidism and/or pediatric onset. The role of the genetic practitioner is to interpret the sequencing data by categorizing the variants into 5 classes of pathogenicity. The aim of genetic analysis is to identify the genetic variant involved in the patient's phenotype, in order to make or refute a diagnosis of hereditary primary hyperparathyroidism, and to adapt management and monitoring. Appropriate genetic counseling should then be provided for patient and family.
Collapse
Affiliation(s)
- Pauline Romanet
- Inserm, MMG, Laboratory of Molecular Biology GEnOPé, BIOGENOPOLE, La Timone University Hospital, Aix-Marseille University, AP-HM, Marseille, France.
| | - Lucie Coppin
- Inserm, CNRS, UMR9020-U1277 - CANTHER - Cancer - Heterogeneity Plasticity and Resistance to Therapies, University of Lille, CHU of Lille, Lille, France
| | - Arnaud Molin
- UNICAEN, RU7450 BioTARGen, Department of Genetics, Reference Center for Developmental Disorders and Malformative Syndromes, Anddi-Rares Network, Caen University Hospital, University of Normandy, Caen, France
| | - Nicolas Santucci
- Department of Digestive, Oncological and Endocrine Surgery, Dijon University Hospital Centre, Dijon, France
| | - Maëlle Le Bras
- Department of Endocrinology, Nantes University Hospital, Nantes, France.
| | - Marie-Françoise Odou
- Inserm, U1286 - Infinite, University of Lille, CHU of Lille, 59045 Lille cedex, France; Department of Biochemistry and Molecular Biology, Lille University Hospital, Lille, France.
| |
Collapse
|
3
|
Mohr I, Mirzaiebadizi A, Sanyal SK, Chuenban P, Ahmadian MR, Ivanov R, Bauer P. Characterization of the small Arabidopsis thaliana GTPase and ADP-ribosylation factor-like 2 protein TITAN 5. J Cell Sci 2024; 137:jcs262315. [PMID: 39056156 PMCID: PMC11361645 DOI: 10.1242/jcs.262315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 07/28/2024] Open
Abstract
Small GTPases switch between GDP- and GTP-bound states during cell signaling. The ADP-ribosylation factor (ARF) family of small GTPases is involved in vesicle trafficking. Although evolutionarily well conserved, little is known about ARF and ARF-like GTPases in plants. We characterized biochemical properties and cellular localization of the essential small ARF-like GTPase TITAN 5 (TTN5; also known as HALLIMASCH, ARL2 and ARLC1) from Arabidopsis thaliana, and two TTN5 proteins with point mutants in conserved residues, TTN5T30N and TTN5Q70L, that were expected to be unable to perform nucleotide exchange and GTP hydrolysis, respectively. TTN5 exhibited very rapid intrinsic nucleotide exchange and remarkably low GTP hydrolysis activity, functioning as a non-classical small GTPase being likely present in a GTP-loaded active form. We analyzed signals from YFP-TTN5 and HA3-TTN5 by in situ immunolocalization in Arabidopsis seedlings and through use of a transient expression system. Colocalization with endomembrane markers and pharmacological treatments suggests that TTN5 can be present at the plasma membrane and that it dynamically associates with membranes of vesicles, Golgi stacks and multivesicular bodies. Although TTN5Q70L mirrored wild-type TTN5 behavior, the TTN5T30N mutant differed in some aspects. Hence, the unusual rapid nucleotide exchange activity of TTN5 is linked with its membrane dynamics, and TTN5 likely has a role in vesicle transport within the endomembrane system.
Collapse
Affiliation(s)
- Inga Mohr
- Institute of Botany, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Amin Mirzaiebadizi
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Sibaji K. Sanyal
- Institute of Botany, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Pichaporn Chuenban
- Institute of Botany, Heinrich Heine University, 40225 Düsseldorf, Germany
- Center for Plant Genome Engineering, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Mohammad R. Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Rumen Ivanov
- Institute of Botany, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Petra Bauer
- Institute of Botany, Heinrich Heine University, 40225 Düsseldorf, Germany
- Cluster of Excellence on Plant Sciences, Heinrich Heine University, 40225 Düsseldorf, Germany
| |
Collapse
|
4
|
Shankar S, Chew TW, Chichili VPR, Low BC, Sivaraman J. Structural basis for the distinct roles of non-conserved Pro116 and conserved Tyr124 of BCH domain of yeast p50RhoGAP. Cell Mol Life Sci 2024; 81:216. [PMID: 38740643 PMCID: PMC11090974 DOI: 10.1007/s00018-024-05238-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 05/16/2024]
Abstract
p50RhoGAP is a key protein that interacts with and downregulates the small GTPase RhoA. p50RhoGAP is a multifunctional protein containing the BNIP-2 and Cdc42GAP Homology (BCH) domain that facilitates protein-protein interactions and lipid binding and the GAP domain that regulates active RhoA population. We recently solved the structure of the BCH domain from yeast p50RhoGAP (YBCH) and showed that it maintains the adjacent GAP domain in an auto-inhibited state through the β5 strand. Our previous WT YBCH structure shows that a unique kink at position 116 thought to be made by a proline residue between alpha helices α6 and α7 is essential for the formation of intertwined dimer from asymmetric monomers. Here we sought to establish the role and impact of this Pro116. However, the kink persists in the structure of P116A mutant YBCH domain, suggesting that the scaffold is not dictated by the proline residue at this position. We further identified Tyr124 (or Tyr188 in HBCH) as a conserved residue in the crucial β5 strand. Extending to the human ortholog, when substituted to acidic residues, Tyr188D or Tyr188E, we observed an increase in RhoA binding and self-dimerization, indicative of a loss of inhibition of the GAP domain by the BCH domain. These results point to distinct roles and impact of the non-conserved and conserved amino acid positions in regulating the structural and functional complexity of the BCH domain.
Collapse
Affiliation(s)
- Srihari Shankar
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore
| | - Ti Weng Chew
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
| | | | - Boon Chuan Low
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore.
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore.
- NUS College, National University of Singapore, Singapore, 138593, Singapore.
| | - J Sivaraman
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore.
| |
Collapse
|
5
|
Wong DCP, Pan CQ, Er SY, Thivakar T, Rachel TZY, Seah SH, Chua PJ, Jiang T, Chew TW, Chaudhuri PK, Mukherjee S, Salim A, Aye TA, Koh CG, Lim CT, Tan PH, Bay BH, Ridley AJ, Low BC. The scaffold RhoGAP protein ARHGAP8/BPGAP1 synchronizes Rac and Rho signaling to facilitate cell migration. Mol Biol Cell 2023; 34:ar13. [PMID: 36598812 PMCID: PMC10011724 DOI: 10.1091/mbc.e21-03-0099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 01/05/2023] Open
Abstract
Rho GTPases regulate cell morphogenesis and motility under the tight control of guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). However, the underlying mechanism(s) that coordinate their spatiotemporal activities, whether separately or together, remain unclear. We show that a prometastatic RhoGAP, ARHGAP8/BPGAP1, binds to inactive Rac1 and localizes to lamellipodia. BPGAP1 recruits the RacGEF Vav1 under epidermal growth factor (EGF) stimulation and activates Rac1, leading to polarized cell motility, spreading, invadopodium formation, and cell extravasation and promotes cancer cell migration. Importantly, BPGAP1 down-regulates local RhoA activity, which influences Rac1 binding to BPGAP1 and its subsequent activation by Vav1. Our results highlight the importance of BPGAP1 in recruiting Vav1 and Rac1 to promote Rac1 activation for cell motility. BPGAP1 also serves to control the timing of Rac1 activation with RhoA inactivation via its RhoGAP activity. BPGAP1, therefore, acts as a dual-function scaffold that recruits Vav1 to activate Rac1 while inactivating RhoA to synchronize both Rho and Rac signaling in cell motility. As epidermal growth factor receptor (EGFR), Vav1, RhoA, Rac1, and BPGAP1 are all associated with cancer metastasis, BPGAP1 could provide a crucial checkpoint for the EGFR-BPGAP1-Vav1-Rac1-RhoA signaling axis for cancer intervention.
Collapse
Affiliation(s)
| | | | - Shi Yin Er
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore 117558
| | - T. Thivakar
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Tan Zi Yi Rachel
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Sock Hong Seah
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Pei Jou Chua
- Department of Anatomy, Yong Loo Lin School of Medicine, National University Health System, Singapore 117594
| | - Tingting Jiang
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore 117558
| | - Ti Weng Chew
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | | | - Somsubhro Mukherjee
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Agus Salim
- Melbourne School of Population and Global Health and School of Mathematics and Statistics, The University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Thike Aye Aye
- Department of Pathology, Singapore General Hospital, Singapore 169856
| | - Cheng Gee Koh
- Division of Molecular Genetics & Cell Biology, School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583
| | - Puay Hoon Tan
- Department of Pathology, Singapore General Hospital, Singapore 169856
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University Health System, Singapore 117594
| | - Anne J. Ridley
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Boon Chuan Low
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore 117558
- NUS College, National University of Singapore, Singapore 138593
| |
Collapse
|
6
|
Jones RB, Farhi J, Adams M, Parwani KK, Cooper GW, Zecevic M, Lee RS, Hong AL, Spangle JM. Targeting MLL Methyltransferases Enhances the Antitumor Effects of PI3K Inhibition in Hormone Receptor-positive Breast Cancer. CANCER RESEARCH COMMUNICATIONS 2022; 2:1569-1578. [PMID: 36970726 PMCID: PMC10036132 DOI: 10.1158/2767-9764.crc-22-0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 10/03/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022]
Abstract
The high frequency of aberrant PI3K pathway activation in hormone receptor-positive (HR+) breast cancer has led to the development, clinical testing, and approval of the p110α-selective PI3K inhibitor alpelisib. The limited clinical efficacy of alpelisib and other PI3K inhibitors is partially attributed to the functional antagonism between PI3K and estrogen receptor (ER) signaling, which is mitigated via combined PI3K inhibition and endocrine therapy. We and others have previously demonstrated chromatin-associated mechanisms by which PI3K supports cancer development and antagonizes ER signaling through the modulation of the H3K4 methylation axis, inhibition of KDM5A promoter H3K4 demethylation and KMT2D/MLL4-directed enhancer H3K4 methylation. Here we show that inhibition of the H3K4 histone methyltransferase MLL1 in combination with PI3K inhibition impairs HR+ breast cancer clonogenicity and cell proliferation. While combined PI3K/MLL1 inhibition reduces PI3K/AKT signaling and H3K4 methylation, MLL1 inhibition increases PI3K/AKT signaling through the dysregulation of gene expression associated with AKT activation. These data reveal a feedback loop between MLL1 and AKT whereby MLL1 inhibition reactivates AKT. We show that combined PI3K and MLL1 inhibition synergizes to cause cell death in in vitro and in vivo models of HR+ breast cancer, which is enhanced by the additional genetic ablation of the H3K4 methyltransferase and AKT target KMT2D/MLL4. Together, our data provide evidence of a feedback mechanism connecting histone methylation with AKT and may support the preclinical development and testing of pan-MLL inhibitors. Significance Here the authors leverage PI3K/AKT-driven chromatin modification to identify histone methyltransferases as a therapeutic target. Dual PI3K and MLL inhibition synergize to reduce clonogenicity and cell proliferation, and promote in vivo tumor regression. These findings suggest patients with PIK3CA-mutant, HR+ breast cancer may derive clinical benefit from combined PI3K/MLL inhibition.
Collapse
Affiliation(s)
- Robert B. Jones
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Jonathan Farhi
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
- Cancer Biology Graduate Program, Emory University School of Medicine, Atlanta, Georgia
| | - Miranda Adams
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
- Cancer Biology Graduate Program, Emory University School of Medicine, Atlanta, Georgia
| | - Kiran K. Parwani
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
- Cancer Biology Graduate Program, Emory University School of Medicine, Atlanta, Georgia
| | - Garrett W. Cooper
- Genetics and Molecular Biology Graduate Program, Emory University School of Medicine, Atlanta, Georgia
| | - Milica Zecevic
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Richard S. Lee
- Department of Biology, Emory University, Atlanta, Georgia
| | - Andrew L. Hong
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Jennifer M. Spangle
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
- Cancer Biology Graduate Program, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
7
|
Wong DCP, Xiao J, Chew TW, Pan M, Lee CJM, Ang JW, Yow I, Thivakar T, Ackers‐Johnson M, Lee NJW, Foo RS, Kanchanawong P, Low BC. BNIP-2 Activation of Cellular Contractility Inactivates YAP for H9c2 Cardiomyoblast Differentiation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202834. [PMID: 35975420 PMCID: PMC9631078 DOI: 10.1002/advs.202202834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/17/2022] [Indexed: 06/15/2023]
Abstract
Rho GTPases and Hippo kinases are key regulators of cardiomyoblast differentiation. However, how these signaling axes are coordinated spatiotemporally remains unclear. Here, the central and multifaceted roles of the BCH domain containing protein, BNIP-2, in orchestrating the expression of two key cardiac genes (cardiac troponin T [cTnT] and cardiac myosin light chain [Myl2]) in H9c2 and human embryonic stem cell-derived cardiomyocytes are delineated. This study shows that BNIP-2 mRNA and protein expression increase with the onset of cTnT and Myl2 and promote the alignment of H9c2 cardiomyocytes. Mechanistically, BNIP-2 is required for the inactivation of YAP through YAP phosphorylation and its cytosolic retention. Turbo-ID proximity labeling corroborated by super-resolution analyses and biochemical pulldown data reveals a scaffolding role of BNIP-2 for LATS1 to phosphorylate and inactivate YAP in a process that requires BNIP-2 activation of cellular contractility. The findings identify BNIP-2 as a pivotal signaling scaffold that spatiotemporally integrates RhoA/Myosin II and LATS1/YAP mechanotransduction signaling to drive cardiomyoblast differentiation, by switching the genetic programming from YAP-dependent growth to YAP-silenced differentiation. These findings offer insights into the importance of scaffolding proteins in bridging the gap between mechanical and biochemical signals in cell growth and differentiation and the prospects in translational applications.
Collapse
Affiliation(s)
- Darren Chen Pei Wong
- Mechanobiology Institute SingaporeNational University of SingaporeSingapore117411Singapore
- Department of Biological SciencesNational University of SingaporeSingapore117558Singapore
| | - Jingwei Xiao
- Mechanobiology Institute SingaporeNational University of SingaporeSingapore117411Singapore
| | - Ti Weng Chew
- Mechanobiology Institute SingaporeNational University of SingaporeSingapore117411Singapore
| | - Meng Pan
- Mechanobiology Institute SingaporeNational University of SingaporeSingapore117411Singapore
| | - Chang Jie Mick Lee
- Genome Institute of SingaporeAgency for ScienceTechnology and ResearchSingapore138672Singapore
| | - Jing Wen Ang
- Department of Biological SciencesNational University of SingaporeSingapore117558Singapore
| | - Ivan Yow
- Mechanobiology Institute SingaporeNational University of SingaporeSingapore117411Singapore
| | - T. Thivakar
- Mechanobiology Institute SingaporeNational University of SingaporeSingapore117411Singapore
| | - Matthew Ackers‐Johnson
- Genome Institute of SingaporeAgency for ScienceTechnology and ResearchSingapore138672Singapore
- Cardiovascular Research InstituteNational University Healthcare SystemsSingapore117599Singapore
| | - Nicole Jia Wen Lee
- Department of Biological SciencesNational University of SingaporeSingapore117558Singapore
| | - Roger Sik‐Yin Foo
- Genome Institute of SingaporeAgency for ScienceTechnology and ResearchSingapore138672Singapore
- Cardiovascular Research InstituteNational University Healthcare SystemsSingapore117599Singapore
- Department of MedicineYong Loo Lin School of MedicineNational University of SingaporeSingapore117597Singapore
| | - Pakorn Kanchanawong
- Mechanobiology Institute SingaporeNational University of SingaporeSingapore117411Singapore
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Boon Chuan Low
- Mechanobiology Institute SingaporeNational University of SingaporeSingapore117411Singapore
- Department of Biological SciencesNational University of SingaporeSingapore117558Singapore
- NUS CollegeNational University of SingaporeSingapore138593Singapore
| |
Collapse
|
8
|
Fixing the GAP: the role of RhoGAPs in cancer. Eur J Cell Biol 2022; 101:151209. [DOI: 10.1016/j.ejcb.2022.151209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/29/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
|
9
|
Li T, Huang S, Yan W, Zhang Y, Guo Q. PRUNE2 inhibits progression of colorectal cancer in vitro and in vivo. Exp Ther Med 2021; 23:169. [PMID: 35069850 PMCID: PMC8764654 DOI: 10.3892/etm.2021.11092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/09/2021] [Indexed: 12/24/2022] Open
Abstract
Prune homolog 2 with BCH domain (PRUNE2) is associated with prostate cancer, neuroblastoma, glioblastoma and melanoma; however, the function of PRUNE2 in colorectal cancer (CRC) remains unknown. The present study aimed to evaluate the effects of PRUNE2 on the development of CRC. The biological function of PRUNE2 in CRC cell lines was investigated by using Cell Counting Kit-8, colony formation, flow cytometry and Transwell assay. Additionally, a mouse model was established to investigate the effect of PRUNE2 on metastasis of CRC cells. The expression levels of PRUNE2 were lower in CRC compared with adjacent normal tissue and this expression pattern was associated with poor relapse-free survival probability. PRUNE2 overexpression significantly decreased cell proliferation and invasion, increased cell apoptosis and arrested the cell cycle. Consistently, it increased the protein expression levels of pro-apoptosis genes and decreased the expression of antiapoptotic proteins. PRUNE2 knockdown had the opposite effects. Furthermore, PRUNE2 overexpression decreased the tumorigenicity of CRC cells. In conclusion, PRUNE2 decreased cell survival, proliferation, invasion and tumorigenicity and promoted apoptosis, suggesting that PRUNE2 may function as a tumor-suppressive gene in CRC.
Collapse
Affiliation(s)
- Ting Li
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Silin Huang
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Wei Yan
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Yu Zhang
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
| | - Qiang Guo
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
10
|
Structural basis for p50RhoGAP BCH domain-mediated regulation of Rho inactivation. Proc Natl Acad Sci U S A 2021; 118:2014242118. [PMID: 34006635 DOI: 10.1073/pnas.2014242118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Spatiotemporal regulation of signaling cascades is crucial for various biological pathways, under the control of a range of scaffolding proteins. The BNIP-2 and Cdc42GAP Homology (BCH) domain is a highly conserved module that targets small GTPases and their regulators. Proteins bearing BCH domains are key for driving cell elongation, retraction, membrane protrusion, and other aspects of active morphogenesis during cell migration, myoblast differentiation, and neuritogenesis. We previously showed that the BCH domain of p50RhoGAP (ARHGAP1) sequesters RhoA from inactivation by its adjacent GAP domain; however, the underlying molecular mechanism for RhoA inactivation by p50RhoGAP remains unknown. Here, we report the crystal structure of the BCH domain of p50RhoGAP Schizosaccharomyces pombe and model the human p50RhoGAP BCH domain to understand its regulatory function using in vitro and cell line studies. We show that the BCH domain adopts an intertwined dimeric structure with asymmetric monomers and harbors a unique RhoA-binding loop and a lipid-binding pocket that anchors prenylated RhoA. Interestingly, the β5-strand of the BCH domain is involved in an intermolecular β-sheet, which is crucial for inhibition of the adjacent GAP domain. A destabilizing mutation in the β5-strand triggers the release of the GAP domain from autoinhibition. This renders p50RhoGAP active, thereby leading to RhoA inactivation and increased self-association of p50RhoGAP molecules via their BCH domains. Our results offer key insight into the concerted spatiotemporal regulation of Rho activity by BCH domain-containing proteins.
Collapse
|
11
|
Cuveillier C, Boulan B, Ravanello C, Denarier E, Deloulme JC, Gory-Fauré S, Delphin C, Bosc C, Arnal I, Andrieux A. Beyond Neuronal Microtubule Stabilization: MAP6 and CRMPS, Two Converging Stories. Front Mol Neurosci 2021; 14:665693. [PMID: 34025352 PMCID: PMC8131560 DOI: 10.3389/fnmol.2021.665693] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/09/2021] [Indexed: 12/21/2022] Open
Abstract
The development and function of the central nervous system rely on the microtubule (MT) and actin cytoskeletons and their respective effectors. Although the structural role of the cytoskeleton has long been acknowledged in neuronal morphology and activity, it was recently recognized to play the role of a signaling platform. Following this recognition, research into Microtubule Associated Proteins (MAPs) diversified. Indeed, historically, structural MAPs—including MAP1B, MAP2, Tau, and MAP6 (also known as STOP);—were identified and described as MT-binding and -stabilizing proteins. Extensive data obtained over the last 20 years indicated that these structural MAPs could also contribute to a variety of other molecular roles. Among multi-role MAPs, MAP6 provides a striking example illustrating the diverse molecular and cellular properties of MAPs and showing how their functional versatility contributes to the central nervous system. In this review, in addition to MAP6’s effect on microtubules, we describe its impact on the actin cytoskeleton, on neuroreceptor homeostasis, and its involvement in signaling pathways governing neuron development and maturation. We also discuss its roles in synaptic plasticity, brain connectivity, and cognitive abilities, as well as the potential relationships between the integrated brain functions of MAP6 and its molecular activities. In parallel, the Collapsin Response Mediator Proteins (CRMPs) are presented as examples of how other proteins, not initially identified as MAPs, fall into the broader MAP family. These proteins bind MTs as well as exhibiting molecular and cellular properties very similar to MAP6. Finally, we briefly summarize the multiple similarities between other classical structural MAPs and MAP6 or CRMPs.In summary, this review revisits the molecular properties and the cellular and neuronal roles of the classical MAPs, broadening our definition of what constitutes a MAP.
Collapse
|
12
|
Pan M, Chew TW, Wong DCP, Xiao J, Ong HT, Chin JFL, Low BC. BNIP-2 retards breast cancer cell migration by coupling microtubule-mediated GEF-H1 and RhoA activation. SCIENCE ADVANCES 2020; 6:eaaz1534. [PMID: 32789168 PMCID: PMC7399486 DOI: 10.1126/sciadv.aaz1534] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 06/18/2020] [Indexed: 06/11/2023]
Abstract
Microtubules display dynamic turnover during cell migration, leading to cell contractility and focal adhesion maturation regulated by Rho guanosine triphosphatase activity. This interplay between microtubules and actomyosin is mediated by guanine nucleotide exchange factor (GEF)-H1 released after microtubule depolymerization or microtubule disconnection from focal adhesions. However, how GEF-H1 activates Rho upon microtubule disassembly remains elusive. Here, we found that BNIP-2, a BCH domain-containing protein that binds both RhoA and GEF-H1 and traffics with kinesin-1 on microtubules, is important for GEF-H1-driven RhoA activation upon microtubule disassembly. Depletion of BNIP-2 in MDA-MB-231 breast cancer cells decreases RhoA activity and promotes cell migration. Upon nocodazole-induced microtubule disassembly, the interaction between BNIP-2 and GEF-H1 increases, while knockdown of BNIP-2 reduces RhoA activation and cell rounding via uncoupling RhoA-GEF-H1 interaction. Together, these findings revealed that BNIP-2 couples microtubules and focal adhesions via scaffolding GEF-H1 and RhoA, fine-tuning RhoA activity and cell migration.
Collapse
Affiliation(s)
- Meng Pan
- Mechanobiology Institute, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Ti Weng Chew
- Mechanobiology Institute, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Darren Chen Pei Wong
- Mechanobiology Institute, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Jingwei Xiao
- Mechanobiology Institute, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Hui Ting Ong
- Mechanobiology Institute, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Jasmine Fei Li Chin
- Mechanobiology Institute, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Boon Chuan Low
- Mechanobiology Institute, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
- Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543, Singapore
- University Scholars Programme, 18 College Avenue East, Singapore 138593, Singapore
| |
Collapse
|
13
|
Park S, Yoo HJ, Jee SH, Lee JH, Kim M. Weighting approaches for a genetic risk score and an oxidative stress score for predicting the incidence of obesity. Diabetes Metab Res Rev 2020; 36:e3230. [PMID: 31654550 DOI: 10.1002/dmrr.3230] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/09/2019] [Accepted: 10/06/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND We aimed to predict the incidence of obesity in a Korean population using a genetic risk score (GRS) constructed with obesity-related single nucleotide polymorphisms (SNPs) along with an oxidative stress score (OSS). METHODS A total of 9460 Korean subjects and 356 974 SNPs were included. The GRS was constructed using three significant obesity-related SNP loci, and the OSS was calculated with three reliable oxidative stress biomarkers. RESULTS The GRS showed a more significant association with increased obesity (OR = 2.879) than did individual SNPs after adjusting for age and sex. Three oxidative stress biomarkers, including malondialdehyde, oxidized low-density lipoprotein, and 8-epi-prostaglandin F2α , showed significantly high levels in the obese group. The OSS, which was the sum of each oxidative stress biomarker score, showed a markedly high association with the incidence of obesity, with an OR of 3.213. Based on the results of the regression tests and a receiver-operating characteristic (ROC) curve analysis, we found that HOMA-IR, high-sensitivity C-reactive protein (hs-CRP), the GRS, and the OSS were the most relevant factors for the increased risk of obesity and were significantly associated with the incidence of obesity. The area under the ROC curve was improved when the GRS was added to the model (from 74.2% to 75.1%). CONCLUSIONS We first identified that subjects with an obesity GRS and a high OSS might have a higher risk of obesity. Our findings and weighting approaches were effective in predicting the incidence of obesity; furthermore, the GRS is a relevant factor that significantly predicts the risk of obesity.
Collapse
Affiliation(s)
- Seonmin Park
- Department of Science for Aging, Graduate School of Yonsei University, Seoul, Korea
| | - Hye Jin Yoo
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, Korea
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, Korea
| | - Sun Ha Jee
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Jong Ho Lee
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, Korea
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, Korea
| | - Minjoo Kim
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, Korea
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, Korea
| |
Collapse
|
14
|
Alsadoun N, MacGrogan G, Truntzer C, Lacroix-Triki M, Bedgedjian I, Koeb MH, El Alam E, Medioni D, Parent M, Wuithier P, Robert I, Boidot R, Arnould L. Solid papillary carcinoma with reverse polarity of the breast harbors specific morphologic, immunohistochemical and molecular profile in comparison with other benign or malignant papillary lesions of the breast: a comparative study of 9 additional cases. Mod Pathol 2018; 31:1367-1380. [PMID: 29785016 DOI: 10.1038/s41379-018-0047-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 02/03/2018] [Accepted: 02/04/2018] [Indexed: 12/26/2022]
Abstract
Solid papillary carcinoma with reverse polarity is a rare breast cancer of favorable prognosis that can be difficult to diagnose. We report here nine additional cases of this tumor, and we describe its morphologic, immunohistochemical and molecular profile in comparison to other types of papillary and micropapillary lesions of the breast that are intraductal papilloma with usual ductal hyperplasia, encapsulated papillary carcinoma, solid papillary carcinoma and invasive micropapillary carcinoma. We studied nine cases of this special papillary tumor and six of each other types mentioned above. We found that solid papillary carcinoma with reverse polarity harbor specific morphologic features as cuboid or tall cells with abundant eosinophilic cytoplasms located at the basal pole giving the impression of reverse nuclear polarity. Nuclei were sometimes grooved. Immunohistochemistry demonstrated the lack of myoepithelial cells, as in encapsulated papillary carcinoma and solid papillary carcinoma, questioning their invasive nature. Seven of nine solid papillary carcinoma with reverse polarity showed a low Ki67 proliferative index (Ki67 <5%). They showed expression of CK5/6 as in intraductal papilloma with usual ductal hyperplasia. They showed expression of calretinin and a low or lack of hormonal receptor (HR) expression that were not observed in other breast tumors studied. By whole-exome analysis, seven of nine solid papillary carcinomas with reverse polarity (78%) harbored a hotspot mutation in IDH2 (R172) that was totally absent in other groups. Six of nine tumors (67%) also harbored PRUNE2 mutation, including the two IDH2 wild-type cases. We also demonstrated for the first time in this breast tumor, immunostaining with a specific antibody IDH1/2 mutant R132/R172 (7/9) that can highlight IDH2 mutation. Moreover, transcriptomic analysis showed that proteoglycan pathway was significantly enriched. Our findings support the fact that solid papillary carcinoma with reverse polarity is a singular breast neoplasm that can be distinguished from other papillary breast tumors.
Collapse
Affiliation(s)
- Nadjla Alsadoun
- Département de Biologie et Pathologie des Tumeurs, Centre Georges-François Leclerc - Unicancer, 1 rue Professeur Marion, 21000, Dijon, France.
| | - Gaëtan MacGrogan
- Département de Biopathologie, Institut Bergonié, 229 cours de l'Argonne, 33076, Bordeaux, France
| | - Caroline Truntzer
- Département de Biologie et Pathologie des Tumeurs, Centre Georges-François Leclerc - Unicancer, 1 rue Professeur Marion, 21000, Dijon, France
| | - Magali Lacroix-Triki
- Département de Pathologie, Institut Gustave Roussy, 114 rue Edouard Vaillant, 94805, Villejuif, France
| | - Isabelle Bedgedjian
- Service d'Anatomie et Cytologie Pathologiques, Centre Hospitalier Universitaire de Besançon, 25030, Besançon Cedex, France
| | | | - Elsy El Alam
- Département de Pathologie, Institut Curie, Saint Cloud, 35 rue Dailly, 92210, Saint Cloud, France
| | - Dan Medioni
- Medipath Cannes-Antibes-Grasse, 80 allée des ormes, 06250, Mougins, France
| | - Michel Parent
- Pathologie Nord Unilabs, 60 boulevard Jean Baptiste Lebas, 59000, Lille, France
| | | | - Isabelle Robert
- Atalante Pathologie, 10 rue J-Louis Bertrand -BP11633, 35016, Rennes cedex, France
| | - Romain Boidot
- Département de Biologie et Pathologie des Tumeurs, Centre Georges-François Leclerc - Unicancer, 1 rue Professeur Marion, 21000, Dijon, France
| | - Laurent Arnould
- Département de Biologie et Pathologie des Tumeurs, Centre Georges-François Leclerc - Unicancer, 1 rue Professeur Marion, 21000, Dijon, France
| |
Collapse
|
15
|
Islam S, Ueda M, Nishida E, Wang MX, Osawa M, Lee D, Itoh M, Nakagawa K, Tana, Nakagawa T. Odor preference and olfactory memory are impaired in Olfaxin-deficient mice. Brain Res 2018; 1688:81-90. [PMID: 29571668 DOI: 10.1016/j.brainres.2018.03.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/19/2018] [Accepted: 03/19/2018] [Indexed: 12/12/2022]
Abstract
Olfaxin, which is a BNIP2 and Cdc42GAP homology (BCH) domain-containing protein, is predominantly expressed in mitral and tufted (M/T) cells in the olfactory bulb (OB). Olfaxin and Caytaxin, which share 56.3% amino acid identity, are similar in their glutamatergic terminal localization, kidney-type glutaminase (KGA) interaction, and caspase-3 substrate. Although the deletion of Caytaxin protein causes human Cayman ataxia and ataxia in the mutant mouse, the function of Olfaxin is largely unknown. In this study, we generated Prune2 gene mutant mice (Prune2Ex16-/-; knock out [KO] mice) using the CRISPR/Cas9 system, during which the exon 16 containing start codon of Olfaxin mRNA was deleted. Exon 16 has 80 nucleotides and is contained in four of five Prune2 isoforms, including PRUNE2, BMCC1, BNIPXL, and Olfaxin/BMCC1s. The levels of Olfaxin mRNA and Olfaxin protein in the OB and piriform cortex of KO mice significantly decreased. Although Prune2 mRNA also significantly decreased in the spinal cord, the gross anatomy of the spinal cord and dorsal root ganglion (DRG) was intact. Further, disturbance of the sensory and motor system was not observed in KO mice. Therefore, in the current study, we examined the role of Olfaxin in the olfactory system where PRUNE2, BMCC1, and BNIPXL are scarcely expressed. Odor preference was impaired in KO mice using opposite-sex urinary scents as well as a non-social odor stimulus (almond). Results of the odor-aversion test demonstrated that odor-associative learning was disrupted in KO mice. Moreover, the NMDAR2A/NMDAR2B subunits switch in the piriform cortex was not observed in KO mice. These results indicated that Olfaxin may play a critical role in odor preference and olfactory memory.
Collapse
Affiliation(s)
- Saiful Islam
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masashi Ueda
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan; Department of Embryology, Institute for Developmental Research, Aichi Human Service Center, Aichi, Japan
| | - Emika Nishida
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Miao-Xing Wang
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masatake Osawa
- Department of Molecular Design and Synthesis, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Dongsoo Lee
- Department of Molecular Design and Synthesis, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masanori Itoh
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Kiyomi Nakagawa
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tana
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Toshiyuki Nakagawa
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan.
| |
Collapse
|
16
|
Anuppalle M, Maddirevula S, Kumar A, Huh TL, Choe J, Rhee M. Expression patterns of prune2 is regulated by Notch and retinoic acid signaling pathways in the zebrafish embryogenesis. Gene Expr Patterns 2017; 23-24:45-51. [DOI: 10.1016/j.gep.2017.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 03/07/2017] [Accepted: 03/08/2017] [Indexed: 01/12/2023]
|
17
|
Transcriptional regulation of BMCC1 mediated by E2F1 in neuroblastoma cells. Biochem Biophys Res Commun 2016; 478:81-86. [DOI: 10.1016/j.bbrc.2016.07.089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 07/20/2016] [Indexed: 01/15/2023]
|
18
|
PRUNE2 is a human prostate cancer suppressor regulated by the intronic long noncoding RNA PCA3. Proc Natl Acad Sci U S A 2015; 112:8403-8. [PMID: 26080435 DOI: 10.1073/pnas.1507882112] [Citation(s) in RCA: 217] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer antigen 3 (PCA3) is the most specific prostate cancer biomarker but its function remains unknown. Here we identify PRUNE2, a target protein-coding gene variant, which harbors the PCA3 locus, thereby classifying PCA3 as an antisense intronic long noncoding (lnc)RNA. We show that PCA3 controls PRUNE2 levels via a unique regulatory mechanism involving formation of a PRUNE2/PCA3 double-stranded RNA that undergoes adenosine deaminase acting on RNA (ADAR)-dependent adenosine-to-inosine RNA editing. PRUNE2 expression or silencing in prostate cancer cells decreased and increased cell proliferation, respectively. Moreover, PRUNE2 and PCA3 elicited opposite effects on tumor growth in immunodeficient tumor-bearing mice. Coregulation and RNA editing of PRUNE2 and PCA3 were confirmed in human prostate cancer specimens, supporting the medical relevance of our findings. These results establish PCA3 as a dominant-negative oncogene and PRUNE2 as an unrecognized tumor suppressor gene in human prostate cancer, and their regulatory axis represents a unique molecular target for diagnostic and therapeutic intervention.
Collapse
|
19
|
BMCC1, which is an interacting partner of BCL2, attenuates AKT activity, accompanied by apoptosis. Cell Death Dis 2015; 6:e1607. [PMID: 25611382 PMCID: PMC4669766 DOI: 10.1038/cddis.2014.568] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 10/19/2014] [Accepted: 10/30/2014] [Indexed: 11/08/2022]
Abstract
BNIP2 and Cdc42GAP homology (BCH) motif-containing molecule at the carboxyl-terminal region 1 (BMCC1) gene is highly expressed in patients with favorable neuroblastoma (NB). It encodes a 340-kDa protein with a conserved BCH scaffold domain that may regulate signaling networks and multiple cellular functions, including apoptosis. In this study, we determined the mechanism by which BMCC1 promotes apoptosis in human NB and non-NB cells, as BMCC1 is normally expressed in various organs, particularly in neuronal and epithelial tissues. We demonstrated in this report that BMCC1 was induced by DNA damage, one of the triggers of intrinsic apoptosis. Accordingly, we investigated whether BMCC1 expression impacts intracellular signals in the regulation of apoptosis via its C-terminal region containing BCH scaffold domain. BMCC1 decreased phosphorylation of survival signals on AKT and its upstream kinase PDK1. BMCC1 upregulation was correlated with the activation of forkhead box-O3a (FOXO3a) (a downstream inducer of apoptosis, which is suppressed by AKT) and induction of BCL2 inhibitor BIM, suggesting that BMCC1 negatively regulates phosphorylation pathway of AKT, resulted in apoptosis. In addition, we found that BNIP2 homology region of BMCC1 interacts with BCL2. Intrinsic apoptosis induced by DNA damage was enhanced by BMCC1 overexpression, and was diminished by knockdown of BMCC1. Taken together, we conclude that BMCC1 promotes apoptosis at multiple steps in AKT-mediated survival signal pathway. These steps include physical interaction with BCL2 and attenuation of AKT-dependent inhibition of FOXO3a functions, such as transcriptional induction of BIM and phosphorylation of ataxia telangiectasia-mutated (ATM) after DNA damage. We propose that downregulation of BMCC1 expression, which is frequently observed in unfavorable NB and epithelial-derived cancers, may facilitate tumor development by abrogating DNA damage repair and apoptosis.
Collapse
|
20
|
Harris JL, Richards RS, Chow CWK, Lee S, Kim M, Buck M, Teng L, Clarke R, Gardiner RA, Lavin MF. BMCC1 is an AP-2 associated endosomal protein in prostate cancer cells. PLoS One 2013; 8:e73880. [PMID: 24040105 PMCID: PMC3765211 DOI: 10.1371/journal.pone.0073880] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 07/23/2013] [Indexed: 12/04/2022] Open
Abstract
The prostate cancer antigen gene 3 (PCA3) is embedded in an intron of a second gene BMCC1 (Bcl2-/adenovirus E1B nineteen kDa-interacting protein 2 (BNIP-2) and Cdc42GAP homology BCH motif-containing molecule at the carboxyl terminal region 1) which is also upregulated in prostate cancer. BMCC1 was initially annotated as two genes (C9orf65/PRUNE and BNIPXL) on either side of PCA3 but our data suggest that it represents a single gene coding for a high molecular weight protein. Here we demonstrate for the first time the expression of a >300 kDa BMCC1 protein (BMCC1-1) in prostate cancer and melanoma cell lines. This protein was found exclusively in the microsomal fraction and localised to cytoplasmic vesicles. We also observed expression of BMCC1 protein in prostate cancer sections using immunohistology. GST pull down, immunoprecipitation and mass spectrometry protein interaction studies identified multiple members of the Adaptor Related Complex 2 (AP-2) as BMCC1 interactors. Consistent with a role for BMCC1 as an AP-2 interacting endosomal protein, BMCC1 co-localised with β-adaptin at the perinuclear region of the cell. BMCC1 also showed partial co-localisation with the early endosome small GTP-ase Rab-5 as well as strong co-localisation with internalised pulse-chase labelled transferrin (Tf), providing evidence that BMCC1 is localised to functional endocytic vesicles. BMCC1 knockdown did not affect Tf uptake and AP-2 knockdown did not disperse BMCC1 vesicular distribution, excluding an essential role for BMCC1 in canonical AP-2 mediated endocytic uptake. Instead, we posit a novel role for BMCC1 in post-endocytic trafficking. This study provides fundamental characterisation of the BMCC1 complex in prostate cancer cells and for the first time implicates it in vesicle trafficking.
Collapse
Affiliation(s)
- Janelle L. Harris
- Queensland Institute of Medical Research, Herston, Brisbane, Queensland, Australia
- * E-mail: (MFL); (JLH)
| | - Renée S. Richards
- Queensland Institute of Medical Research, Herston, Brisbane, Queensland, Australia
- University of Queensland Centre for Clinical Research, Herston, Brisbane, Queensland, Australia
| | - Clement W. K. Chow
- Queensland Institute of Medical Research, Herston, Brisbane, Queensland, Australia
- University of Queensland Centre for Clinical Research, Herston, Brisbane, Queensland, Australia
| | - Soon Lee
- School of Medicine, University of Western Sydney, Liverpool, Sydney, Australia
| | - Misook Kim
- University of Queensland Centre for Clinical Research, Herston, Brisbane, Queensland, Australia
| | - Marion Buck
- Queensland Institute of Medical Research, Herston, Brisbane, Queensland, Australia
- University of Queensland Centre for Clinical Research, Herston, Brisbane, Queensland, Australia
| | - Linda Teng
- University of Queensland Centre for Clinical Research, Herston, Brisbane, Queensland, Australia
| | - Raymond Clarke
- School of Medicine, University of Western Sydney, Liverpool, Sydney, Australia
| | - Robert A. Gardiner
- University of Queensland Centre for Clinical Research, Herston, Brisbane, Queensland, Australia
| | - Martin F. Lavin
- Queensland Institute of Medical Research, Herston, Brisbane, Queensland, Australia
- University of Queensland Centre for Clinical Research, Herston, Brisbane, Queensland, Australia
- * E-mail: (MFL); (JLH)
| |
Collapse
|
21
|
Zhao LR, Tian W, Wang GW, Chen KX, Yang JL. The prognostic role of PRUNE2 in leiomyosarcoma. CHINESE JOURNAL OF CANCER 2013; 32:648-52. [PMID: 23731771 PMCID: PMC3870848 DOI: 10.5732/cjc.013.10069] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PRUNE2 plays an important role in regulating tumor cell differentiation, proliferation, and invasiveness in neuroblastoma. Our previous study revealed that PRUNE2/OBSCN two-gene relative expression classifer accurately differentiated leiomyosarcoma from gastrointestinal stromal tumor. However, the association between PRUNE2 expression and prognosis in leiomyosarcoma is poorly understood. In this study, we evaluated the prognostic role of PRUNE2 in leiomyosarcoma. PRUNE2 expression was detected using immunohistochemistry in 30 formalin-fixed, paraffin-embedded leiomyosarcoma tissues from MD Anderson Cancer Center, and high expression was detected in 36.7% (11/30) of the samples. To validate these results, immunohistochemistry was performed on another cohort of 45 formalin-fixed, paraffin-embedded leiomyosarcoma tissues from Tianjin Medical University Cancer Institute & Hospital, and high PRUNE2 protein expression was detected in 37.8% (17/45) of the samples. Moreover, elevated PRUNE2 expression was significantly associated with tumor size (P = 0.03) and hemorrhage/cyst (P = 0.014), and was an independent favorable prognostic factor for overall survival in leiomyosarcoma patients from Tianjin Medical University Cancer Institute & Hospital (P < 0.05). These data suggest that increased PRUNE2 protein expression may serve as a favorable prognostic marker in human leiomyosarcoma.
Collapse
Affiliation(s)
- Lin-Ru Zhao
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Hospital & Institute, Tianjin 300060, P. R. China. .
| | | | | | | | | |
Collapse
|
22
|
Song Y, Ahn J, Suh Y, Davis ME, Lee K. Identification of novel tissue-specific genes by analysis of microarray databases: a human and mouse model. PLoS One 2013; 8:e64483. [PMID: 23741331 PMCID: PMC3669334 DOI: 10.1371/journal.pone.0064483] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 04/15/2013] [Indexed: 12/15/2022] Open
Abstract
Understanding the tissue-specific pattern of gene expression is critical in elucidating the molecular mechanisms of tissue development, gene function, and transcriptional regulations of biological processes. Although tissue-specific gene expression information is available in several databases, follow-up strategies to integrate and use these data are limited. The objective of the current study was to identify and evaluate novel tissue-specific genes in human and mouse tissues by performing comparative microarray database analysis and semi-quantitative PCR analysis. We developed a powerful approach to predict tissue-specific genes by analyzing existing microarray data from the NCBI′s Gene Expression Omnibus (GEO) public repository. We investigated and confirmed tissue-specific gene expression in the human and mouse kidney, liver, lung, heart, muscle, and adipose tissue. Applying our novel comparative microarray approach, we confirmed 10 kidney, 11 liver, 11 lung, 11 heart, 8 muscle, and 8 adipose specific genes. The accuracy of this approach was further verified by employing semi-quantitative PCR reaction and by searching for gene function information in existing publications. Three novel tissue-specific genes were discovered by this approach including AMDHD1 (amidohydrolase domain containing 1) in the liver, PRUNE2 (prune homolog 2) in the heart, and ACVR1C (activin A receptor, type IC) in adipose tissue. We further confirmed the tissue-specific expression of these 3 novel genes by real-time PCR. Among them, ACVR1C is adipose tissue-specific and adipocyte-specific in adipose tissue, and can be used as an adipocyte developmental marker. From GEO profiles, we predicted the processes in which AMDHD1 and PRUNE2 may participate. Our approach provides a novel way to identify new sets of tissue-specific genes and to predict functions in which they may be involved.
Collapse
Affiliation(s)
- Yan Song
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Jinsoo Ahn
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio, United States of America
- The Ohio State University Interdisciplinary PhD Program in Nutrition (OSUN), The Ohio State University, Columbus, Ohio, United States of America
| | - Yeunsu Suh
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Michael E. Davis
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Kichoon Lee
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio, United States of America
- The Ohio State University Interdisciplinary PhD Program in Nutrition (OSUN), The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
23
|
Li S, Hayakawa-Yano Y, Itoh M, Ueda M, Ohta K, Suzuki Y, Mizuno A, Ohta E, Hida Y, Wang MX, Nakagawa T. Olfaxin as a novel Prune2 isoform predominantly expressed in olfactory system. Brain Res 2012; 1488:1-13. [DOI: 10.1016/j.brainres.2012.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 09/05/2012] [Accepted: 10/01/2012] [Indexed: 01/01/2023]
|
24
|
Ravichandran A, Low BC. SmgGDS antagonizes BPGAP1-induced Ras/ERK activation and neuritogenesis in PC12 cell differentiation. Mol Biol Cell 2012; 24:145-56. [PMID: 23155002 PMCID: PMC3541961 DOI: 10.1091/mbc.e12-04-0300] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BPGAP1 controls morphogenesis, migration, and ERK signaling by the concerted action of its multiple domains. Its BCH domain targets K-Ras and induces robust ERK activation and neuronal differentiation in a process antagonized by SmgGDS. The results highlight unique cross-talk of two regulators of GTPases in Ras/ERK signaling and differentiation. BPGAP1 is a Rho GTPase-activating protein (RhoGAP) that regulates cell morphogenesis, cell migration, and ERK signaling by the concerted action of its proline-rich region (PRR), RhoGAP domain, and the BNIP-2 and Cdc42GAP homology (BCH) domain. Although multiple cellular targets for the PRR and RhoGAP have been identified, and their functions delineated, the mechanism by which the BCH domain regulates functions of BPGAP1 remains unclear. Here we show that its BCH domain induced robust ERK activation leading to PC12 cell differentiation by targeting specifically to K-Ras. Such stimulatory effect was inhibited, however, by both dominant-negative mutants of Mek2 (Mek2-K101A) and K-Ras (K-Ras-S17N) and also by the small G-protein GDP dissociation stimulator (SmgGDS). Consequently SmgGDS knockdown released this inhibition and resulted in a superinduction of K-Ras activation and PC12 differentiation mediated by BCH domain. These results demonstrate the versatility of the BCH domain of BPGAP1 in regulating ERK signaling by involving K-Ras and SmgGDS and support the unique role of BPGAP1 as a dual regulator for Ras and Rho signaling in cell morphogenesis and differentiation.
Collapse
Affiliation(s)
- Aarthi Ravichandran
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore 117543, Republic of Singapore
| | | |
Collapse
|
25
|
Pan CQ, Sudol M, Sheetz M, Low BC. Modularity and functional plasticity of scaffold proteins as p(l)acemakers in cell signaling. Cell Signal 2012; 24:2143-65. [PMID: 22743133 DOI: 10.1016/j.cellsig.2012.06.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 05/22/2012] [Accepted: 06/16/2012] [Indexed: 01/14/2023]
Abstract
Cells coordinate and integrate various functional modules that control their dynamics, intracellular trafficking, metabolism and gene expression. Such capacity is mediated by specific scaffold proteins that tether multiple components of signaling pathways at plasma membrane, Golgi apparatus, mitochondria, endoplasmic reticulum, nucleus and in more specialized subcellular structures such as focal adhesions, cell-cell junctions, endosomes, vesicles and synapses. Scaffold proteins act as "pacemakers" as well as "placemakers" that regulate the temporal, spatial and kinetic aspects of protein complex assembly by modulating the local concentrations, proximity, subcellular dispositions and biochemical properties of the target proteins through the intricate use of their modular protein domains. These regulatory mechanisms allow them to gate the specificity, integration and crosstalk of different signaling modules. In addition to acting as physical platforms for protein assembly, many professional scaffold proteins can also directly modify the properties of their targets while they themselves can be regulated by post-translational modifications and/or mechanical forces. Furthermore, multiple scaffold proteins can form alliances of higher-order regulatory networks. Here, we highlight the emerging themes of scaffold proteins by analyzing their common and distinctive mechanisms of action and regulation, which underlie their functional plasticity in cell signaling. Understanding these mechanisms in the context of space, time and force should have ramifications for human physiology and for developing new therapeutic approaches to control pathological states and diseases.
Collapse
Affiliation(s)
- Catherine Qiurong Pan
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Republic of Singapore.
| | | | | | | |
Collapse
|
26
|
Pan CQ, Low BC. Functional plasticity of the BNIP-2 and Cdc42GAP Homology (BCH) domain in cell signaling and cell dynamics. FEBS Lett 2012; 586:2674-91. [DOI: 10.1016/j.febslet.2012.04.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 04/16/2012] [Accepted: 04/16/2012] [Indexed: 10/28/2022]
|
27
|
Arama J, Boulay AC, Bosc C, Delphin C, Loew D, Rostaing P, Amigou E, Ezan P, Wingertsmann L, Guillaud L, Andrieux A, Giaume C, Cohen-Salmon M. Bmcc1s, a novel brain-isoform of Bmcc1, affects cell morphology by regulating MAP6/STOP functions. PLoS One 2012; 7:e35488. [PMID: 22523599 PMCID: PMC3327665 DOI: 10.1371/journal.pone.0035488] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 03/16/2012] [Indexed: 12/21/2022] Open
Abstract
The BCH (BNIP2 and Cdc42GAP Homology) domain-containing protein Bmcc1/Prune2 is highly enriched in the brain and is involved in the regulation of cytoskeleton dynamics and cell survival. However, the molecular mechanisms accounting for these functions are poorly defined. Here, we have identified Bmcc1s, a novel isoform of Bmcc1 predominantly expressed in the mouse brain. In primary cultures of astrocytes and neurons, Bmcc1s localized on intermediate filaments and microtubules and interacted directly with MAP6/STOP, a microtubule-binding protein responsible for microtubule cold stability. Bmcc1s overexpression inhibited MAP6-induced microtubule cold stability by displacing MAP6 away from microtubules. It also resulted in the formation of membrane protrusions for which MAP6 was a necessary cofactor of Bmcc1s. This study identifies Bmcc1s as a new MAP6 interacting protein able to modulate MAP6-induced microtubule cold stability. Moreover, it illustrates a novel mechanism by which Bmcc1 regulates cell morphology.
Collapse
Affiliation(s)
- Jessica Arama
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
| | - Anne-Cécile Boulay
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
| | - Christophe Bosc
- Equipe Physiopathologie du Cytosquelette, Institut National de la Santé et de la Recherche Médicale U836, Institut des Neurosciences, Université Joseph Fourier, Faculté de Médecine, Domaine de la Merci, La Tronche, France
| | - Christian Delphin
- Equipe Physiopathologie du Cytosquelette, Institut National de la Santé et de la Recherche Médicale U836, Institut des Neurosciences, Université Joseph Fourier, Faculté de Médecine, Domaine de la Merci, La Tronche, France
| | - Damarys Loew
- Institut Curie, Laboratory of Proteomic Mass Spectrometry, Paris, France
| | - Philippe Rostaing
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Institut National de la Santé et de la Recherche Médicale U1024, Paris, France
| | - Edwige Amigou
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
| | - Pascal Ezan
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
| | - Laure Wingertsmann
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Institut National de la Santé et de la Recherche Médicale U1024, Paris, France
| | - Laurent Guillaud
- Cell and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| | - Annie Andrieux
- Equipe Physiopathologie du Cytosquelette, Institut National de la Santé et de la Recherche Médicale U836, Institut des Neurosciences, Université Joseph Fourier, Faculté de Médecine, Domaine de la Merci, La Tronche, France
| | - Christian Giaume
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
| | - Martine Cohen-Salmon
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
- * E-mail:
| |
Collapse
|
28
|
Gupta AB, Wee LE, Zhou YT, Hortsch M, Low BC. Cross-species analyses identify the BNIP-2 and Cdc42GAP homology (BCH) domain as a distinct functional subclass of the CRAL_TRIO/Sec14 superfamily. PLoS One 2012; 7:e33863. [PMID: 22479462 PMCID: PMC3313917 DOI: 10.1371/journal.pone.0033863] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 02/18/2012] [Indexed: 11/19/2022] Open
Abstract
The CRAL_TRIO protein domain, which is unique to the Sec14 protein superfamily, binds to a diverse set of small lipophilic ligands. Similar domains are found in a range of different proteins including neurofibromatosis type-1, a Ras GTPase-activating Protein (RasGAP) and Rho guanine nucleotide exchange factors (RhoGEFs). Proteins containing this structural protein domain exhibit a low sequence similarity and ligand specificity while maintaining an overall characteristic three-dimensional structure. We have previously demonstrated that the BNIP-2 and Cdc42GAP Homology (BCH) protein domain, which shares a low sequence homology with the CRAL_TRIO domain, can serve as a regulatory scaffold that binds to Rho, RhoGEFs and RhoGAPs to control various cell signalling processes. In this work, we investigate 175 BCH domain-containing proteins from a wide range of different organisms. A phylogenetic analysis with ∼100 CRAL_TRIO and similar domains from eight representative species indicates a clear distinction of BCH-containing proteins as a novel subclass within the CRAL_TRIO/Sec14 superfamily. BCH-containing proteins contain a hallmark sequence motif R(R/K)h(R/K)(R/K)NL(R/K)xhhhhHPs (‘h’ is large and hydrophobic residue and ‘s’ is small and weekly polar residue) and can be further subdivided into three unique subtypes associated with BNIP-2-N, macro- and RhoGAP-type protein domains. A previously unknown group of genes encoding ‘BCH-only’ domains is also identified in plants and arthropod species. Based on an analysis of their gene-structure and their protein domain context we hypothesize that BCH domain-containing genes evolved through gene duplication, intron insertions and domain swapping events. Furthermore, we explore the point of divergence between BCH and CRAL-TRIO proteins in relation to their ability to bind small GTPases, GAPs and GEFs and lipid ligands. Our study suggests a need for a more extensive analysis of previously uncharacterized BCH, ‘BCH-like’ and CRAL_TRIO-containing proteins and their significance in regulating signaling events involving small GTPases.
Collapse
Affiliation(s)
- Anjali Bansal Gupta
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Liang En Wee
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Yi Ting Zhou
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Michael Hortsch
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Boon Chuan Low
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
29
|
Li S, Itoh M, Ohta K, Ueda M, Mizuno A, Ohta E, Hida Y, Wang MX, Takeuchi K, Nakagawa T. The expression and localization of Prune2 mRNA in the central nervous system. Neurosci Lett 2011; 503:208-14. [PMID: 21893162 DOI: 10.1016/j.neulet.2011.08.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 07/28/2011] [Accepted: 08/18/2011] [Indexed: 11/24/2022]
Abstract
A family of Bcl-2/adenovirus E1B 19kDa-interacting proteins (BNIPs) plays critical roles in several cellular processes such as cellular transformation, apoptosis, neuronal differentiation, and synaptic function, which are mediated by the BNIP2 and Cdc42GAP homology (BCH) domain. Prune homolog 2 (Drosophila) (PRUNE2) and its isoforms -C9orf65, BCH motif-containing molecule at the carboxyl terminal region 1 (BMCC1), and BNIP2 Extra Long (BNIPXL) - have been shown to be a susceptibility gene for Alzheimer's disease, a biomarker for leiomyosarcomas, a proapoptotic protein in neuronal cells, and an antagonist of cellular transformation, respectively. However, precise localization of PRUNE2 in the brain remains unclear. Here, we identified the distribution of Prune2 mRNA in the adult mouse brain. Prune2 mRNA is predominantly expressed in the neurons of the cranial nerve motor nuclei and the motor neurons of the spinal cord. The expression in the dorsal root ganglia (DRG) is consistent with the previously described reports. In addition, we observed the expression in another sensory neuron in the mesencephalic trigeminal nucleus. These results suggest that Prune2 may be functional in these restricted brain regions.
Collapse
Affiliation(s)
- Shimo Li
- Department of Neurobiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Iwama E, Tsuchimoto D, Iyama T, Sakumi K, Nakagawara A, Takayama K, Nakanishi Y, Nakabeppu Y. Cancer-related PRUNE2 protein is associated with nucleotides and is highly expressed in mature nerve tissues. J Mol Neurosci 2011; 44:103-14. [PMID: 21234814 DOI: 10.1007/s12031-010-9490-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 12/28/2010] [Indexed: 10/18/2022]
Abstract
Human PRUNE is thought to enhance the metastasis of tumor cells. We found that a hypothetical paralog of PRUNE, PRUNE2, binds to 8-oxo-GTP, an oxidized form of GTP. Hypothetical PRUNE2 gene consists of C9orf65 and BMCC1/BNIPXL, both of which are malignant tumor-associated genes. We isolated PRUNE2 complementary DNA and revealed that the protein is composed of 3,062 residues. C9orf65 and BMCC1/BNIPXL encode the N-terminal part (259 residues) and C-terminal part (2,729 residues) of PRUNE2, respectively. We demonstrated the endogenous full-length PRUNE2 protein (338 kDa) by Western blot and mass spectrometry. PRUNE2 bound to 8-oxo-GTP as well as GTP. The expression levels of human PRUNE2 and mouse Prune2 messenger RNA (mRNA) were highest in the dorsal root ganglia (DRG) and, to a lesser extent, in other nerve tissues. DRG neurons express higher levels of PRUNE2 in their soma compared with adjacent cells. In addition, their expression levels in the adult nerve tissues were higher than those in fetal or neonatal nerve tissues. The present study indicates that C9orf65 and BMCC1/BNIPXL are transcribed as PRUNE2 mRNA, which is translated to a large PRUNE2 protein. The nerve tissue-specific and post-development expression of PRUNE2/Prune2 suggests that PRUNE2 may contribute to the maintenance of mature nervous systems.
Collapse
Affiliation(s)
- Eiji Iwama
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Nile AH, Bankaitis VA, Grabon A. Mammalian diseases of phosphatidylinositol transfer proteins and their homologs. CLINICAL LIPIDOLOGY 2010; 5:867-897. [PMID: 21603057 PMCID: PMC3097519 DOI: 10.2217/clp.10.67] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Inositol and phosphoinositide signaling pathways represent major regulatory systems in eukaryotes. The physiological importance of these pathways is amply demonstrated by the variety of diseases that involve derangements in individual steps in inositide and phosphoinositide production and degradation. These diseases include numerous cancers, lipodystrophies and neurological syndromes. Phosphatidylinositol transfer proteins (PITPs) are emerging as fascinating regulators of phosphoinositide metabolism. Recent advances identify PITPs (and PITP-like proteins) to be coincidence detectors, which spatially and temporally coordinate the activities of diverse aspects of the cellular lipid metabolome with phosphoinositide signaling. These insights are providing new ideas regarding mechanisms of inherited mammalian diseases associated with derangements in the activities of PITPs and PITP-like proteins.
Collapse
Affiliation(s)
- Aaron H Nile
- Department of Cell & Developmental Biology, Lineberger Comprehensive Cancer Center School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-27090, USA
| | - Vytas A Bankaitis
- Department of Cell & Developmental Biology, Lineberger Comprehensive Cancer Center School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-27090, USA
| | - Aby Grabon
- Department of Cell & Developmental Biology, Lineberger Comprehensive Cancer Center School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-27090, USA
| |
Collapse
|
32
|
Zhou YT, Chew LL, Lin SC, Low BC. The BNIP-2 and Cdc42GAP homology (BCH) domain of p50RhoGAP/Cdc42GAP sequesters RhoA from inactivation by the adjacent GTPase-activating protein domain. Mol Biol Cell 2010; 21:3232-46. [PMID: 20660160 PMCID: PMC2938388 DOI: 10.1091/mbc.e09-05-0408] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The BNIP-2 and Cdc42GAP Homology (BCH) domain from p50RhoGAP sequesters RhoA from inactivation by the adjacent GAP domain and it confers unique Rho-binding profile from that of GAP domain. This suppression is further augmented by an intramolecular interaction, adding to a new paradigm for regulating p50RhoGAP signaling. The BNIP-2 and Cdc42GAP homology (BCH) domain is a novel regulator for Rho GTPases, but its impact on p50-Rho GTPase-activating protein (p50RhoGAP or Cdc42GAP) in cells remains elusive. Here we show that deletion of the BCH domain from p50RhoGAP enhanced its GAP activity and caused drastic cell rounding. Introducing constitutively active RhoA or inactivating GAP domain blocked such effect, whereas replacing the BCH domain with endosome-targeting SNX3 excluded requirement of endosomal localization in regulating the GAP activity. Substitution with homologous BCH domain from Schizosaccharomyces pombe, which does not bind mammalian RhoA, also led to complete loss of suppression. Interestingly, the p50RhoGAP BCH domain only targeted RhoA, but not Cdc42 or Rac1, and it was unable to distinguish between GDP and the GTP-bound form of RhoA. Further mutagenesis revealed a RhoA-binding motif (residues 85-120), which when deleted, significantly reduced BCH inhibition on GAP-mediated cell rounding, whereas its full suppression also required an intramolecular interaction motif (residues 169-197). Therefore, BCH domain serves as a local modulator in cis to sequester RhoA from inactivation by the adjacent GAP domain, adding to a new paradigm for regulating p50RhoGAP signaling.
Collapse
Affiliation(s)
- Yi Ting Zhou
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore 117543, Republic of Singapore.
| | | | | | | |
Collapse
|
33
|
Salagierski M, Verhaegh GW, Jannink SA, Smit FP, Hessels D, Schalken JA. Differential expression of PCA3 and its overlapping PRUNE2 transcript in prostate cancer. Prostate 2010; 70:70-8. [PMID: 19760627 DOI: 10.1002/pros.21040] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND PCA3 is one of the most prostate cancer (PrCa)-specific markers described so far. Recently, a new genomic structure of PCA3 as well as new flanking and overlapping gene transcripts has been identified. Furthermore, a co-regulation of PCA3 and its overlapping gene PRUNE2(BMCC1) has been suggested. Our aim was to assess the diagnostic performance of a new PCA3 isoform (PCA3-TS4) and to study the interactions between PCA3 and BMCC1 in PrCa. METHODS We used SYBR Green quantitative (q)PCR with specific primers to compare PCA3 and BMCC1 expression of normal versus tumor tissue of human prostate. PCA3-TS4 plasmid was created to calculate the absolute amounts of PCA3 transcripts. The androgen regulation of PCA3 and BMCC1 expression was studied in LNCaP and 22Rv1 cells stimulated with 5alpha-dihydrotestosterone. RESULTS We have not found any relevant diagnostic advantage of the PCA3-TS4 isoform over the "classical" PCA3 isoform in our group of PrCa patients. Additionally, PCA3-TS4 appears to be only a minor PCA3 transcript. We were also unable to confirm the hypothesis that BMCC1 isoforms are androgen-induced in vitro. CONCLUSIONS Despite the presence of the recently identified marginal PCA3 transcripts in human PrCa, the previously described major PCA3 isoform still constitutes the best target for diagnostic purposes. PCA3 and BMCC1 are overlapping genes in reverse orientation that do not appear to be co-regulated.
Collapse
Affiliation(s)
- Maciej Salagierski
- Department of Urology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | | | | | | | | | | |
Collapse
|
34
|
Edelman LB, Toia G, Geman D, Zhang W, Price ND. Two-transcript gene expression classifiers in the diagnosis and prognosis of human diseases. BMC Genomics 2009; 10:583. [PMID: 19961616 PMCID: PMC2797819 DOI: 10.1186/1471-2164-10-583] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Accepted: 12/05/2009] [Indexed: 11/15/2022] Open
Abstract
Background Identification of molecular classifiers from genome-wide gene expression analysis is an important practice for the investigation of biological systems in the post-genomic era - and one with great potential for near-term clinical impact. The 'Top-Scoring Pair' (TSP) classification method identifies pairs of genes whose relative expression correlates strongly with phenotype. In this study, we sought to assess the effectiveness of the TSP approach in the identification of diagnostic classifiers for a number of human diseases including bacterial and viral infection, cardiomyopathy, diabetes, Crohn's disease, and transformed ulcerative colitis. We examined transcriptional profiles from both solid tissues and blood-borne leukocytes. Results The algorithm identified multiple predictive gene pairs for each phenotype, with cross-validation accuracy ranging from 70 to nearly 100 percent, and high sensitivity and specificity observed in most classification tasks. Performance compared favourably with that of pre-existing transcription-based classifiers, and in some cases was comparable to the accuracy of current clinical diagnostic procedures. Several diseases of solid tissues could be reliably diagnosed through classifiers based on the blood-borne leukocyte transcriptome. The TSP classifier thus represents a simple yet robust method to differentiate between diverse phenotypic states based on gene expression profiles. Conclusion Two-transcript classifiers have the potential to reliably classify diverse human diseases, through analysis of both local diseased tissue and the immunological response assayed through blood-borne leukocytes. The experimental simplicity of this method results in measurements that can be easily translated to clinical practice.
Collapse
Affiliation(s)
- Lucas B Edelman
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | | | | | | | | |
Collapse
|
35
|
Sall A, Zhang HM, Qiu D, Liu Z, Yuan J, Liu Z, Lim T, Ye X, Marchant D, McManus B, Yang D. Pro-apoptotic activity of mBNIP-21 depends on its BNIP-2 and Cdc42GAP homology (BCH) domain and is enhanced by coxsackievirus B3 infection. Cell Microbiol 2009; 12:599-614. [PMID: 19951366 DOI: 10.1111/j.1462-5822.2009.01416.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Our previous study reported that mouse BNIP-21 (mBNIP-21) induces apoptosis through a mitochondria-dependent pathway. To map the functional domains of mBNIP-21, we performed mutational analyses and demonstrated that the BNIP-2 and Cdc42GAP homology (BCH) domain is required for apoptosis induction by mBNIP-21 targeting the mitochondria and inducing cytochrome c release. This pro-apoptotic activity was enhanced by coxsackievirus infection. However, deletion of the Bcl-2 homology 3 (BH3)-like domain, a well-known cell 'death domain' in proapoptotic Bcl-2 family proteins, did not affect the activity of mBNIP-21. These data were further supported by transfection of a mouse Bax (mBax) mutant, whose BH3 was replaced by the mBNIP-21 BH3-like domain. This replacement significantly reduced the pro-apoptotic activity of mBax. We also found that the predicted calcium binding domain has no contribution to the mBNIP-21-induced apoptosis. Further mapping of the motifs of BCH domain demonstrated that deletion of the hydrophobic motif proximal to the C-terminal of the BCH significantly reduced its proapoptotic activity. These findings suggest that mBNIP-21, as a member of the BNIP subgroup of the Bcl-2-related proteins, functions without need of BH3 but its BCH domain is critical for its activity in inducing cell elongation, membrane protrusions and apoptotic cell death.
Collapse
Affiliation(s)
- Alhousseynou Sall
- Department of Pathology and Laboratory Medicine, University of British Columbia-Providence Heart and Lung Institute, the iCapture Center, St. Paul's Hospital, Vancouver, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Clarke RA, Zhao Z, Guo AY, Roper K, Teng L, Fang ZM, Samaratunga H, Lavin MF, Gardiner RA. New genomic structure for prostate cancer specific gene PCA3 within BMCC1: implications for prostate cancer detection and progression. PLoS One 2009; 4:e4995. [PMID: 19319183 PMCID: PMC2655648 DOI: 10.1371/journal.pone.0004995] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Accepted: 02/05/2009] [Indexed: 11/20/2022] Open
Abstract
Background The prostate cancer antigen 3 (PCA3/DD3) gene is a highly specific biomarker upregulated in prostate cancer (PCa). In order to understand the importance of PCA3 in PCa we investigated the organization and evolution of the PCA3 gene locus. Methods/Principal Findings We have employed cDNA synthesis, RTPCR and DNA sequencing to identify 4 new transcription start sites, 4 polyadenylation sites and 2 new differentially spliced exons in an extended form of PCA3. Primers designed from these novel PCA3 exons greatly improve RT-PCR based discrimination between PCa, PCa metastases and BPH specimens. Comparative genomic analyses demonstrated that PCA3 has only recently evolved in an anti-sense orientation within a second gene, BMCC1/PRUNE2. BMCC1 has been shown previously to interact with RhoA and RhoC, determinants of cellular transformation and metastasis, respectively. Using RT-PCR we demonstrated that the longer BMCC1-1 isoform - like PCA3 – is upregulated in PCa tissues and metastases and in PCa cell lines. Furthermore PCA3 and BMCC1-1 levels are responsive to dihydrotestosterone treatment. Conclusions/Significance Upregulation of two new PCA3 isoforms in PCa tissues improves discrimination between PCa and BPH. The functional relevance of this specificity is now of particular interest given PCA3's overlapping association with a second gene BMCC1, a regulator of Rho signalling. Upregulation of PCA3 and BMCC1 in PCa has potential for improved diagnosis.
Collapse
Affiliation(s)
- Raymond A. Clarke
- Prostate Cancer Institute, Cancer Care Centre, St George Hospital Clinical School of Medicine, University of New South Wales, Kogarah, New South Wales, Australia
- Division of Cancer and Cell Biology, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Zhongming Zhao
- Department of Psychiatry and Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, United States of Amerca
| | - An-Yuan Guo
- Department of Psychiatry and Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, United States of Amerca
| | - Kathrein Roper
- Hopkins Marine Station, Stanford University, Stanford, California, United States of America
| | - Linda Teng
- Division of Cancer and Cell Biology, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Zhi-Ming Fang
- Prostate Cancer Institute, Cancer Care Centre, St George Hospital Clinical School of Medicine, University of New South Wales, Kogarah, New South Wales, Australia
| | | | - Martin F. Lavin
- Division of Cancer and Cell Biology, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
- University of Queensland Centre for Clinical Research, Brisbane, Australia
- * E-mail: (MFL); (RAG)
| | - Robert A. Gardiner
- University of Queensland Centre for Clinical Research, Brisbane, Australia
- * E-mail: (MFL); (RAG)
| |
Collapse
|